Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 442
Filter
Add more filters

Publication year range
1.
Genes Dev ; 32(13-14): 929-943, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29950492

ABSTRACT

While a mutation in C9ORF72 is the most common genetic contributor to amyotrophic lateral sclerosis (ALS), much remains to be learned concerning the function of the protein normally encoded at this locus. To elaborate further on functions for C9ORF72, we used quantitative mass spectrometry-based proteomics to identify interacting proteins in motor neurons and found that its long isoform complexes with and stabilizes SMCR8, which further enables interaction with WDR41. To study the organismal and cellular functions for this tripartite complex, we generated Smcr8 loss-of-function mutant mice and found that they developed phenotypes also observed in C9orf72 loss-of-function animals, including autoimmunity. Along with a loss of tolerance for many nervous system autoantigens, we found increased lysosomal exocytosis in Smcr8 mutant macrophages. In addition to elevated surface Lamp1 (lysosome-associated membrane protein 1) expression, we also observed enhanced secretion of lysosomal components-phenotypes that we subsequently observed in C9orf72 loss-of-function macrophages. Overall, our findings demonstrate that C9ORF72 and SMCR8 have interdependent functions in suppressing autoimmunity as well as negatively regulating lysosomal exocytosis-processes of potential importance to ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Autoimmunity/genetics , Carrier Proteins/metabolism , Exocytosis/genetics , Lysosomes/metabolism , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Carrier Proteins/genetics , Gene Expression Regulation/genetics , Humans , Lymph Nodes/pathology , Lysosomal-Associated Membrane Protein 1/genetics , Macrophages/pathology , Mice , Mice, Knockout , Mutation , Protein Isoforms , Protein Stability , Splenomegaly/genetics
2.
J Neurosci ; 44(31)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38951039

ABSTRACT

The release of neurotransmitters (NTs) at central synapses is dependent on a cascade of protein interactions, specific to the presynaptic compartment. Among those dedicated molecules, the cytosolic complexins play an incompletely defined role as synaptic transmission regulators. Complexins are multidomain proteins that bind soluble N-ethylmaleimide sensitive factor attachment protein receptor complexes, conferring both inhibitory and stimulatory functions. Using systematic mutagenesis and comparing reconstituted in vitro membrane fusion assays with electrophysiology in cultured neurons from mice of either sex, we deciphered the function of the N-terminus of complexin (Cpx) II. The N-terminus (amino acid 1-27) starts with a region enriched in hydrophobic amino acids (1-12), which binds lipids. Mutants maintaining this hydrophobic character retained the stimulatory function of Cpx, whereas exchanges introducing charged residues perturbed both spontaneous and evoked exocytosis. Mutants in the more distal region of the N-terminal domain (amino acid 11-18) showed a spectrum of effects. On the one hand, mutation of residue A12 increased spontaneous release without affecting evoked release. On the other hand, replacing D15 with amino acids of different shapes or hydrophobic properties (but not charge) not only increased spontaneous release but also impaired evoked release. Most surprising, this substitution reduced the size of the readily releasable pool, a novel function for Cpx at mammalian synapses. Thus, the exact amino acid composition of the Cpx N-terminus fine-tunes the degree of spontaneous and evoked NT release.


Subject(s)
Nerve Tissue Proteins , Synaptic Vesicles , Animals , Synaptic Vesicles/metabolism , Synaptic Vesicles/genetics , Mice , Male , Female , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/chemistry , Mutation , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Membrane Fusion/physiology , Membrane Fusion/genetics , Cells, Cultured , Phenotype , Neurons/metabolism , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Mice, Inbred C57BL , Exocytosis/physiology , Exocytosis/genetics
3.
Plant Cell ; 34(10): 3961-3982, 2022 09 27.
Article in English | MEDLINE | ID: mdl-35766888

ABSTRACT

AP-1 and AP-2 adaptor protein (AP) complexes mediate clathrin-dependent trafficking at the trans-Golgi network (TGN) and the plasma membrane, respectively. Whereas AP-1 is required for trafficking to plasma membrane and vacuoles, AP-2 mediates endocytosis. These AP complexes consist of four subunits (adaptins): two large subunits (ß1 and γ for AP-1 and ß2 and α for AP-2), a medium subunit µ, and a small subunit σ. In general, adaptins are unique to each AP complex, with the exception of ß subunits that are shared by AP-1 and AP-2 in some invertebrates. Here, we show that the two putative Arabidopsis thaliana AP1/2ß adaptins co-assemble with both AP-1 and AP-2 subunits and regulate exocytosis and endocytosis in root cells, consistent with their dual localization at the TGN and plasma membrane. Deletion of both ß adaptins is lethal in plants. We identified a critical role of ß adaptins in pollen wall formation and reproduction, involving the regulation of membrane trafficking in the tapetum and pollen germination. In tapetal cells, ß adaptins localize almost exclusively to the TGN and mediate exocytosis of the plasma membrane transporters such as ATP-binding cassette (ABC)G9 and ABCG16. This study highlights the essential role of AP1/2ß adaptins in plants and their specialized roles in specific cell types.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Adaptor Protein Complex beta Subunits/metabolism , Adenosine Triphosphate/metabolism , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Clathrin/genetics , Clathrin/metabolism , Exocytosis/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Pollen/genetics , Pollen/metabolism , Transcription Factor AP-1/metabolism
4.
PLoS Genet ; 18(5): e1010194, 2022 05.
Article in English | MEDLINE | ID: mdl-35587496

