Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 523
Filtrer
1.
J Cell Mol Med ; 28(10): e18402, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-39008328

RÉSUMÉ

Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.


Sujet(s)
Athérosclérose , Autophagie , Cellules endothéliales de la veine ombilicale humaine , Inflammation , Lipoprotéines LDL , Protéines Qa-SNARE , Autophagie/génétique , Animaux , Humains , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Souris , Lipoprotéines LDL/métabolisme , Techniques de knock-down de gènes , Lysosomes/métabolisme , Souris knockout , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Alimentation riche en graisse/effets indésirables , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/déficit
2.
J Pediatr Hematol Oncol ; 46(6): e393-e401, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38968556

RÉSUMÉ

OBJECTIVE: Neonatal sepsis and familial hemophagocytic lymphohistiocytosis (fHLH) have similar clinical and laboratory symptoms and the possibility of overlooking fHLH diagnosis is high in newborns with sepsis. History of consanguineous marriage and/or sibling death, hepatomegaly/splenomegaly, and hyperferritinemia (>500 ng/mL) are likely to support fHLH in newborns with sepsis. Therefore, in newborns with sepsis in whom at least 2 of these 3 criteria were detected, genetic variants was investigated for the definitive diagnosed of fHLH. According to the results of genetic examination, we investigated whether these criteria supporting fHLH could be used as a screening test in fHLH. MATERIALS AND METHODS: fHLH-associated genetic variants were investigated in 22 patients diagnosed with neonatal sepsis who fulfilled at least 2 of the following criteria (1) history of consanguineous marriage and/or sibling death, (2) hepatomegaly/splenomegaly, and (3) hyperferritinemia (>500 ng/mL). RESULTS: Heterozygous variants were determined in 6 patients (27.2%): 3 STXBP2 , 1 STX11 , 1 UNC13D , and 1 PRF1 . Polymorphisms associated with the clinical symptoms and signs of HLH were determined in 5 patients (22.7%): 4 UNC13D , 1 PRF1 . Two patients were in the heterozygous variants and polymorphism associated with the clinical symptoms and signs of HLH groups. In 12 patients, benign polymorphisms were detected in STXBP2 and UNC13D genes. No change in fHLH associated genes were found in 1 patient. CONCLUSION: Some variants and/or polymorphisms identified in our patients have been previously reported in patients with HLH. Therefore, we recommend further investigation of fHLH in patients with neonatal sepsis who fulfill at least 2 of the above 3 criteria.


Sujet(s)
Lymphohistiocytose hémophagocytaire , Sepsis néonatal , Humains , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/diagnostic , Nouveau-né , Mâle , Femelle , Sepsis néonatal/diagnostic , Sepsis néonatal/génétique , Perforine/génétique , Protéines Qa-SNARE/génétique , Protéines membranaires/génétique , Dépistage génétique/méthodes
3.
Elife ; 122024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38831696

RÉSUMÉ

During macroautophagy, cytoplasmic constituents are engulfed by autophagosomes. Lysosomes fuse with closed autophagosomes but not with unclosed intermediate structures. This is achieved in part by the late recruitment of the autophagosomal SNARE syntaxin 17 (STX17) to mature autophagosomes. However, how STX17 recognizes autophagosome maturation is not known. Here, we show that this temporally regulated recruitment of STX17 depends on the positively charged C-terminal region of STX17. Consistent with this finding, mature autophagosomes are more negatively charged compared with unclosed intermediate structures. This electrostatic maturation of autophagosomes is likely driven by the accumulation of phosphatidylinositol 4-phosphate (PI4P) in the autophagosomal membrane. Accordingly, dephosphorylation of autophagosomal PI4P prevents the association of STX17 to autophagosomes. Furthermore, molecular dynamics simulations support PI4P-dependent membrane insertion of the transmembrane helices of STX17. Based on these findings, we propose a model in which STX17 recruitment to mature autophagosomes is temporally regulated by a PI4P-driven change in the surface charge of autophagosomes.


