51
|
Larisch N, Schulze C, Galione A, Dietrich P. An N-Terminal Dileucine Motif Directs Two-Pore Channels to the Tonoplast of Plant Cells. Traffic 2012; 13:1012-22. [DOI: 10.1111/j.1600-0854.2012.01366.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 04/06/2012] [Accepted: 04/10/2012] [Indexed: 12/23/2022]
Affiliation(s)
- Nina Larisch
- Department Biology; Friedrich-Alexander-Universität Erlangen-Nürnberg, Molecular Plant Physiology and Erlangen Center of Plant Science; Staudtstrasse 5; 91058; Erlangen; Germany
| | - Christina Schulze
- Department Biology; Friedrich-Alexander-Universität Erlangen-Nürnberg, Molecular Plant Physiology and Erlangen Center of Plant Science; Staudtstrasse 5; 91058; Erlangen; Germany
| | - Antony Galione
- Department of Pharmacology; Oxford University; Oxford; OX1 3QT; UK
| | - Petra Dietrich
- Department Biology; Friedrich-Alexander-Universität Erlangen-Nürnberg, Molecular Plant Physiology and Erlangen Center of Plant Science; Staudtstrasse 5; 91058; Erlangen; Germany
| |
Collapse
|
52
|
Pietrancosta N, Anne C, Prescher H, Ruivo R, Sagné C, Debacker C, Bertrand HO, Brossmer R, Acher F, Gasnier B. Successful prediction of substrate-binding pocket in SLC17 transporter sialin. J Biol Chem 2012; 287:11489-97. [PMID: 22334707 DOI: 10.1074/jbc.m111.313056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Secondary active transporters from the SLC17 protein family are required for excitatory and purinergic synaptic transmission, sialic acid metabolism, and renal function, and several members are associated with inherited neurological or metabolic diseases. However, molecular tools to investigate their function or correct their genetic defects are limited or absent. Using structure-activity, homology modeling, molecular docking, and mutagenesis studies, we have located the substrate-binding site of sialin (SLC17A5), a lysosomal sialic acid exporter also recently implicated in exocytotic release of aspartate. Human sialin is defective in two inherited sialic acid storage diseases and is responsible for metabolic incorporation of the dietary nonhuman sialic acid N-glycolylneuraminic acid. We built cytosol-open and lumen-open three-dimensional models of sialin based on weak, but significant, sequence similarity with the glycerol-3-phosphate and fucose permeases from Escherichia coli, respectively. Molecular docking of 31 synthetic sialic acid analogues to both models was consistent with inhibition studies. Narrowing the sialic acid-binding site in the cytosol-open state by two phenylalanine to tyrosine mutations abrogated recognition of the most active analogue without impairing neuraminic acid transport. Moreover, a pilot virtual high-throughput screening of the cytosol-open model could identify a pseudopeptide competitive inhibitor showing >100-fold higher affinity than the natural substrate. This validated model of human sialin and sialin-guided models of other SLC17 transporters should pave the way for the identification of inhibitors, glycoengineering tools, pharmacological chaperones, and fluorescent false neurotransmitters targeted to these proteins.
Collapse
Affiliation(s)
- Nicolas Pietrancosta
- Centre National de la Recherche Scientifique, UMR 8601, Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, F-75006 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Blakely RD, Edwards RH. Vesicular and plasma membrane transporters for neurotransmitters. Cold Spring Harb Perspect Biol 2012; 4:a005595. [PMID: 22199021 PMCID: PMC3281572 DOI: 10.1101/cshperspect.a005595] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The regulated exocytosis that mediates chemical signaling at synapses requires mechanisms to coordinate the immediate response to stimulation with the recycling needed to sustain release. Two general classes of transporter contribute to release, one located on synaptic vesicles that loads them with transmitter, and a second at the plasma membrane that both terminates signaling and serves to recycle transmitter for subsequent rounds of release. Originally identified as the target of psychoactive drugs, these transport systems have important roles in transmitter release, but we are only beginning to understand their contribution to synaptic transmission, plasticity, behavior, and disease. Recent work has started to provide a structural basis for their activity, to characterize their trafficking and potential for regulation. The results indicate that far from the passive target of psychoactive drugs, neurotransmitter transporters undergo regulation that contributes to synaptic plasticity.
Collapse
Affiliation(s)
- Randy D Blakely
- Department of Pharmacology and Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548, USA
| | | |
Collapse
|
54
|
Kim E, Shino S, Yoon J, Leung HT. In search of proteins that are important for synaptic functions in Drosophila visual system. J Neurogenet 2012; 26:151-7. [PMID: 22283835 DOI: 10.3109/01677063.2011.648290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This is the second of two reviews that include some of the studies we, members of the Pak laboratory and collaborators, did from 2000 to 2010 on the mutants that affect synaptic transmission in the Drosophila visual system. Of the five mutants we discuss, two turned out to also play roles in the larval neuromuscular junction. This review complements the one on phototransduction to give a fairly complete account of what we focused on during the 10-year period, although we also did some studies on photoreceptor degeneration in the early part of the decade. Besides showing the power of using a genetic approach to the study of synaptic transmission, the review contains some unexpected results that illustrate the serendipitous nature of research.
Collapse
Affiliation(s)
- Eunju Kim
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | | | |
Collapse
|
55
|
Molecular pathogenesis of sialic acid storage diseases: insight gained from four missense mutations and a putative polymorphism of human sialin. Biol Cell 2012; 100:551-9. [DOI: 10.1042/bc20070166] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
56
|
Abstract
Synapse formation is driven by precisely orchestrated intercellular communication between the presynaptic and the postsynaptic cell, involving a cascade of anterograde and retrograde signals. At the neuromuscular junction (NMJ), both neuron and muscle secrete signals into the heavily glycosylated synaptic cleft matrix sandwiched between the two synapsing cells. These signals must necessarily traverse and interact with the extracellular environment, for the ligand-receptor interactions mediating communication to occur. This complex synaptomatrix, rich in glycoproteins and proteoglycans, comprises heterogeneous, compartmentalized domains where specialized glycans modulate trans-synaptic signaling during synaptogenesis and subsequent synapse modulation. The general importance of glycans during development, homeostasis and disease is well established, but this important molecular class has received less study in the nervous system. Glycan modifications are now understood to play functional and modulatory roles as ligands and co-receptors in numerous tissues; however, roles at the synapse are relatively unexplored. We highlight here properties of synaptomatrix glycans and glycan-interacting proteins with key roles in synaptogenesis, with a particular focus on recent advances made in the Drosophila NMJ genetic system. We discuss open questions and interesting new findings driving this investigation of complex, diverse, and largely understudied glycan mechanisms at the synapse.
Collapse
Affiliation(s)
- Neil Dani
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232 USA
| | - Kendal Broadie
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232 USA
| |
Collapse
|
57
|
Broadie K, Baumgartner S, Prokop A. Extracellular matrix and its receptors in Drosophila neural development. Dev Neurobiol 2011; 71:1102-30. [PMID: 21688401 PMCID: PMC3192297 DOI: 10.1002/dneu.20935] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Extracellular matrix (ECM) and matrix receptors are intimately involved in most biological processes. The ECM plays fundamental developmental and physiological roles in health and disease, including processes underlying the development, maintenance, and regeneration of the nervous system. To understand the principles of ECM-mediated functions in the nervous system, genetic model organisms like Drosophila provide simple, malleable, and powerful experimental platforms. This article provides an overview of ECM proteins and receptors in Drosophila. It then focuses on their roles during three progressive phases of neural development: (1) neural progenitor proliferation, (2) axonal growth and pathfinding, and (3) synapse formation and function. Each section highlights known ECM and ECM-receptor components and recent studies done in mutant conditions to reveal their in vivo functions, all illustrating the enormous opportunities provided when merging work on the nervous system with systematic research into ECM-related gene functions.