ABSTRACT

In the ciliate Tetrahymena thermophila, lysosome-related organelles called mucocysts accumulate at the cell periphery where they secrete their contents in response to extracellular events, a phenomenon called regulated exocytosis. The molecular bases underlying regulated exocytosis have been extensively described in animals but it is not clear whether similar mechanisms exist in ciliates or their sister lineage, the Apicomplexan parasites, which together belong to the ecologically and medically important superphylum Alveolata. Beginning with a T. thermophila mutant in mucocyst exocytosis, we used a forward genetic approach to uncover MDL1 (Mucocyst Discharge with a LamG domain), a novel gene that is essential for regulated exocytosis of mucocysts. Mdl1p is a 40 kDa membrane glycoprotein that localizes to mucocysts, and specifically to a tip domain that contacts the plasma membrane when the mucocyst is docked. This sub-localization of Mdl1p, which occurs prior to docking, underscores a functional asymmetry in mucocysts that is strikingly similar to that of highly polarized secretory organelles in other Alveolates. A mis-sense mutation in the LamG domain results in mucocysts that dock but only undergo inefficient exocytosis. In contrast, complete knockout of MDL1 largely prevents mucocyst docking itself. Mdl1p is physically associated with 9 other proteins, all of them novel and largely restricted to Alveolates, and sedimentation analysis supports the idea that they form a large complex. Analysis of three other members of this putative complex, called MDD (for Mucocyst Docking and Discharge), shows that they also localize to mucocysts. Negative staining of purified MDD complexes revealed distinct particles with a central channel. Our results uncover a novel macromolecular complex whose subunits are conserved within alveolates but not in other lineages, that is essential for regulated exocytosis in T. thermophila.


Subject(s)
Tetrahymena thermophila , Tetrahymena , Animals , Exocytosis/genetics , Lysosomes/metabolism , Organelles/metabolism , Secretory Vesicles/genetics , Secretory Vesicles/metabolism , Tetrahymena thermophila/genetics
5.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33431696

ABSTRACT

Neurotransmitter release occurs by regulated exocytosis from synaptic vesicles (SVs). Evolutionarily conserved proteins mediate the essential aspects of this process, including the membrane fusion step and priming steps that make SVs release-competent. Unlike the proteins constituting the core fusion machinery, the SV protein Mover does not occur in all species and all synapses. Its restricted expression suggests that Mover may modulate basic aspects of transmitter release and short-term plasticity. To test this hypothesis, we analyzed synaptic transmission electrophysiologically at the mouse calyx of Held synapse in slices obtained from wild-type mice and mice lacking Mover. Spontaneous transmission was unaffected, indicating that the basic release machinery works in the absence of Mover. Evoked release and vesicular release probability were slightly reduced, and the paired pulse ratio was increased in Mover knockout mice. To explore whether Mover's role is restricted to certain subpools of SVs, we analyzed our data in terms of two models of priming. A model assuming two SV pools in parallel showed a reduced release probability of so-called "superprimed vesicles" while "normally primed" ones were unaffected. For the second model, which holds that vesicles transit sequentially from a loosely docked state to a tightly docked state before exocytosis, we found that knocking out Mover selectively decreased the release probability of tight state vesicles. These results indicate that Mover regulates a subclass of primed SVs in the mouse calyx of Held.