Sujet(s)
Autophagosomes , Phosphates phosphatidylinositol , Protéines Qa-SNARE , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Autophagosomes/métabolisme , Phosphates phosphatidylinositol/métabolisme , Humains , Simulation de dynamique moléculaire , Autophagie/physiologie
4.
Pestic Biochem Physiol ; 202: 105934, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38879326

RÉSUMÉ

Syntaxin5 (Syx5) belongs to SNAREs family, which play important roles in fusion of vesicles to target membranes. Most of what we know about functions of Syx5 originates from studies in fungal or vertebrate cells, how Syx5 operates during the development of insects is poorly understood. In this study, we investigated the role of LmSyx5 in the gut development of the hemimetabolous insect Locusta migratoria. LmSyx5 was expressed in many tissues, with higher levels in the gut. Knockdown of LmSyx5 by RNA interference (RNAi) considerably suppressed feeding in both nymphs and adults. The dsLmSyx5-injected locusts lost body weight and finally died at a mortality of 100%. Furthermore, hematoxylin-eosin staining indicated that the midgut is deformed in dsLmSyx5-treated nymphs and the brush border in midgut epithelial cells is severely damaged, suggesting that LmSyx5 is involved in morphogenesis of the midgut. TEM further showed that the endoplasmic reticulum of midgut cells have a bloated appearance. Taken together, these results suggest that LmSyx5 is essential for midgut epithelial homeostsis that affects growth and development of L. migratoria. Thus, Syx5 is a promising RNAi target for controlling L. migratoria, and even other pests.


Sujet(s)
Comportement alimentaire , Protéines d'insecte , Muqueuse intestinale , Locusta Migratoria , Protéines Qa-SNARE , Locusta Migratoria/génétique , Locusta Migratoria/croissance et développement , Locusta Migratoria/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Muqueuse intestinale/croissance et développement , Protéines d'insecte/génétique , Protéines d'insecte/métabolisme , Comportement alimentaire/physiologie , Techniques de knock-down de gènes , Similitude de séquences d'acides aminés , Distribution tissulaire , Poids/génétique , Régulation de l'expression des gènes au cours du développement
5.
Curr Biol ; 34(14): 3102-3115.e6, 2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-38944035

RÉSUMÉ

By modulating stomatal opening and closure, plants control gas exchange, water loss, and photosynthesis in response to various environmental signals. During light-induced stomatal opening, the transport of ions and solutes across the plasma membrane (PM) of the surrounding guard cells results in an increase in turgor pressure, leading to cell swelling. Simultaneously, vesicles for exocytosis are delivered via membrane trafficking to compensate for the enlarged cell surface area and maintain an appropriate ion-channel density in the PM. In eukaryotic cells, soluble N-ethylmaleimide-sensitive factor adaptor protein receptors (SNAREs) mediate membrane fusion between vesicles and target compartments by pairing the cognate glutamine (Q)- and arginine (R)-SNAREs to form a core SNARE complex. Syntaxin of plants 121 (SYP121) is a known Q-SNARE involved in stomatal movement, which not only facilitates the recycling of K+ channels to the PM but also binds to the channels to regulate their activity. In this study, we found that the expression of a receptor-like cytoplasmic kinase, low-K+ sensitive 4/schengen 1 (LKS4/SGN1), was induced by light; it directly interacted with SYP121 and phosphorylated T270 within the SNARE motif. Further investigation revealed that LKS4-dependent phosphorylation of SYP121 facilitated the interaction between SYP121 and R-SNARE vesicle-associated membrane protein 722 (VAMP722), promoting the assembly of the SNARE complex. Our findings demonstrate that the phosphorylation of SNARE proteins is an important strategy adopted by plants to regulate the SNARE complex assembly as well as membrane fusion. Additionally, we discovered the function of LKS4/SGN1 in light-induced stomatal opening via the phosphorylation of SYP121.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Lumière , Stomates de plante , Protéines Qa-SNARE , Arabidopsis/métabolisme , Arabidopsis/physiologie , Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Stomates de plante/physiologie , Stomates de plante/métabolisme , Stomates de plante/effets des radiations , Phosphorylation , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Protéines SNARE/métabolisme , Protéines SNARE/génétique , Protéines du cycle cellulaire
6.
J Exp Med ; 221(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38722309