Collapse
Affiliation(s)
- Kendal Broadie
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232 USA
| | - Stefan Baumgartner
- Department of Experimental Medical Sciences, Lund University, BMC B12, 22184 Lund, Sweden
| | - Andreas Prokop
- Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
58
|
Shim MS, Kim JY, Lee KH, Jung HK, Carlson BA, Xu XM, Hatfield DL, Lee BJ. l(2)01810 is a novel type of glutamate transporter that is responsible for megamitochondrial formation. Biochem J 2011; 439:277-86. [PMID: 21728998 PMCID: PMC3460806 DOI: 10.1042/bj20110582] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
l(2)01810 causes glutamine-dependent megamitochondrial formation when it is overexpressed in Drosophila cells. In the present study, we elucidated the function of l(2)01810 during megamitochondrial formation. The overexpression of l(2)01810 and the inhibition of glutamine synthesis showed that l(2)01810 is involved in the accumulation of glutamate. l(2)01810 was predicted to contain transmembrane domains and was found to be localized to the plasma membrane. By using (14)C-labelled glutamate, l(2)01810 was confirmed to uptake glutamate into Drosophila cells with high affinity (K(m)=69.4 μM). Also, l(2)01810 uptakes glutamate in a Na(+)-independent manner. Interestingly, however, this uptake was not inhibited by cystine, which is a competitive inhibitor of Na(+)-independent glutamate transporters, but by aspartate. A signal peptide consisting of 34 amino acid residues targeting to endoplasmic reticulum was predicted at the N-terminus of l(2)01810 and this signal peptide is essential for the protein's localization to the plasma membrane. In addition, l(2)01810 has a conserved functional domain of a vesicular-type glutamate transporter, and Arg(146) in this domain was found to play a key role in glutamate transport and megamitochondrial formation. These results indicate that l(2)01810 is a novel type of glutamate transporter and that glutamate uptake is a rate-limiting step for megamitochondrial formation.
Collapse
Affiliation(s)
- Myoung Sup Shim
- Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Jin Young Kim
- Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Kwang Hee Lee
- Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Hee Kyoung Jung
- Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Bradley A. Carlson
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, U.S.A
| | - Xue-Ming Xu
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, U.S.A
| | - Dolph L. Hatfield
- Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, U.S.A
| | - Byeong Jae Lee
- Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|
59
|
Savalas LRT, Gasnier B, Damme M, Lübke T, Wrocklage C, Debacker C, Jézégou A, Reinheckel T, Hasilik A, Saftig P, Schröder B. Disrupted in renal carcinoma 2 (DIRC2), a novel transporter of the lysosomal membrane, is proteolytically processed by cathepsin L. Biochem J 2011; 439:113-28. [PMID: 21692750 DOI: 10.1042/bj20110166] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
DIRC2 (Disrupted in renal carcinoma 2) has been initially identified as a breakpoint-spanning gene in a chromosomal translocation putatively associated with the development of renal cancer. The DIRC2 protein belongs to the MFS (major facilitator superfamily) and has been previously detected by organellar proteomics as a tentative constituent of lysosomal membranes. In the present study, lysosomal residence of overexpressed as well as endogenous DIRC2 was shown by several approaches. DIRC2 is proteolytically processed into a N-glycosylated N-terminal and a non-glycosylated C-terminal fragment respectively. Proteolytic cleavage occurs in lysosomal compartments and critically depends on the activity of cathepsin L which was found to be indispensable for this process in murine embryonic fibroblasts. The cleavage site within DIRC2 was mapped between amino acid residues 214 and 261 using internal epitope tags, and is presumably located within the tentative fifth intralysosomal loop, assuming the typical MFS topology. Lysosomal targeting of DIRC2 was demonstrated to be mediated by a N-terminal dileucine motif. By disrupting this motif, DIRC2 can be redirected to the plasma membrane. Finally, in a whole-cell electrophysiological assay based on heterologous expression of the targeting mutant at the plasma membrane of Xenopus oocytes, the application of a complex metabolic mixture evokes an outward current associated with the surface expression of full-length DIRC2. Taken together, these data strongly support the idea that DIRC2 is an electrogenic lysosomal metabolite transporter which is subjected to and presumably modulated by limited proteolytic processing.
Collapse
|
60
|
Miyaji T, Omote H, Moriyama Y. Functional characterization of vesicular excitatory amino acid transport by human sialin. J Neurochem 2011; 119:1-5. [PMID: 21781115 DOI: 10.1111/j.1471-4159.2011.07388.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sialin, the protein coded by SLC17A5, is responsible for membrane potential (Δψ)-driven aspartate and glutamate transport into synaptic vesicles in addition to H+/sialic acid co-transport in lysosomes. Rodent sialin mutants harboring the mutations associated with Salla disease in humans did not transport aspartate and glutamate whereas H+/sialic acid co-transport activity was about one-third of the wild-type protein. In this study, we investigate the effects of various mutations on the transport activities of human sialin. Proteoliposomes containing purified heterologously expressed human sialin exhibited both Δψ-driven aspartate and glutamate transport activity and H+/sialic acid co-transport activity. Aspartate and glutamate transport was not detected in the R39C and K136E mutant forms of SLC17A5 protein associated with Salla disease, whereas H+/sialic acid co-transport activity corresponded to 30-50% of the recombinant wild-type protein. In contrast, SLC17A5 protein harboring the mutations associated with infantile sialic acid storage disease, H183R and Δ268SSLRN272 still showed normal levels of Δψ-driven aspartate and glutamate transport even though H+/sialic acid co-transport activity was absent. Human sialin carrying the G328E mutation that causes both phenotypes, and P334R and G378V mutations that cause infantile sialic acid storage disease showed no transport activity. These results support the idea that people suffering from Salla disease have been defective in aspartergic and glutamatergic neurotransmissions.
Collapse
Affiliation(s)
- Takaaki Miyaji
- Advanced Science Research Center, Okayama University, Okayama, Japan.
| | | | | |
Collapse
|
61
|
Miyaji T, Omote H, Moriyama Y. A vesicular transporter that mediates aspartate and glutamate neurotransmission. Biol Pharm Bull 2011; 33:1783-5. [PMID: 21048299 DOI: 10.1248/bpb.33.1783] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aspartate, an excitatory amino acid, is known to be stored in synaptic vesicles and exocytosed from some neurons to perform aspartergic neurotransmission. Through in vitro reconstitution, we found that sialin, a lysosomal sialic acid exporter, is responsible for the vesicular storage of aspartate in hippocampal neurons and pinealocytes. Mutations found in Salla disease cause decreased aspartate transport activity without affecting sialic acid transport. Thus, sialin is a multifunctional transporter. It is possible that people with Salla disease lose the ability of aspartergic neurotransmission, and this could explain why Salla disease involves severe neurological defects.
