1
|
Jia Q, Sieburth D. Mitochondrial hydrogen peroxide positively regulates neuropeptide secretion during diet-induced activation of the oxidative stress response. Nat Commun 2021; 12:2304. [PMID: 33863916 PMCID: PMC8052458 DOI: 10.1038/s41467-021-22561-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
Mitochondria play a pivotal role in the generation of signals coupling metabolism with neurotransmitter release, but a role for mitochondrial-produced ROS in regulating neurosecretion has not been described. Here we show that endogenously produced hydrogen peroxide originating from axonal mitochondria (mtH2O2) functions as a signaling cue to selectively regulate the secretion of a FMRFamide-related neuropeptide (FLP-1) from a pair of interneurons (AIY) in C. elegans. We show that pharmacological or genetic manipulations that increase mtH2O2 levels lead to increased FLP-1 secretion that is dependent upon ROS dismutation, mitochondrial calcium influx, and cysteine sulfenylation of the calcium-independent PKC family member PKC-1. mtH2O2-induced FLP-1 secretion activates the oxidative stress response transcription factor SKN-1/Nrf2 in distal tissues and protects animals from ROS-mediated toxicity. mtH2O2 levels in AIY neurons, FLP-1 secretion and SKN-1 activity are rapidly and reversibly regulated by exposing animals to different bacterial food sources. These results reveal a previously unreported role for mtH2O2 in linking diet-induced changes in mitochondrial homeostasis with neuropeptide secretion.
Collapse
Affiliation(s)
- Qi Jia
- PIBBS program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Derek Sieburth
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Calmodulin Bidirectionally Regulates Evoked and Spontaneous Neurotransmitter Release at Retinal Ribbon Synapses. eNeuro 2021; 8:ENEURO.0257-20.2020. [PMID: 33293457 PMCID: PMC7808332 DOI: 10.1523/eneuro.0257-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 11/21/2022] Open
Abstract
For decades, a role for the Ca2+-binding protein calmodulin (CaM) in Ca2+-dependent presynaptic modulation of synaptic transmission has been recognized. Here, we investigated the influence of CaM on evoked and spontaneous neurotransmission at rod bipolar (RB) cell→AII amacrine cell synapses in the mouse retina. Our work was motivated by the observations that expression of CaM in RB axon terminals is extremely high and that [Ca2+] in RB terminals normally rises sufficiently to saturate endogenous buffers, making tonic CaM activation likely. Taking advantage of a model in which RBs can be stimulated by expressed channelrhodopsin-2 (ChR2) to avoid dialysis of the presynaptic terminal, we found that inhibition of CaM dramatically decreased evoked release by inhibition of presynaptic Ca channels while at the same time potentiating both Ca2+-dependent and Ca2+-independent spontaneous release. Remarkably, inhibition of myosin light chain kinase (MLCK), but not other CaM-dependent targets, mimicked the effects of CaM inhibition on evoked and spontaneous release. Importantly, initial antagonism of CaM occluded the effect of subsequent inhibition of MLCK on spontaneous release. We conclude that CaM, by acting through MLCK, bidirectionally regulates evoked and spontaneous release at retinal ribbon synapses.
Collapse
|
3
|
Stephens DC, Powell TW, Taraska JW, Harris DA. Imaging the rapid yet transient accumulation of regulatory lipids, lipid kinases, and protein kinases during membrane fusion, at sites of exocytosis of MMP-9 in MCF-7 cells. Lipids Health Dis 2020; 19:195. [PMID: 32829709 PMCID: PMC7444259 DOI: 10.1186/s12944-020-01374-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/17/2020] [Indexed: 11/10/2022] Open
Abstract
Background The regulation of exocytosis is physiologically vital in cells and requires a variety of distinct proteins and lipids that facilitate efficient, fast, and timely release of secretory vesicle cargo. Growing evidence suggests that regulatory lipids act as important lipid signals and regulate various biological processes including exocytosis. Though functional roles of many of these regulatory lipids has been linked to exocytosis, the dynamic behavior of these lipids during membrane fusion at sites of exocytosis in cell culture remains unknown. Methods Total internal reflection fluorescence microscopy (TIRF) was used to observe the spatial organization and temporal dynamics (i.e. spatial positioning and timing patterns) of several lipids, and accessory proteins, like lipid kinases and protein kinases, in the form of protein kinase C (PRKC) associated with sites of exocytosis of matrix metalloproteinase-9 (MMP-9) in living MCF-7 cancer cells. Results Following stimulation with phorbol myristate acetate (PMA) to promote exocytosis, a transient accumulation of several distinct regulatory lipids, lipid kinases, and protein kinases at exocytic sites was observed. This transient accumulation centered at the time of membrane fusion is followed by a rapid diffusion away from the fusion sites. Additionally, the synthesis of these regulatory lipids, degradation of these lipids, and the downstream effectors activated by these lipids, are also achieved by the recruitment and accumulation of key enzymes at exocytic sites (during the moment of cargo release). This includes key enzymes like lipid kinases, protein kinases, and phospholipases that facilitate membrane fusion and exocytosis of MMP-9. Conclusions This work suggests that these regulatory lipids and associated effector proteins are locally synthesized and/or recruited to sites of exocytosis, during membrane fusion and cargo release. More importantly, their enrichment at fusion sites serves as an important spatial and temporal organizing “element” defining individual exocytic sites.
Collapse
Affiliation(s)
- Dominique C Stephens
- Department of Chemistry, Howard University, 525 College Street NW, Washington, D.C, 20059, USA
| | - Tyrel W Powell
- Department of Chemistry, Howard University, 525 College Street NW, Washington, D.C, 20059, USA
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dinari A Harris
- Department of Chemistry, Howard University, 525 College Street NW, Washington, D.C, 20059, USA.
| |
Collapse
|
4
|
Taleat Z, Larsson A, Ewing AG. Anticancer Drug Tamoxifen Affects Catecholamine Transmitter Release and Storage from Single Cells. ACS Chem Neurosci 2019; 10:2060-2069. [PMID: 30763068 DOI: 10.1021/acschemneuro.8b00714] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Electrochemical measurements of exocytosis combined with intracellular vesicle impact electrochemical cytometry have been used to evaluate the effect of an anticancer drug, tamoxifen, on catecholamine release at the single-cell level. Tamoxifen has been used for over 40 years to treat estrogen receptor-positive breast cancers during both early stages of the disease and in the adjuvant setting. Tamoxifen causes memory and cognitive dysfunction, but the reasons for the cognitive impairment and memory problems induced by this anticancer drug are not well-known. We show that tamoxifen, through a nongenomic mechanism, can modulate both exocytosis and vesicle catecholamine storage in a model cell line. The results indicate that exocytosis is inhibited at high concentrations of tamoxifen and is stimulated at low levels. Tamoxifen also elicits a significant concentration-dependent change in total catecholamine content of single vesicles, while sub-nanomolar concentrations of the drug have stimulatory activity on the catecholamine content of vesicles. In addition, it has profound effects on storage at higher concentrations. Tamoxifen also reduces the intracellular free Ca2+ but only at micromolar concentration, by acting on voltage-gated Ca2+ channels, which likely affects neurotransmitter secretion.
Collapse
Affiliation(s)
- Zahra Taleat
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Gothenburg, Sweden
| | - Anna Larsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Gothenburg, Sweden
| | - Andrew G. Ewing
- Department of Chemistry and Molecular Biology, University of Gothenburg, SE-412 96 Gothenburg, Sweden
| |
Collapse
|
5
|
Armant DR. Intracellular Ca2+ signaling and preimplantation development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 843:151-71. [PMID: 25956298 PMCID: PMC10412982 DOI: 10.1007/978-1-4939-2480-6_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The key, versatile role of intracellular Ca2+ signaling during egg activation after fertilization has been appreciated for several decades. More recently, evidence has accumulated supporting the concept that cytoplasmic Ca2+ is also a major signaling nexus during subsequent development of the fertilized ovum. This chapter will review the molecular reactions that regulate intracellular Ca2+ levels and cell function, the role of Ca2+ signaling during egg activation and specific examples of repetitive Ca2+ signaling found throughout pre- and peri-implantation development. Many of the upstream and downstream pathways utilized during egg activation are also critical for specific processes that take place during embryonic development. Much remains to be done to elucidate the full complexity of Ca2+ signaling mechanisms in preimplantation embryos to the level of detail accomplished for egg activation. However, an emerging concept is that because this second messenger can be modulated downstream of numerous receptors and is able to bind and activate multiple cytoplasmic signaling proteins, it can help the coordination of development through up- and downstream pathways that change with each embryonic stage.
Collapse
Affiliation(s)
- D Randall Armant
- Department of Obstetrics and Gynecology, Wayne State University C.S. Mott Center for Human Growth and Development, 275 E. Hancock Street, 48201-1405, Detroit, MI, USA,
| |
Collapse
|
6
|
Fang P, Xu W, Li D, Zhao X, Dai J, Wang Z, Yan X, Qin M, Zhang Y, Xu C, Wang L, Qiao Z. A novel acrosomal protein, IQCF1, involved in sperm capacitation and the acrosome reaction. Andrology 2014; 3:332-44. [PMID: 25380116 DOI: 10.1111/andr.296] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 09/08/2014] [Accepted: 09/20/2014] [Indexed: 12/11/2022]
Abstract
On the basis of the unknown tags in the mature human sperm serial analysis of gene expression library constructed by our laboratory, some transcripts were cloned, including Iqcf1 (IQ motif containing F1). To investigate the function of sperm-retained Iqcf1 in spermatogenesis and fertilization of mice, we investigated the spatial and temporal expression of IQCF1. By using the (transcription activator-like effector nuclease) strategy, Iqcf1-knockout mice were produced, and the phenotypes of the Iqcf1(-/-) mice were analyzed. The results showed that IQCF1 was localized in the acrosome of spermatozoa and spermatids; the expression of IQCF1 in testes was associated with spermatogenic capacity. The Iqcf1(-/-) mice were significantly less fertile than the wild-type mice (p = 0.0057) because of reduced sperm motility (p = 0.0094) and the acrosome reaction (AR) (p = 0.0093). In spermatozoa, IQCF1 interacted with calmodulin (CaM) and possibly participated in the tyrosine phosphorylation of sperm proteins during capacitation. In conclusion, a newly identified acrosomal protein, IQCF1, is closely related to sperm capacitation and AR; in particular, it is involved in tyrosine phosphorylation of sperm proteins through interaction with CaM. Research into the function of IQCF1 during fertilization could facilitate the investigation of the molecular mechanism of capacitation, which is unclear.