Subject(s)
Exocytosis/genetics , Nerve Tissue Proteins/genetics , Synaptic Transmission/genetics , Synaptic Vesicles/genetics , Animals , Brain Stem/metabolism , Brain Stem/physiology , Calcium/metabolism , Excitatory Postsynaptic Potentials , Humans , Membrane Fusion/genetics , Membrane Fusion/physiology , Mice , Mice, Knockout , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Synapses/genetics , Synapses/metabolism , Synapses/physiology , Synaptic Vesicles/metabolism , Synaptic Vesicles/physiology
6.
J Assist Reprod Genet ; 41(1): 49-61, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37993579

ABSTRACT

PURPOSE: Patients with polycystic ovarian morphology (PCOM) make up 20% cases for assisted reproductive technology (ART). Folliculogenesis is impaired in PCOS. Signaling molecules are involved in follicle development. Dysregulations of intrafollicular environment and signaling molecules are observed in PCOS. Granulosa cells (GCs) and oocytes secrete molecules into follicular fluid by exocytosis of SNAREs. The aim of this study is to evaluate vesicle transport and vesicle fusion proteins (SNAREs) in GCs from PCOS patients who have undergone IVF treatment. METHODS: Follicular fluids were collected from patients who undergo IVF/ICSI with the diagnosis of male factor (n = 10) and PCOS (n = 10) patients. GCs were separated and cultured. Each group of GCs was stimulated with FSH-hCG. The cells were examined under electron microscope. Immunofluorescent labeling was performed on cells for Stx6, SNAP25, StxBP1, FSHr, and KITL. Integrated density was analyzed from images of Stx6, SNAP25, StxBP1, FSHr, and KITL. RESULTS: Intercellular communication occurs by signal molecules; Stx6, SNAP25, and StxBP1 fusion proteins involved in exocytosis were decreased in the GCs of PCOS. There was no increase in in vitro stimulation with FSH-hCG either. In the electron microscope, it was observed that exocytosis of the vesicles was disrupted. CONCLUSIONS: Exocytosis and vesicular dynamics are among the basic physiological functions of human steroidogenic granulosa cells. Follicle development is necessary for production of competent oocytes and ovulation. Understanding the pathophysiology of PCOS at follicular level is important for disease management. According to our findings, deficits in vesicular dynamics of human granulosa cells in may be central to the treatment strategy for PCOS patients.


Subject(s)
Polycystic Ovary Syndrome , Female , Humans , Male , Granulosa Cells/metabolism , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , Exocytosis/genetics , Cell Communication
7.
Hum Genet ; 142(8): 1263-1270, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37085629

ABSTRACT

Exocytosis is the process by which secretory vesicles fuse with the plasma membrane to deliver materials to the cell surface or to release cargoes to the extracellular space. The exocyst-an evolutionarily conserved octameric protein complex-mediates spatiotemporal control of SNARE complex assembly for vesicle fusion and tethering the secretory vesicles to the plasma membrane. The exocyst participates in diverse cellular functions, including protein trafficking to the plasma membrane, membrane extension, cell polarity, neurite outgrowth, ciliogenesis, cytokinesis, cell migration, autophagy, host defense, and tumorigenesis. Exocyst subunits are essential for cell viability; and mutations or variants in several exocyst subunits have been implicated in human diseases, mostly neurodevelopmental disorders and ciliopathies. These conditions often share common features such as developmental delay, intellectual disability, and brain abnormalities. In this review, we summarize the mutations and variants in exocyst subunits that have been linked to disease and discuss the implications of exocyst dysfunction in other disorders.


Subject(s)
Nervous System Diseases , Vesicular Transport Proteins , Humans , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Cytoplasm/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Exocytosis/genetics , Nervous System Diseases/genetics
8.
PLoS Pathog ; 17(4): e1009551, 2021 04.
Article in English | MEDLINE | ID: mdl-33909710

ABSTRACT

Lipid transfer proteins (LTPs) are the key contributor of organelle-specific lipid distribution and cellular lipid homeostasis. Here, we report a novel implication of LTPs in phagocytosis, trogocytosis, pinocytosis, biosynthetic secretion, recycling of pinosomes, and motility of the parasitic protist E. histolytica, the etiological agent of human amoebiasis. We show that two StAR-related lipid transfer (START) domain-containing LTPs (named as EhLTP1 and 3) are involved in these biological pathways in an LTP-specific manner. Our findings provide novel implications of LTPs, which are relevant to the elucidation of pathophysiology of the diseases caused by parasitic protists.


Subject(s)
Carrier Proteins/physiology , Endocytosis/genetics , Entamoeba histolytica/physiology , Exocytosis/genetics , Animals , CHO Cells , Cell Movement/genetics , Cricetulus , Entamoeba histolytica/genetics , Entamoeba histolytica/metabolism , Entamoebiasis/genetics , Entamoebiasis/metabolism , Entamoebiasis/parasitology , Membrane Transport Proteins/physiology , Metabolic Networks and Pathways/genetics , Organisms, Genetically Modified , Phagocytosis/genetics , Phosphoproteins/chemistry
9.
Proc Natl Acad Sci U S A ; 117(40): 25150-25158, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32968023