Collapse
Affiliation(s)
- Takaaki Miyaji
- Department of Genomics and Proteomics, Advanced Science Research Center, Okayama University, Okayama 700–8530, Japan
| | | | | |
Collapse
|
62
|
He M, Lin F, Qin L, Zhou J, Yang G, Yang X, Wang S. Postnatal expression of sialin in the mouse submandibular gland. Arch Oral Biol 2011; 56:1333-8. [PMID: 21620373 DOI: 10.1016/j.archoralbio.2011.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 03/31/2011] [Accepted: 04/29/2011] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Sialin has been identified as a sialic acid and aspartate/glutamate transporter. Both cytoplasmic localization and the plasma membrane labelling pattern suggested that sialin may possess multiple transport functions in different cell types. In mouse embryos, sialin expression was primarily detected in the central nervous system. However, sialin shows widespread and high-level expression in adult tissues. Despite its ubiquitous expression and important functions, the postnatal expression profile and subcellular localization of sialin in the salivary gland remains elusive. The aim of the present study was to investigate the expression and subcellular distribution of sialin during postnatal development in the mouse submandibular gland (SMG). DESIGN Six SMGs from both female and male C57BL/6 mice were collected at P10, P30 and P90, and the material from each littermate of either sex was pooled to extract total RNA and tissue protein. The remaining tissues were immediately fixed in 10% neutral buffered formalin for histological analysis. The mRNA and protein expression levels of sialin were examined by quantitative real-time RT-PCR and Western blot analysis. The subcellular distribution of sialin was analysed by immunohistochemistry and immunofluorescence. RESULTS The postnatal expression level of sialin in the mouse SMG was comparable with that in brain at each time point tested. The temporal expression of sialin in the SMG gradually increased during postnatal maturation. Immunohistochemical and immunofluorescence analysis demonstrated that sialin was predominantly expressed on the basal cytoplasmic membrane of acini and ducts, as well as in some myoepithelial cells in the SMG. CONCLUSIONS The high-level expression and subcellular distribution pattern of sialin in the SMG suggest that sialin may play an important role in the transport and secretion of saliva.
Collapse
Affiliation(s)
- Miao He
- Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy & Tooth Regeneration, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing 100050, China
| | | | | | | | | | | | | |
Collapse
|
63
|
Two dileucine motifs mediate late endosomal/lysosomal targeting of transmembrane protein 192 (TMEM192) and a C-terminal cysteine residue is responsible for disulfide bond formation in TMEM192 homodimers. Biochem J 2011; 434:219-31. [PMID: 21143193 DOI: 10.1042/bj20101396] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
TMEM192 (transmembrane protein 192) is a novel constituent of late endosomal/lysosomal membranes with four potential transmembrane segments and an unknown function that was initially discovered by organellar proteomics. Subsequently, localization in late endosomes/lysosomes has been confirmed for overexpressed and endogenous TMEM192, and homodimers of TMEM192 linked by disulfide bonds have been reported. In the present study the molecular determinants of TMEM192 mediating its transport to late endosomes/lysosomes were analysed by using CD4 chimaeric constructs and mutagenesis of potential targeting motifs in TMEM192. Two directly adjacent N-terminally located dileucine motifs of the DXXLL-type were found to be critical for transport of TMEM192 to late endosomes/lysosomes. Whereas disruption of both dileucine motifs resulted in mistargeting of TMEM192 to the plasma membrane, each of the two motifs was sufficient to ensure correct targeting of TMEM192. In order to study disulfide bond formation, mutagenesis of cysteine residues was performed. Mutation of Cys266 abolished disulfide bridge formation between TMEM192 molecules, indicating that TMEM192 dimers are linked by a disulfide bridge between their C-terminal tails. According to the predicted topology, Cys266 would be localized in the reductive milieu of the cytosol where disulfide bridges are generally uncommon. Using immunogold labelling and proteinase protection assays, the localization of the N- and C-termini of TMEM192 on the cytosolic side of the late endosomal/lysosomal membrane was experimentally confirmed. These findings may imply close proximity of the C-termini in TMEM192 dimers and a possible involvement of this part of the protein in dimer assembly.
Collapse
|
64
|
Abstract
Lysosomes are organelles of eukaryotic cells that are critically involved in the degradation of macromolecules mainly delivered by endocytosis and autophagocytosis. Degradation is achieved by more than 60 hydrolases sequestered by a single phospholipid bilayer. The lysosomal membrane facilitates interaction and fusion with other compartments and harbours transport proteins catalysing the export of catabolites, thereby allowing their recycling. Lysosomal proteins have been addressed in various proteomic studies that are compared in this review regarding the source of material, the organelle/protein purification scheme, the proteomic methodology applied and the proteins identified. Distinguishing true constituents of an organelle from co-purifying contaminants is a central issue in subcellular proteomics, with additional implications for lysosomes as being the site of degradation of many cellular and extracellular proteins. Although many of the lysosomal hydrolases were identified by classical biochemical approaches, the knowledge about the protein composition of the lysosomal membrane has remained fragmentary for a long time. Using proteomics many novel lysosomal candidate proteins have been discovered and it can be expected that their functional characterisation will help to understand functions of lysosomes at a molecular level that have been characterised only phenomenologically so far and to generally deepen our understanding of this indispensable organelle.
Collapse
Affiliation(s)
- Bernd A Schröder
- Biochemical Institute, Christian-Albrechts University, Kiel, Germany.
| | | | | | | |
Collapse
|
65
|
Abstract
The Ca(2+)-dependent release of aspartate from hippocampal preparations was first reported 35 years ago, but the functional significance of this process remains uncertain. Aspartate satisfies all the criteria normally required for identification of a CNS transmitter. It is synthesized in nerve terminals, is accumulated and stored in synaptic vesicles, is released by exocytosis upon nerve terminal depolarization, and activates postsynaptic NMDA receptors. Aspartate may be employed as a neuropeptide-like co-transmitter by pathways that release either glutamate or GABA as their principal transmitter. Aspartate mechanisms include vesicular transport by sialin, vesicular content sensitive to glucose concentration, release mainly outside the presynaptic active zones, and selective activation of extrasynaptic NR1-NR2B NMDA receptors. Possible neurobiological functions of aspartate in immature neurons include activation of cAMP-dependent gene transcription and in mature neurons inhibition of CREB function, reduced BDNF expression, and induction of excitotoxic neuronal death. Recent findings suggest new experimental approaches toward resolving the functional significance of aspartate release.
Collapse
Affiliation(s)
- J Victor Nadler
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
66
|
Sharifi A, Kousi M, Sagné C, Bellenchi GC, Morel L, Darmon M, Hulková H, Ruivo R, Debacker C, El Mestikawy S, Elleder M, Lehesjoki AE, Jalanko A, Gasnier B, Kyttälä A. Expression and lysosomal targeting of CLN7, a major facilitator superfamily transporter associated with variant late-infantile neuronal ceroid lipofuscinosis. Hum Mol Genet 2010; 19:4497-514. [PMID: 20826447 DOI: 10.1093/hmg/ddq381] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neuronal ceroid lipofuscinoses (NCLs) constitute a group of progressive neurodegenerative disorders resulting from mutations in at least eight different genes. Mutations in the most recently identified NCL gene, MFSD8/CLN7, underlie a variant of late-infantile NCL (vLINCL). The MFSD8/CLN7 gene encodes a polytopic protein with unknown function, which shares homology with ion-coupled membrane transporters. In this study, we confirmed the lysosomal localization of the native CLN7 protein. This localization of CLN7 is not impaired by the presence of pathogenic missense mutations or after genetic ablation of the N-glycans. Expression of chimeric and full-length constructs showed that lysosomal targeting of CLN7 is mainly determined by an N-terminal dileucine motif, which specifically binds to the heterotetrameric adaptor AP-1 in vitro. We also show that CLN7 mRNA is more abundant in neurons than astrocytes and microglia, and that it is expressed throughout rat brain, with increased levels in the granular layer of cerebellum and hippocampal pyramidal cells. Interestingly, this cellular and regional distribution is in good agreement with the autofluorescent lysosomal storage and cell loss patterns found in brains from CLN7-defective patients. Overall, these data highlight lysosomes as the primary site of action for CLN7, and suggest that the pathophysiology underpinning CLN7-associated vLINCL is a cell-autonomous process.
