Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 19(11): e1011029, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38011217

ABSTRACT

Mammalian evolution has been influenced by viruses for millions of years, leaving signatures of adaptive evolution within genes encoding for viral interacting proteins. Synaptogyrin-2 (SYNGR2) is a transmembrane protein implicated in promoting bacterial and viral infections. A genome-wide association study of pigs experimentally infected with porcine circovirus type 2b (PCV2b) uncovered a missense mutation (SYNGR2 p.Arg63Cys) associated with viral load. In this study, CRISPR/Cas9-mediated gene editing of the porcine kidney 15 (PK15, wtSYNGR2+p.63Arg) cell line generated clones homozygous for the favorable SYNGR2 p.63Cys allele (emSYNGR2+p.63Cys). Infection of edited clones resulted in decreased PCV2 replication compared to wildtype PK15 (P<0.05), with consistent effects across genetically distinct PCV2b and PCV2d isolates. Sequence analyses of wild and domestic pigs (n>700) revealed the favorable SYNGR2 p.63Cys allele is unique to domestic pigs and more predominant in European than Asian breeds. A haplotype defined by the SYNGR2 p.63Cys allele was likely derived from an ancestral haplotype nearly fixed within European (0.977) but absent from Asian wild boar. We hypothesize that the SYNGR2 p.63Cys allele arose post-domestication in ancestral European swine. Decreased genetic diversity in homozygotes for the SYNGR2 p.63Cys allele compared to SYNGR2 p.63Arg, corroborates a rapid increase in frequency of SYGNR2 p.63Cys via positive selection. Signatures of adaptive evolution across mammalian species were also identified within SYNGR2 intraluminal loop domains, coinciding with the location of SYNGR2 p.Arg63Cys. Therefore, SYNGR2 may reflect a novel component of the host-virus evolutionary arms race across mammals with SYNGR2 p.Arg63Cys representing a species-specific example of putative adaptive evolution.


Subject(s)
Circovirus , Swine Diseases , Swine/genetics , Animals , Circovirus/genetics , Synaptogyrins/genetics , Genome-Wide Association Study , Swine Diseases/genetics , Genotype , Sus scrofa/genetics
2.
Brain Behav ; 13(2): e2886, 2023 02.
Article in English | MEDLINE | ID: mdl-36624932

ABSTRACT

BACKGROUND: Leucine-rich repeat kinase 2 (LRRK2) mutation is a common genetic risk factor of Parkinson's disease (PD). Presynaptic dysfunction is an early pathogenic event associated with dopamine (DA) dysregulation in striatum of the brain. DA uptake activity of DA uptake transporter (DAT) affects synaptic plasticity and motor and non-motor behavior. Synaptogyrin-3 (SYNGR3) is part of the synaptogyrin family, especially abundant in brain. Previous in vitro studies demonstrated interaction between SYNGR3 and DAT. Reduced SYNGR3 expression was observed in human PD brains with unclear reasons. METHODS: Here, we further explored whether inducing SYNGR3 expression can influence (i) cellular DA uptake using differentiated human SH-SY5Y neuronal cells, (ii) striatal synaptosomal DA uptake in a mutant LRRK2R1441G  knockin mouse model of PD, and (iii) innate rodent behavior using the marble burying test. RESULTS: Young LRRK2 mutant mice exhibited significantly lower SYNGR3 levels in striatum compared to age-matched wild-type (WT) controls, resembling level in aged WT mice. SYNGR3 is spatially co-localized with DAT at striatal presynaptic terminals, visualized by immuno-gold transmission electron microscopy and immunohistochemistry. Their protein-protein interaction was confirmed by co-immunoprecipitation. Transient overexpression of SYNGR3 in differentiated SH-SY5Y cells increased cellular DA uptake activity without affecting total DAT levels. Inducing SYNGR3 overexpression by adeno-associated virus-7 (AAV7) injection in vivo into striatum increased ex vivo synaptosomal DA uptake in LRRK2 mutant mice and improved their innate marble burying behavior. CONCLUSION: Brain SYNGR3 expression may be an important determinant to striatal DA homeostasis and synaptic function. Our preliminary behavioral test showed improved innate behavior after SYNGR3 overexpression in LRRK2 mutant mice, advocating further studies to determine the influence of SYNGR3 in the pathophysiology of DA neurons in PD.