Collapse
Affiliation(s)
- P Fang
- School of Life Sciences and Biotechnology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Ando K, Kudo Y, Aoyagi K, Ishikawa R, Igarashi M, Takahashi M. Calmodulin-dependent regulation of neurotransmitter release differs in subsets of neuronal cells. Brain Res 2013; 1535:1-13. [DOI: 10.1016/j.brainres.2013.08.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/25/2013] [Accepted: 08/08/2013] [Indexed: 02/05/2023]
|
8
|
Activity-dependent regulation of synaptic vesicle exocytosis and presynaptic short-term plasticity. Neurosci Res 2011; 70:16-23. [DOI: 10.1016/j.neures.2011.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 01/25/2011] [Accepted: 03/15/2011] [Indexed: 11/23/2022]
|
9
|
Saransaari P, Oja SS. Mechanisms of Inhibitory Amino Acid Release in the Brain Stem Under Normal and Ischemic Conditions. Neurochem Res 2010; 35:1948-56. [DOI: 10.1007/s11064-010-0265-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2010] [Indexed: 12/23/2022]
|
10
|
Di Giovanni J, Iborra C, Maulet Y, Lévêque C, El Far O, Seagar M. Calcium-dependent regulation of SNARE-mediated membrane fusion by calmodulin. J Biol Chem 2010; 285:23665-75. [PMID: 20519509 DOI: 10.1074/jbc.m109.096073] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuroexocytosis requires SNARE proteins, which assemble into trans complexes at the synaptic vesicle/plasma membrane interface and mediate bilayer fusion. Ca(2+) sensitivity is thought to be conferred by synaptotagmin, although the ubiquitous Ca(2+)-effector calmodulin has also been implicated in SNARE-dependent membrane fusion. To examine the molecular mechanisms involved, we examined the direct action of calmodulin and synaptotagmin in vitro, using fluorescence resonance energy transfer to assay lipid mixing between target- and vesicle-SNARE liposomes. Ca(2+)/calmodulin inhibited SNARE assembly and membrane fusion by binding to two distinct motifs located in the membrane-proximal regions of VAMP2 (K(D) = 500 nm) and syntaxin 1 (K(D) = 2 microm). In contrast, fusion was increased by full-length synaptotagmin 1 anchored in vesicle-SNARE liposomes. When synaptotagmin and calmodulin were combined, synaptotagmin overcame the inhibitory effects of calmodulin. Furthermore, synaptotagmin displaced calmodulin binding to target-SNAREs. These findings suggest that two distinct Ca(2+) sensors act antagonistically in SNARE-mediated fusion.
Collapse
|
11
|
Calmodulin controls synaptic strength via presynaptic activation of calmodulin kinase II. J Neurosci 2010; 30:4132-42. [PMID: 20237283 DOI: 10.1523/jneurosci.3129-09.2010] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Calmodulin regulates multifarious cellular processes via a panoply of target interactions. However, the central role, multiple isoforms, and complex target interactions of calmodulin make it difficult to examine its precise functions. Here, we analyzed calmodulin function in neurons using lentivirally delivered short-hairpin RNAs that suppressed expression of all calmodulin isoforms by approximately 70%. Calmodulin knockdown did not significantly alter neuronal survival or synapse formation but depressed spontaneous neuronal network activity. Strikingly, calmodulin knockdown decreased the presynaptic release probability almost twofold, without altering the presynaptic readily-releasable vesicle pool or postsynaptic neurotransmitter reception. In calmodulin knockdown neurons, presynaptic release was restored to wild-type levels by expression of constitutively active calmodulin-dependent kinase-IIalpha (CaMKIIalpha); in contrast, in control neurons, expression of constitutively active CaMKIIalpha had no effect on presynaptic release. Viewed together, these data suggest that calmodulin performs a major function in boosting synaptic strength via direct activation of presynaptic calmodulin-dependent kinase II.
Collapse
|
12
|
Ca2+-dependent release of Munc18-1 from presynaptic mGluRs in short-term facilitation. Proc Natl Acad Sci U S A 2009; 106:18385-9. [PMID: 19822743 DOI: 10.1073/pnas.0910088106] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Short-term synaptic facilitation plays an important role in information processing in the central nervous system. Although the crucial requirement of presynaptic Ca(2+) in the expression of this plasticity has been known for decades, the molecular mechanisms underlying the plasticity remain controversial. Here, we show that presynaptic metabotropic glutamate receptors (mGluRs) bind and release Munc18-1 (also known as rbSec1/nSec1), an essential protein for synaptic transmission, in a Ca(2+)-dependent manner, whose actions decrease and increase synaptic vesicle release, respectively. We found that mGluR4 bound Munc18-1 with an EC(50) for Ca(2+) of 168 nM, close to the resting Ca(2+) concentration, and that the interaction was disrupted by Ca(2+)-activated calmodulin (CaM) at higher concentrations of Ca(2+). Consistently, the Munc18-1-interacting domain of mGluR4 suppressed both dense-core vesicle secretion from permeabilized PC12 cells and synaptic transmission in neuronal cells. Furthermore, this domain was sufficient to induce paired-pulse facilitation. Obviously, the role of mGluR4 in these processes was independent of its classical function of activation by glutamate. On the basis of these experimental data, we propose the following model: When neurons are not active, Munc18-1 is sequestered by mGluR4, and therefore the basal synaptic transmission is kept low. After the action potential, the increase in the Ca(2+) level activates CaM, which in turn liberates Munc18-1 from mGluR4, causing short-term synaptic facilitation. Our findings unite and provide a new insight into receptor signaling and vesicular transport, which are pivotal activities involved in a variety of cellular processes.
Collapse
|
13
|
Russo LC, Goñi CN, Castro LM, Asega AF, Camargo ACM, Trujillo CA, Ulrich H, Glucksman MJ, Scavone C, Ferro ES. Interaction with calmodulin is important for the secretion of thimet oligopeptidase following stimulation. FEBS J 2009; 276:4358-71. [PMID: 19614740 DOI: 10.1111/j.1742-4658.2009.07144.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thimet oligopeptidase (EC 3.4.24.15; EP24.15) was originally described as a neuropeptide-metabolizing enzyme, highly expressed in the brain, kidneys and neuroendocrine tissue. EP24.15 lacks a typical signal peptide sequence for entry into the secretory pathway and is secreted by cells via an unconventional and unknown mechanism. In this study, we identified a novel calcium-dependent interaction between EP24.15 and calmodulin, which is important for the stimulated, but not constitutive, secretion of EP24.15. We demonstrated that, in vitro, EP24.15 and calmodulin physically interact only in the presence of Ca2+, with an estimated Kd value of 0.52 mum. Confocal microscopy confirmed that EP24.15 colocalizes with calmodulin in the cytosol of resting HEK293 cells. This colocalization markedly increases when cells are treated with either the calcium ionophore A23187 or the protein kinase A activator forskolin. Overexpression of calmodulin in HEK293 cells is sufficient to greatly increase the A23187-stimulated secretion of EP24.15, which can be inhibited by the calmodulin inhibitor calmidazolium. The specific inhibition of protein kinase A with KT5720 reduces the A23187-stimulated secretion of EP24.15 and inhibits the synergistic effects of forskolin with A23187. Treatment with calmidazolium and KT5720 nearly abolishes the stimulatory effects of A23187 on EP24.15 secretion. Together, these data suggest that the interaction between EP24.15 and calmodulin is regulated within cells and is important for the stimulated secretion of EP24.15 from HEK293 cells.
Collapse
Affiliation(s)
- Lilian C Russo
- Department of Cell Biology and Development, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Signal transduction pathways that regulate sperm capacitation and the acrosome reaction. Arch Biochem Biophys 2009; 485:72-81. [DOI: 10.1016/j.abb.2009.02.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 02/06/2009] [Accepted: 02/07/2009] [Indexed: 01/28/2023]
|
15
|
Delarue-Cochin S, McCort-Tranchepain I. Synthesis of new aza-analogs of staurosporine, K-252a and rebeccamycin by nucleophilic opening of C2-symmetric bis-aziridines. Org Biomol Chem 2009; 7:706-16. [DOI: 10.1039/b815737e] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
16
|
Vesicle priming and recruitment by ubMunc13-2 are differentially regulated by calcium and calmodulin. J Neurosci 2008; 28:1949-60. [PMID: 18287511 DOI: 10.1523/jneurosci.5096-07.2008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ca2+ regulates multiple processes in nerve terminals, including synaptic vesicle recruitment, priming, and fusion. Munc13s, the mammalian homologs of Caenorhabditis elegans Unc13, are essential vesicle-priming proteins and contain multiple regulatory domains that bind second messengers such as diacylglycerol and Ca2+/calmodulin (Ca2+/CaM). Binding of Ca2+/CaM is necessary for the regulatory effect that allows Munc13-1 and ubMunc13-2 to promote short-term synaptic plasticity. However, the relative contributions of Ca2+ and Ca2+/CaM to vesicle priming and recruitment by Munc13 are not known. Here, we investigated the effect of Ca2+/CaM binding on ubMunc13-2 activity in chromaffin cells via membrane-capacitance measurements and a detailed simulation of the exocytotic machinery. Stimulating secretion under various basal Ca2+ concentrations from cells overexpressing either ubMunc13-2 or a ubMunc13-2 mutant deficient in CaM binding enabled a distinction between the effects of Ca2+ and Ca2+/CaM. We show that vesicle priming by ubMunc13-2 is Ca2+ dependent but independent of CaM binding to ubMunc13-2. However, Ca2+/CaM binding to ubMunc13-2 specifically promotes vesicle recruitment during ongoing stimulation. Based on the experimental data and our simulation, we propose that ubMunc13-2 is activated by two Ca2+-dependent processes: a slow activation mode operating at low Ca2+ concentrations, in which ubMunc13-2 acts as a priming switch, and a fast mode at high Ca2+ concentrations, in which ubMunc13-2 is activated in a Ca2+/CaM-dependent manner and accelerates vesicle recruitment and maturation during stimulation. These different Ca2+ activation steps determine the kinetic properties of exocytosis and vesicle recruitment and can thus alter plasticity and efficacy of transmitter release.