ABSTRACT

The plasma membrane (PM) acts as the interface between intra- and extracellular environments and exhibits a tightly regulated molecular composition. The composition and amount of PM proteins are regulated by balancing endocytic and exocytic trafficking in a cargo-specific manner, according to the demands of specific cellular states and developmental processes. In plant cells, retrieval of membrane proteins from the PM depends largely on clathrin-mediated endocytosis (CME). However, the mechanisms for sorting PM proteins during CME remain ambiguous. In this study, we identified a homologous pair of ANTH domain-containing proteins, PICALM1a and PICALM1b, as adaptor proteins for CME of the secretory vesicle-associated longin-type R-SNARE VAMP72 group. PICALM1 interacted with the SNARE domain of VAMP72 and clathrin at the PM. The loss of function of PICALM1 resulted in faulty retrieval of VAMP72, whereas general endocytosis was not considerably affected by this mutation. The double mutant of PICALM1 exhibited impaired vegetative development, indicating the requirement of VAMP72 recycling for normal plant growth. In the mammalian system, VAMP7, which is homologous to plant VAMP72, is retrieved from the PM via the interaction with a clathrin adaptor HIV Rev-binding protein in the longin domain during CME, which is not functional in the plant system, whereas retrieval of brevin-type R-SNARE members is dependent on a PICALM1 homolog. These results indicate that ANTH domain-containing proteins have evolved to be recruited distinctly for recycling R-SNARE proteins and are critical to eukaryote physiology.


Subject(s)
Endocytosis/genetics , Membrane Proteins/genetics , Protein Transport/genetics , R-SNARE Proteins/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Cell Membrane/genetics , Clathrin/metabolism , Eukaryota/genetics , Exocytosis/genetics , Gene Expression Regulation, Plant/genetics , Plant Cells/metabolism , Protein Domains/genetics
10.
J Neurosci ; 41(16): 3563-3578, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33664131

ABSTRACT

Synaptophysin (syp) is a major integral membrane protein of secretory vesicles. Previous work has demonstrated functions for syp in synaptic vesicle cycling, endocytosis, and synaptic plasticity, but the role of syp in the process of membrane fusion during Ca2+-triggered exocytosis remains poorly understood. Furthermore, although syp resides on both large dense-core and small synaptic vesicles, its role in dense-core vesicle function has received less attention compared with synaptic vesicle function. To explore the role of syp in membrane fusion and dense-core vesicle function, we used amperometry to measure catecholamine release from single vesicles in male and female mouse chromaffin cells with altered levels of syp and the related tetraspanner protein synaptogyrin (syg). Knocking out syp slightly reduced the frequency of vesicle fusion events below wild-type (WT) levels, but knocking out both syp and syg reduced the frequency 2-fold. Knocking out both proteins stabilized initial fusion pores, promoted fusion pore closure (kiss-and-run), and reduced late-stage fusion pore expansion. Introduction of a syp construct lacking its C-terminal dynamin-binding domain in syp knock-outs (KOs) increased the duration and fraction of kiss-and-run events, increased total catecholamine release per event, and reduced late-stage fusion pore expansion. These results demonstrated that syp and syg regulate dense-core vesicle function at multiple stages to initiate fusion, control the choice of mode between full-fusion and kiss-and-run, and influence the dynamics of both initial and late-stage fusion pores. The transmembrane domain (TMD) influences small initial fusion pores, and the C-terminal domain influences large late-stage fusion pores, possibly through an interaction with dynamin.SIGNIFICANCE STATEMENT The secretory vesicle protein synaptophysin (syp) is known to function in synaptic vesicle cycling, but its roles in dense-core vesicle functions, and in controlling membrane fusion during Ca2+-triggered exocytosis remain unclear. The present study used amperometry recording of catecholamine release from endocrine cells to assess the impact of syp and related proteins on membrane fusion. A detailed analysis of amperometric spikes arising from the exocytosis of single vesicles showed that these proteins influence fusion pores at multiple stages and control the choice between kiss-and-run and full-fusion. Experiments with a syp construct lacking its C terminus indicated that the transmembrane domain (TMD) influences the initial fusion pore, while the C-terminal domain influences later stages after fusion pore expansion.