RÉSUMÉ

SYNTAXIN-11 (STX11) is a SNARE protein that mediates the fusion of cytotoxic granules with the plasma membrane at the immunological synapses of CD8 T or NK cells. Autosomal recessive inheritance of deleterious STX11 variants impairs cytotoxic granule exocytosis, causing familial hemophagocytic lymphohistiocytosis type 4 (FHL-4). In several FHL-4 patients, we also observed hypogammaglobulinemia, elevated frequencies of naive B cells, and increased double-negative DN2:DN1 B cell ratios, indicating a hitherto unrecognized role of STX11 in humoral immunity. Detailed analysis of Stx11-deficient mice revealed impaired CD4 T cell help for B cells, associated with disrupted germinal center formation, reduced isotype class switching, and low antibody avidity. Mechanistically, Stx11-/- CD4 T cells exhibit impaired membrane fusion leading to reduced CD107a and CD40L surface mobilization and diminished IL-2 and IL-10 secretion. Our findings highlight a critical role of STX11 in SNARE-mediated membrane trafficking and vesicle exocytosis in CD4 T cells, important for successful CD4 T cell-B cell interactions. Deficiency in STX11 impairs CD4 T cell-dependent B cell differentiation and humoral responses.


Sujet(s)
Lymphocytes B , Lymphocytes T CD4+ , Protéines Qa-SNARE , Animaux , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Souris , Humains , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphohistiocytose hémophagocytaire/immunologie , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/métabolisme , Souris knockout , Souris de lignée C57BL , Femelle , Mâle , Centre germinatif/immunologie , Centre germinatif/métabolisme , Immunité humorale , Exocytose
7.
Proc Natl Acad Sci U S A ; 121(16): e2309211121, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38593081

RÉSUMÉ

Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and ß-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.


Sujet(s)
Fusion membranaire , Secretagogins , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Secretagogins/métabolisme , Membrane cellulaire/métabolisme , Protéine SNAP-25/métabolisme , Exocytose , Communication cellulaire , Syntaxine-1/métabolisme , Liaison aux protéines
8.
Ups J Med Sci ; 1292024.
Article de Anglais | MEDLINE | ID: mdl-38571883

RÉSUMÉ

The Grey allele in horses is causing premature hair greying and susceptibility to melanoma. The causal mutation is a 4.6 kb tandem duplication in intron 6 of the Syntaxin 17 gene. A recent study demonstrated that the most common allele at the Grey locus (G3) involves three tandem copies of this sequence, whilst a more rare allele (G2) has two tandem copies and the wild-type allele (G1) only one copy. The G3 allele is causing fast greying and high incidence of skin melanoma, whereas the G2 allele is causing slow greying and no obvious increase in melanoma incidence. Further somatic copy number expansion has been documented in melanoma tissue from Grey horses. Functional studies showed that this intronic sequence acts as a weak melanocyte-specific enhancer that becomes substantially stronger by the copy number expansion. The Grey mutation is associated with upregulated expression of both Syntaxin 17 and the neighbouring NR4A3 gene in Grey horse melanomas. It is still an open question which of these genes is most important for the phenotypic effects or if causality is due to the combined effect of upregulation of both genes. Interestingly, RNAseq data in the Human Protein Atlas give support for a possible role of NR4A3 because it is particularly upregulated in human skin cancer, and it belongs to a cluster of genes associated with skin cancer and melanin biosynthesis. The Grey mutation and its association with melanoma provide a possibility to study the path to tumour development in numerous Grey horses carrying exactly the same predisposing mutation.


Sujet(s)
Mélanome , Tumeurs cutanées , Humains , Mélanome/génétique , Mélanome/médecine vétérinaire , Tumeurs cutanées/génétique , Tumeurs cutanées/médecine vétérinaire , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Mutation , Poils/métabolisme , Poils/anatomopathologie
9.
J Microbiol ; 62(4): 315-325, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38451450

RÉSUMÉ

Acinetobacter baumannii (A. baumannii) causes autophagy flux disorder by degrading STX17, resulting in a serious inflammatory response. It remains unclear whether STX17 can alter the inflammatory response process by controlling autolysosome function. This study aimed to explore the role of STX17 in the regulation of pyroptosis induced by A. baumannii. Our findings indicate that overexpression of STX17 enhances autophagosome degradation, increases LAMP1 expression, reduces Cathepsin B release, and improves lysosomal function. Conversely, knockdown of STX17 suppresses autophagosome degradation, reduces LAMP1 expression, augments Cathepsin B release, and accelerates lysosomal dysfunction. In instances of A. baumannii infection, overexpression of STX17 was found to improve lysosomal function and reduce the expression of mature of GSDMD and IL-1ß, along with the release of LDH, thus inhibiting pyroptosis caused by A. baumannii. Conversely, knockdown of STX17 led to increased lysosomal dysfunction and further enhanced the expression of mature of GSDMD and IL-1ß, and increased the release of LDH, exacerbating pyroptosis induced by A. baumannii. These findings suggest that STX17 regulates pyroptosis induced by A. baumannii by modulating lysosomal function.