Collapse
Affiliation(s)
- A Sharifi
- Institut de Biologie Physico-Chimique, Université Paris Descartes, Centre National de la Recherche Scientifique, UMR 8192, Institut de Biologie Physico-Chimique, 13 Rue P. et M. Curie, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Steenhuis P, Herder S, Gelis S, Braulke T, Storch S. Lysosomal targeting of the CLN7 membrane glycoprotein and transport via the plasma membrane require a dileucine motif. Traffic 2010; 11:987-1000. [PMID: 20406422 DOI: 10.1111/j.1600-0854.2010.01073.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CLN7 is a polytopic lysosomal membrane protein deficient in variant late infantile neuronal ceroid lipofuscinosis, a neurodegenerative lysosomal storage disorder. In this study fluorescence protease protection assays and mutational analyses revealed the N- and C-terminal tails of CLN7 in the cytosol and two N-glycosylation sites at N371 and N376. Both partially and non-glycosylated CLN7 were correctly transported to lysosomes. To identify lysosomal targeting motifs, we generated CD4-chimera fused to the N- and C-terminal domains of CLN7. Lysosomal localization of the chimeric proteins requires a consensus acidic dileucine-based motif in the N-terminus and two tandem tyrosine-based signals in the C-terminus. Mutation of these sorting motifs resulted in cell surface redistribution of CD4 chimeras. However, the dileucine-based motif is of critical importance for lysosomal localization of the full-length CLN7 in different cell lines. Cell surface biotinylation revealed that at equilibrium 22% of total CLN7 is localized at the plasma membrane. Mutation of the dileucine motif or the co-expression of dominant-negative mutant dynamin K44A led to a further increase of CLN7 at the plasma membrane. Our data demonstrate that CLN7 contains several cytoplasmic lysosomal targeting signals of which the N-terminal dileucine-based motif is required for the predominant lysosomal targeting along the indirect pathway and clathrin-mediated endocytosis of CLN7.
Collapse
Affiliation(s)
- Pieter Steenhuis
- Department of Biochemistry, Children's Hospital, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246 Hamburg, Germany
| | | | | | | | | |
Collapse
|
68
|
Synaptic and vesicular coexistence of VGLUT and VGAT in selected excitatory and inhibitory synapses. J Neurosci 2010; 30:7634-45. [PMID: 20519538 DOI: 10.1523/jneurosci.0141-10.2010] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The segregation between vesicular glutamate and GABA storage and release forms the molecular foundation between excitatory and inhibitory neurons and guarantees the precise function of neuronal networks. Using immunoisolation of synaptic vesicles, we now show that VGLUT2 and VGAT, and also VGLUT1 and VGLUT2, coexist in a sizeable pool of vesicles. VGAT immunoisolates transport glutamate in addition to GABA. Furthermore, VGLUT activity enhances uptake of GABA and monoamines. Postembedding immunogold double labeling revealed that VGLUT1, VGLUT2, and VGAT coexist in mossy fiber terminals of the hippocampal CA3 area. Similarly, cerebellar mossy fiber terminals harbor VGLUT1, VGLUT2, and VGAT, while parallel and climbing fiber terminals exclusively contain VGLUT1 or VGLUT2, respectively. VGLUT2 was also observed in cerebellar GABAergic basket cells terminals. We conclude that the synaptic coexistence of vesicular glutamate and GABA transporters allows for corelease of both glutamate and GABA from selected nerve terminals, which may prevent systemic overexcitability by downregulating synaptic activity. Furthermore, our data suggest that VGLUT enhances transmitter storage in nonglutamatergic neurons. Thus, synaptic and vesicular coexistence of VGLUT and VGAT is more widespread than previously anticipated, putatively influencing fine-tuning and control of synaptic plasticity.
Collapse
|
69
|
Courville P, Quick M, Reimer RJ. Structure-function studies of the SLC17 transporter sialin identify crucial residues and substrate-induced conformational changes. J Biol Chem 2010; 285:19316-23. [PMID: 20424173 PMCID: PMC2885210 DOI: 10.1074/jbc.m110.130716] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Revised: 04/26/2010] [Indexed: 11/06/2022] Open
Abstract
Salla disease and infantile sialic acid storage disorder are human diseases caused by loss of function of sialin, a lysosomal transporter that mediates H(+)-coupled symport of acidic sugars N-acetylneuraminic acid and glucuronic acid out of lysosomes. Along with the closely related vesicular glutamate transporters, sialin belongs to the SLC17 transporter family. Despite their critical role in health and disease, these proteins remain poorly understood both structurally and mechanistically. Here, we use substituted cysteine accessibility screening and radiotracer flux assays to evaluate experimentally a computationally generated three-dimensional structure model of sialin. According to this model, sialin consists of 12 transmembrane helices (TMs) with an overall architecture similar to that of the distantly related glycerol 3-phosphate transporter GlpT. We show that TM4 in sialin lines a large aqueous cavity that forms a part of the substrate permeation pathway and demonstrate substrate-induced alterations in accessibility of substituted cysteine residues in TM4. In addition, we demonstrate that one mutant, F179C, has a dramatically different effect on the apparent affinity and transport rate for N-acetylneuraminic acid and glucuronic acid, suggesting that it may be directly involved in substrate recognition and/or translocation. These findings offer a basis for further defining the transport mechanism of sialin and other SLC17 family members.
Collapse
Affiliation(s)
- Pascal Courville
- From the Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305 and
| | - Matthias Quick
- the Center for Molecular Recognition, Department of Psychiatry, Columbia University College of Physicians and Surgeons, and Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032
| | - Richard J. Reimer
- From the Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305 and
| |
Collapse
|
70
|
Abstract
Salla disease and infantile sialic acid storage disease are autosomal recessive lysosomal storage disorders caused by mutations in the gene encoding sialin, a membrane protein that transports free sialic acid out of the lysosome after it is cleaved from sialoglycoconjugates undergoing degradation. Accumulation of sialic acid in lysosomes defines these disorders, and the clinical phenotype is characterized by neurodevelopmental defects, including severe CNS hypomyelination. In this study, we used a sialin-deficient mouse to address how loss of sialin leads to the defect in myelination. Behavioral analysis of the sialin(-/-) mouse demonstrates poor coordination, seizures, and premature death. Analysis by histology, electron microscopy, and Western blotting reveals a decrease in myelination of the CNS but normal neuronal cytoarchitecture and normal myelination of the PNS. To investigate potential mechanisms underlying CNS hypomyelination, we studied myelination and oligodendrocyte development in optic nerves. We found reduced numbers of myelinated axons in optic nerves from sialin(-/-) mice, but the myelin that was present appeared grossly normal. Migration and density of oligodendrocyte precursor cells were normal; however, a marked decrease in the number of postmitotic oligodendrocytes and an associated increase in the number of apoptotic cells during the later stages of myelinogenesis were observed. These findings suggest that a defect in maturation of cells in the oligodendrocyte lineage leads to increased apoptosis and underlies the myelination defect associated with sialin loss.
Collapse
|
71
|
Mochel F, Yang B, Barritault J, Thompson JN, Engelke UFH, McNeill NH, Benko WS, Kaneski CR, Adams DR, Tsokos M, Abu-Asab M, Huizing M, Seguin F, Wevers RA, Ding J, Verheijen FW, Schiffmann R. Free sialic acid storage disease without sialuria. Ann Neurol 2009; 65:753-7. [PMID: 19557856 PMCID: PMC3508714 DOI: 10.1002/ana.21624] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We performed high-resolution in vitro proton nuclear magnetic resonance spectroscopy on cerebrospinal fluid and urine samples of 44 patients with leukodystrophies of unknown cause. Free sialic acid concentration was increased in cerebrospinal fluid of two siblings with mental retardation and mild hypomyelination. By contrast, urinary excretion of free sialic acid in urine was normal on repeated testing by two independent methods. Both patients were homozygous for the K136E mutation in SLC17A5, the gene responsible for the free sialic acid storage diseases. Our findings demonstrate that mutations in the SLC17A5 gene have to be considered in patients with hypomyelination, even in the absence of sialuria.
Collapse
Affiliation(s)
- Fanny Mochel
- Institut National de la Sante et de la Recherche Médicale UMR S679, Hôpital La Salpêtrière, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Ruivo R, Anne C, Sagné C, Gasnier B. Molecular and cellular basis of lysosomal transmembrane protein dysfunction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:636-49. [DOI: 10.1016/j.bbamcr.2008.12.008] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/10/2008] [Accepted: 12/11/2008] [Indexed: 02/04/2023]
|
73
|
Boonen M, Rezende de Castro R, Cuvelier G, Hamer I, Jadot M. A dileucine signal situated in the C-terminal tail of the lysosomal membrane protein p40 is responsible for its targeting to lysosomes. Biochem J 2008; 414:431-40. [PMID: 18479248 DOI: 10.1042/bj20071626] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Transport of newly synthesized lysosomal membrane proteins from the TGN (trans-Golgi network) to the lysosomes is due to the presence of specific signals in their cytoplasmic domains that are recognized by cytosolic adaptors. p40, a hypothetical transporter of 372 amino acids localized in the lysosomal membrane, contains four putative lysosomal sorting motifs in its sequence: three of the YXXphi-type (Y(6)QLF, Y(106)VAL, Y(333)NGL) and one of the [D/E]XXXL[L/I]-type (EQERL(360)L(361)). To test the role of these motifs in the biosynthetic transport of p40, we replaced the most critical residues of these consensus sequences, the tyrosine residue or the leucine-leucine pair, by alanine or alanine-valine respectively. We analysed the subcellular localization of the mutated p40 proteins in transfected HeLa cells by confocal microscopy and by biochemical approaches (subcellular fractionation on self-forming Percoll density gradients and cell surface biotinylation). The results of the present study show that p40 is mistargeted to the plasma membrane when its dileucine motif is disrupted. No role of the tyrosine motifs could be put forward. Taken together, our results provide evidence that the sorting of p40 from the TGN to the lysosomes is directed by the dileucine EQERL(360)L(361) motif situated in its C-terminal tail.
Collapse
Affiliation(s)
- Marielle Boonen
- URPhiM, Laboratoire de Chimie Physiologique, FUNDP, B-5000 Namur, Belgium
| | | | | | | | | |
Collapse
|
74
|
Lysosomes are the major vesicular compartment undergoing Ca2+-regulated exocytosis from cortical astrocytes. J Neurosci 2008; 28:7648-58. [PMID: 18650341 DOI: 10.1523/jneurosci.0744-08.2008] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although Ca(2+)-dependent exocytosis is considered to be a pathway for gliotransmitter release from astrocytes, the structural and functional bases of this process remain controversial. We studied the relationship between near-membrane Ca(2+) elevations and the dynamics of single astroglial vesicles with styryl (FM) dyes. We show that cultured astrocytes, unlike neurons, spontaneously internalize FM dyes, resulting in the labeling of the entire acidic vesicle population within minutes. Interestingly, metabotropic glutamate receptor activation did not affect the FM labeling. Most FM-stained vesicles expressed sialin, CD63/LAMP3, and VAMP7, three markers for lysosomes and late endosomes. A subset of lysosomes underwent asynchronous exocytosis that required both Ca(2+) mobilization from intracellular stores and Ca(2+) influx across the plasma membrane. Lysosomal fusion occurred within seconds and was complete with no evidence for kiss and run. Our experiments suggest that astroglial Ca(2+)-regulated exocytosis is carried by lysosomes and operates on a timescale orders of magnitude slower than synaptic transmission.
Collapse
|
75
|
Abstract
Aspartate is an excitatory amino acid that is costored with glutamate in synaptic vesicles of hippocampal neurons and synaptic-like microvesicles (SLMVs) of pinealocytes and is exocytosed and stimulates neighboring cells by binding to specific cell receptors. Although evidence increasingly supports the occurrence of aspartergic neurotransmission, this process is still debated because the mechanism for the vesicular storage of aspartate is unknown. Here, we show that sialin, a lysosomal H(+)/sialic acid cotransporter, is present in hippocampal synaptic vesicles and pineal SLMVs. RNA interference of sialin expression decreased exocytosis of aspartate and glutamate in pinealocytes. Proteoliposomes containing purified sialin actively accumulated aspartate and glutamate to a similar extent when inside positive membrane potential is imposed as the driving force. Sialin carrying a mutation found in people suffering from Salla disease (R39C) was completely devoid of aspartate and glutamate transport activity, although it retained appreciable H(+)/sialic acid cotransport activity. These results strongly suggest that sialin possesses dual physiological functions and acts as a vesicular aspartate/glutamate transporter. It is possible that people with Salla disease lose aspartergic (and also the associated glutamatergic) neurotransmission, and this could provide an explanation for why Salla disease causes severe neurological defects.
Collapse
|
76
|
Hippert C, Dubois G, Morin C, Disson O, Ibanes S, Jacquet C, Schwendener R, Antignac C, Kremer EJ, Kalatzis V. Gene Transfer May Be Preventive But Not Curative for a Lysosomal Transport Disorder. Mol Ther 2008; 16:1372-81. [DOI: 10.1038/mt.2008.126] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
77
|
Long AA, Kim E, Leung HT, Woodruff E, An L, Doerge RW, Pak WL, Broadie K. Presynaptic calcium channel localization and calcium-dependent synaptic vesicle exocytosis regulated by the Fuseless protein. J Neurosci 2008; 28:3668-82. [PMID: 18385325 PMCID: PMC2769928 DOI: 10.1523/jneurosci.5553-07.2008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2007] [Revised: 02/29/2008] [Accepted: 03/01/2008] [Indexed: 11/21/2022] Open
Abstract
A systematic forward genetic Drosophila screen for electroretinogram mutants lacking synaptic transients identified the fuseless (fusl) gene, which encodes a predicted eight-pass transmembrane protein in the presynaptic membrane. Null fusl mutants display >75% reduction in evoked synaptic transmission but, conversely, an approximately threefold increase in the frequency and amplitude of spontaneous synaptic vesicle fusion events. These neurotransmission defects are rescued by a wild-type fusl transgene targeted only to the presynaptic cell, demonstrating a strictly presynaptic requirement for Fusl function. Defects in FM dye turnover at the synapse show a severely impaired exo-endo synaptic vesicle cycling pool. Consistently, ultrastructural analyses reveal accumulated vesicles arrested in clustered and docked pools at presynaptic active zones. In the absence of Fusl, calcium-dependent neurotransmitter release is dramatically compromised and there is little enhancement of synaptic efficacy with elevated external Ca(2+) concentrations. These defects are causally linked with severe loss of the Cacophony voltage-gated Ca(2+) channels, which fail to localize normally at presynaptic active zone domains in the absence of Fusl. These data indicate that Fusl regulates assembly of the presynaptic active zone Ca(2+) channel domains required for efficient coupling of the Ca(2+) influx and synaptic vesicle exocytosis during neurotransmission.
Collapse
Affiliation(s)
- A. Ashleigh Long
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235-1634, and
| | - Eunju Kim
- Departments of Biological Sciences and
| | | | - Elvin Woodruff
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235-1634, and
| | - Lingling An
- Statistics, Purdue University, West Lafayette, Indiana 47907
| | - R. W. Doerge
- Statistics, Purdue University, West Lafayette, Indiana 47907
| | | | - Kendal Broadie
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235-1634, and
| |
Collapse
|
78
|
Sagné C, Gasnier B. Molecular physiology and pathophysiology of lysosomal membrane transporters. J Inherit Metab Dis 2008; 31:258-66. [PMID: 18425435 DOI: 10.1007/s10545-008-0879-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2008] [Revised: 02/19/2008] [Accepted: 02/20/2008] [Indexed: 10/22/2022]
Abstract
In contrast to lysosomal hydrolytic enzymes, the lysosomal membrane remains poorly characterized. In particular, although the genetic study of cystinosis and sialic acid storage disorders led to the identification of two lysosomal transporters for cystine and sialic acids, respectively, ten years ago, most transporters responsible for exporting lysosomal hydrolysis products to the cytosol are still unknown at the molecular level. However, two lines of investigation recently started to fill this gap in the knowledge of lysosomal biology. First, novel proteomic approaches are now able to provide a reliable inventory of lysosomal membrane proteins. On the other hand, a novel functional approach based on intracellular trafficking mechanisms allows direct transport measurement in whole cells by redirecting recombinant lysosomal transporters to the cell surface. After surveying the current state of knowledge in this field, the review focuses on the sialic acid transporter sialin and shows how recent functional data using the above whole-cell approach shed new light on the pathogenesis of sialic acid storage disorders by revealing the existence of a residual transport activity associated with Salla disease.
Collapse
Affiliation(s)
- C Sagné
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Université Paris 7 Denis Diderot, Paris, France
| | | |
Collapse
|
79
|
Guo B, Jin Y, Wussler C, Blancaflor EB, Motes CM, Versaw WK. Functional analysis of the Arabidopsis PHT4 family of intracellular phosphate transporters. THE NEW PHYTOLOGIST 2008; 177:889-898. [PMID: 18086223 DOI: 10.1111/j.1469-8137.2007.02331.x] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The transport of phosphate (Pi) between subcellular compartments is central to metabolic regulation. Although some of the transporters involved in controlling the intracellular distribution of Pi have been identified in plants, others are predicted from genetic, biochemical and bioinformatics studies. Heterologous expression in yeast, and gene expression and localization in plants were used to characterize all six members of an Arabidopsis thaliana membrane transporter family designated here as PHT4. PHT4 proteins share similarity with SLC17/type I Pi transporters, a diverse group of animal proteins involved in the transport of Pi, organic anions and chloride. All of the PHT4 proteins mediate Pi transport in yeast with high specificity. Bioinformatic analysis and localization of PHT4-GFP fusion proteins indicate that five of the proteins are targeted to the plastid envelope, and the sixth resides in the Golgi apparatus. PHT4 genes are expressed in both roots and leaves, although two of the genes are expressed predominantly in leaves and one mostly in roots. These expression patterns, together with Pi transport activities and subcellular locations, suggest roles for PHT4 proteins in the transport of Pi between the cytosol and chloroplasts, heterotrophic plastids and the Golgi apparatus.
Collapse
Affiliation(s)
| | | | | | - E B Blancaflor
- Plant Biology Division, Samuel Roberts Noble Foundation, Ardmore, OK 73401, USA
| | - C M Motes
- Plant Biology Division, Samuel Roberts Noble Foundation, Ardmore, OK 73401, USA
| | - W K Versaw
- Department of Biology
- Molecular and Environmental Plant Sciences Program, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
80
|
Laridon B, Callaerts P, Norga K. Embryonic expression patterns of Drosophila ACS family genes related to the human sialin gene. Gene Expr Patterns 2007; 8:275-83. [PMID: 18255354 DOI: 10.1016/j.gep.2007.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 12/11/2007] [Accepted: 12/12/2007] [Indexed: 10/22/2022]
Abstract
The anion/cation symporter (ACS) family is a large subfamily of the major facilitator superfamily (MFS) of transporters. ACS family permeases are widely distributed in nature and transport either organic or inorganic anions in response to chemiosmotic cation gradients. The only protein in the ACS family to which a human disease has been linked, is sialin, the proton-driven lysosomal carrier for sialic acid. Genetic defects in sialin cause a lysosomal storage disease in humans. Here we have identified a group of conserved Drosophila ACS family genes related to sialin and studied their expression patterns throughout embryogenesis. Drosophila sialin-related genes are expressed in a wide variety of tissues. Expression is frequently observed throughout various parts of the intestinal tract, including Malpighian tubules and salivary glands. Additionally, some genes are expressed in vitellophages (yolk nuclei), nervous system, respiratory tract and a number of other embryonic tissues. These data will aid the establishment of a fruitfly model of human lysosomal storage disorders, the most common cause of neurodegeneration in childhood.
Collapse
Affiliation(s)
- Bram Laridon
- Laboratory of Developmental Genetics, V.I.B., Herestraat 49, Mailbox 602, B-3000 Leuven, Belgium
| | | | | |
Collapse
|
81
|
Kawakubo T, Okamoto K, Iwata JI, Shin M, Okamoto Y, Yasukochi A, Nakayama KI, Kadowaki T, Tsukuba T, Yamamoto K. Cathepsin E prevents tumor growth and metastasis by catalyzing the proteolytic release of soluble TRAIL from tumor cell surface. Cancer Res 2007; 67:10869-78. [PMID: 18006832 DOI: 10.1158/0008-5472.can-07-2048] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aspartic proteinase cathepsin E is expressed predominantly in cells of the immune system and highly secreted by activated phagocytes, and deficiency of cathepsin E in mice results in a phenotype affecting immune responses. However, because physiologic substrates for cathepsin E have not yet been identified, the relevance of these observations to the physiologic functions of this protein remains speculative. Here, we show that cathepsin E specifically induces growth arrest and apoptosis in human prostate carcinoma tumor cell lines without affecting normal cells by catalyzing the proteolytic release of soluble tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) from the cell surface. The antitumor activity of cathepsin E was corroborated by in vivo studies with mice bearing human and mouse tumor transplants. Administration of purified cathepsin E into human tumor xenografts in nude mice dose-dependently induced apoptosis in the tumor cells to inhibit tumor growth. The growth, viability, and metastasis of mouse B16 melanoma cells were also more profound in cathepsin E-deficient mice compared with those in the syngeneic wild-type and transgenic mice overexpressing cathepsin E. Taken together, the number of apoptotic tumor cells, as well as tumor-infiltrating activated macrophages, was apparently reduced in cathepsin E-deficient mice compared with those in the other two groups, implying the positive correlation of endogenous cathepsin E levels with the extent of tumor suppression in vivo. These results thus indicate that cathepsin E plays a substantial role in host defense against tumor cells through TRAIL-dependent apoptosis and/or tumor-associated macrophage-mediated cytotoxicity.
