Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Exp Physiol ; 104(7): 1029-1037, 2019 07.
Article in English | MEDLINE | ID: mdl-31025768

ABSTRACT

NEW FINDINGS: What is the central question of this study? Does protein restriction in early life modify glucose-induced insulin secretion by altering [Ca2+ ]i and the expression of SNARE proteins in pancreatic islets from pregnant rats? What is the main finding and its importance? Protein restriction in early life increased the first phase of glucose-induced insulin secretion and [Ca2+ ]i without altering the expression of SNARE proteins during pregnancy. This finding contributes to our understanding of the mechanisms of altered insulin secretion and might provide new perspectives for the development of therapeutic tools for gestational diabetes. ABSTRACT: We investigated the kinetics of glucose-induced insulin secretion and their relationship with [Ca2+ ]i and the expression of protein from exocytotic machinery in islets from recovered pregnant and long-term protein-deficient pregnant rats. Isolated islets were evaluated from control-fed pregnant (CP), protein-deficient pregnant (DP), control-fed non-pregnant (CNP) and protein-deficient non-pregnant (DNP) female adult rats, and from protein-deficient pregnant (RP) and non-pregnant (RNP) rats that were recovered after weaning. The insulin responses to glucose during the first phase of secretion were higher in RP than in CP groups, and both were higher than in the DP group. Islets from RP rats displayed a rapid increase in insulin release (first phase), followed by a plateau that was maintained thereafter. The [Ca2+ ]i in islets from the protein-deficient groups was lower than in the control groups, and both were lower than in the RP and RNP groups. SNAP-25 was increased in islets from pregnant rats independently of their nutritional status, and the syntaxin-1A content was reduced in islets from the RP rats compared with the RNP rats. The VAMP2 content was similar among the groups. Thus, protein restriction during intrauterine life and lactation increased insulin secretion during pregnancy, attributable, in part, to increased [Ca2+ ]i , and independent of an alteration of expression of SNARE proteins.


Subject(s)
Calcium/metabolism , Diet, Protein-Restricted/trends , Gene Expression Regulation, Developmental , Insulin Secretion/physiology , Intracellular Fluid/metabolism , SNARE Proteins/biosynthesis , Animals , Blood Glucose/metabolism , Female , Islets of Langerhans/metabolism , Male , Pregnancy , Rats , Rats, Wistar , SNARE Proteins/genetics
2.
Alcohol Clin Exp Res ; 42(9): 1661-1673, 2018 09.
Article in English | MEDLINE | ID: mdl-29944190

ABSTRACT

BACKGROUND: Recent work with long-term ethanol (EtOH) self-administration in nonhuman primate models has revealed a complex array of behavioral and physiological effects that closely mimic human alcohol abuse. Detailed neurophysiological analysis in these models suggests a myriad of pre- and postsynaptic neurobiological effects that may contribute to the behavioral manifestations of long-term EtOH drinking. The molecular mechanisms regulating presynaptic effects of this chronic EtOH exposure are largely unknown. To this end, we analyzed the effects of long-term EtOH self-administration on the levels of presynaptic SNARE complex proteins in Macaca mulatta basolateral amygdala, a brain region known to regulate both aversive and reward-seeking behaviors. METHODS: Basolateral amygdala samples from control and EtOH-drinking male and female monkeys were processed. Total basolateral amygdala protein was analyzed by Western blotting using antibodies directed against both core SNARE and SNARE-associated proteins. We also performed correlational analyses between protein expression levels and a number of EtOH drinking parameters, including lifetime grams of EtOH consumed, preference, and blood alcohol concentration. RESULTS: Significant interactions or main effects of sex/drinking were seen for a number of SNARE core and SNARE-associated proteins. Across the range of EtOH-drinking phenotypes, SNAP25 and Munc13-1 proteins levels were significantly different between males and females, and Munc13-2 levels were significantly lower in animals with a history of EtOH drinking. A separate analysis of very heavy-drinking individuals revealed significant decreases in Rab3c (females) and complexin 2 (males). CONCLUSIONS: Protein expression analysis of basolateral amygdala total protein from controls and animals following long-term EtOH self-administration suggests a number of alterations in core SNARE or SNARE-associated components that could dramatically alter presynaptic function. A number of proteins or multiprotein components were also correlated with EtOH drinking behavior, which suggest a potentially heritable role for presynaptic SNARE proteins.


Subject(s)
Alcohol Drinking/metabolism , Alcohol Drinking/trends , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/metabolism , Ethanol/administration & dosage , SNARE Proteins/biosynthesis , Alcohol Drinking/adverse effects , Animals , Basolateral Nuclear Complex/chemistry , Ethanol/adverse effects , Female , Macaca mulatta , Male , SNARE Proteins/analysis , Self Administration , Time Factors
3.
Neuron ; 98(1): 127-141.e7, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29621484

ABSTRACT

Dysfunction of the neuronal RNA binding protein RBFOX1 has been linked to epilepsy and autism spectrum disorders. Rbfox1 loss in mice leads to neuronal hyper-excitability and seizures, but the physiological basis for this is unknown. We identify the vSNARE protein Vamp1 as a major Rbfox1 target. Vamp1 is strongly downregulated in Rbfox1 Nes-cKO mice due to loss of 3' UTR binding by RBFOX1. Cytoplasmic Rbfox1 stimulates Vamp1 expression in part by blocking microRNA-9. We find that Vamp1 is specifically expressed in inhibitory neurons, and that both Vamp1 knockdown and Rbfox1 loss lead to decreased inhibitory synaptic transmission and E/I imbalance. Re-expression of Vamp1 selectively within interneurons rescues the electrophysiological changes in the Rbfox1 cKO, indicating that Vamp1 loss is a major contributor to the Rbfox1 Nes-cKO phenotype. The regulation of interneuron-specific Vamp1 by Rbfox1 provides a paradigm for broadly expressed RNA-binding proteins performing specialized functions in defined neuronal subtypes.


Subject(s)
Neural Inhibition/physiology , Neurons/metabolism , RNA Splicing Factors/physiology , Synaptic Transmission/physiology , Vesicle-Associated Membrane Protein 1/biosynthesis , Animals , Cells, Cultured , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/chemistry , RNA Splicing Factors/analysis , RNA Splicing Factors/deficiency , SNARE Proteins/analysis , SNARE Proteins/biosynthesis , Vesicle-Associated Membrane Protein 1/analysis
4.
Curr Biol ; 28(8): R397-R401, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29689222

ABSTRACT

A fundamental hallmark of eukaryotic cells is their compartmentalization into functionally distinct organelles, including those of the secretory and endocytic pathways. Transport of cargo between these compartments and to/from the cell surface is mediated by membrane-bound vesicles and tubules. Delivery of cargo is facilitated by SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)-mediated membrane fusion of vesicles with their target compartments. Vesicles contain a variety of cargos, including lipids, membrane proteins, signaling molecules, biosynthetic and hydrolytic enzymes, and the trafficking machinery itself. Proper function of membrane trafficking is required for cellular growth, division, movement, and cell-cell communication. Defects in these processes have been implicated in a variety of human diseases, such as cancer, diabetes, neurodegenerative disorders, ciliopathies, and infections. The elucidation of the mechanisms of SNARE assembly and disassembly is key to understanding how membrane fusion is regulated throughout eukaryotes. Here, we introduce the SNARE proteins, their structures and functions in eukaryotic cells, and discuss recent breakthroughs in elucidating the regulation of SNARE assembly and disassembly through the use of high-resolution structural biology and biophysical techniques.


Subject(s)
Membrane Fusion/physiology , SNARE Proteins/biosynthesis , SNARE Proteins/metabolism , Animals , Biological Transport/physiology , Cell Membrane/metabolism , Humans , Protein Binding , Protein Transport/physiology , SNARE Proteins/physiology
5.
Cancer Res ; 78(7): 1685-1699, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29363545

ABSTRACT

Restoring antigen presentation for efficient and durable activation of tumor-specific CD8+ T-cell responses is pivotal to immunotherapy, yet the mechanisms that cause subversion of dendritic cell (DC) functions are not entirely understood, limiting the development of targeted approaches. In this study, we show that bona fide DCs resident in lung tumor tissues or DCs exposed to factors derived from whole lung tumors become refractory to endosomal and cytosolic sensor stimulation and fail to secrete IL12 and IFNI. Tumor-conditioned DC exhibited downregulation of the SNARE VAMP3, a regulator of endosomes trafficking critical for cross-presentation of tumor antigens and DC-mediated tumor rejection. Dissection of cell-extrinsic suppressive pathways identified lactic acid in the tumor microenvironment as sufficient to inhibit type-I IFN downstream of TLR3 and STING. DC conditioning by lactate also impacted adaptive function, accelerating antigen degradation and impairing cross-presentation. Importantly, DCs conditioned by lactate failed to prime antitumor responses in vivo These findings provide a new mechanistic viewpoint to the concept of DC suppression and hold potential for future therapeutic approaches.Significance: These findings provide insight into the cell-intrinsic and cell-extrinsic mechanisms that cause loss of presentation of tumor-specific antigens in lung cancer tissues. Cancer Res; 78(7); 1685-99. ©2018 AACR.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lung Neoplasms/immunology , Membrane Transport Proteins/biosynthesis , Animals , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Down-Regulation , Endosomes/metabolism , Immunotherapy , Interferon Type I/antagonists & inhibitors , Lactic Acid/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , SNARE Proteins/biosynthesis , Tumor Microenvironment/immunology , Vesicle-Associated Membrane Protein 3/biosynthesis
6.
Am J Clin Pathol ; 145(5): 604-16, 2016 May.
Article in English | MEDLINE | ID: mdl-27247366