Sujet(s)
Acinetobacter baumannii , Interleukine-1 bêta , Lysosomes , Pyroptose , Protéines Qa-SNARE , Lysosomes/métabolisme , Acinetobacter baumannii/métabolisme , Acinetobacter baumannii/génétique , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Humains , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Protéines de liaison aux phosphates/métabolisme , Protéines de liaison aux phosphates/génétique , Autophagie , Animaux , Cathepsine B/métabolisme , Cathepsine B/génétique , Infections à Acinetobacter/microbiologie , Souris , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Autophagosomes/métabolisme , Protéine de membrane-1 associée au lysosome/métabolisme , Gasdermines
10.
Crit Rev Eukaryot Gene Expr ; 34(4): 55-68, 2024.
Article de Anglais | MEDLINE | ID: mdl-38505873

RÉSUMÉ

As a primary liver malignancy, hepatocellular carcinoma (HCC) is commonly induced by chronic liver disease and cirrhosis. Bioinformatics analysis reveals that long noncoding RNA KDM4A antisense RNA 1 (KDM4A-AS1) may be aberrantly expressed in HCC and its abnormal expression might influence prognosis in patients. We conducted this study to illustrate the functions and mechanism of KDM4A-AS1 in regulating HCC malignant cell behavior. KD-M4A-AS1, microRNA (miR)-4306 and messenger RNA syntaxin 6 (STX6) expression was examined by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). HCC cell proliferation, apoptosis, migration, and invasion were measured by colony forming assays, flow cytometry, wound healing and Transwell assays. The interaction between genes was verified by RNA immunoprecipitation and luciferase reporter assays. Western blotting was performed to quantify protein expression of STX6 or apoptotic markers. KDM4A-AS1 was highly expressed in HCC cells and tissues. KDM4A-AS1 knockdown led to enhanced HCC cell apoptosis and suppressed HCC cell proliferation, migration, and invasion. MiR-4306 bound to and negatively regulated STX6. KDM4A-AS1 directly bound to miR-4306 and thus up-regulated STX6. STX6 overexpression reversed the inhibitory influence of KDM4A-AS1 depletion on HCC malignant behavior. KDM4A-AS1 promotes HCC cell migration, invasion, and growth by upregulating STX6 via miR-4306.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , microARN , ARN long non codant , Humains , Carcinome hépatocellulaire/anatomopathologie , microARN/génétique , microARN/métabolisme , Tumeurs du foie/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Jumonji Domain-Containing Histone Demethylases/génétique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme
11.
J Biol Chem ; 300(4): 107170, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38492777

RÉSUMÉ

Intercellular miRNA exchange acts as a key mechanism to control gene expression post-transcriptionally in mammalian cells. Regulated export of repressive miRNAs allows the expression of inflammatory cytokines in activated macrophages. Intracellular trafficking of miRNAs from the endoplasmic reticulum to endosomes is a rate-determining step in the miRNA export process and plays an important role in controlling cellular miRNA levels and inflammatory processes in macrophages. We have identified the SNARE protein Syntaxin 5 (STX5) to show a synchronized expression pattern with miRNA activity loss in activated mammalian macrophage cells. STX5 is both necessary and sufficient for macrophage activation and clearance of the intracellular pathogen Leishmania donovani from infected macrophages. Exploring the mechanism of how STX5 acts as an immunostimulant, we have identified the de novo RNA-binding property of this SNARE protein that binds specific miRNAs and facilitates their accumulation in endosomes in a cooperative manner with human ELAVL1 protein, Human antigen R. This activity ensures the export of miRNAs and allows the expression of miRNA-repressed cytokines. Conversely, in its dual role in miRNA export, this SNARE protein prevents lysosomal targeting of endosomes by enhancing the fusion of miRNA-loaded endosomes with the plasma membrane to ensure accelerated release of extracellular vesicles and associated miRNAs.