Subject(s)
Chromaffin Cells/physiology , Exocytosis/physiology , Synaptophysin/physiology , Animals , Animals, Newborn , Catecholamines/metabolism , Dynamins/metabolism , Dynamins/physiology , Electrophysiological Phenomena , Exocytosis/genetics , Female , Membrane Fusion , Mice , Mice, Knockout , Pregnancy , Primary Cell Culture , Synaptic Vesicles/metabolism , Synaptic Vesicles/physiology , Synaptogyrins/genetics , Synaptogyrins/physiology , Synaptophysin/genetics
11.
J Biol Chem ; 296: 100268, 2021.
Article in English | MEDLINE | ID: mdl-33837726

ABSTRACT

Degranulation, a fundamental effector response from mast cells (MCs) and platelets, is an example of regulated exocytosis. This process is mediated by SNARE proteins and their regulators. We have previously shown that several of these proteins are essential for exocytosis in MCs and platelets. Here, we assessed the role of the SNARE protein SNAP23 using conditional knockout mice, in which SNAP23 was selectively deleted from either the megakaryocyte/platelet or connective tissue MC lineages. We found that removal of SNAP23 in platelets results in severe defects in degranulation of all three platelet secretory granule types, i.e., alpha, dense, and lysosomal granules. The mutation also induces thrombocytopenia, abnormal platelet morphology and activation, and reduction in the number of alpha granules. Therefore, the degranulation defect might not be secondary to an intrinsic failure of the machinery mediating regulated exocytosis in platelets. When we removed SNAP23 expression in MCs, there was a complete developmental failure in vitro and in vivo. The developmental defects in platelets and MCs and the abnormal translocation of membrane proteins to the surface of platelets indicate that SNAP23 is also involved in constitutive exocytosis in these cells. The MC conditional deletant animals lacked connective tissue MCs, but their mucosal MCs were normal and expanded in response to an antigenic stimulus. We used this mouse to show that connective tissue MCs are required and mucosal MCs are not sufficient for an anaphylactic response.


Subject(s)
Anaphylaxis/immunology , Blood Platelets/immunology , Connective Tissue/immunology , Mast Cells/immunology , Qb-SNARE Proteins/immunology , Qc-SNARE Proteins/immunology , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Blood Platelets/pathology , Connective Tissue/pathology , Exocytosis/genetics , Exocytosis/immunology , Mast Cells/pathology , Mice , Mice, Knockout , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , Secretory Vesicles/genetics , Secretory Vesicles/immunology
12.
Plant J ; 106(3): 831-843, 2021 05.
Article in English | MEDLINE | ID: mdl-33599020

ABSTRACT

Spatially directed cell division and expansion is important for plant growth and morphogenesis and relies on cooperation between the cytoskeleton and the secretory pathway. The phylogenetically conserved octameric complex exocyst mediates exocytotic vesicle tethering at the plasma membrane. Unlike other exocyst subunits of land plants, the core exocyst subunit SEC6 exists as a single paralog in Physcomitrium patens and Arabidopsis thaliana genomes. Arabidopsis SEC6 (AtSEC6) loss-of-function (LOF) mutation causes male gametophytic lethality. Our attempts to inactivate the P. patens SEC6 gene, PpSEC6, using targeted gene replacement produced two independent partial LOF ('weak allele') mutants via perturbation of the PpSEC6 gene locus. These mutants exhibited the same pleiotropic developmental defects: protonema with dominant chloronema stage; diminished caulonemal filament elongation rate; and failure in post-initiation gametophore development. Mutant gametophore buds, mostly initiated from chloronema cells, exhibited disordered cell file organization and cross-wall perforations, resulting in arrested development at the eight- to 10-cell stage. Complementation of both sec6 moss mutant lines by both PpSEC6 and AtSEC6 cDNA rescued gametophore development, including sexual organ differentiation. However, regular sporophyte formation and viable spore production were recovered only by the expression of PpSEC6, whereas the AtSEC6 complementants were only rarely fertile, indicating moss-specific SEC6 functions.


Subject(s)
Bryopsida/growth & development , Exocytosis , Plant Proteins/physiology , Bryopsida/genetics , Exocytosis/genetics , Genes, Plant/genetics , Germ Cells, Plant , Mutation , Plant Proteins/genetics
13.
Cell Mol Life Sci ; 78(4): 1689-1708, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32734583

ABSTRACT

OSBP-homologous proteins (ORPs, Oshp) are lipid binding/transfer proteins. Several ORP/Oshp localize to membrane contacts between the endoplasmic reticulum (ER) and the plasma membrane, where they mediate lipid transfer or regulate lipid-modifying enzymes. A common way in which they target contacts is by binding to the ER proteins, VAP/Scs2p, while the second membrane is targeted by other interactions with lipids or proteins.We have studied the cross-talk of secretory SNARE proteins and their regulators with ORP/Oshp and VAPA/Scs2p at ER-plasma membrane contact sites in yeast and murine primary neurons. We show that Oshp-Scs2p interactions depend on intact secretory SNARE proteins, especially Sec9p. SNAP-25/Sec9p directly interact with ORP/Osh proteins and their disruption destabilized the ORP/Osh proteins, associated with dysfunction of VAPA/Scs2p. Deleting OSH1-3 in yeast or knocking down ORP2 in primary neurons reduced the oligomerization of VAPA/Scs2p and affected their multiple interactions with SNAREs. These observations reveal a novel cross-talk between the machineries of ER-plasma membrane contact sites and those driving exocytosis.