Collapse
Affiliation(s)
- Tomoyo Kawakubo
- Department of Pharmacology, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Myall NJ, Wreden CC, Wlizla M, Reimer RJ. G328E and G409E sialin missense mutations similarly impair transport activity, but differentially affect trafficking. Mol Genet Metab 2007; 92:371-4. [PMID: 17933575 PMCID: PMC2171360 DOI: 10.1016/j.ymgme.2007.08.121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Revised: 08/25/2007] [Accepted: 08/25/2007] [Indexed: 10/22/2022]
Abstract
Two disease-associated missense mutations in the sialin gene (G328E and G409E) have recently been identified in patients with lysosomal free sialic acid storage disease. We have assessed the effect of these mutations and find complete loss of measurable transport activity with both and impaired trafficking of the G409E protein. These results suggest that the two residues are important for proper function of sialin and confirm the association of loss of transport with disease causative mutations.
Collapse
Affiliation(s)
- Nathaniel J. Myall
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Christopher C. Wreden
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Richard J. Reimer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
83
|
Desnos C, Huet S, Fanget I, Chapuis C, Böttiger C, Racine V, Sibarita JB, Henry JP, Darchen F. Myosin va mediates docking of secretory granules at the plasma membrane. J Neurosci 2007; 27:10636-45. [PMID: 17898234 PMCID: PMC6673143 DOI: 10.1523/jneurosci.1228-07.2007] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Myosin Va (MyoVa) is a prime candidate for controlling actin-based organelle motion in neurons and neuroendocrine cells. Its function in secretory granule (SG) trafficking was investigated in enterochromaffin cells by wide-field and total internal reflection fluorescence microscopy. The distribution of endogenous MyoVa partially overlapped with SGs and microtubules. Impairing MyoVa function by means of a truncated construct (MyoVa tail) or RNA interference prevented the formation of SG-rich regions at the cell periphery and reduced SG density in the subplasmalemmal region. Individual SG trajectories were tracked to analyze SG mobility. A wide distribution of their diffusion coefficient, D(xy), was observed. Almost immobile SGs (D(xy) < 5 x 10(-4) microm2 x s(-1)) were considered as docked at the plasma membrane based on two properties: (1) SGs that undergo exocytosis have a D(xy) below this threshold value for at least 2 s before fusion; (2) a negative autocorrelation of the vertical motion was found in subtrajectories with a D(xy) below the threshold. Using this criterion of docking, we found that the main effect of MyoVa inhibition was to reduce the number of docked granules, leading to reduced secretory responses. Surprisingly, this reduction was not attributable to a decreased transport of SGs toward release sites. In contrast, MyoVa silencing reduced the occurrence of long-lasting, but not short-lasting, docking periods. We thus propose that, despite its known motor activity, MyoVa directly mediates stable attachment of SGs at the plasma membrane.
Collapse
Affiliation(s)
- Claire Desnos
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| | - Sébastien Huet
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| | - Isabelle Fanget
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| | - Catherine Chapuis
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| | - Caroline Böttiger
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| | - Victor Racine
- Institut Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75248 Paris Cedex 05, France
| | - Jean-Baptiste Sibarita
- Institut Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 144, 75248 Paris Cedex 05, France
| | - Jean-Pierre Henry
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| | - François Darchen
- Institut de Biologie Physico-Chimique, Centre National de la Recherche Scientifique, Unité Propre de Recherche 1929, Université Paris 7 Denis Diderot, 75005 Paris, France, and
| |
Collapse
|
84
|
Abstract
Changes in the response to release of a single synaptic vesicle have generally been attributed to postsynaptic modification of receptor sensitivity, but considerable evidence now demonstrates that alterations in vesicle filling also contribute to changes in quantal size. Receptors are not saturated at many synapses, and changes in the amount of transmitter per vesicle contribute to the physiological regulation of release. On the other hand, the presynaptic factors that determine quantal size remain poorly understood. Aside from regulation of the fusion pore, these mechanisms fall into two general categories: those that affect the accumulation of transmitter inside a vesicle and those that affect vesicle size. This review will summarize current understanding of the neurotransmitter cycle and indicate basic, unanswered questions about the presynaptic regulation of quantal size.
Collapse
Affiliation(s)
- Robert H Edwards
- Department of Neurology and Physiology, UCSF School of Medicine, San Francisco, CA 94158-2517, USA.
| |
Collapse
|
85
|
Juge N, Yoshida Y, Yatsushiro S, Omote H, Moriyama Y. Vesicular glutamate transporter contains two independent transport machineries. J Biol Chem 2006; 281:39499-506. [PMID: 17046815 DOI: 10.1074/jbc.m607670200] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vesicular glutamate transporters (VGLUTs) are responsible for the vesicular storage of l-glutamate and play an essential role in glutamatergic signal transmission in the central nervous system. The molecular mechanism of the transport remains unknown. Here, we established a novel in vitro assay procedure, which includes purification of wild and mutant VGLUT2 and their reconstitution with purified bacterial F(o)F(1)-ATPase (F-ATPase) into liposomes. Upon the addition of ATP, the proteoliposomes facilitated l-glutamate uptake in a membrane potential (DeltaPsi)-dependent fashion. The ATP-dependent l-glutamate uptake exhibited an absolute requirement for approximately 4 mm Cl(-), was sensitive to Evans blue, but was insensitive to d,l-aspartate. VGLUT2s with mutations in the transmembrane-located residues Arg(184), His(128), and Glu(191) showed a dramatic loss in l-glutamate transport activity, whereas Na(+)-dependent inorganic phosphate (P(i)) uptake remained comparable to that of the wild type. Furthermore, P(i) transport did not require Cl(-) and was not inhibited by Evans blue. Thus, VGLUT2 appears to possess two intrinsic transport machineries that are independent of each other: a DeltaPsi-dependent l-glutamate uptake and a Na(+)-dependent P(i) uptake.
Collapse
Affiliation(s)
- Narinobu Juge
- Laboratory of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | | | | | | | | |
Collapse
|
86
|
Abstract
The expression of vesicular glutamate transporters (VGLUTs) 1 and 2 accounts for the ability of most traditionally accepted excitatory neurons to release glutamate by exocytosis. However, several cell populations (serotonin and dopamine neurons) have been demonstrated to release glutamate in vitro and do not obviously express these transporters. Rather, these neurons express a novel, third isoform that in fact appears confined to neurons generally associated with a transmitter other than glutamate. They include serotonin and possibly dopamine neurons, cholinergic interneurons in the striatum, and GABAergic interneurons of the hippocampus and cortex. Although the physiological role of VGLUT3 remains largely conjectural, several observations in vivo suggest that the glutamate release mediated by VGLUT3 has an important role in synaptic transmission, plasticity, and development.