Sujet(s)
Protéine-1 similaire à ELAV , Macrophages , microARN , Protéines Qa-SNARE , Animaux , Humains , Souris , Endosomes/métabolisme , Leishmania donovani/métabolisme , Leishmania donovani/génétique , Activation des macrophages , Macrophages/métabolisme , microARN/métabolisme , microARN/génétique , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Transport des ARN , Protéine-1 similaire à ELAV/métabolisme
12.
Plant J ; 118(4): 1036-1053, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38289468

RÉSUMÉ

In plants so-called plasma membrane intrinsic proteins (PIPs) are major water channels governing plant water status. Membrane trafficking contributes to functional regulation of major PIPs and is crucial for abiotic stress resilience. Arabidopsis PIP2;1 is rapidly internalised from the plasma membrane in response to high salinity to regulate osmotic water transport, but knowledge of the underlying mechanisms is fragmentary. Here we show that PIP2;1 occurs in complex with SYNTAXIN OF PLANTS 132 (SYP132) together with the plasma membrane H+-ATPase AHA1 as evidenced through in vivo and in vitro analysis. SYP132 is a multifaceted vesicle trafficking protein, known to interact with AHA1 and promote endocytosis to impact growth and pathogen defence. Tracking native proteins in immunoblot analysis, we found that salinity stress enhances SYP132 interactions with PIP2;1 and PIP2;2 isoforms to promote redistribution of the water channels away from the plasma membrane. Concurrently, AHA1 binding within the SYP132-complex was significantly reduced under salinity stress and increased the density of AHA1 proteins at the plasma membrane in leaf tissue. Manipulating SYP132 function in Arabidopsis thaliana enhanced resilience to salinity stress and analysis in heterologous systems suggested that the SNARE influences PIP2;1 osmotic water permeability. We propose therefore that SYP132 coordinates AHA1 and PIP2;1 abundance at the plasma membrane and influences leaf hydraulics to regulate plant responses to abiotic stress signals.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Protéines Qa-SNARE , Stress salin , Aquaporines/métabolisme , Aquaporines/génétique , Arabidopsis/physiologie , Arabidopsis/génétique , Arabidopsis/métabolisme , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Membrane cellulaire/métabolisme , Transport des protéines , Proton-Translocating ATPases/métabolisme , Proton-Translocating ATPases/génétique , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Protéines SNARE/métabolisme , Protéines SNARE/génétique
13.
J Biol Chem ; 300(3): 105687, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38280430

RÉSUMÉ

HIV-1 Gag protein is synthesized in the cytosol and is transported to the plasma membrane, where viral particle assembly and budding occur. Endosomes are alternative sites of Gag accumulation. However, the intracellular transport pathways and carriers for Gag have not been clarified. We show here that Syntaxin6 (Syx6), a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) involved in membrane fusion in post-Golgi networks, is a molecule responsible for Gag trafficking and also for tumor necrosis factor-α (TNFα) secretion and that Gag and TNFα are cotransported via Syx6-positive compartments/vesicles. Confocal and live-cell imaging revealed that Gag colocalized and cotrafficked with Syx6, a fraction of which localizes in early and recycling endosomes. Syx6 knockdown reduced HIV-1 particle production, with Gag distributed diffusely throughout the cytoplasm. Coimmunoprecipitation and pulldown show that Gag binds to Syx6, but not its SNARE partners or their assembly complexes, suggesting that Gag preferentially binds free Syx6. The Gag matrix domain and the Syx6 SNARE domain are responsible for the interaction and cotrafficking. In immune cells, Syx6 knockdown/knockout similarly impaired HIV-1 production. Interestingly, HIV-1 infection facilitated TNFα secretion, and this enhancement did not occur in Syx6-depleted cells. Confocal and live-cell imaging revealed that TNFα and Gag partially colocalized and were cotransported via Syx6-positive compartments/vesicles. Biochemical analyses indicate that TNFα directly binds the C-terminal domain of Syx6. Altogether, our data provide evidence that both Gag and TNFα make use of Syx6-mediated trafficking machinery and suggest that Gag expression does not inhibit but rather facilitates TNFα secretion in HIV-1 infection.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Protéines Qa-SNARE , Vésicules de transport , Facteur de nécrose tumorale alpha , Produits du gène gag du virus de l'immunodéficience humaine , Endosomes/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Transport des protéines/génétique , Produits du gène gag du virus de l'immunodéficience humaine/génétique , Produits du gène gag du virus de l'immunodéficience humaine/métabolisme , Liaison aux protéines , Domaines protéiques , Infections à VIH/métabolisme , Infections à VIH/virologie , Humains , Lignée cellulaire , Vésicules de transport/métabolisme , Réplication virale/génétique
14.
Neurobiol Dis ; 190: 106363, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37996040