Subject(s)
Carrier Proteins/genetics , Endoplasmic Reticulum/genetics , Membrane Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Vesicular Transport Proteins/genetics , Animals , Biological Transport/genetics , Carrier Proteins/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Exocytosis/genetics , Humans , Lipid Metabolism/genetics , Mice , Qc-SNARE Proteins/genetics , Receptors, Steroid/genetics , SNARE Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sterols/metabolism , Synaptosomal-Associated Protein 25/genetics
14.
Proc Natl Acad Sci U S A ; 116(28): 13952-13957, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31235584

ABSTRACT

R-SNAREs (soluble N-ethylmaleimide-sensitive factor receptor), Q-SNAREs, and Sec1/Munc18 (SM)-family proteins are essential for membrane fusion in exocytic and endocytic trafficking. The yeast vacuolar tethering/SM complex HOPS (homotypic fusion and vacuole protein sorting) increases the fusion of membranes bearing R-SNARE to those with 3Q-SNAREs far more than it enhances their trans-SNARE pairings. We now report that the fusion of these proteoliposomes is also supported by GST-PX or GST-FYVE, recombinant dimeric proteins which tether by binding the phosphoinositides in both membranes. GST-PX is purely a tether, as it supports fusion without SNARE recognition. GST-PX tethering supports the assembly of new, active SNARE complexes rather than enhancing the function of the fusion-inactive SNARE complexes which had spontaneously formed in the absence of a tether. When SNAREs are more disassembled, as by Sec17, Sec18, and ATP (adenosine triphosphate), HOPS is required, and GST-PX does not suffice. We propose a working model where tethering orients SNARE domains for parallel, active assembly.


Subject(s)
Adenosine Triphosphatases/chemistry , Glutathione Peroxidase/chemistry , Membrane Fusion Proteins/chemistry , R-SNARE Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Vesicular Transport Proteins/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/chemistry , Endocytosis/genetics , Exocytosis/genetics , Glutathione Peroxidase/genetics , Membrane Fusion/genetics , Membrane Fusion Proteins/genetics , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Protein Multimerization/genetics , Protein Transport/genetics , R-SNARE Proteins/genetics , Recombinant Proteins/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/genetics , Vacuoles/chemistry , Vacuoles/genetics , Vesicular Transport Proteins/genetics
15.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232740

ABSTRACT

The nerve growth factor (NGF) and calcitonin gene-related peptide (CGRP) play a crucial role in the regulation of orofacial pain. It has been demonstrated that CGRP increases orofacial pain induced by NGF. V-type proton ATPase subunit an isoform 1 (Atp6v0a1) is involved in the exocytosis pathway, especially in vesicular transport in neurons. The objective was to examine the role of Atp6v0a1 in NGF-induced upregulation of CGRP in orofacial pain induced by experimental tooth movement. Orofacial pain was elicited by ligating closed-coil springs between incisors and molars in Sprague-Dawley rats. Gene and protein expression levels were determined through real-time polymerase chain reaction, immunostaining, and fluorescence in situ hybridization. Lentivirus vectors carrying Atp6v0a1 shRNA were used to knockdown the expression of Atp6v0a1 in TG and SH-SY5Y neurons. The release of vesicles in SH-SY5Y neurons was observed by using fluorescence dye FM1-43, and the release of CGRP was detected by Enzyme-Linked Immunosorbent Assy. Orofacial pain was evaluated through the rat grimace scale. Our results revealed that intraganglionic administration of NGF and Atp6v0a1 shRNA upregulated and downregulated CGRP in trigeminal ganglia (TG) and trigeminal subnucleus caudalis (Vc), respectively, and the orofacial pain was also exacerbated and alleviated, respectively, following administration of NGF and Atp6v0a1 shRNA. Besides, intraganglionic administration of NGF simultaneously caused the downregulation of Atp6v0a1 in TG. Moreover, the release of vesicles and CGRP in SH-SY5Y neurons was interfered by NGF and Atp6v0a1 shRNA. In conclusion, in the orofacial pain induced by experimental tooth movement, NGF induced the upregulation of CGRP in TG and Vc, and this process is dependent on Atp6v0a1 and vesicle release, suggesting that they are involved in the transmission of nociceptive information in orofacial pain.


Subject(s)
Calcitonin Gene-Related Peptide , Facial Pain , Nerve Growth Factor , Tooth Movement Techniques , Vacuolar Proton-Translocating ATPases , Adenosine Triphosphatases/metabolism , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Exocytosis/genetics , Exocytosis/physiology , Facial Pain/etiology , Facial Pain/genetics , Facial Pain/metabolism , Immunosorbents , In Situ Hybridization, Fluorescence , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Neuroblastoma , Neurons/metabolism , Nociception/physiology , Protons , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Tooth Movement Techniques/methods , Up-Regulation , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
16.
Genes Cells ; 25(6): 391-401, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32167217

ABSTRACT

Vesicular transport serves as an important mechanism for cell shape regulation during development. Although the semaphorin signaling molecule, a well-known regulator of axon guidance, induces endocytosis in the growth cone and the axonal transport of vertebrate neurons, the underlying molecular mechanisms remain largely unclear. Here, we show that the Caenorhabditis elegans SNT-1/synaptotagmin-UNC-41/stonin2 system, whose role in synaptic vesicle recycling in neurons has been studied extensively, is involved in semaphorin-regulated vesicular transport in larval epidermal cells. Mutations in the snt-1/unc-41 genes strongly suppressed the cell shape defects of semaphorin mutants. The null mutation in the semaphorin receptor gene, plx-1, altered the expression and localization pattern of endocytic and exocytic markers in the epidermal cells while repressing the transport of SNT-1-containing vesicles toward late endosome/lysosome pathways. Our findings suggest that the nematode semaphorins regulate the vesicular transport in epidermal cells in a manner distinct from that of vertebrate semaphorins in neurons.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Epidermal Cells/metabolism , Semaphorins/metabolism , Synaptic Vesicles/metabolism , Synaptotagmins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Animals, Genetically Modified , Biological Transport, Active/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Endocytosis/genetics , Endosomes/genetics , Endosomes/metabolism , Exocytosis/genetics , Larva/growth & development , Larva/metabolism , Lysosomes/genetics , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA Interference , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Semaphorins/genetics , Signal Transduction/genetics , Synaptotagmins/genetics , Vesicular Transport Proteins/genetics
17.
Plant Physiol ; 184(4): 1823-1839, 2020 12.
Article in English | MEDLINE | ID: mdl-33051268

ABSTRACT

Pollen development, pollen grain germination, and pollen tube elongation are crucial biological processes in angiosperm plants that need precise regulation to deliver sperm cells to ovules for fertilization. Highly polarized secretion at a growing pollen tube tip requires the exocyst tethering complex responsible for specific targeting of secretory vesicles to the plasma membrane. Here, we demonstrate that Arabidopsis (Arabidopsis thaliana) EXO70A2 (At5g52340) is the main exocyst EXO70 isoform in the male gametophyte, governing the conventional secretory function of the exocyst, analogous to EXO70A1 (At5g03540) in the sporophyte. Our analysis of a CRISPR-generated exo70a2 mutant revealed that EXO70A2 is essential for efficient pollen maturation, pollen grain germination, and pollen tube growth. GFP:EXO70A2 was localized to the nucleus and cytoplasm in developing pollen grains and later to the apical domain in growing pollen tube tips characterized by intensive exocytosis. Moreover, EXO70A2 could substitute for EXO70A1 function in the sporophyte, but not vice versa, indicating partial functional redundancy of these two closely related isoforms and higher specificity of EXO70A2 for pollen development-related processes. Phylogenetic analysis revealed that the ancient duplication of EXO70A, one of which is always highly expressed in pollen, occurred independently in monocots and dicots. In summary, EXO70A2 is a crucial component of the exocyst complex in Arabidopsis pollen that is required for efficient plant sexual reproduction.


Subject(s)
Arabidopsis/growth & development , Arabidopsis/genetics , Exocytosis/genetics , Exocytosis/physiology , Pollen Tube/growth & development , Pollen Tube/genetics , Gene Expression Regulation, Plant , Genes, Plant , Genetic Variation , Genotype , Phylogeny
18.
Mol Cell Biochem ; 476(10): 3745-3756, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34100174

ABSTRACT

Sepsis is one of the leading causes of morbidity and mortality and a major cause of acute lung injury (ALI). carried by exosomes play a role in a variety of diseases. However,there are not many studies of exosomal miRNAs in sepsis and sepsis lung injury.miR-1298-5p and suppressor of cytokine signaling 6 (SOCS6) were silenced or overexpressed in human bronchial epithelial cells (BEAS-2B). PKH-67 Dye was used to trace exosome endocytosis. Cell permeability was evaluated by measuring trans-epithelial electrical resistance (TEER) and FITC dextran flux. ELISA kits were used for cytokine detection. Quantitative RT-PCR and western blots were used to evaluate gene expression. miR-1298-5p was elevated in exosomes from patients with sepsis lung injury (Sepsis_exo). Treatment of BEAS-2B cells using Sepsis_exo significantly inhibited cell proliferation, and induced cell permeability and inflammatory response. miR-1298-5p directly targeted SOCS6. Overexpressing SOCS6 reversed miR-1298-5p-induced cell permeability and inflammatory response. Inhibition of STAT3 blocked SOCS6-silencing caused significant increase of cell permeability and inflammation. Exosomes isolated from patients of sepsis lung injury increased cell permeability and inflammatory response in BEAS-2B cells through exosomal miR-1298-5p which targeted SOCS6 via STAT3 pathway. The findings highlight the importance of miR-1298-5p/SOCS6/STAT3 axis in sepsis lung injury and provide new insights into therapeutic strategies for sepsis lung injury.


Subject(s)
Acute Lung Injury/metabolism , MicroRNAs/metabolism , Sepsis/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Acute Lung Injury/genetics , Cell Line , Exocytosis/genetics , Exosomes/metabolism , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Sepsis/genetics , Suppressor of Cytokine Signaling Proteins/genetics
19.
PLoS Genet ; 14(6): e1007432, 2018 06.
Article in English | MEDLINE | ID: mdl-29912942

ABSTRACT

Axonal growth and guidance rely on correct growth cone responses to guidance cues. Unlike the signaling cascades that link axonal growth to cytoskeletal dynamics, little is known about the crosstalk mechanisms between guidance and membrane dynamics and turnover. Recent studies indicate that whereas axonal attraction requires exocytosis, chemorepulsion relies on endocytosis. Indeed, our own studies have shown that Netrin-1/Deleted in Colorectal Cancer (DCC) signaling triggers exocytosis through the SNARE Syntaxin-1 (STX1). However, limited in vivo evidence is available about the role of SNARE proteins in axonal guidance. To address this issue, here we systematically deleted SNARE genes in three species. We show that loss-of-function of STX1 results in pre- and post-commissural axonal guidance defects in the midline of fly, chick, and mouse embryos. Inactivation of VAMP2, Ti-VAMP, and SNAP25 led to additional abnormalities in axonal guidance. We also confirmed that STX1 loss-of-function results in reduced sensitivity of commissural axons to Slit-2 and Netrin-1. Finally, genetic interaction studies in Drosophila show that STX1 interacts with both the Netrin-1/DCC and Robo/Slit pathways. Our data provide evidence of an evolutionarily conserved role of STX1 and SNARE proteins in midline axonal guidance in vivo, by regulating both pre- and post-commissural guidance mechanisms.


Subject(s)
Neurogenesis/genetics , Syntaxin 1/genetics , Syntaxin 1/physiology , Animals , Axons/metabolism , Chemotaxis/genetics , Chick Embryo , Drosophila/genetics , Drosophila Proteins/genetics , Exocytosis/genetics , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Netrin-1/genetics , Netrin-1/metabolism , Neurogenesis/physiology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , SNARE Proteins/genetics , SNARE Proteins/metabolism , Signal Transduction/genetics , Spinal Cord/embryology , Spinal Cord/metabolism
20.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203249

ABSTRACT

By providing ~70% of the eye's refractive power, the preocular tear film is essential for optimal vision. However, its integrity is often jeopardized by environmental and pathologic conditions that accelerate evaporation and cause sight-impairing dry eye. A key adaptive response to evaporation-induced tear film hyperosmolarity is the reflex-triggered release of tear-stabilizing mucin from conjunctival goblet cells. Here, we review progress in elucidating the roles of ion channels in mediating this important exocytotic response. Much is now known about the modulatory impact of ATP-sensitive potassium channels, nonspecific cation channels and voltage-gated calcium channels. Recently, we discovered that during unremitting extracellular hyperosmolarity, P2X7 receptor/channels also become activated and markedly impair goblet cell viability. However, our understanding of possible adaptive benefits of this P2X7 activation remains limited. In the present study, we utilized high-temporal resolution membrane capacitance measurements to monitor the exocytotic activity of single goblet cells located in freshly excised rat conjunctiva. We now report that activation of P2X7 purinoceptors boosts neural-evoked exocytosis and accelerates replenishment of mucin-filled granules after exocytotic depletion. Thus, P2X7 activation exerts a yin-yang effect on conjunctival goblet cells: the high-gain benefit of enhancing the supply of tear-stabilizing mucin is implemented at the high-risk of endangering goblet cell survival.


Subject(s)
Goblet Cells/metabolism , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X/metabolism , Animals , Cell Survival/genetics , Cell Survival/physiology , Exocytosis/genetics , Exocytosis/physiology , Humans , Receptors, Purinergic P2X/genetics
SELECTION OF CITATIONS
SEARCH DETAIL