Collapse
Affiliation(s)
- R P Seal
- Department of Neurology, UCSF School of Medicine, 600 16th St., GH-N272B, San Francisco CA, 94143-2140, USA
| | | |
Collapse
|
87
|
Boonen M, Hamer I, Boussac M, Delsaute AF, Flamion B, Garin J, Jadot M. Intracellular localization of p40, a protein identified in a preparation of lysosomal membranes. Biochem J 2006; 395:39-47. [PMID: 16367739 PMCID: PMC1409702 DOI: 10.1042/bj20051647] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Unlike lysosomal soluble proteins, few lysosomal membrane proteins have been identified. Rat liver lysosomes were purified by centrifugation on a Nycodenz density gradient. The most hydrophobic proteins were extracted from the lysosome membrane preparation and were identified by MS. We focused our attention on a protein of approx. 40 kDa, p40, which contains seven to ten putative transmembrane domains and four lysosomal consensus sorting motifs in its sequence. Knowing that preparations of lysosomes obtained by centrifugation always contain contaminant membranes, we combined biochemical and morphological methods to analyse the subcellular localization of p40. The results of subcellular fractionation of mouse liver homogenates validate the lysosomal residence of p40. In particular, a density shift of lysosomes induced by Triton WR-1339 similarly affected the distributions of p40 and beta-galactosidase, a lysosomal marker protein. We confirmed by fluorescence microscopy on eukaryotic cells transfected with p40 or p40-GFP (green fluorescent protein) constructs that p40 is localized in lysosomes. A first molecular characterization of p40 in transfected Cos-7 cells revealed that it is an unglycosylated protein tightly associated with membranes. Taken together, our results strongly support the hypothesis that p40 is an authentic lysosomal membrane protein.
Collapse
Affiliation(s)
- Marielle Boonen
- *URΦM, Laboratoire de Chimie Physiologique, Facultés Universitaires Notre-Dame de la Paix, B-5000 Namur, Belgium
| | - Isabelle Hamer
- *URΦM, Laboratoire de Chimie Physiologique, Facultés Universitaires Notre-Dame de la Paix, B-5000 Namur, Belgium
| | - Muriel Boussac
- †Laboratoire de Chimie des Protéines, CEA/INSERM/UJF, F-38054 Grenoble, France
| | - Anne-Françoise Delsaute
- *URΦM, Laboratoire de Chimie Physiologique, Facultés Universitaires Notre-Dame de la Paix, B-5000 Namur, Belgium
| | - Bruno Flamion
- ‡URΦM, Laboratoire de Physiologie/Pharmacologie, Facultés Universitaires Notre-Dame de la Paix, B-5000 Namur, Belgium
| | - Jérôme Garin
- †Laboratoire de Chimie des Protéines, CEA/INSERM/UJF, F-38054 Grenoble, France
| | - Michel Jadot
- *URΦM, Laboratoire de Chimie Physiologique, Facultés Universitaires Notre-Dame de la Paix, B-5000 Namur, Belgium
- To whom correspondence should be addressed (email )
| |
Collapse
|
88
|
Nadrigny F, Rivals I, Hirrlinger PG, Koulakoff A, Personnaz L, Vernet M, Allioux M, Chaumeil M, Ropert N, Giaume C, Kirchhoff F, Oheim M. Detecting fluorescent protein expression and co-localisation on single secretory vesicles with linear spectral unmixing. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2006; 35:533-47. [PMID: 16568270 DOI: 10.1007/s00249-005-0040-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Accepted: 12/07/2005] [Indexed: 10/24/2022]
Abstract
Many questions in cell biology and biophysics involve the quantitation of co-localisation and the interaction of proteins tagged with different fluorophores. However, the incomplete separation of the different colour channels due to the presence of autofluorescence, along with cross-excitation and emission "bleed-through" of one colour channel into the other, all combine to render the interpretation of multi-band images ambiguous. Here we introduce a new live-cell epifluorescence spectral imaging and linear unmixing technique for classifying resolution-limited point objects containing multiple fluorophores. We demonstrate the performance of our technique by detecting, at the single-vesicle level, the co-expression of the vesicle-associated membrane protein, VAMP-2 (also called synaptobrevin-2), linked to either enhanced green fluorescent protein (EGFP) or citrine [a less pH-sensitive variant of enhanced yellow fluorescent protein (EYFP)], in mouse cortical astrocytes. In contrast, the co-expression of VAMP-2-citrine and the lysosomal transporter sialine fused to EGFP resulted in little overlap. Spectral imaging and linear unmixing permit us to fingerprint the expression of spectrally overlapping fluorescent proteins on single secretory organelles in the presence of a spectrally broad autofluorescence. Our technique provides a robust alternative to error-prone dual- or triple colour co-localisation studies.
Collapse
Affiliation(s)
- Fabien Nadrigny
- Molecular and Cellular Biophysics of Synaptic Transmission, Laboratory of Neurophysiology and New Microscopies, INSERM U603, CNRS FRE 2500, Université René Descartes (Paris 5), 45 rue des Saints Pères, 75 006, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Biancheri R, Rossi A, Verbeek HA, Schot R, Corsolini F, Assereto S, Mancini GMS, Verheijen FW, Minetti C, Filocamo M. Homozygosity for the p.K136E mutation in the SLC17A5 gene as cause of an Italian severe Salla disease. Neurogenetics 2005; 6:195-9. [PMID: 16170568 DOI: 10.1007/s10048-005-0011-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Accepted: 07/28/2005] [Indexed: 11/29/2022]
Abstract
Lysosomal free sialic acid storage diseases are recessively inherited allelic neurodegenerative disorders that include Salla disease (SD) and infantile sialic acid storage disease (ISSD) caused by mutations in the SLC17A5 gene encoding for a lysosomal membrane protein, sialin, transporting sialic acid from lysosomes. The classical form of SD, enriched in the Finnish population, is related to the p.R39C designed Salla(FIN) founder mutation. A more severe phenotype is due both to compound heterozygosity for the p.R39C mutation and to different mutations. The p.R39C has not been reported in ISSD. We identified the first case of SD caused by the homozygosity for p.K136E (c.406A>G) mutation, showing a severe clinical picture, as demonstrated by the early age at onset, the degree of motor retardation, the occurrence of peripheral nerve involvement, as well as cerebral hypomyelination. Recently, in vitro functional studies have shown that the p.K136E mutant produces a mislocalization and a reduced activity of the intracellular sialin. We discuss the in vivo phenotypic consequence of the p.K136E in relation to the results obtained by the in vitro functional characterization of the p.K136E mutant. The severity of the clinical picture, in comparison with the classical SD, may be explained by the fact that the p.K136E mutation mislocalizes the protein to a greater degree than p.R39C. On the other hand, the presence of a residual transport activity may account for the absence of hepatosplenomegaly, dysostosis multiplex, and early lethality typical of ISSD and related to the abolished transport activity found in this latter form.
Collapse
Affiliation(s)
- R Biancheri
- Department of Neuroscience and Rehabilitation, Muscular and Neurodegenerative Disease Unit, G. Gaslini Institute, University of Genova, Genova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|