RÉSUMÉ

Sporadic Creutzfeldt-Jakob disease (sCJD), the most common human prion disease, is thought to occur when the cellular prion protein (PrPC) spontaneously misfolds and assembles into prion fibrils, culminating in fatal neurodegeneration. In a genome-wide association study of sCJD, we recently identified risk variants in and around the gene STX6, with evidence to suggest a causal increase of STX6 expression in disease-relevant brain regions. STX6 encodes syntaxin-6, a SNARE protein primarily involved in early endosome to trans-Golgi network retrograde transport. Here we developed and characterised a mouse model with genetic depletion of Stx6 and investigated a causal role of Stx6 expression in mouse prion disease through a classical prion transmission study, assessing the impact of homozygous and heterozygous syntaxin-6 knockout on disease incubation periods and prion-related neuropathology. Following inoculation with RML prions, incubation periods in Stx6-/- and Stx6+/- mice differed by 12 days relative to wildtype. Similarly, in Stx6-/- mice, disease incubation periods following inoculation with ME7 prions also differed by 12 days. Histopathological analysis revealed a modest increase in astrogliosis in ME7-inoculated Stx6-/- animals and a variable effect of Stx6 expression on microglia activation, however no differences in neuronal loss, spongiform change or PrP deposition were observed at endpoint. Importantly, Stx6-/- mice are viable and fertile with no gross impairments on a range of neurological, biochemical, histological and skeletal structure tests. Our results provide some support for a pathological role of Stx6 expression in prion disease, which warrants further investigation in the context of prion disease but also other neurodegenerative diseases considering syntaxin-6 appears to have pleiotropic risk effects in progressive supranuclear palsy and Alzheimer's disease.


Sujet(s)
Maladie de Creutzfeldt-Jakob , Maladies à prions , Prions , Souris , Humains , Animaux , Maladie de Creutzfeldt-Jakob/génétique , Maladie de Creutzfeldt-Jakob/anatomopathologie , Prions/génétique , Prions/métabolisme , Étude d'association pangénomique , Souris transgéniques , Encéphale/métabolisme , Maladies à prions/génétique , Maladies à prions/anatomopathologie , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme
15.
J Infect Dis ; 228(12): 1776-1788, 2023 12 20.
Article de Anglais | MEDLINE | ID: mdl-37926090

RÉSUMÉ

Neisseria gonorrhoeae establishes tight interactions with mucosal epithelia through activity of its type IV pilus, while pilus retraction forces activate autophagic responses toward invading gonococci. Here we studied pilus-independent epithelial cell responses and showed that pilus-negative gonococci residing in early and late endosomes are detected and targeted by nucleotide-binding oligomerization domain 1 (NOD1). NOD1 subsequently forms a complex with immunity-related guanosine triphosphatase M (IRGM) and autophagy-related 16-like 1 (ATG16L1) to activate autophagy and recruit microtubule-associated protein light chain 3 (LC3) to the intracellular bacteria. IRGM furthermore directly recruits syntaxin 17 (STX17), which is able to form tethering complexes with the lysosome. Importantly, IRGM-STX17 interactions are enhanced by LC3 but were still observed at lower levels in an LC3 knockout cell line. These findings demonstrate key roles for NOD1 and IRGM in the sensing of intracellular N gonorrhoeae and subsequent directing of the bacterium to the lysosome for degradation.