Subject(s)
Neuroblastoma , Parkinson Disease , Aged , Animals , Humans , Mice , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mutation , Synaptogyrins/genetics , Synaptogyrins/metabolism
3.
Int J Mol Sci ; 23(7)2022 Mar 26.
Article in English | MEDLINE | ID: mdl-35409005

ABSTRACT

Synaptogyrin-3 (SYNGR3) is a synaptic vesicular membrane protein. Amongst four homologues (SYNGR1 to 4), SYNGR1 and 3 are especially abundant in the brain. SYNGR3 interacts with the dopamine transporter (DAT) to facilitate dopamine (DA) uptake and synaptic DA turnover in dopaminergic transmission. Perturbed SYNGR3 expression is observed in Parkinson's disease (PD). The regulatory elements which affect SYNGR3 expression are unknown. Nuclear-receptor-related-1 protein (NURR1) can regulate dopaminergic neuronal differentiation and maintenance via binding to NGFI-B response elements (NBRE). We explored whether NURR1 can regulate SYNGR3 expression using an in silico analysis of the 5'-flanking region of the human SYNGR3 gene, reporter gene activity and an electrophoretic mobility shift assay (EMSA) of potential cis-acting sites. In silico analysis of two genomic DNA segments (1870 bp 5'-flanking region and 1870 + 159 bp of first exon) revealed one X Core Promoter Element 1 (XCPE1), two SP1, and three potential non-canonical NBRE response elements (ncNBRE) but no CAAT or TATA box. The longer segment exhibited gene promoter activity in luciferase reporter assays. Site-directed mutagenesis of XCPE1 decreased promoter activity in human neuroblastoma SH-SY5Y (↓43.2%) and human embryonic kidney HEK293 cells (↓39.7%). EMSA demonstrated NURR1 binding to these three ncNBRE. Site-directed mutagenesis of these ncNBRE reduced promoter activity by 11-17% in SH-SY5Y (neuronal) but not in HEK293 (non-neuronal) cells. C-DIM12 (Nurr1 activator) increased SYNGR3 protein expression in SH-SY5Y cells and its promoter activity using a real-time luciferase assay. As perturbed vesicular function is a feature of major neurodegenerative diseases, inducing SYNGR3 expression by NURR1 activators may be a potential therapeutic target to attenuate synaptic dysfunction in PD.


Subject(s)
Synaptic Vesicles , Transcription Factors , Gene Expression Regulation , HEK293 Cells , Humans , Luciferases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Synaptic Vesicles/metabolism , Synaptogyrins/genetics , Synaptogyrins/metabolism , Transcription Factors/metabolism
4.
Front Immunol ; 12: 713611, 2021.
Article in English | MEDLINE | ID: mdl-34867944

ABSTRACT

Background: Methotrexate (MTX) is the first line treatment of rheumatoid arthritis (RA), and methylation changes in bulk T cells have been reported after treatment with MTX. We have investigated cell-type specific DNA methylation changes across the genome in naïve and memory CD4+ T cells before and after MTX treatment of RA patients. DNA methylation profiles of newly diagnosed RA patients (N=9) were assessed by reduced representation bisulfite sequencing. Results: We found that MTX treatment significantly influenced DNA methylation levels at multiple CpG sites in both cell populations. Interestingly, we identified differentially methylated sites annotated to two genes; TRIM15 and SORC2, previously reported to predict treatment outcome in RA patients when measured in bulk T cells. Furthermore, several of the genes, including STAT3, annotated to the significant CpG sites are relevant for RA susceptibility or the action of MTX. Conclusion: We detected CpG sites that were associated with MTX treatment in CD4+ naïve and memory T cells isolated from RA patients. Several of these sites overlap genetic regions previously associated with RA risk and MTX treatment outcome.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , DNA Methylation/drug effects , Methotrexate/therapeutic use , Adult , Aged , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CpG Islands , DNA-Binding Proteins/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Memory T Cells/drug effects , Memory T Cells/immunology , Methotrexate/pharmacology , Middle Aged , Receptors, CCR6/genetics , Receptors, Cell Surface/genetics , STAT3 Transcription Factor/genetics , Synaptogyrins/genetics
5.
Nat Commun ; 12(1): 1611, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712590

ABSTRACT

Genome-wide association studies of Systemic Lupus Erythematosus (SLE) nominate 3073 genetic variants at 91 risk loci. To systematically screen these variants for allelic transcriptional enhancer activity, we construct a massively parallel reporter assay (MPRA) library comprising 12,396 DNA oligonucleotides containing the genomic context around every allele of each SLE variant. Transfection into the Epstein-Barr virus-transformed B cell line GM12878 reveals 482 variants with enhancer activity, with 51 variants showing genotype-dependent (allelic) enhancer activity at 27 risk loci. Comparison of MPRA results in GM12878 and Jurkat T cell lines highlights shared and unique allelic transcriptional regulatory mechanisms at SLE risk loci. In-depth analysis of allelic transcription factor (TF) binding at and around allelic variants identifies one class of TFs whose DNA-binding motif tends to be directly altered by the risk variant and a second class of TFs that bind allelically without direct alteration of their motif by the variant. Collectively, our approach provides a blueprint for the discovery of allelic gene regulation at risk loci for any disease and offers insight into the transcriptional regulatory mechanisms underlying SLE.


Subject(s)
Alleles , Genetic Predisposition to Disease/genetics , Lupus Erythematosus, Systemic/genetics , B-Lymphocytes , Cell Line , Chromatin , Gene Expression Regulation , Genome-Wide Association Study , Genotype , Herpesvirus 4, Human , Humans , Quantitative Trait Loci , Synaptogyrins/genetics , T-Lymphocytes
6.
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
7.
Neuron ; 109(5): 767-777.e5, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33472038

ABSTRACT

Tau is a major driver of neurodegeneration and is implicated in over 20 diseases. Tauopathies are characterized by synaptic loss and neuroinflammation, but it is unclear if these pathological events are causally linked. Tau binds to Synaptogyrin-3 on synaptic vesicles. Here, we interfered with this function to determine the role of pathogenic Tau at pre-synaptic terminals. We show that heterozygous knockout of synaptogyrin-3 is benign in mice but strongly rescues mutant Tau-induced defects in long-term synaptic plasticity and working memory. It also significantly rescues the pre- and post-synaptic loss caused by mutant Tau. However, Tau-induced neuroinflammation remains clearly upregulated when we remove the expression of one allele of synaptogyrin-3. Hence neuroinflammation is not sufficient to cause synaptic loss, and these processes are separately induced in response to mutant Tau. In addition, the pre-synaptic defects caused by mutant Tau are enough to drive defects in cognitive tasks.


Subject(s)
Memory Disorders/physiopathology , Microglia/physiology , Presynaptic Terminals/physiology , Synaptogyrins/physiology , tau Proteins/physiology , Animals , Encephalitis/physiopathology , Female , Hippocampus/physiopathology , Hippocampus/ultrastructure , Male , Mice, Knockout , Neuronal Plasticity , Presynaptic Terminals/ultrastructure , Synaptogyrins/genetics
8.
Hum Mol Genet ; 29(16): 2647-2661, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32686835

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an incurable neurological disease with progressive loss of motor neuron (MN) function in the brain and spinal cord. Mutations in TARDBP, encoding the RNA-binding protein TDP-43, are one cause of ALS, and TDP-43 mislocalization in MNs is a key pathological feature of >95% of ALS cases. While numerous studies support altered RNA regulation by TDP-43 as a major cause of disease, specific changes within MNs that trigger disease onset remain unclear. Here, we combined translating ribosome affinity purification (TRAP) with RNA sequencing to identify molecular changes in spinal MNs of TDP-43-driven ALS at motor symptom onset. By comparing the MN translatome of hTDP-43A315T mice to littermate controls and to mice expressing wild type hTDP-43, we identified hundreds of mRNAs that were selectively up- or downregulated in MNs. We validated the deregulated candidates Tex26, Syngr4, and Plekhb1 mRNAs in an independent TRAP experiment. Moreover, by quantitative immunostaining of spinal cord MNs, we found corresponding protein level changes for SYNGR4 and PLEKHB1. We also observed these changes in spinal MNs of an independent ALS mouse model caused by a different patient mutant allele of TDP-43, suggesting that they are general features of TDP-43-driven ALS. Thus, we identified SYNGR4 and PLEKHB1 to be deregulated in MNs at motor symptom onset in TDP-43-driven ALS models. This spatial and temporal pattern suggests that these proteins could be functionally important for driving the transition to the symptomatic phase of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Membrane Proteins/genetics , Synaptogyrins/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Protein Biosynthesis/genetics , RNA-Seq , Spinal Cord/metabolism , Spinal Cord/pathology
9.
Int J Dermatol ; 58(3): 302-310, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30229893

ABSTRACT

BACKGROUND: The pathogenesis of oral lichen planus (OLP) may be related to mental factors. In this study, we investigated the molecular mechanism of mental factors in the development of OLP. METHODS: The normal control group and OLP patients were tested and evaluated by Zung self-rating anxiety scale and self-rating depression scale. Secondly, Agilent human LncRNA chip technology was used to detect differential genes in the total RNA of the normal control group and OLP patients, and to screen out the differentially expressed genes related to anxiety and depression. Thirdly, we verified the genes at gene level and protein level, respectively. RESULTS: Zung self-rating anxiety scale and self-rating depression scale showed that tendency of anxiety and depression in OLP patients is significantly higher than that in normal controls, but there was no significant difference between the erosion form group and the reticular form group; the duration of OLP showed significant negative correlations between degree of anxiety and depression. Microarray analysis showed there were four differential genes (PDE4B, RGS2, SYNGR1, and SYNE1) related to anxiety and depression in OLP patients; real-time qPCR confirmed the expression of PDE4B mRNA was lower in the peripheral blood of normal control group (P < 0.001). The expression of RGS2, SYNGR1, and SYNE1 mRNA was higher in the normal control group (P < 0.05, P < 0.05, P < 0.05). Wes automatic western blot confirmed that the expression of PDE4B protein was lower in the peripheral blood of the normal control group (P < 0.01). CONCLUSION: PDE4B gene may play a role in the pathogenesis of OLP.


Subject(s)
Anxiety/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/blood , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Depression/genetics , Lichen Planus, Oral/blood , Lichen Planus, Oral/genetics , RNA, Messenger/blood , Adult , Case-Control Studies , Cytoskeletal Proteins , Female , Gene Expression , Humans , Lichen Planus, Oral/psychology , Male , Middle Aged , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Psychiatric Status Rating Scales , RGS Proteins/genetics , Synaptogyrins/genetics
10.
PLoS Genet ; 14(10): e1007750, 2018 10.
Article in English | MEDLINE | ID: mdl-30379811

ABSTRACT

Porcine circovirus 2 (PCV2) is a circular single-stranded DNA virus responsible for a group of diseases collectively known as PCV2 Associated Diseases (PCVAD). Variation in the incidence and severity of PCVAD exists between pigs suggesting a host genetic component involved in pathogenesis. A large-scale genome-wide association study of experimentally infected pigs (n = 974), provided evidence of a host genetic role in PCV2 viremia, immune response and growth during challenge. Host genotype explained 64% of the phenotypic variation for overall viral load, with two major Quantitative Trait Loci (QTL) identified on chromosome 7 (SSC7) near the swine leukocyte antigen complex class II locus and on the proximal end of chromosome 12 (SSC12). The SNP having the strongest association, ALGA0110477 (SSC12), explained 9.3% of the genetic and 6.2% of the phenotypic variance for viral load. Dissection of the SSC12 QTL based on gene annotation, genomic and RNA-sequencing, suggested that a missense mutation in the SYNGR2 (SYNGR2 p.Arg63Cys) gene is potentially responsible for the variation in viremia. This polymorphism, located within a protein domain conserved across mammals, results in an amino acid variant SYNGR2 p.63Cys only observed in swine. PCV2 titer in PK15 cells decreased when the expression of SYNGR2 was silenced by specific-siRNA, indicating a role of SYNGR2 in viral replication. Additionally, a PK15 edited clone generated by CRISPR-Cas9, carrying a partial deletion of the second exon that harbors a key domain and the SYNGR2 p.Arg63Cys, was associated with a lower viral titer compared to wildtype PK15 cells (>24 hpi) and supernatant (>48hpi)(P < 0.05). Identification of a non-conservative substitution in this key domain of SYNGR2 suggests that the SYNGR2 p.Arg63Cys variant may underlie the observed genetic effect on viral load.


Subject(s)
Circovirus/genetics , Synaptogyrins/genetics , Synaptogyrins/metabolism , Animals , Circovirus/pathogenicity , DNA Replication , Genome-Wide Association Study , Swine/genetics , Synaptogyrins/physiology , Viral Load/genetics , Viremia/genetics , Virus Replication/genetics
11.
Sci Rep ; 7: 45494, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28382934

ABSTRACT

Combining genome-wide mapping of SNP-rich regions in schizophrenics and gene expression data in all brain compartments across the human life span revealed that genes with promoters most frequently mutated in schizophrenia are expression hubs interacting with far more genes than the rest of the genome. We summed up the differentially methylated "expression neighbors" of genes that fall into one of 108 distinct schizophrenia-associated loci with high number of SNPs. Surprisingly, the number of expression neighbors of the genes in these loci were 35 times higher for the positively correlating genes (32 times higher for the negatively correlating ones) than for the rest of the ~16000 genes. While the genes in the 108 loci have little known impact in schizophrenia, we identified many more known schizophrenia-related important genes with a high degree of connectedness (e.g. MOBP, SYNGR1 and DGCR6), validating our approach. Both the most connected positive and negative hubs affected synapse-related genes the most, supporting the synaptic origin of schizophrenia. At least half of the top genes in both the correlating and anti-correlating categories are cancer-related, including oncogenes (RRAS and ALDOA), providing further insight into the observed inverse relationship between the two diseases.


Subject(s)
Myelin Proteins/metabolism , Polymorphism, Single Nucleotide , Schizophrenia/pathology , Synapses/metabolism , Brain/metabolism , DNA Methylation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Regulatory Networks/genetics , Genetic Loci , Humans , Myelin Proteins/genetics , Nuclear Proteins , Promoter Regions, Genetic , Schizophrenia/genetics , Synaptogyrins/genetics , Synaptogyrins/metabolism
12.
J Biol Chem ; 291(31): 16138-49, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27226560

ABSTRACT

Synaptogyrin-2 is a non-neuronal member of the synaptogyrin family involved in synaptic vesicle biogenesis and trafficking. Little is known about the function of synaptogyrin-2. Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease characterized by high fever, thrombocytopenia, and leukocytopenia with high mortality, caused by a novel tick-borne phlebovirus in the family Bunyaviridae. Our previous studies have shown that the viral nonstructural protein NSs forms inclusion bodies (IBs) that are involved in viral immune evasion, as well as viral RNA replication. In this study, we sought to elucidate the mechanism by which NSs formed the IBs, a lipid droplet-based structure confirmed by NSs co-localization with perilipin A and adipose differentiation-related protein (ADRP). Through a high throughput screening, we identified synaptogyrin-2 to be highly up-regulated in response to SFTS bunyavirus (SFTSV) infection and to be a promoter of viral replication. We demonstrated that synaptogyrin-2 interacted with NSs and was translocated into the IBs, which were reconstructed from lipid droplets into large structures in infection. Viral RNA replication decreased, and infectious virus titers were lowered significantly when synaptogyrin-2 was silenced in specific shRNA-expressing cells, which correlated with the reduced number of the large IBs restructured from regular lipid droplets. We hypothesize that synaptogyrin-2 is essential to promoting the formation of the IBs to become virus factories for viral RNA replication through its interaction with NSs. These findings unveil the function of synaptogyrin-2 as an enhancer in viral infection.


Subject(s)
Bunyaviridae Infections/metabolism , Phlebovirus/physiology , Synaptogyrins/metabolism , Tick-Borne Diseases/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Animals , Bunyaviridae Infections/genetics , Chlorocebus aethiops , HeLa Cells , Humans , Inclusion Bodies, Viral/genetics , Inclusion Bodies, Viral/metabolism , Inclusion Bodies, Viral/virology , RNA, Viral/biosynthesis , RNA, Viral/genetics , Synaptogyrins/genetics , Tick-Borne Diseases/genetics , Vero Cells , Viral Nonstructural Proteins/genetics
13.
Biochem J ; 471(1): 89-99, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26221025

ABSTRACT

RNAi acts as a host immune response against non-self molecules, including viruses. Viruses evolved to neutralize this response by expressing suppressor proteins. In the present study, we investigated dengue virus non structural protein 3 (dvNS3), for its RNAi-suppressor activity in human cell lines. Dengue virus (DV) NS3 reverts the GFP expression in GFP-silenced cell lines. Pull-down assays of dvNS3 revealed that it interacts with the host factor human heat shock cognate 70 (hHSC70). Down-regulation of hHSC70 resulted in accumulation of dengue viral genomic RNA. Also, the interaction of dvNS3 with hHSC70 perturbs the formation of RISC (RNA-induced silencing complex)-loading complex (RLC), by displacing TRBP (TAR RNA-binding protein) and possibly impairing the downstream activity of miRNAs. Interestingly, some of these miRNAs have earlier been reported to be down-regulated upon DV infection in Huh7 cells. Further studies on the miRNA-mRNA relationship along with mRNA profiling of samples overexpressing dvNS3 revealed up-regulation of TAZ (tafazzin) and SYNGR1 (synaptogyrin 1), known dengue viral host factors (DVHFs). Importantly, overexpression of dvNS3 in human embryonic kidney (HEK) 293T cells resulted in modulation of both mature and precursor miRNAs in human cell lines. Subsequent analysis suggested that dvNS3 induced stage-specific down-regulation of miRNAs. Taken together, these results suggest that dvNS3 affects biogenesis and function of host miRNAs to regulate DVHFs for favouring DV replication.