ABSTRACT

OBJECTIVES: Five proteins from the soluble N-ethylmaleimide-sensitive factor activating protein receptor (SNARE) complex family were studied in normal hematopoietic cells in bone marrow; normal lymphocytes at different stages of maturation and differentiation in bone marrow, thymus, tonsil, and lymph node; malignant lymphomas; and leukemias. METHODS: Sixty-eight reactive and 380 hematopoietic and lymphoid neoplasms were immunohistochemically stained for syntaxin 7 (STX7), vesicle-associated membrane proteins (VAMP2, VAMP7, VAMP8), and synaptosomal-associated protein 23 (SNAP23). RESULTS: STX7 has potential for being a useful marker for distinguishing between normal B precursors (hematogones) vs B lymphoblasts, as well as between the "popcorn" cells of nodular lymphocyte-predominant Hodgkin lymphoma vs the Reed-Sternberg cells of classic Hodgkin lymphoma or the B cells of T-cell, histiocyte-rich B-cell lymphoma. VAMP2 is uniquely expressed by both reactive and malignant plasma cells, in contrast to B-cell non-Hodgkin lymphoma. There is differential expression of SNARE proteins in normal and neoplastic lymphoid tissue depending on lymphocyte maturation stage. CONCLUSIONS: Differential SNARE protein expression in the lymphoid system may have potential use in diagnosis and may offer clues to lymphoma biology. VAMP2 is a promising new plasma cell marker.


Subject(s)
Biomarkers, Tumor/analysis , Hematologic Neoplasms/pathology , SNARE Proteins/biosynthesis , Bone Marrow/metabolism , Humans , Immunohistochemistry , Lymphoid Tissue/metabolism , SNARE Proteins/analysis , Tissue Array Analysis
7.
Appl Microbiol Biotechnol ; 100(1): 505-18, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26450509

ABSTRACT

Soluble N-ethylmaleimide-sensitive factor attachment receptor proteins (SNAREs) are essential components of the yeast protein-trafficking machinery and are required at the majority of membrane fusion events in the cell, where they facilitate SNARE-mediated fusion between the protein transport vesicles, the various membrane-enclosed organelles and, ultimately, the plasma membrane. We have demonstrated an increase in secretory titers for the Talaromyces emersonii Cel7A (Te-Cel7A, a cellobiohydrolase) and the Saccharomycopsis fibuligera Cel3A (Sf-Cel3A, a ß-glucosidase) expressed in Saccharomyces cerevisiae through single and co-overexpression of some of the endoplasmic reticulum (ER)-to-Golgi SNAREs (BOS1, BET1, SEC22 and SED5). Overexpression of SED5 yielded the biggest improvements for both of the cellulolytic reporter proteins tested, with maximum increases in extracellular enzyme activity of 22 % for the Sf-Cel3A and 68 % for the Te-Cel7A. Co-overexpression of the ER-to-Golgi SNAREs yielded proportionately smaller increases for the Te-Cel7A (46 %), with the Sf-Cel3A yielding no improvement. Co-overexpression of the most promising exocytic SNARE components identified in literature for secretory enhancement of the cellulolytic proteins tested (SSO1 for Sf-Cel3A and SNC1 for Te-Cel7A) with the most effective ER-to-Golgi SNARE components identified in this study (SED5 for both Sf-Cel3A and Te-Cel7A) yielded variable results, with Sf-Cel3A improved by 131 % and Te-Cel7A yielding no improvement. Improvements were largely independent of gene dosage as all strains only integrated single additional SNARE gene copies, with episomal variance between the most improved strains shown to be insignificant. This study has added further credence to the notion that SNARE proteins fulfil an essential role within a larger cascade of secretory machinery components that could contribute significantly to future improvements to S. cerevisiae as protein production host.


Subject(s)
Cellulase/metabolism , Gene Expression , SNARE Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Cellulase/genetics , Eurotiales/enzymology , Eurotiales/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SNARE Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomycopsis/enzymology , Saccharomycopsis/genetics
8.
J Neurosci ; 34(50): 16594-604, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25505312

ABSTRACT

Experimental advances in the study of neuroglia signaling have been greatly accelerated by the generation of transgenic mouse models. In particular, an elegant manipulation that interferes with astrocyte vesicular release of gliotransmitters via overexpression of a dominant-negative domain of vesicular SNARE (dnSNARE) has led to documented astrocytic involvement in processes that were traditionally considered strictly neuronal, including the sleep-wake cycle, LTP, cognition, cortical slow waves, depression, and pain. A key premise leading to these conclusions was that expression of the dnSNARE was specific to astrocytes. Inconsistent with this premise, we report here widespread expression of the dnSNARE transgene in cortical neurons. We further demonstrate that the activity of cortical neurons is reversibly suppressed in dnSNARE mice. These findings highlight the need for independent validation of astrocytic functions identified in dnSNARE mice and thus question critical evidence that astrocytes contribute to neurotransmission through SNARE-dependent vesicular release of gliotransmitters.


Subject(s)
Gene Expression Regulation , Neurons/metabolism , SNARE Proteins/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Electroencephalography/methods , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , SNARE Proteins/genetics , Sleep Stages/physiology
9.
Neuron ; 84(6): 1112-5, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25521372

ABSTRACT

Gliotransmission, a process involving active vesicular release of glutamate and other neurotransmitters by astrocytes, is thought to play a critical role in many brain functions. A new paper by Nedergaard et al. (2014) identifies an experimental flaw in these previous studies suggesting that astrocytes may not perform active vesicular release after all.


Subject(s)
Gene Expression Regulation , Neurons/metabolism , SNARE Proteins/biosynthesis , Animals , Female , Male
10.
Appl Microbiol Biotechnol ; 98(12): 5567-78, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24639206

ABSTRACT

SNAREs (soluble NSF [N-ethylmaleimide-sensitive factor] attachment receptor proteins) are required at the majority of fusion events during intracellular membrane transport and play crucial roles in facilitating protein trafficking between the various membrane-enclosed organelles and the plasma membrane. We demonstrate increases in the secretion of the Talaromyces emersonii Cel7A (a cellobiohydrolase) and the Saccharomycopsis fibuligera Cel3A (a ß-glucosidase), through the separate and simultaneous over-expression of different components of the exocytic SNARE complex in Saccharomyces cerevisiae. Over-expression of SNC1 yielded the biggest improvement in Te-Cel7A secretion (71 %), whilst SSO1 over-expression lead to the highest increases in Sf-Cel3A secretion (43.8 %). Simultaneous over-expression of differential combinations of these SNARE components yielded maximal increases of ~52 % and ~49 % for the secretion of Te-Cel7A and Sf-Cel3A, respectively. These increases generally did not cause deleterious growth effects, whilst differential improvement patterns were observed for the two reporter proteins (Sf-Cel3A and Te-Cel7A). Simultaneous over-expression of up to three of these components, in strains secreting the more efficiently expressed Sf-Cel3A, illustrated a slight decrease in osmotic tolerance at elevated NaCl concentrations, as well as a detectable decrease in ethanol tolerance at increased concentrations. This work illustrates the potential of engineering components of the anterograde secretory pathway, particularly its SNARE components, for the improvement of heterologous cellulase secretion.


Subject(s)
Cellulase/metabolism , SNARE Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Cellulase/genetics , Eurotiales/enzymology , Eurotiales/genetics , Gene Expression , Genes, Reporter , Metabolic Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SNARE Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
11.
Glia ; 61(5): 724-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23378051

ABSTRACT

Activation of the immune system elicits several behavioral changes collectively called sickness. Among the behavioral changes, systemic infections induce an increase in time spent in nonrapid-eye-movement (NREM) sleep and an increase of slow wave activity (or "sleep pressure"). Using an inducible, astrocyte-specific transgenic dominant negative SNARE (dnSNARE) mouse line we recently demonstrated that gliotransmission plays an important role in sleep homeostasis through an adenosine receptor 1 (A1R)-sensitive pathway. It has been hypothesized that systemic infection, mimicked by peripheral administration of lipopolysaccharide (LPS), increases sleeping behavior in part through upregulation of central adenosine levels. Moreover, as a source of immunologically relevant factors, astrocytes play a pivotal role in the central nervous system immune and inflammatory responses. However, little is known about the role of astrocytes in the CNS response to a peripheral immune challenge. We hypothesize that LPS impacts sleep homeostasis through the modulation of astrocyte-derived adenosine accumulation. We therefore used dnSNARE mice to determine whether astrocytes contribute to the increased sleep pressure under immune challenge and whether this is a result of changes in adenosine signaling. We demonstrate that dnSNARE-mediated gliotransmission is required for the ability of LPS to elevate sleep pressure as measured by the power of slow wave activity during NREM sleep. Moreover, in agreement with a role of astrocyte-derived adenosine in modulating sleep homeostasis, we find that intracerebroventricular infusion of the A1R antagonist 8-cyclopentyl-1,3-dimethylxanthine (CPT) mimics this dnSNARE phenotype. Taken together, our data demonstrate that astrocytic adenosine acting through A1 receptors contributes to the modulation of sleep pressure by LPS.


Subject(s)
Adenosine/physiology , Astrocytes/pathology , Sleep/physiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Electroencephalography/methods , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Adenosine A1/physiology , SNARE Proteins/biosynthesis , SNARE Proteins/genetics , Sleep/drug effects
12.
Planta ; 235(4): 841-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22089884

ABSTRACT

The bacterial merC gene from the Tn21-encoded mer operon is a potential molecular tool for improving the efficiency of metal phytoremediation. Arabidopsis SNARE molecules, including SYP111, SYP121, and AtVAM3 (SYP22), were attached to the C-terminus of MerC to target the protein to various organelles. The subcellular localization of transiently expressed GFP-fused MerC-SYP111, MerC-SYP121, and MerC-AtVAM3 was examined in Arabidopsis suspension-cultured cells. We found that GFP-MerC-SYP111 and GFP-MerC-SYP121 localized to the plasma membrane, whereas GFP-AtVAM3 localized to the vacuolar membranes. These results demonstrate that SYP111/SYP121 and AtVAM3 target foreign molecules to the plasma membrane and vacuolar membrane, respectively. To enhance the efficiency and potential of plants to sequester and accumulate cadmium from contaminated sites, transgenic Arabidopsis plants expressing MerC, MerC-SYP111, MerC-SYP121, or MerC-AtVAM3 were generated. The transgenic plants that expressed MerC, MerC-SYP121, or MerC-AtVAM3 appeared to be normal, whereas the transgenic that expressed MerC-SYP111 exhibited severe growth defects. The transgenic plants expressing merC-SYP121 were more resistant to cadmium than the wild type and accumulated significantly more cadmium. Thus, the expression of MerC-SYP121 in the plant plasma membrane may provide an ecologically compatible approach for the phytoremediation of cadmium pollution.


Subject(s)
Arabidopsis Proteins/biosynthesis , Arabidopsis/metabolism , Cadmium/metabolism , Cation Transport Proteins/biosynthesis , Qa-SNARE Proteins/biosynthesis , Recombinant Fusion Proteins/biosynthesis , SNARE Proteins/biosynthesis , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Biodegradation, Environmental , Cation Transport Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Plant , Genes, Plant , Genetic Engineering , Genetic Variation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Plant Roots/metabolism , Plants, Genetically Modified , Qa-SNARE Proteins/genetics , Recombinant Fusion Proteins/genetics , SNARE Proteins/genetics , Transformation, Genetic
13.
Brain Res ; 1385: 26-37, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21354112

ABSTRACT

Prolonged stress has been associated with altered synaptic plasticity but little is known about the molecular components and mechanisms involved in the stress response. In this study, we examined the effect of chronic restraint stress (CRS) on the expression of genes associated with synaptic vesicle exocytosis in rat prefrontal cortex and hippocampus. Rats were stressed daily using a 21day restraint stress paradigm, with durations of half an hour or 6h. RNA and protein were extracted from the same tissue sample and used for real-time quantitative polymerase chain reaction (real-time qPCR) and immunoblotting, respectively. Focusing on the SNARE complex, we investigated the expression of the SNARE core components syntaxin 1A, SNAP-25, and VAMP2 at both transcriptional and protein levels. In addition, the expression of 10 SNARE regulatory proteins was investigated at the transcriptional level. Overall, the prefrontal cortex was more sensitive to CRS compared to the hippocampus. In prefrontal cortex, CRS induced increased mRNA levels of VAMP2, VAMP1, syntaxin 1A, snapin, synaptotagmins I and III, and synapsins I and II, whereas SNAP-25 was down-regulated after CRS. Immunoblotting demonstrated equivalent changes in protein levels of VAMP2, syntaxin 1A, and SNAP-25. In hippocampus, we found increased mRNA levels of VAMP2 and SNAP-29 and a decrease in VAMP1 levels. Immunoblotting revealed decreased VAMP2 protein levels despite increased mRNA levels. Changes in the expression of synaptic proteins may accompany or contribute to the morphological, functional, and behavioral changes observed in experimental models of stress and may have relevance to the pathophysiology of stress-related disorders.


Subject(s)
Gene Expression Regulation , Hippocampus/metabolism , Prefrontal Cortex/metabolism , SNARE Proteins/biosynthesis , Stress, Psychological/metabolism , Animals , Chronic Disease , Male , Rats , Rats, Sprague-Dawley , Restraint, Physical/psychology , Stress, Psychological/psychology
14.
Mol Biol Cell ; 22(2): 216-29, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21119004

ABSTRACT

The role of specific membrane lipids in transport between endoplasmic reticulum (ER) and Golgi compartments is poorly understood. Using cell-free assays that measure stages in ER-to-Golgi transport, we screened a variety of enzyme inhibitors, lipid-modifying enzymes, and lipid ligands to investigate requirements in yeast. The pleckstrin homology (PH) domain of human Fapp1, which binds phosphatidylinositol-4-phosphate (PI(4)P) specifically, was a strong and specific inhibitor of anterograde transport. Analysis of wild type and mutant PH domain proteins in addition to recombinant versions of the Sac1p phosphoinositide-phosphatase indicated that PI(4)P was required on Golgi membranes for fusion with coat protein complex II (COPII) vesicles. PI(4)P inhibition did not prevent vesicle tethering but significantly reduced formation of soluble n-ethylmaleimide sensitive factor adaptor protein receptor (SNARE) complexes between vesicle and Golgi SNARE proteins. Moreover, semi-intact cell membranes containing elevated levels of the ER-Golgi SNARE proteins and Sly1p were less sensitive to PI(4)P inhibitors. Finally, in vivo analyses of a pik1 mutant strain showed that inhibition of PI(4)P synthesis blocked anterograde transport from the ER to early Golgi compartments. Together, the data presented here indicate that PI(4)P is required for the SNARE-dependent fusion stage of COPII vesicles with the Golgi complex.


Subject(s)
COP-Coated Vesicles/metabolism , Golgi Apparatus/metabolism , Phosphatidylinositol Phosphates/metabolism , SNARE Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Humans , Intracellular Membranes/metabolism , Membrane Fusion , Membrane Proteins/metabolism , Mutation , Phosphatidylinositol Phosphates/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Transport , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
15.
J Neurosci ; 30(10): 3624-33, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20219996

ABSTRACT

Glycogen synthase kinase-3 (GSK-3), a Ser/Thr protein kinase abundantly expressed in neurons, plays diverse functions in physiological and neurodegenerative conditions. Our recent study shows that upregulation of GSK-3 suppresses long-term potentiation and presynaptic release of glutamate; however, the underlying mechanism is elusive. Here, we show that activation of GSK-3beta retards the synaptic vesicle exocytosis in response to membrane depolarization. Using calcium imaging, whole-cell patch-clamp, as well as specific Ca(2+) channel inhibitors, we demonstrate that GSK-3beta phosphorylates the intracellular loop-connecting domains II and III (L(II-III)) of P/Q-type Ca(2+) channels, which leads to a decrease of intracellular Ca(2+) rise through the P/Q-type voltage-dependent calcium channel. To further illustrate the mechanisms of GSK-3beta's action, we show that activation of GSK-3beta interferes with the formation of the soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) complex through: (1) weakening the association of synaptobrevin with SNAP25 and syntaxin; (2) reducing the interactions among the phosphorylated L(II-III) and synaptotagmin, SNAP25, and syntaxin; and (3) inhibiting dissociation of synaptobrevin from synaptophysin I. These results indicate that GSK-3beta negatively regulates synaptic vesicle fusion events via interfering with Ca(2+)-dependent SNARE complex formation.


Subject(s)
Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Glycogen Synthase Kinase 3/physiology , Presynaptic Terminals/metabolism , SNARE Proteins/antagonists & inhibitors , Synaptic Vesicles/metabolism , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Exocytosis/physiology , Glycogen Synthase Kinase 3/biosynthesis , Glycogen Synthase Kinase 3 beta , Molecular Sequence Data , Neural Inhibition/physiology , Phosphorylation , Rats , SNARE Proteins/biosynthesis
16.
Anat Rec (Hoboken) ; 293(6): 1070-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20186959

ABSTRACT

Distribution of three soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor (SNARE) proteins, syntaxin-1, synaptosomal-associated protein of 25 kDa (SNAP-25), and vesicle-associated membrane protein-2 (VAMP-2), was examined in dental pulp and periodontal ligament of the rat incisor. In the trigeminal ganglion, syntaxin-1 and SNAP-25 immunoreactivity was predominately detected in medium- to large-sized neurons. Most syntaxin-1 immunoreactive neurons expressed SNAP-25. In contrast, VAMP-2 was localized in small- to medium-sized neurons and in slender-shaped cells surrounding SNAP-25-immunopositive neurons. When the inferior alveolar nerve, one of the mandibular nerve branches innervating the dental pulp and periodontal ligament, was ligated, SNARE proteins accumulated at the site proximal to the ligation. In the incisor dental pulp, all nerve fibers displayed immunoreactivity for syntaxin-1, SNAP-25, and VAMP-2. In the periodontal ligament of the incisor, almost all nerve fibers displayed both syntaxin-1 and SNAP-25 immunoreactivity, but lacked VAMP-2 immunoreactivity. SNAP-25 protein expression was localized around the vesicle membranes at the axon terminal of the periodontal mechanoreceptors. These present data suggest that these three SNARE proteins are synthesized at the trigeminal ganglion, transported centrally and peripherally, and expressed in sensory endings where apparent synapses are not present. Because those proteins participate in docking and exocytosis of synapse vesicles in the central nervous system, they might also contribute to vesicle exocytosis at receptive fields where apparent synapses are not present.


Subject(s)
Dental Pulp/chemistry , Dental Pulp/metabolism , Incisor/chemistry , Incisor/metabolism , Periodontal Ligament/chemistry , Periodontal Ligament/metabolism , SNARE Proteins/chemistry , SNARE Proteins/metabolism , Animals , Dental Pulp/innervation , Immunohistochemistry , Incisor/innervation , Male , Nerve Fibers/chemistry , Nerve Fibers/metabolism , Nerve Fibers/ultrastructure , Periodontal Ligament/innervation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , SNARE Proteins/biosynthesis , Sensory Receptor Cells/chemistry , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Synapses/chemistry , Synapses/metabolism , Synapses/ultrastructure , Synaptosomal-Associated Protein 25/biosynthesis , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/genetics , Syntaxin 1/biosynthesis , Syntaxin 1/chemistry , Syntaxin 1/genetics , Trigeminal Nerve/chemistry , Trigeminal Nerve/metabolism , Trigeminal Nerve/ultrastructure , Vesicle-Associated Membrane Protein 2/biosynthesis , Vesicle-Associated Membrane Protein 2/chemistry , Vesicle-Associated Membrane Protein 2/genetics
17.
J Neurosci ; 29(39): 12292-301, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19793988

ABSTRACT

The soluble N-ethylmaleimide-sensitive fusion (NSF) attachment protein (SNAP) receptor (SNARE) protein syntaxin 1A forms nano-sized clusters (membrane rafts) on the plasma membrane (PM) that are in equilibrium with freely diffusing syntaxin molecules. SNARE-complex formation between syntaxin 1A and SNAP-25 (synaptosome-associated protein of 25 kDa) on the PM and synaptobrevin 2 on the vesicles (trans-SNAREs) is crucial for vesicle priming and fusion. This process might be impeded by the spontaneous accumulation of non-fusogenic cis-SNARE complexes formed when all three SNARE proteins reside on the PM. We investigated the kinetics of cis-SNARE complex assembly and disassembly and both exhibited biphasic behavior. The experimental measurements were analyzed through integration of differential rate equations pertinent to the reaction mechanism and through the application of a heuristic search for time constants and concentrations using a genetic algorithm. Reconstruction of the measurements necessitated the partitioning of syntaxin into two phases that might represent the syntaxin clusters and free syntaxin outside the clusters. The analysis suggests that most of the syntaxin in the clusters is concentrated in a nonreactive form. Consequently, cis-SNARE complex assembly in the clusters is substantially slower than outside the rafts. Interestingly, the clusters also mediate efficient disassembly of cis-SNARE complexes possibly attributable to the high local concentration of complexes in the clusters area that allows efficient disassembly by the enzymatic reaction of NSF.


Subject(s)
Membrane Microdomains/metabolism , Qa-SNARE Proteins/metabolism , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Intracellular Membranes/metabolism , Intracellular Membranes/physiology , Kinetics , Membrane Microdomains/physiology , Multigene Family/physiology , PC12 Cells , Protein Binding/physiology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , Rats , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/biosynthesis , SNARE Proteins/metabolism
18.
Biochemistry ; 47(32): 8342-9, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18636750

ABSTRACT

Previously, we have demonstrated physical and functional interactions of the voltage-gated potassium channel Kv2.1 with the plasma membrane protein components of the exocytotic SNARE complex, syntaxin 1A, and the t-SNARE, syntaxin 1A/SNAP-25, complex. Importantly, the physical interaction of Kv2.1 with syntaxin was shown to be involved in the facilitation of secretion from PC12 cells, which was independent of potassium currents. Recently, we showed that also VAMP2, the vesicular SNARE, interacts physically and functionally with Kv2.1. Here, we first set out to test the interaction of the full SNARE, syntaxin/SNAP-25/VAMP2, complex with the channel. Using the interaction of VAMP2 with Kv2.1 in Xenopus oocytes as a probe, we showed that coexpression of the t-SNARE complex with VAMP2 abolished the VAMP2 effect on channel inactivation and reduced the amount of VAMP2 that coprecipitated with Kv2.1. Further, in vitro pull down assays showed that the full SNARE complex failed to interact with Kv2.1 N- and C-termini in tandem, in contrast to the individual SNARE components. This suggests that the interactions of the SNARE components with Kv2.1 are abolished upon their recruitment into a full SNARE complex, which does not interact with the channel. Other important findings arising from the in vitro study are that the t-SNARE complex, in addition to syntaxin, interacts with a specific C-terminal channel domain, C1a, shown to mediate the facilitation of release by Kv2.1 and that the presence of Kv2.1 N-terminus has crucial contribution to these interactions. These findings provide important insights into the understanding of the complex molecular events involved in the novel phenomenon of secretion facilitation in neuroendocrine cells by Kv2.1.


Subject(s)
Protein Subunits/metabolism , SNARE Proteins/biosynthesis , SNARE Proteins/metabolism , Shab Potassium Channels/metabolism , Animals , Female , Oocytes/metabolism , Protein Binding/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Rats , SNARE Proteins/genetics , Shab Potassium Channels/genetics , Vesicle-Associated Membrane Protein 2/metabolism , Xenopus laevis
19.
Somatosens Mot Res ; 24(1-2): 85-95, 2007.
Article in English | MEDLINE | ID: mdl-17558925

ABSTRACT

The role of the capsule encasing the Pacinian corpuscle's (PC's) neurite, where mechanotransduction occurs, may be more than mechanical. The inner core of the PC's capsule consists of lamellar cells that are of Schwann-cell origin. Previously, we found both voltage-gated Na+ and K+ channels in these inner-core lamellae. Research on astrocytes and Schwann cells shows bidirectional signaling between glia and neurons, a major component of which is glutamate. Furthermore, Merkel cells show positive immunoreactivity for glutamate receptor mGluR5, and the glutamate-receptor antagonist kynurenate greatly decreases the static activity of the slowly adapting neurons of Merkel cell-neurite complexes. To investigate the possibility of glutaminergic interaction in PCs, we applied antibodies to glutamate, glutamate receptors, glutamate transporters, and SNARE proteins to cat mesenteric PC sections. Positive labeling was seen in the inner-core lamellae, at inter-lamellar connections, where the lamellae contact the membrane of the neurite and at the lamellar tips. The presence of these proteins on the lamellae and neurite membranes, demonstrated both with immunofluorescent light microscopy as well as immunogold electron microscopy, suggests a chemical, possibly bidirectional, interaction between the lamellar cells and the neurite. Thus, the capsule of the PC, apart from having a mechanical filtering function, may also provide an environment for lamellar-neurite interaction, perhaps acting as a neuro-modulator of the initiation, and/or continuation, of the mechanical-electrical transduction process. At the very least, the presence of the aforementioned proteins suggest some sort of "synaptic-like" activity in these mechanoreceptors, which up until now has not been considered possible.


Subject(s)
Glutamic Acid/metabolism , Mechanotransduction, Cellular/physiology , Neurites/metabolism , Neurites/ultrastructure , Pacinian Corpuscles/metabolism , Pacinian Corpuscles/ultrastructure , Animals , Cats , Female , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Receptors, Metabotropic Glutamate/biosynthesis , SNARE Proteins/biosynthesis , Vesicular Glutamate Transport Protein 1/biosynthesis
20.
Biochem Biophys Res Commun ; 356(3): 756-62, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17379187

ABSTRACT

Type 2 diabetes is characterised by elevated blood glucose and fatty acid concentrations, and aberrant expression of exocytotic soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. Restoration of normoglycaemia is often accomplished through use of the thiazolidinedione drug rosiglitazone (RSG), although little is known of its actions on the pancreas. Here we report that high glucose resulted in 96.6+/-0.2% inhibition of secretagogue-stimulated insulin secretion and 44.9+/-6.2% reduction in beta-cell insulin content. High glucose and lipid resulted in altered target-SNARE expression, syntaxin 1 becoming barely detectable whilst SNAP-25 was greatly up-regulated. RSG intervention further increased the expression of SNAP-25, but did not up-regulate syntaxin 1 expression. In summary, high glucose results in almost total attenuation of stimulated insulin secretion, partial depletion of beta-cell insulin stores and dysregulation of SNARE protein expression. RSG up-regulates SNAP-25 expression, but crucially not syntaxin 1 and hence fails to enhance insulin secretion.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Thiazolidinediones/pharmacology , Animals , Cells, Cultured , Glucose/toxicity , Insulin Secretion , Insulin-Secreting Cells/drug effects , Oleic Acid/toxicity , Palmitic Acid/toxicity , Rats , Rosiglitazone , SNARE Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...