Collapse
|
17
|
Staal RGW, Hananiya A, Sulzer D. PKC theta activity maintains normal quantal size in chromaffin cells. J Neurochem 2008; 105:1635-41. [PMID: 18248621 DOI: 10.1111/j.1471-4159.2008.05264.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Protein kinase C (PKC) activity mediates multiple neurosecretory processes, but these are poorly understood due in part to the existence of at least 12 PKC isoforms. Using amperometry to record quantal catecholamine release from chromaffin cells, we found that both broad spectrum PKC antagonists and rottlerin, a selective inhibitor of the novel isoforms PKC theta and PKC delta, decreased quantal size and the number of secretory events recorded per stimulus. In contrast, drugs that selectively inhibit the atypical and conventional PKC isoforms had no effect on these parameters. While both PKC theta and delta were expressed in chromaffin cells, mice deficient for PKC theta, but not for PKC delta, exhibited lower quantal size than wild-type and were insensitive to rottlerin. Finally, an inhibitory PKC theta pseudosubstrate produced rottlerin-like responses in wild-type mice, indicating that the lack of rottlerin response in the PKC theta mutants was not the result of a form of compensation. These findings demonstrate neurosecretory regulation by a novel PKC isoform, PKC theta, and should contribute to defining mechanisms of activity-dependent regulation of neurosecretion.
Collapse
Affiliation(s)
- Roland G W Staal
- Department of Neurology, Columbia University Medical Center, New York, New York 10032, USA
| | | | | |
Collapse
|
18
|
Abstract
G-proteins (guanine nucleotide-binding proteins) are membrane-attached proteins composed of three subunits, alpha, beta, and gamma. They transduce signals from G-protein coupled receptors (GPCRs) to target effector proteins. The agonistactivated receptor induces a conformational change in the G-protein trimer so that the alpha-subunit binds GTP in exchange for GDP and alpha-GTP, and betagamma-subunits separate to interact with the target effector. Effector-interaction is terminated by the alpha-subunit GTPase activity, whereby bound GTP is hydrolyzed to GDP. This is accelerated in situ by RGS proteins, acting as GTPase-activating proteins (GAPs). Galpha-GDP and Gbetagamma then reassociate to form the Galphabetagamma trimer. G-proteins primarily involved in the modulation of neurotransmitter release are G(o), G(q) and G(s). G(o) mediates the widespread presynaptic auto-inhibitory effect of many neurotransmitters (e.g., via M2/M4 muscarinic receptors, alpha(2) adrenoreceptors, micro/delta opioid receptors, GABAB receptors). The G(o) betagamma-subunit acts in two ways: first, and most ubiquitously, by direct binding to CaV2 Ca(2+) channels, resulting in a reduced sensitivity to membrane depolarization and reduced Ca(2+) influx during the terminal action potential; and second, through a direct inhibitory effect on the transmitter release machinery, by binding to proteins of the SNARE complex. G(s) and G(q) are mainly responsible for receptor-mediated facilitatory effects, through activation of target enzymes (adenylate cyclase, AC and phospholipase-C, PLC respectively) by the GTP-bound alpha-subunits.
Collapse
Affiliation(s)
- David A Brown
- Department of Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| | | |
Collapse
|
19
|
Pungercar J, Krizaj I. Understanding the molecular mechanism underlying the presynaptic toxicity of secreted phospholipases A2. Toxicon 2007; 50:871-92. [PMID: 17905401 DOI: 10.1016/j.toxicon.2007.07.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 07/13/2007] [Accepted: 07/20/2007] [Indexed: 11/24/2022]
Abstract
An important group of toxins, whose action at the molecular level is still a matter of debate, is secreted phospholipases A(2) (sPLA(2)s) endowed with presynaptic or beta-neurotoxicity. The current belief is that these beta-neurotoxins (beta-ntxs) exert their toxicity primarily due to their extracellular enzymatic action on the plasma membrane of motoneurons at the neuromuscular junction. However, the discovery of several extra- and intracellular proteins, with high binding affinity for snake venom beta-ntxs, has raised the question as to whether this explanation is adequate to account for all the observed phenomena in the process of presynaptic toxicity. The purpose of this review is to critically examine the various published studies, including the most recent results on internalization of a beta-ntx into motor nerve terminals, in order to contribute to a better understanding of the molecular mechanism of beta-neurotoxicity. As a result, we propose that presynaptic neurotoxicity of sPLA(2)s is a result of both extra- and intracellular actions of beta-ntxs, involving enzymatic activity as well as interaction of the toxins with intracellular proteins affecting the cycling of synaptic vesicles in the axon terminals of vertebrate motoneurons.
Collapse
Affiliation(s)
- Joze Pungercar
- Department of Molecular and Biomedical Sciences, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | | |
Collapse
|
20
|
Igarashi M, Watanabe M. Roles of calmodulin and calmodulin-binding proteins in synaptic vesicle recycling during regulated exocytosis at submicromolar Ca2+ concentrations. Neurosci Res 2007; 58:226-33. [PMID: 17601619 DOI: 10.1016/j.neures.2007.03.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Revised: 03/14/2007] [Accepted: 03/14/2007] [Indexed: 11/28/2022]
Abstract
Calcium ion is required at various concentrations for vesicular recycling in the presynaptic terminal. Although calmodulin (CaM) is the most abundant Ca2+-binding protein and has a submicromolar affinity for Ca2+, it is not the Ca2+ sensor for vesicular fusion because this process requires Ca2+ concentrations above 1 microM. Several lines of evidence, however, suggest that CaM mediates the regulation of vesicular recycling by submicromolar Ca2+ via novel protein-protein interactions. In this review, we discuss recent findings on how CaM regulates synaptic vesicle recycling by controlling the SNARE mechanism, which is the molecular machinery that mediates exocytosis.
Collapse
Affiliation(s)
- Michihiro Igarashi
- Division of Molecular and Cellular Biology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahi-Machi, Niigata 951-8510, Japan.
| | | |
Collapse
|
21
|
Saransaari P, Oja SS. Modulation of GABA release by second messenger substances and NO in mouse brain stem slices under normal and ischemic conditions. Neurochem Res 2006; 31:1317-25. [PMID: 17053971 DOI: 10.1007/s11064-006-9174-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Accepted: 09/14/2006] [Indexed: 11/27/2022]
Abstract
GABA is the inhibitory neurotransmitter in most brain stem nuclei. The properties of release of preloaded [(3)H]GABA were now investigated with slices from the mouse brain stem under normal and ischemic (oxygen and glucose deprivation) conditions, using a superfusion system. The ischemic GABA release increased about fourfold in comparison with normal conditions. The tyrosine kinase inhibitor genistein had no effect on GABA release, while the phospholipase inhibitor quinacrine reduced both the basal and K(+)-evoked release in normoxia and ischemia. The activator of protein kinase C (PKC) 4beta-phorbol 12-myristate 13-acetate had no effects on the releases, whereas the PKC inhibitor chelerythrine reduced the basal release in ischemia. When the cyclic guanosine monophosphate (cGMP) levels were increased by superfusion with zaprinast and other phosphodiesterase inhibitors, GABA release was reduced under normal conditions. The NO donors S-nitroso-N-acetylpenicillamine (SNAP) and hydroxylamine (HA) enhanced the basal and K(+)-stimulated release by acting directly on presynaptic terminals. Under ischemic conditions GABA release was enhanced when cGMP levels were increased by zaprinast. This effect was confirmed by inhibition of the release by the guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ). The NO-producing agents SNAP, HA, and sodium nitroprusside potentiated GABA release in ischemia. These effects were reduced by the NO synthase inhibitor N(G)-nitro-L: -arginine, but not by ODQ. The results show that particularly NO and cGMP regulate both normal and ischemic GABA release in the brain stem. Their effects are however complex.