Sujet(s)
Autophagie , Neisseria gonorrhoeae , Neisseria gonorrhoeae/métabolisme , Cellules épithéliales/métabolisme , Lysosomes/métabolisme , Protéines associées aux microtubules/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Endosomes/métabolisme
16.
Cancer Med ; 12(24): 22185-22195, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-38014487

RÉSUMÉ

BACKGROUND: Some conventional prognostic biomarkers for esophageal squamous cell carcinoma (ESCC) have the disadvantage that they have only been investigated at the level of either mRNA or protein levels or only in individual cohorts. Associations between Syntaxin 3 (STX3) expression and malignancy have been reported in several tumor types but not in ESCC. Here, we investigated the levels of both STX3 mRNA and protein, and its prognostic potential in two independent cohorts of patients with ESCC. METHODS: STX3 mRNA levels were examined in surgical specimens by quantitative PCR in a cohort that included 176 ESCC patients. STX3 protein levels were investigated in surgically resected ESCC tissues by immunohistochemistry using tissue microarrays in a different cohort of 177 ESCC patients. Correlations were analyzed between the expression of STX3 mRNA and protein with clinicopathological factors and long-term prognosis. RESULTS: Quantitative PCR indicated a significant association between high level of STX3 mRNA expression and lymph node involvement, pathological stage, and poor overall survival. The multivariate analysis demonstrated that high STX3 mRNA expression was independently associated with poor overall survival outcomes. Immunohistochemistry revealed that STX3 protein expression in ESCC tissues and high STX3 protein expression were also significantly correlated with unfavorable overall survival. CONCLUSIONS: Overexpression of STX3 mRNA and protein may serve as potential prognostic biomarkers for ESCC patients.


Sujet(s)
Carcinome épidermoïde , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Protéines Qa-SNARE , Humains , Marqueurs biologiques tumoraux/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/chirurgie , Carcinome épidermoïde/métabolisme , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/chirurgie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Stadification tumorale , Pronostic , Protéines Qa-SNARE/génétique , ARN messager/génétique , ARN messager/analyse
17.
J Bone Miner Res ; 38(11): 1718-1730, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37718532

RÉSUMÉ

SNARE proteins comprise a conserved protein family responsible for catalyzing membrane fusion during vesicle traffic. Syntaxin18 (STX18) is a poorly characterized endoplasmic reticulum (ER)-resident t-SNARE. Recently, together with TANGO1 and SLY1, its involvement was shown in ER to Golgi transport of collagen II during chondrogenesis. We report a fetus with a severe osteochondrodysplasia in whom we identified a homozygous substitution of the highly conserved p.Arg10 to Pro of STX18. CRISPR/Cas9-mediated Stx18 deficiency in zebrafish reveals a crucial role for Stx18 in cartilage and bone development. Furthermore, increased expression of multiple components of the Stx18 SNARE complex and of COPI and COPII proteins suggests that Stx18 deficiency impairs antero- and retrograde vesicular transport in the crispant stx18 zebrafish. Taken together, our studies highlight a new candidate gene for a recessive form of osteochondrodysplasia, thereby possibly broadening the SNAREopathy phenotypic spectrum and opening new doors toward future research avenues. © 2023 American Society for Bone and Mineral Research (ASBMR).


Sujet(s)
Ostéochondrodysplasies , Danio zébré , Animaux , Humains , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Danio zébré/génétique , Danio zébré/métabolisme , Ostéochondrodysplasies/métabolisme , Appareil de Golgi/métabolisme , Cartilage/métabolisme , Développement osseux , Transport des protéines
18.
J Allergy Clin Immunol ; 152(6): 1597-1606, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37595757

RÉSUMÉ

BACKGROUND: Inborn errors of immunity are mostly monogenic. However, disease phenotype and outcome may be modified by the coexistence of a second gene defect. OBJECTIVE: We sought to identify the genetic basis of the disease in a patient who experienced bleeding episodes, pancytopenia, hepatosplenomegaly, and recurrent pneumonia that resulted in death. METHODS: Genetic analysis was done using next-generation sequencing. Protein expression and phosphorylation were determined by immunoblotting. T-cell proliferation and F-actin levels were studied by flow cytometry. RESULTS: The patient harbored 2 homozygous deletions in STX11 (c.369_370del, c.374_376del; p.V124fs60∗) previously associated with familial hemophagocytic lymphohistiocytosis and a novel homozygous missense variant in SLP76 (c.767C>T; p.T256I) that resulted in an approximately 85% decrease in SLP76 levels and absent T-cell proliferation. The patient's heterozygous family members showed an approximately 50% decrease in SLP76 levels but normal immune function. SLP76-deficient J14 Jurkat cells did not express SLP76 and had decreased extracellular signal-regulated kinase signaling, basal F-actin levels, and polymerization following T-cell receptor stimulation. Reconstitution of J14 cells with T256I mutant SLP76 resulted in low protein expression and abnormal extracellular signal-regulated kinase phosphorylation and F-actin polymerization after T-cell receptor activation compared with normal expression and J14 function when wild-type SLP76 was introduced. CONCLUSIONS: The hypomorphic mutation in SLP76 tones down the hyperinflammation due to STX11 deletion, resulting in a combined immunodeficiency that overshadows the hemophagocytic lymphohistiocytosis phenotype. To our knowledge, this study represents the first report of the opposing effects of 2 gene defects on the disease in a patient with an inborn error of immunity.