Subject(s)
Dengue Virus/metabolism , Dengue/metabolism , MicroRNAs/metabolism , RNA Interference , Serine Endopeptidases/metabolism , Acyltransferases , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Dengue/genetics , Dengue/pathology , Dengue Virus/genetics , HEK293 Cells , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , Humans , MicroRNAs/genetics , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Serine Endopeptidases/genetics , Synaptogyrins/biosynthesis , Synaptogyrins/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
14.
J Cell Sci ; 128(14): 2423-9, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26071524

ABSTRACT

Insulin-stimulated delivery of glucose transporters (GLUT4, also known as SLC2A4) from specialized intracellular GLUT4 storage vesicles (GSVs) to the surface of fat and muscle cells is central to whole-body glucose regulation. This translocation and subsequent internalization of GLUT4 back into intracellular stores transits through numerous small membrane-bound compartments (internal GLUT4-containing vesicles; IGVs) including GSVs, but the function of these different compartments is not clear. Cellugyrin (also known as synaptogyrin-2) and sortilin define distinct populations of IGV; sortilin-positive IGVs represent GSVs, but the function of cellugyrin-containing IGVs is unknown. Here, we demonstrate a role for cellugyrin in intracellular sequestration of GLUT4 in HeLa cells and have used a proximity ligation assay to follow changes in pairwise associations between cellugyrin, sortilin, GLUT4 and membrane trafficking machinery following insulin-stimulation of 3T3-L1 adipoctyes. Our data suggest that insulin stimulates traffic from cellugyrin-containing to sortilin-containing membranes, and that cellugyrin-containing IGVs provide an insulin-sensitive reservoir to replenish GSVs following insulin-stimulated exocytosis of GLUT4. Furthermore, our data support the existence of a pathway from cellugyrin-containing membranes to the surface of 3T3-L1 adipocytes that bypasses GSVs under basal conditions, and that insulin diverts traffic away from this into GSVs.


Subject(s)
Cell Membrane/metabolism , Exocytosis/drug effects , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Secretory Vesicles/metabolism , 3T3-L1 Cells , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Membrane/genetics , Exocytosis/genetics , Glucose Transporter Type 4/genetics , Mice , Protein Transport/drug effects , Protein Transport/genetics , Secretory Vesicles/genetics , Synaptogyrins/genetics , Synaptogyrins/metabolism
15.
J Neurosci ; 32(50): 18054-67, 18067a, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23238721

ABSTRACT

Sustained neuronal communication relies on the coordinated activity of multiple proteins that regulate synaptic vesicle biogenesis and cycling within the presynaptic terminal. Synaptogyrin and synaptophysin are conserved MARVEL domain-containing transmembrane proteins that are among the most abundant synaptic vesicle constituents, although their role in the synaptic vesicle cycle has remained elusive. To further investigate the function of these proteins, we generated and characterized a synaptogyrin (gyr)-null mutant in Drosophila, whose genome encodes a single synaptogyrin isoform and lacks a synaptophysin homolog. We demonstrate that Drosophila synaptogyrin plays a modulatory role in synaptic vesicle biogenesis at larval neuromuscular junctions. Drosophila lacking synaptogyrin are viable and fertile and have no overt deficits in motor function. However, ultrastructural analysis of gyr larvae revealed increased synaptic vesicle diameter and enhanced variability in the size of synaptic vesicles. In addition, the resolution of endocytic cisternae into synaptic vesicles in response to strong stimulation is defective in gyr mutants. Electrophysiological analysis demonstrated an increase in quantal size and a concomitant decrease in quantal content, suggesting functional consequences for transmission caused by the loss of synaptogyrin. Furthermore, high-frequency stimulation resulted in increased facilitation and a delay in recovery from synaptic depression, indicating that synaptic vesicle exo-endocytosis is abnormally regulated during intense stimulation conditions. These results suggest that synaptogyrin modulates the synaptic vesicle exo-endocytic cycle and is required for the proper biogenesis of synaptic vesicles at nerve terminals.


Subject(s)
Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Synaptogyrins/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Blotting, Western , Drosophila , Immunohistochemistry , Microscopy, Electron, Transmission , Molecular Sequence Data , Mutation , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Patch-Clamp Techniques , Synaptic Vesicles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...