Collapse
Affiliation(s)
- Pirjo Saransaari
- Brain Research Center, Medical School, 33014 University of Tampere, Tampere, Finland.
| | | |
Collapse
|
22
|
Abstract
Embryonic development is initiated after the fertilizing spermatozoon enters the egg and triggers a series of events known as egg activation. Activation results in an increase in intracellular calcium concentration, cortical granule exocytosis (CGE), cell cycle resumption and recruitment of maternal mRNA. CGE is an evolutionary developed mechanism that causes modification of the zona pellucida to prevent penetration of additional spermatozoa, ensuring successful egg activation and embryo development. The egg CGE is a unique and convenient mammalian model for studying the different proteins participating at the membrane fusion cascade, which, unlike other secretory cells, occurs only once in the egg's lifespan. This article highlights a number of proteins, ascribed to participate in CGE and thus the block to polyspermy. CGE can be triggered either by a calcium dependent pathway, or via protein kinase C (PKC) activation that requires a very low calcium concentration. In a recent study, we suggested that the filamentous actin (F-actin) at the egg's cortex is a dynamic network. It can be maneuvered towards allowing CGE by activated actin associated proteins and/or by activated PKC and its down stream proteins, such as myristoylated alanine-rich C kinase substrate (MARCKS). MARCKS, a protein known to cross-link F-actin in other cell types, was found to be expressed and colocalized with actin in non-activated MII eggs. We further demonstrated MARCKS dissociation from actin after activation by ionomycin, a process that can lead to the breakdown of the actin network, thus allowing CGE. The more we know of the intricate process of CGE and of the proteins participating in it, the more the assisted reproductive procedures might benefit from that knowledge.
Collapse
Affiliation(s)
- Alina Tsaadon
- Department of Cell & Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
23
|
Kubista H, Boehm S. Molecular mechanisms underlying the modulation of exocytotic noradrenaline release via presynaptic receptors. Pharmacol Ther 2006; 112:213-42. [PMID: 16730801 DOI: 10.1016/j.pharmthera.2006.04.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Accepted: 04/10/2006] [Indexed: 10/24/2022]
Abstract
The release of noradrenaline from nerve terminals is modulated by a variety of presynaptic receptors. These receptors belong to one of the following three receptor superfamilies: transmitter-gated ion channels, G protein-coupled receptors (GPCR), and membrane receptors with intracellular enzymatic activities. For representatives of each of these three superfamilies, receptor activation has been reported to cause either an enhancement or a reduction of noradrenaline release. As these receptor classes display greatly diverging structures and functions, a multitude of different molecular mechanisms are involved in the regulation of noradrenaline release via presynaptic receptors. This review gives a short overview of the presynaptic receptors on noradrenergic nerve terminals and summarizes the events involved in vesicle exocytosis in order to finally delineate the most important signaling cascades that mediate the modulation via presynaptic receptors. In addition, the interactions between the various presynaptic receptors are described and the underlying molecular mechanisms are elucidated. Together, these presynaptic signaling mechanisms form a sophisticated network that precisely adapts the amount of noradrenaline being released to a given situation.
Collapse
Affiliation(s)
- Helmut Kubista
- Institute of Pharmacology, Centre of Biomolecular Medicine and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, A-1090 Vienna, Austria
| | | |
Collapse
|
24
|
Hilfiker S, Benfenati F, Doussau F, Nairn AC, Czernik AJ, Augustine GJ, Greengard P. Structural domains involved in the regulation of transmitter release by synapsins. J Neurosci 2006; 25:2658-69. [PMID: 15758176 PMCID: PMC6725186 DOI: 10.1523/jneurosci.4278-04.2005] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Synapsins are a family of neuron-specific phosphoproteins that regulate neurotransmitter release by associating with synaptic vesicles. Synapsins consist of a series of conserved and variable structural domains of unknown function. We performed a systematic structure-function analysis of the various domains of synapsin by assessing the actions of synapsin fragments on neurotransmitter release, presynaptic ultrastructure, and the biochemical interactions of synapsin. Injecting a peptide derived from domain A into the squid giant presynaptic terminal inhibited neurotransmitter release in a phosphorylation-dependent manner. This peptide had no effect on vesicle pool size, synaptic depression, or transmitter release kinetics. In contrast, a peptide fragment from domain C reduced the number of synaptic vesicles in the periphery of the active zone and increased the rate and extent of synaptic depression. This peptide also slowed the kinetics of neurotransmitter release without affecting the number of docked vesicles. The domain C peptide, as well as another peptide from domain E that is known to have identical effects on vesicle pool size and release kinetics, both specifically interfered with the binding of synapsins to actin but not with the binding of synapsins to synaptic vesicles. This suggests that both peptides interfere with release by preventing interactions of synapsins with actin. Thus, interactions of domains C and E with the actin cytoskeleton may allow synapsins to perform two roles in regulating release, whereas domain A has an actin-independent function that regulates transmitter release in a phosphorylation-sensitive manner.
Collapse
Affiliation(s)
- Sabine Hilfiker
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10021, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Barclay JW, Morgan A, Burgoyne RD. Calcium-dependent regulation of exocytosis. Cell Calcium 2005; 38:343-53. [PMID: 16099500 DOI: 10.1016/j.ceca.2005.06.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Accepted: 06/28/2005] [Indexed: 11/30/2022]
Abstract
A rapid increase in intracellular calcium directly triggers regulated exocytosis. In addition, changes in intracellular calcium concentration can adjust the extent of exocytosis (quantal content) or the magnitude of individual release events (quantal size) in both the short- and long-term. It is generally agreed that calcium achieves this regulation via an interaction with a number of different molecular targets located at or near to the site of membrane fusion. We review here the synaptic proteins with defined calcium-binding domains and protein kinases activated by calcium, summarize what is known about their function in membrane fusion and the experimental evidence in support of their involvement in synaptic plasticity.
Collapse
Affiliation(s)
- Jeff W Barclay
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | | | | |
Collapse
|
26
|
Vitale N, Mawet J, Camonis J, Regazzi R, Bader MF, Chasserot-Golaz S. The Small GTPase RalA controls exocytosis of large dense core secretory granules by interacting with ARF6-dependent phospholipase D1. J Biol Chem 2005; 280:29921-8. [PMID: 15980073 DOI: 10.1074/jbc.m413748200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RalA and RalB constitute a family of highly similar Ras-related GTPases widely distributed in different tissues. Recently, active forms of Ral proteins have been shown to bind to the exocyst complex, implicating them in the regulation of cellular secretion. Since RalA is present on the plasma membrane in neuroendocrine chromaffin and PC12 cells, we investigated the potential role of RalA in calcium-regulated exocytotic secretion. We show here that endogenous RalA is activated during exocytosis. Expression of the constitutively active RalA (G23V) mutant enhances secretagogue-evoked secretion from PC12 cells. Conversely, expression of the constitutively inactive GDP-bound RalA (G26A) or silencing of the RalA gene by RNA interference led to a strong impairment of the exocytotic response. RalA was found to co-localize with phospholipase D1 (PLD1) at the plasma membrane in PC12 cells. We demonstrate that cell stimulation triggers a direct interaction between RalA and ARF6-activated PLD1. Moreover, reduction of endogenous RalA expression level interfered with the activation of PLD1 observed in secretagogue-stimulated cells. Finally, using various RalA mutants selectively impaired in their ability to activate downstream effectors, we show that PLD1 activation is essential for the activation of secretion by GTP-loaded RalA. Together, these results provide evidence that RalA is a positive regulator of calcium-evoked exocytosis of large dense core secretory granules and suggest that stimulation of PLD1 and consequent changes in plasma membrane phospholipid composition is the major function RalA undertakes in calcium-regulated exocytosis.
Collapse
Affiliation(s)
- Nicolas Vitale
- Centre National de la Recherche Scientifique, Unité Propre de Recherche 2356 Neurotransmission and Sécrétion Neuroendocrine, Strasbourg France
| | | | | | | | | | | |
Collapse
|
27
|
Armant DR. Blastocysts don't go it alone. Extrinsic signals fine-tune the intrinsic developmental program of trophoblast cells. Dev Biol 2005; 280:260-80. [PMID: 15882572 PMCID: PMC2715296 DOI: 10.1016/j.ydbio.2005.02.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2004] [Revised: 01/16/2005] [Accepted: 02/08/2005] [Indexed: 01/02/2023]
Abstract
The preimplantation embryo floats freely within the oviduct and is capable of developing into a blastocyst independently of the maternal reproductive tract. While establishment of the trophoblast lineage is dependent on expression of developmental regulatory genes, further differentiation leading to blastocyst implantation in the uterus requires external cues emanating from the microenvironment. Recent studies suggest that trophoblast differentiation requires intracellular signaling initiated by uterine-derived growth factors and integrin-binding components of the extracellular matrix. The progression of trophoblast development from the early blastocyst stage through the onset of implantation appears to be largely independent of new gene expression. Instead, extrinsic signals direct the sequential trafficking of cell surface receptors to orchestrate the developmental program that initiates blastocyst implantation. The dependence on external cues could coordinate embryonic activities with the developing uterine endometrium. Biochemical events that regulate trophoblast adhesion to fibronectin are presented to illustrate a developmental strategy employed by the peri-implantation blastocyst.
Collapse
Affiliation(s)
- D Randall Armant
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201-1415, USA.
| |
Collapse
|
28
|
Suszkiw JB. Presynaptic disruption of transmitter release by lead. Neurotoxicology 2004; 25:599-604. [PMID: 15183013 DOI: 10.1016/j.neuro.2003.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2003] [Accepted: 09/09/2003] [Indexed: 11/24/2022]
Abstract
Low concentrations of inorganic lead ions (Pb2+) disrupt transmitter release by causing aberrant augmentation of spontaneous and suppression of evoked release. These effects result from high affinity interactions of Pb2+ with the voltage-gated calcium channels (VGCC) as well as Ca2+ binding proteins which regulate the synaptic vesicle mobilization, docking, and exocytosis processes. Augmentation of spontaneous release may involve stimulation of vesicle mobilization consequent to Pb2+ activation of CaMKII-dependent phosphorylation of synapsin I and/or stimulation of asynchronous exocytosis via direct Pb2+ activation of the putative exocytotic Ca2+-sensor protein synaptotagmin I. In addition, synergistic stimulation of PLC and DAG/Pb2+-dependent activation of PKC may enhance the secretagogue effects of Pb2+ by increasing metal sensitivity of exocytosis and/or modulating calcium channel activity. In contrast to intracellularly-mediated actions of Pb2+ resulting in augmentation of spontaneous release, the inhibition of evoked transmitter release by Pb2+ is largely attributable to extracellular block of the voltage-gated calcium channels.
Collapse
Affiliation(s)
- Janusz B Suszkiw
- Department of Cellular and Molecular Physiology, College of Medicine, University of Cincinnati, P.O. Box 670576, Cincinnati, OH 45267-0576, USA.
| |
Collapse
|
29
|
Tulsiani DRP, Abou-Haila A. Is sperm capacitation analogous to early phases of Ca2+-triggered membrane fusion in somatic cells and viruses? Bioessays 2004; 26:281-90. [PMID: 14988929 DOI: 10.1002/bies.20003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
An important feature of male fertility is the physiological priming of spermatozoa by a multifaceted process collectively referred to as capacitation. The end point of this evasive process is the hyperactivated spermatozoa capable of binding to terminal sugar residues on the egg's extracellular coat, the zona pellucida (ZP), and undergoing acrosomal exocytosis (i.e., induction of the acrosome reaction). The hydrolytic action of acrosomal enzymes released at the site of zona binding, along with the enhanced thrust generated by the hyperactivated beat pattern of the bound spermatozoa, are important factors that regulate the penetration of ZP and fertilization of the egg. Despite many advances in identifying sperm components that promote capacitation, the mechanism underlying the calcium-triggered process remains elusive. The purpose of this review article is to focus on new advances that have enhanced our understanding of in vivo/in vitro capacitation, a prerequisite event resulting from a dramatic modification and reorganization of the sperm membrane molecules. Special emphasis has been laid on accumulating evidence suggesting potential similarities between the sperm capacitation and early phases of calcium-triggered membrane fusion (i.e., tethering and docking) during secretory and endocytotic pathways among eukaryotes.
Collapse
Affiliation(s)
- Daulat R P Tulsiani
- Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
30
|
Martin TF, Grishanin RN. PC12 cells as a model for studies of regulated secretion in neuronal and endocrine cells. Methods Cell Biol 2004; 71:267-86. [PMID: 12884694 DOI: 10.1016/s0091-679x(03)01012-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pheochromocytoma-derived cell lines such as PC12 cells maintain a differentiated neuroendocrine phenotype and have been widely used as a convenient model system for a wide variety of cell biological studies on neurotrophin action, monoamine biogenesis, protein trafficking, and secretory vesicle dynamics. This chapter reviews a number of methods that are useful for studies of the regulated dense core vesicle secretory pathway. This includes protocols for maintaining cells and preserving their phenotype. A variety of assays are discussed for monitoring secretion in intact or permeable cells and in transfected cells. Specific methods for immunocytochemical studies in permeable cells are discussed. Finally, protocols for high-efficiency PC12 cell transfections and the isolation of stably transfected cell lines are provided.
Collapse
Affiliation(s)
- T F Martin
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
31
|
de Haro L, Ferracci G, Opi S, Iborra C, Quetglas S, Miquelis R, Lévêque C, Seagar M. Ca2+/calmodulin transfers the membrane-proximal lipid-binding domain of the v-SNARE synaptobrevin from cis to trans bilayers. Proc Natl Acad Sci U S A 2004; 101:1578-83. [PMID: 14757830 PMCID: PMC341777 DOI: 10.1073/pnas.0303274101] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor (SNARE) protein interactions at the synaptic vesicle/plasma membrane interface play an essential role in neurotransmitter release. The membrane-proximal region (amino acids 77-90) of the v-SNARE vesicle-associated membrane protein 2 (VAMP 2, synaptobrevin) binds acidic phospholipids or Ca(2+)/calmodulin in a mutually exclusive manner, processes that are required for Ca(2+)-dependent exocytosis. To address the mechanisms involved, we asked whether this region of VAMP can interact with cis (outer vesicle leaflet) and/or trans (inner plasma membrane leaflet) lipids. To evaluate cis lipid binding, recombinant VAMP was reconstituted into liposomes and accessibility to site-directed antibodies was probed by surface plasmon resonance. Data indicated that the membrane-proximal domain of VAMP dips into the cis lipid bilayer, sequestering epitopes between the tetanus toxin cleavage site and the membrane anchor. These epitopes were unmasked by VAMP double mutation W89A, W90A, which abolishes lipid interactions. To evaluate trans lipid binding, VAMP was reconstituted in cis liposomes, which were then immobilized on beads. The ability of VAMP to capture protein-free (3)H-labeled trans liposomes was then measured. When cis lipid interactions were eliminated by omitting negatively charged lipids, trans lipid binding to VAMP was revealed. In contrast, when cis and trans liposomes both contained acidic headgroups (i.e., approximating physiological conditions), cis lipid interactions totally occluded trans lipid binding. In these conditions Ca(2+)/calmodulin displaced cis inhibition, transferring the lipid-binding domain of VAMP from the cis to the trans bilayer. Our results suggest that calmodulin acts as a unidirectional Ca(2+)-activated shuttle that docks the juxtamembrane portion of the v-SNARE in the target membrane to prepare fusion.
Collapse
Affiliation(s)
- Luc de Haro
- Institut National de la Santé et de la Recherche Médicale/Université de la Méditerranée, Unité Mixte de Recherche 464, Faculté de Médecine Secteur Nord, Boulevard Pierre Dramard, 13916 Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Yaney GC, Corkey BE. Fatty acid metabolism and insulin secretion in pancreatic beta cells. Diabetologia 2003; 46:1297-312. [PMID: 13680127 DOI: 10.1007/s00125-003-1207-4] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2003] [Revised: 07/14/2003] [Indexed: 01/16/2023]
Abstract
Increases in glucose or fatty acids affect metabolism via changes in long-chain acyl-CoA formation and chronically elevated fatty acids increase total cellular CoA. Understanding the response of pancreatic beta cells to increased amounts of fuel and the role that altered insulin secretion plays in the development and maintenance of obesity and Type 2 diabetes is important. Data indicate that the activated form of fatty acids acts as an effector molecule in stimulus-secretion coupling. Glucose increases cytosolic long-chain acyl-CoA because it increases the "switch" compound malonyl-CoA that blocks mitochondrial beta-oxidation, thus implementing a shift from fatty acid to glucose oxidation. We present arguments in support of the following: (i) A source of fatty acid either exogenous or endogenous (derived by lipolysis of triglyceride) is necessary to support normal insulin secretion; (ii) a rapid increase of fatty acids potentiates glucose-stimulated secretion by increasing fatty acyl-CoA or complex lipid concentrations that act distally by modulating key enzymes such as protein kinase C or the exocytotic machinery; (iii) a chronic increase of fatty acids enhances basal secretion by the same mechanism, but promotes obesity and a diminished response to stimulatory glucose; (iv) agents which raise cAMP act as incretins, at least in part, by stimulating lipolysis via beta-cell hormone-sensitive lipase activation. Furthermore, increased triglyceride stores can give higher rates of lipolysis and thus influence both basal and stimulated insulin secretion. These points highlight the important roles of NEFA, LC-CoA, and their esterified derivatives in affecting insulin secretion in both normal and pathological states.
Collapse
Affiliation(s)
- G C Yaney
- Boston University School of Medicine, Obesity Research Center, 650 Albany Street, Boston, MA 02118, USA
| | | |
Collapse
|
33
|
Aikawa Y, Martin TFJ. ARF6 regulates a plasma membrane pool of phosphatidylinositol(4,5)bisphosphate required for regulated exocytosis. J Cell Biol 2003; 162:647-59. [PMID: 12925709 PMCID: PMC2173784 DOI: 10.1083/jcb.200212142] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ADP-ribosylation factor (ARF) 6 regulates endosomal plasma membrane trafficking in many cell types, but is also suggested to play a role in Ca2+-dependent dense-core vesicle (DCV) exocytosis in neuroendocrine cells. In the present work, expression of the constitutively active GTPase-defective ARF6Q67L mutant in PC12 cells was found to inhibit Ca2+-dependent DCV exocytosis. The inhibition of exocytosis was accompanied by accumulation of ARFQ67L, phosphatidylinositol 4,5-bisphosphate (PIP2), and the phosphatidylinositol 4-phosphate 5-kinase type I (PIP5KI) on endosomal membranes with their corresponding depletion from the plasma membrane. That the depletion of PIP2 and PIP5K from the plasma membrane caused the inhibition of DCV exocytosis was demonstrated directly in permeable cell reconstitution studies in which overexpression or addition of PIP5KIgamma restored Ca2+-dependent exocytosis. The restoration of exocytosis in ARF6Q67L-expressing permeable cells unexpectedly exhibited a Ca2+ dependence, which was attributed to the dephosphorylation and activation of PIP5K. Increased Ca2+ and dephosphorylation stimulated the association of PIP5KIgamma with ARF6. The results reveal a mechanism by which Ca2+ influx promotes increased ARF6-dependent synthesis of PIP2. We conclude that ARF6 plays a role in Ca2+-dependent DCV exocytosis by regulating the activity of PIP5K for the synthesis of an essential plasma membrane pool of PIP2.
Collapse
Affiliation(s)
- Yoshikatsu Aikawa
- Department of Biochemistry, University of Wisconsin, 433 Babcock Drive, Madison, WI 53706, USA
| | | |
Collapse
|
34
|
Baldwin ML, Rostas JAP, Sim ATR. Two modes of exocytosis from synaptosomes are differentially regulated by protein phosphatase types 2A and 2B. J Neurochem 2003; 85:1190-9. [PMID: 12753078 DOI: 10.1046/j.1471-4159.2003.01779.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The inhibitors okadaic acid (OA), fostriecin (FOS) and cyclosporin A (CsA), were used to investigate the roles of protein phosphatases in regulating exocytosis in rat brain synaptosomes by measuring glutamate release and the release of the styryl dye FM 2-10. Depolarization was induced by 30 mM KCl, or 0.3 mM or 1 mM 4-aminopyridine (4AP). OA and FOS produced a similar partial inhibition of KCl- and 0.3 mM 4AP- evoked exocytosis in both assays, but had little effect upon exocytosis evoked by 1 mM 4AP. In contrast, CsA had no effect upon KCl- and 0.3 mM 4AP-evoked exocytosis, but significantly enhanced glutamate release but not FM 2-10 dye release evoked by 1 mM 4AP. None of the phosphatase inhibitors changed calcium signals from FURA-2-loaded synaptosomes either before or after depolarization. Pretreatment with 100 nM phorbol 12-myristate 13-acetate abolished the inhibitory effect of OA on exocytosis induced by 0.3 mM 4AP. Taken together, these results show that exocytosis from synaptosomes has a phosphatase-sensitive and phosphatase-insensitive component, and that there are two modes of phosphatase-sensitive exocytosis that can be elicited by different depolarization conditions. Moreover, these two modes are differentially sensitive to phosphatase 2A and 2B.
Collapse
Affiliation(s)
- Monique L Baldwin
- School of Biomedical Sciences, University of Newcastle and Clinical Neuroscience Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | | | | |
Collapse
|
35
|
Abstract
Regulated exocytosis of secretory granules or dense-core granules has been examined in many well-characterized cell types including neurons, neuroendocrine, endocrine, exocrine, and hemopoietic cells and also in other less well-studied cell types. Secretory granule exocytosis occurs through mechanisms with many aspects in common with synaptic vesicle exocytosis and most likely uses the same basic protein components. Despite the widespread expression and conservation of a core exocytotic machinery, many variations occur in the control of secretory granule exocytosis that are related to the specialized physiological role of particular cell types. In this review we describe the wide range of cell types in which regulated secretory granule exocytosis occurs and assess the evidence for the expression of the conserved fusion machinery in these cells. The signals that trigger and regulate exocytosis are reviewed. Aspects of the control of exocytosis that are specific for secretory granules compared with synaptic vesicles or for particular cell types are described and compared to define the range of accessory control mechanisms that exert their effects on the core exocytotic machinery.
Collapse
Affiliation(s)
- Robert D Burgoyne
- The Physiological Laboratory, University of Liverpool, United Kingdom.
| | | |
Collapse
|
36
|
Rozenvayn N, Flaumenhaft R. Protein kinase C mediates translocation of type II phosphatidylinositol 5-phosphate 4-kinase required for platelet alpha-granule secretion. J Biol Chem 2003; 278:8126-34. [PMID: 12509423 DOI: 10.1074/jbc.m206493200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To better understand the molecular mechanisms of platelet granule secretion, we have evaluated the role of type II phosphatidylinositol (PtdIns) 5-phosphate 4-kinase in agonist-induced platelet alpha-granule secretion. SFLLRN-stimulated alpha-granule secretion from SL-O-permeabilized platelets was inhibited by either antibodies directed at type II PtdIns 5-phosphate 4-kinase or by a kinase-impaired point mutant of type IIbeta PtdIns 5-phosphate 4-kinase. In contrast, recombinant type IIbeta PtdIns 5-phosphate 4-kinase augmented SFLLRN-stimulated alpha-granule secretion from SL-O-permeabilized platelets. SFLLRN-stimulated alpha-granule secretion was inhibited by a protein kinase C-specific inhibitor peptide or bisindolylmaleimide I. Phorbol 12-myristate 13-acetate-stimulated alpha-granule secretion was inhibited by anti-type II PtdIns 5-phosphate 4-kinase antibodies or the kinase-impaired point mutant of type IIbeta PtdIns 5-phosphate 4-kinase and augmented by recombinant type IIbeta PtdIns 5-phosphate 4-kinase. Immunoblot analysis demonstrated that type II PtdIns 5-phosphate 4-kinase remained associated with SL-O-permeabilized platelets when incubated in the presence, but not the absence, of SFLLRN. This SFLLRN-induced translocation of type II PtdIns 5-phosphate 4-kinase was blocked by either the protein kinase C-specific inhibitor peptide or bisindolylmaleimide I. In addition to stimulating alpha-granule secretion, both SFLLRN and PMA enhanced the association of a fluorescein isothiocyanate-labeled peptide derived from the PtdIns (4,5)P(2)-binding domain of gelsolin to permeabilized platelets. Agonist-induced recruitment of the PtdIns (4,5)P(2)-binding domain was inhibited by neomycin, bisindolylmaleimide I, and anti-type II PtdIns 5-phosphate 4-kinase antibody. These results suggest a mechanism whereby protein kinase C-mediated translocation of type II PtdIns 5-phosphate 4-kinase leads to the recruitment of PtdIns (4,5)P(2)-binding proteins.
Collapse
Affiliation(s)
- Nataliya Rozenvayn
- Center for Hemostasis and Thrombosis Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
37
|
Matveeva EA, Whiteheart SW, Slevin JT. Accumulation of 7S SNARE complexes in hippocampal synaptosomes from chronically kindled rats. J Neurochem 2003; 84:621-4. [PMID: 12558983 DOI: 10.1046/j.1471-4159.2003.01589.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Kindling is a model of complex partial epilepsy wherein periodic application of an initially subconvulsive stimulus leads to first limbic and then generalized tonic-clonic seizures. Several laboratories have reported that augmented neurotransmitter release of l-glutamate is associated with the chronically kindled state. Neurotransmitter release requires membrane proteins called SNAREs, which form transmembrane complexes that participate in vesicle docking and are required for membrane fusion. We show here that kindling by entorhinal stimulation is associated with an accumulation of 7S SNARE complexes in the ipsilateral hippocampus. This increase of 7S SNARE complexes appears to begin early in the kindling process, achieves a peak with full kindling, and remains at this level for at least a month following cessation of further kindling stimuli. The increase is focal and permanently limited to the ipsilateral hippocampus despite progression to generalized electrographic and behavioral seizures. It is not seen in animals that receive electroconvulsive seizures, suggesting it is related to the kindling process itself. The duration and focality of increased 7S SNARE complexes with entorhinal kindling suggest that this is an altered molecular process associated with epileptogenesis.
Collapse
Affiliation(s)
- E A Matveeva
- Department of Molecular and Cellular Biochemistry, University of Kentucky Medical Center, Lexington, Kentucky, USA
| | | | | |
Collapse
|
38
|
Takahashi M, Itakura M, Kataoka M. New Aspects of Neurotransmitter Releasee and Exocytosis: Regulation of Neurotransmitter Release by Phosphorylation. J Pharmacol Sci 2003; 93:41-5. [PMID: 14501150 DOI: 10.1254/jphs.93.41] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Synaptic transmission is conducted by neurotransmitters released from nerve terminals. Neurotransmitter release is regulated both positively and negatively by multiple mechanisms, and its regulation is believed to be one of the important mechanisms of synaptic plasticity underlying learning and memory. Various protein kinases play important roles in the regulation, and candidates for protein substrates essential for the regulation have been identified.
Collapse
Affiliation(s)
- Masami Takahashi
- Department of Biochemistry, Kitasato University School of Medicine, Kanagawa, Japan.
| | | | | |
Collapse
|
39
|
Barg S. Mechanisms of exocytosis in insulin-secreting B-cells and glucagon-secreting A-cells. PHARMACOLOGY & TOXICOLOGY 2003; 92:3-13. [PMID: 12710591 DOI: 10.1034/j.1600-0773.2003.920102.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In pancreatic B- and A-cells, metabolic stimuli regulate biochemical and electrical processes that culminate in Ca2+-influx and release of insulin or glucagon, respectively. Like in other (neuro)endocrine cells, Ca2+-influx triggers the rapid exocytosis of hormone-containing secretory granules. Only a small fraction of granules (<1% in insulin-secreting B-cells) can be released immediately, while the remainder requires translocation to the plasma membrane and further "priming" for release by several ATP- and Ca2+-dependent reactions. Such functional organization may account for systemic features such as the biphasic time course of glucose-stimulated insulin secretion. Since this release pattern is altered in type-2 diabetes mellitus, it is conceivable that disturbances in the exocytotic machinery underlie the disease. Here I will review recent data from our laboratory relevant for the understanding of these processes in insulin-secreting B-cells and glucagon-secreting A-cells and for the identification of novel targets for antidiabetic drug action. Two aspects are discussed in detail: 1) The importance of a tight interaction between L-type Ca2+-channels and the exocytotic machinery for efficient secretion; and 2) the role of intragranular acidification for the priming of secretory granules and its regulation by a granular 65-kDa sulfonylurea-binding protein.
Collapse
Affiliation(s)
- Sebastian Barg
- Department of Physiological Sciences, Molecular and Cellular Physiology, Lund University, Sölvegatan 19, BMC F11, S-221 84 Lund, Sweden.
| |
Collapse
|
40
|
Abstract
Activation of protein kinase C (PKC) increases vesicular secretion in many cell types. We determined the calcium dependence of secretion and the size of the readily releasable pool of secretory granules in pituitary gonadotropes by photorelease of caged-calcium. The calcium affinity for exocytosis was roughly doubled by activation of PKC by a phorbol ester, whereas the size of the readily releasable pool was not greatly increased. The effect was due to activation of PKC, because it was blocked by a PKC inhibitor and was not mimicked by an inactive phorbol ester analogue. A similar increase in calcium sensitivity was induced by preincubation with gonadotropin-releasing hormone, the physiological releasing hormone. These findings provide direct evidence for physiological regulation of secretion by enhancement of Ca2+-sensing steps. Because exocytosis depends on the third- to fourth-power of intracellular free Ca2+ concentration, this mechanism ensures a powerful up-regulation of hormone release and may explain how PKC can stimulate exocytosis without an increase of Ca2+ above the resting level.
Collapse
Affiliation(s)
- Hongliang Zhu
- Institute of Biophysics and Biochemistry, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | | | | |
Collapse
|
41
|
Westerink RHS, Klompmakers AA, Westenberg HGM, Vijverberg HPM. Signaling pathways involved in Ca2+- and Pb2+-induced vesicular catecholamine release from rat PC12 cells. Brain Res 2002; 957:25-36. [PMID: 12443976 DOI: 10.1016/s0006-8993(02)03580-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Since Pb(2+) substitutes for Ca(2+) in essential steps leading to exocytosis, we have investigated whether Ca(2+) and Pb(2+) induce exocytosis through similar pathways. Vesicular catecholamine release was measured from dexamethasone-differentiated PC12 cells using carbon fiber microelectrode amperometry. Effects of drugs known to modulate PKC (PMA, staurosporine), calcineurin (cyclosporin A), calmodulin (W7), and CaM kinase II (KN-62) activity were investigated in intact and in ionomycin-permeabilized PC12 cells. Activation of PKC and inhibition of calmodulin decrease the frequency of exocytotic events evoked by high K(+) stimulation in intact cells. In addition, inhibition of calmodulin enhances the frequency of basal exocytosis from intact cells. Activation of PKC and inhibition of calcineurin enhance the frequency of basal exocytosis in intact as well as in ionomycin-permeabilized cells. Inhibition of PKC and of CaM kinase II cause no significant effects. None of the treatments has a significant effect on vesicle contents. The combined results indicate that PKC and calcineurin enhance and inhibit exocytosis through direct effects on the exocytotic machinery, whereas calmodulin and CaM kinase II exert indirect effects only. Conversely, Pb(2+)-evoked exocytosis in permeabilized cells is strongly reduced by inhibition of CaM kinase II, but is not sensitive to modulation of PKC and calcineurin activity. Inhibition of calmodulin only reduces the delay to onset of Pb(2+)-evoked exocytosis. Synaptotagmin I- and II-deficient PC12-F7 cells exhibit vesicular catecholamine release following depolarization or superfusion with Pb(2+). However, the frequency of exocytosis and the contents of vesicles released are strongly reduced as compared to PC12 cells. It is concluded that Ca(2+)-evoked exocytosis is modulated mainly by PKC and calcineurin, whereas Pb(2+)-evoked exocytosis is mainly modulated by CaM kinase II.
Collapse
Affiliation(s)
- Remco H S Westerink
- Institute for Risk Assessment Sciences, Utrecht University, PO Box 80176, 3508 TD, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Scales SJ, Hesser BA, Masuda ES, Scheller RH. Amisyn, a novel syntaxin-binding protein that may regulate SNARE complex assembly. J Biol Chem 2002; 277:28271-9. [PMID: 12145319 DOI: 10.1074/jbc.m204929200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of SNARE complex assembly likely plays an important role in governing the specificity as well as the timing of membrane fusion. Here we identify a novel brain-enriched protein, amisyn, with a tomosyn- and VAMP-like coiled-coil-forming domain that binds specifically to syntaxin 1a and syntaxin 4 both in vitro and in vivo, as assessed by co-immunoprecipitation from rat brain. Amisyn is mostly cytosolic, but a fraction co-sediments with membranes. The amisyn coil domain can form SNARE complexes of greater thermostability than can VAMP2 with syntaxin 1a and SNAP-25 in vitro, but it lacks a transmembrane anchor and so cannot act as a v-SNARE in this complex. The amisyn coil domain prevents the SNAP-25 C-terminally mediated rescue of botulinum neurotoxin E inhibition of norepinephrine exocytosis in permeabilized PC12 cells to a greater extent than it prevents the regular exocytosis of these vesicles. We propose that amisyn forms nonfusogenic complexes with syntaxin 1a and SNAP-25, holding them in a conformation ready for VAMP2 to replace it to mediate the membrane fusion event, thereby contributing to the regulation of SNARE complex formation.
Collapse
Affiliation(s)
- Suzie J Scales
- Howard Hughes Medical Institute, Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305-5345, USA
| | | | | | | |
Collapse
|
43
|
Quetglas S, Iborra C, Sasakawa N, De Haro L, Kumakura K, Sato K, Leveque C, Seagar M. Calmodulin and lipid binding to synaptobrevin regulates calcium-dependent exocytosis. EMBO J 2002; 21:3970-9. [PMID: 12145198 PMCID: PMC126150 DOI: 10.1093/emboj/cdf404] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2002] [Revised: 06/06/2002] [Accepted: 06/11/2002] [Indexed: 11/13/2022] Open
Abstract
Neurotransmitter release involves the assembly of a heterotrimeric SNARE complex composed of the vesicle protein synaptobrevin (VAMP 2) and two plasma membrane partners, syntaxin 1 and SNAP-25. Calcium influx is thought to control this process via Ca(2+)-binding proteins that associate with components of the SNARE complex. Ca(2+)/calmodulin or phospholipids bind in a mutually exclusive fashion to a C-terminal domain of VAMP (VAMP(77-90)), and residues involved were identified by plasmon resonance spectroscopy. Microinjection of wild-type VAMP(77-90), but not mutant peptides, inhibited catecholamine release from chromaffin cells monitored by carbon fibre amperometry. Pre-incubation of PC12 pheochromocytoma cells with the irreversible calmodulin antagonist ophiobolin A inhibited Ca(2+)-dependent human growth hormone release in a permeabilized cell assay. Treatment of permeabilized cells with tetanus toxin light chain (TeNT) also suppressed secretion. In the presence of TeNT, exocytosis was restored by transfection of TeNT-resistant (Q(76)V, F(77)W) VAMP, but additional targeted mutations in VAMP(77-90) abolished its ability to rescue release. The calmodulin- and phospholipid-binding domain of VAMP 2 is thus required for Ca(2+)-dependent exocytosis, possibly to regulate SNARE complex assembly.
Collapse
Affiliation(s)
- Stephanie Quetglas
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Cecile Iborra
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Nobuyuki Sasakawa
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Luc De Haro
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Konosuke Kumakura
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Kazuki Sato
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Christian Leveque
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| | - Michael Seagar
- Institut National de la Santé et de la Recherche Médicale Unité 464, Université de la Méditerranée and Unité de Méthodologie des Interactions Moléculaires, Institut Jean-Roche, Faculté de Médecine Secteur Nord, Bd. Pierre Dramard, 13916 Marseille Cedex 20, France, Laboratory of Neurochemistry and Neuropharmacology, Life Science Institute, Sophia University, Chiyoda-ku, Tokyo 102-8554 and Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan Corresponding author e-mail:
| |
Collapse
|
44
|
Glasova H, Berghaus TM, Kullak-Ublick GA, Paumgartner G, Beuers U. Tauroursodeoxycholic acid mobilizes alpha-PKC after uptake in human HepG2 hepatoma cells. Eur J Clin Invest 2002; 32:437-42. [PMID: 12059989 DOI: 10.1046/j.1365-2362.2002.01002.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Tauroursodeoxycholic acid (TUDCA) may exert anticholestatic effects via Ca(++)- and alpha-protein kinase C (alpha-PKC)-dependent apical vesicular insertion of canalicular transporters in cholestatic hepatocytes (Hepatology 2001; 33: 1206-16). Tauroursodeoxycholic acid is mainly taken up into liver cells by Na(+)-taurocholate cotransporting polypeptide (Ntcp). Tauroursodeoxycholic acid selectively translocates alpha-PKC, a key mediator of regulated exocytosis, to hepatocellular membranes. It is unclear whether TUDCA exerts its effects on alpha-PKC after carrier-mediated uptake into liver cells or by interaction with extracellular/membraneous structures. MATERIALS AND METHODS Human hepatoblastoma HepG2 cells lacking Ntcp were stably transfected with pcDNA3.1/Ntcp or sham-transfected with pcDNA3.1 [+]. Distribution of alpha-PKC was studied using a Western blotting technique. Uptake of [(3)H]taurocholic acid (TCA) was determined radiochemically. RESULTS [(3)H]taurocholic acid uptake was approximately 180-fold higher in Ntcp-transfected than in sham-transfected cells. Phorbol 12-myristate 13-acetate (1 micromol L(-1); positive control) increased membrane binding of alpha-PKC by 34% in Ntcp-transfected and by 37% in sham-transfected cells. Tauroursodeoxycholic acid (10 micromol L(-1)) increased membrane-associated alpha-PKC by 19% in Ntcp-transfected, but not in sham-transfected cells (-13%). Taurocholic acid (10 micromol L(-1)) did not affect the distribution of alpha-PKC. CONCLUSION Carrier-mediated uptake is a prerequisite for TUDCA-induced translocation of alpha-PKC to hepatocellular membranes.
Collapse
Affiliation(s)
- Helena Glasova
- Department of Medicine II, Klinikum Grosshadern, University of Munich, Germany
| | | | | | | | | |
Collapse
|
45
|
Regulation of exocytosis through Ca2+/ATP-dependent binding of autophosphorylated Ca2+/calmodulin-activated protein kinase II to syntaxin 1A. J Neurosci 2002. [PMID: 11978810 DOI: 10.1523/jneurosci.22-09-03342.2002] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Syntaxin 1A/HPC-1 is a key component of the exocytotic molecular machinery, namely, the soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor mechanism. Although >10 syntaxin-binding proteins have been identified, they cannot completely explain the regulation of exocytosis. Thus, novel proteins may interact with syntaxin. Because exocytosis requires both Ca2+ and ATP, we searched for Ca2+/ATP-dependent syntaxin-binding proteins from the rat brain and discovered Ca2+/calmodulin-activated protein kinase II (CaMKII)-alpha. At Ca2+ concentrations of >10(-6) m, only autophosphorylated CaMKII bound to syntaxin. Bound CaMKII was released from syntaxin by EGTA or by phosphatase, indicating that the binding is reversible. CaMKII bound to the linker domain of syntaxin, unlike any other known syntaxin-binding proteins. CaMKII-syntaxin complexes were also detected in synaptosomes by immunoprecipitation, and when reconstituted in vitro, they recruited larger amounts of synaptotagmin and SNAP-25 than syntaxin alone. The microinjected CaMKII-binding domain of syntaxin specifically affected exocytosis in chromaffin cells and in neurons. These results indicate that the Ca2+/ATP-dependent binding of CaMKII to syntaxin is an important process in the regulation of exocytosis.
Collapse
|
46
|
Wang J, Mayernik L, Armant DR. Integrin signaling regulates blastocyst adhesion to fibronectin at implantation: intracellular calcium transients and vesicle trafficking in primary trophoblast cells. Dev Biol 2002; 245:270-9. [PMID: 11977980 DOI: 10.1006/dbio.2002.0644] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Accumulating evidence indicates that the endometrial extracellular matrix (ECM) modulates trophoblast adhesion during mouse blastocyst implantation. In previous studies of adhesion-competent mouse blastocysts, we have demonstrated that integrin-mediated fibronectin (FN)-binding activity on the apical surface of trophoblast cells is initially low, but becomes strengthened after embryos are exposed to FN. In the present study, we have examined whether the ligand-induced upregulation of trophoblast adhesion to FN is mediated by integrin signaling. The strengthening of adhesion to FN required integrin ligation, which rapidly elevated cytoplasmic-free Ca(2+). Chelation of intracellular Ca(2+) using BAPTA-AM, or inhibition of the Ca(2+)-dependent proteins, protein kinase C or calmodulin, significantly attenuated the effect of FN on binding activity. Furthermore, direct elevation of cytoplasmic Ca(2+) levels with ionomycin upregulated FN-binding activity, demonstrating that Ca(2+) signaling is required and sufficient for strong adhesion to FN. Ca(2+) signaling may induce protein trafficking, a known requirement for ligand-induced upregulation of FN-binding activity. Indeed, intracellular vesicles accumulated in adhesion-competent blastocysts, but were absent after exposure to either FN or ionomycin. These findings suggest that, during implantation, contact between peri-implantation blastocysts and FN elevates intracellular Ca(2+), which strengthens trophoblast adhesion to ECM through protein redistribution.
Collapse
Affiliation(s)
- Jun Wang
- C. S. Mott Center for Human Growth and Development, Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
47
|
Zhang J, Suneja SK, Potashner SJ. Protein kinase C regulates [3H]D-aspartate release in auditory brain stem nuclei. Exp Neurol 2002; 175:245-56. [PMID: 12009776 DOI: 10.1006/exnr.2002.7890] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We previously found that unilateral cochlear ablation altered transmitter release from glutamatergic synaptic endings in several brain stem auditory nuclei. To determine if this release activity could be regulated by protein kinase C (PKC), which has been associated with regulation of transmitter release, the electrically evoked release of [3H]d-aspartate ([3H]d-Asp) was quantified in vitro as an index of exocytosis from glutamatergic presynaptic endings in the major subdivisions of the cochlear nucleus (CN) and in the main nuclei of the superior olivary complex (SOC). Treating dissected tissues with a PKC activator, such as phorbol 12,13-diacetate (PDA) or phorbol 12,13-dibutyrate (PDBu) (3 microM), elevated the evoked release of [3H]d-Asp by 1.5- to 3.3-fold. The PKC inhibitor Ro31-8220 (50 nM) did not alter the evoked release but blocked the stimulatory effects of PDA and PDBu. These findings suggested that PKC could positively regulate transmitter release from glutamatergic presynaptic endings in brain stem auditory pathways. Seven days after unilateral cochlear ablation, when cochlear nerve endings had degenerated in the ipsilateral CN, PDBu elevated the evoked release bilaterally in each CN subdivision and SOC nucleus, implying that PKC could regulate glutamatergic release in the noncochlear pathways remaining in the ipsilateral CN and in the other pathways after unilateral hearing loss. After 145 postlesion days, Ro31-8220 blocked endogenous elevations in the evoked release in the ipsilateral SOC but did not alter the elevated or upregulated release in the other tissues. This suggested that the elevations of glutamatergic release activity in the ipsilateral SOC that appeared after unilateral cochlear ablation depended on endogenous activation of PKC.
Collapse
Affiliation(s)
- J Zhang
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
48
|
Fukuda M, Kowalchyk JA, Zhang X, Martin TFJ, Mikoshiba K. Synaptotagmin IX regulates Ca2+-dependent secretion in PC12 cells. J Biol Chem 2002; 277:4601-4. [PMID: 11751925 DOI: 10.1074/jbc.c100588200] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Synaptotagmin (Syt) I-deficient phaeochromocytoma (PC12) cell lines show normal Ca(2+)-dependent norepinephrine (NE) release (Shoji-Kasai, Y., Yoshida, A., Sato, K., Hoshino, T., Ogura, A., Kondo, S., Fujimoto, Y., Kuwahara, R., Kato, R., and Takahashi, M. (1992) Science 256, 1821-1823). To identify an alternative Ca(2+) sensor, we searched for other Syt isoforms in Syt I-deficient PC12 cells and identified Syt IX, an isoform closely related to Syt I, as an abundantly expressed dense-core vesicle protein. Here we show that Syt IX is required for the Ca(2+)-dependent release of NE from PC12 cells. Antibodies directed against the C2A domain of either Syt IX or Syt I inhibited Ca(2+)-dependent NE release in permeable PC12 cells indicating that both Syt proteins function in dense-core vesicle exocytosis. Our results support the idea that Syt family proteins that co-reside on secretory vesicles may function cooperatively and redundantly as potential Ca(2+) sensors for exocytosis.
Collapse
Affiliation(s)
- Mitsunori Fukuda
- Laboratory for Developmental Neurobiology, Brain Science Institute, RIKEN (The Institute of Physical and Chemical Research), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| | | | | | | | | |
Collapse
|
49
|
Sugita S, Shin OH, Han W, Lao Y, Südhof TC. Synaptotagmins form a hierarchy of exocytotic Ca(2+) sensors with distinct Ca(2+) affinities. EMBO J 2002; 21:270-80. [PMID: 11823420 PMCID: PMC125835 DOI: 10.1093/emboj/21.3.270] [Citation(s) in RCA: 219] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Synaptotagmins constitute a large family of membrane proteins implicated in Ca(2+)-triggered exocytosis. Structurally similar synaptotagmins are differentially localized either to secretory vesicles or to plasma membranes, suggesting distinct functions. Using measurements of the Ca(2+) affinities of synaptotagmin C2-domains in a complex with phospholipids, we now show that different synaptotagmins exhibit distinct Ca(2+) affinities, with plasma membrane synaptotagmins binding Ca(2+) with a 5- to 10-fold higher affinity than vesicular synaptotagmins. To test whether these differences in Ca(2+) affinities are functionally important, we examined the effects of synaptotagmin C2-domains on Ca(2+)-triggered exocytosis in permeabilized PC12 cells. A precise correlation was observed between the apparent Ca(2+) affinities of synaptotagmins in the presence of phospholipids and their action in PC12 cell exocytosis. This was extended to PC12 cell exocytosis triggered by Sr(2+), which was also selectively affected by high-affinity C2-domains of synaptotagmins. Together, our results suggest that Ca(2+) triggering of exocytosis involves tandem Ca(2+) sensors provided by distinct plasma membrane and vesicular synaptotagmins. According to this hypothesis, plasma membrane synaptotagmins represent high-affinity Ca(2+) sensors involved in slow Ca(2+)-dependent exocytosis, whereas vesicular synaptotagmins function as low-affinity Ca(2+) sensors specialized for fast Ca(2+)-dependent exocytosis.
Collapse
Affiliation(s)
- Shuzo Sugita
- The Center for Basic Neuroscience, Department of Molecular Genetics, and Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
Present address: Division of Cellular and Molecular Biology, Toronto Western Research Institute, Ontario, Canada Corresponding author e-mail:
| | | | | | | | - Thomas C. Südhof
- The Center for Basic Neuroscience, Department of Molecular Genetics, and Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
Present address: Division of Cellular and Molecular Biology, Toronto Western Research Institute, Ontario, Canada Corresponding author e-mail:
| |
Collapse
|
50
|
Abstract
The fusion of biological membranes is the terminal step of all vesicular trafficking reactions in eukaryotic cells. Therefore, this fusion is fundamental for the transfer of proteins and lipids between different compartments, for exocytosis and for the structural integrity of organelles. In the past decade, many parts of the molecular machinery involved in fusion have been uncovered. Although the mechanisms responsible for mutual recognition and binding of membranes inside eukaryotes are becoming reasonably well known, there is considerable uncertainty as to what causes the actual merging of the lipid bilayer. Two classes of mechanisms have been proposed. Proximity models postulate that very close apposition of membranes suffices to induce fusion. By contrast, pore models propose that continuous proteinaceous pores between apposed membranes could be the basis for fusion.
Collapse
Affiliation(s)
- A Mayer
- Friedrich-Miescher-Laboratorium der Max-Planck-Gesellschaft, Spemannstr. 37-39, 72076, Tübingen, Germany.
| |
Collapse
|