Sujet(s)
Actines , Lymphohistiocytose hémophagocytaire , Humains , Extracellular Signal-Regulated MAP Kinases , Lymphohistiocytose hémophagocytaire/génétique , Mutation , Protéines Qa-SNARE/génétique , Récepteurs aux antigènes des cellules T/génétique , Transduction du signal
19.
Int J Biol Sci ; 19(12): 3892-3907, 2023.
Article de Anglais | MEDLINE | ID: mdl-37564208

RÉSUMÉ

Syntaxin-6 (STX6), a protein of the syntaxin family, is located in the trans-Golgi network and is involved in a variety of intracellular membrane transport events. STX6 is overexpressed in different human malignant tumors. However, little is known about its exact function and molecular mechanism in hepatocellular carcinoma (HCC). In this study, we found that the expression of STX6 was significantly increased in HCC tissues and was associated with poor survival. Gain- and loss-of-function experiments showed that STX6 promotes cell proliferation and metastasis of HCC cells both in vitro and in vivo. Mechanistically, STX6 was negatively regulated by the upstream stimulatory factor 2 (USF2). In addition, STX6 facilitates the association of autophagosomes with lysosomes. Importantly, we demonstrated that STX6 overexpression, despite enhanced resistance to lenvatinib, sensitizes HCC cells to the autophagy activator rapamycin. This study revealed that, under the control of USF2, STX6 accelerates the degradation of microtubule-associated protein 1 light chain 3 beta (LC3) by promoting autophagic flux, ultimately promoting HCC progression. Collectively, we suggest that the USF2-STX6-LC3B axis is a potential therapeutic target in liver cancer.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Protéines Qa-SNARE , Humains , Autophagie/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Lignée cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Facteurs de transcription USF/métabolisme
20.
Elife ; 122023 07 21.
Article de Anglais | MEDLINE | ID: mdl-37477116

RÉSUMÉ

Although budding yeast has been extensively used as a model organism for studying organelle functions and intracellular vesicle trafficking, whether it possesses an independent endocytic early/sorting compartment that sorts endocytic cargos to the endo-lysosomal pathway or the recycling pathway has long been unclear. The structure and properties of the endocytic early/sorting compartment differ significantly between organisms; in plant cells, the trans-Golgi network (TGN) serves this role, whereas in mammalian cells a separate intracellular structure performs this function. The yeast syntaxin homolog Tlg2p, widely localizing to the TGN and endosomal compartments, is presumed to act as a Q-SNARE for endocytic vesicles, but which compartment is the direct target for endocytic vesicles remained unanswered. Here we demonstrate by high-speed and high-resolution 4D imaging of fluorescently labeled endocytic cargos that the Tlg2p-residing compartment within the TGN functions as the early/sorting compartment. After arriving here, endocytic cargos are recycled to the plasma membrane or transported to the yeast Rab5-residing endosomal compartment through the pathway requiring the clathrin adaptors GGAs. Interestingly, Gga2p predominantly localizes at the Tlg2p-residing compartment, and the deletion of GGAs has little effect on another TGN region where Sec7p is present but suppresses dynamics of the Tlg2-residing early/sorting compartment, indicating that the Tlg2p- and Sec7p-residing regions are discrete entities in the mutant. Thus, the Tlg2p-residing region seems to serve as an early/sorting compartment and function independently of the Sec7p-residing region within the TGN.


Sujet(s)
Saccharomyces cerevisiae , Réseau trans-golgien , Animaux , Réseau trans-golgien/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Transport des protéines , Endosomes/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Endocytose , Mammifères/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE