Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 918
Filtrer
1.
Elife ; 132024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39109999

RÉSUMÉ

Prions replicate via the autocatalytic conversion of cellular prion protein (PrPC) into fibrillar assemblies of misfolded PrP. While this process has been extensively studied in vivo and in vitro, non-physiological reaction conditions of fibril formation in vitro have precluded the identification and mechanistic analysis of cellular proteins, which may alter PrP self-assembly and prion replication. Here, we have developed a fibril formation assay for recombinant murine and human PrP (23-231) under near-native conditions (NAA) to study the effect of cellular proteins, which may be risk factors or potential therapeutic targets in prion disease. Genetic screening suggests that variants that increase syntaxin-6 expression in the brain (gene: STX6) are risk factors for sporadic Creutzfeldt-Jakob disease. Analysis of the protein in NAA revealed, counterintuitively, that syntaxin-6 is a potent inhibitor of PrP fibril formation. It significantly delayed the lag phase of fibril formation at highly sub-stoichiometric molar ratios. However, when assessing toxicity of different aggregation time points to primary neurons, syntaxin-6 prolonged the presence of neurotoxic PrP species. Electron microscopy and super-resolution fluorescence microscopy revealed that, instead of highly ordered fibrils, in the presence of syntaxin-6 PrP formed less-ordered aggregates containing syntaxin-6. These data strongly suggest that the protein can directly alter the initial phase of PrP self-assembly and, uniquely, can act as an 'anti-chaperone', which promotes toxic aggregation intermediates by inhibiting fibril formation.


Sujet(s)
Protéines Qa-SNARE , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Animaux , Souris , Humains , Protéines prion/métabolisme , Protéines prion/génétique , Protéines prion/composition chimique , Neurones/métabolisme , Agrégats de protéines , Maladie de Creutzfeldt-Jakob/métabolisme , Maladie de Creutzfeldt-Jakob/génétique
2.
Cell Rep ; 43(8): 114624, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39154341

RÉSUMÉ

Chlamydia trachomatis, a leading cause of bacterial sexually transmitted infections, creates a specialized intracellular replicative niche by translocation and insertion of a diverse array of effectors (Incs [inclusion membrane proteins]) into the inclusion membrane. Here, we characterize IncE, a multifunctional Inc that encodes two non-overlapping short linear motifs (SLiMs) within its short cytosolic C terminus. The proximal SLiM, by mimicking just a small portion of an R-N-ethylmaleimide-sensitive factor adaptor protein receptor (SNARE) motif, binds and recruits syntaxin (STX)7- and STX12-containing vesicles to the inclusion. The distal SLiM mimics the sorting nexin (SNX)5 and SNX6 cargo binding site to recruit SNX6-containing vesicles to the inclusion. By simultaneously binding two distinct vesicle classes, IncE brings these vesicles in close apposition with each other at the inclusion to facilitate C. trachomatis intracellular development. Our work suggests that Incs may have evolved SLiMs to enable rapid evolution in a limited protein space to disrupt host cell processes.


Sujet(s)
Protéines bactériennes , Chlamydia trachomatis , Chlamydia trachomatis/métabolisme , Humains , Protéines bactériennes/métabolisme , Cellules HeLa , Motifs d'acides aminés , Transport des protéines , Nexines de tri/métabolisme , Nexines de tri/génétique , Protéines Qa-SNARE/métabolisme , Liaison aux protéines
3.
Nat Commun ; 15(1): 7510, 2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39209879

RÉSUMÉ

The Greying with age phenotype in horses involves loss of hair pigmentation whereas skin pigmentation is not reduced, and a predisposition to melanoma. The causal mutation was initially reported as a duplication of a 4.6 kb intronic sequence in Syntaxin 17. The speed of greying varies considerably among Grey horses. Here we demonstrate the presence of two different Grey alleles, G2 carrying two tandem copies of the duplicated sequence and G3 carrying three. The latter is by far the most common allele, probably due to strong selection for the striking white phenotype. Our results reveal a remarkable dosage effect where the G3 allele is associated with fast greying and high incidence of melanoma whereas G2 is associated with slow greying and low incidence of melanoma. The copy number expansion transforms a weak enhancer to a strong melanocyte-specific enhancer that underlies hair greying (G2 and G3) and a drastically elevated risk of melanoma (G3 only). Our direct pedigree-based observation of the origin of a G2 allele from a G3 allele by copy number contraction demonstrates the dynamic evolution of this locus and provides the ultimate evidence for causality of the copy number variation of the 4.6 kb intronic sequence.


Sujet(s)
Allèles , Variations de nombre de copies de segment d'ADN , Couleur des cheveux , Introns , Mélanome , Protéines Qa-SNARE , Equus caballus/génétique , Animaux , Variations de nombre de copies de segment d'ADN/génétique , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/métabolisme , Mélanome/génétique , Mélanome/médecine vétérinaire , Mélanome/épidémiologie , Introns/génétique , Couleur des cheveux/génétique , Pedigree , Mâle , Femelle , Phénotype , Incidence , Maladies des chevaux/génétique , Maladies des chevaux/épidémiologie , Pigmentation de la peau/génétique
4.
J Biol Chem ; 300(8): 107591, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39032647

RÉSUMÉ

Neuronal exocytosis requires the assembly of three SNARE proteins, syntaxin and SNAP25 on the plasma membrane and synaptobrevin on the vesicle membrane. However, the precise steps in this process and the points at which assembly and fusion are controlled by regulatory proteins are unclear. In the present work, we examine the kinetics and intermediate states during SNARE assembly in vitro using a combination of time resolved fluorescence and EPR spectroscopy. We show that syntaxin rapidly forms a dimer prior to forming the kinetically stable 2:1 syntaxin:SNAP25 complex and that the 2:1 complex is not diminished by the presence of excess SNAP25. Moreover, the 2:1 complex is temperature-dependent with a reduced concentration at 37 °C. The two segments of SNAP25 behave differently. The N-terminal SN1 segment of SNAP25 exhibits a pronounced increase in backbone ordering from the N- to the C-terminus that is not seen in the C-terminal SNAP25 segment SN2. Both the SN1 and SN2 segments of SNAP25 will assemble with syntaxin; however, while the association of the SN1 segment with syntaxin produces a stable 2:2 (SN1:syntaxin) complex, the complex formed between SN2 and syntaxin is largely disordered. Synaptobrevin fails to bind syntaxin alone but will associate with syntaxin in the presence of either the SN1 or SN2 segments; however, the synaptobrevin:syntaxin:SN2 complex remains disordered. Taken together, these data suggest that synaptobrevin and syntaxin do not assemble in the absence of SNAP25 and that the SN2 segment of SNAP25 is the last to enter the SNARE complex.


Sujet(s)
Neurones , Protéines Qa-SNARE , Protéine SNAP-25 , Protéine SNAP-25/métabolisme , Protéine SNAP-25/génétique , Protéine SNAP-25/composition chimique , Neurones/métabolisme , Animaux , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Protéines Qa-SNARE/composition chimique , Cinétique , Protéines SNARE/métabolisme , Protéines SNARE/génétique , Rats , Multimérisation de protéines
5.
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
6.
Cell Mol Life Sci ; 81(1): 249, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836877

RÉSUMÉ

Protein ubiquitination is one of the most important posttranslational modifications (PTMs) in eukaryotes and is involved in the regulation of almost all cellular signaling pathways. The intracellular bacterial pathogen Legionella pneumophila translocates at least 26 effectors to hijack host ubiquitination signaling via distinct mechanisms. Among these effectors, SidC/SdcA are novel E3 ubiquitin ligases with the adoption of a Cys-His-Asp catalytic triad. SidC/SdcA are critical for the recruitment of endoplasmic reticulum (ER)-derived vesicles to the Legionella-containing vacuole (LCV). However, the ubiquitination targets of SidC/SdcA are largely unknown, which restricts our understanding of the mechanisms used by these effectors to hijack the vesicle trafficking pathway. Here, we demonstrated that multiple Rab small GTPases and target soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) proteins are bona fide ubiquitination substrates of SidC/SdcA. SidC/SdcA-mediated ubiquitination of syntaxin 3 and syntaxin 4 promotes their unconventional pairing with the vesicle-SNARE protein Sec22b, thereby contributing to the membrane fusion of ER-derived vesicles with the phagosome. In addition, our data reveal that ubiquitination of Rab7 by SidC/SdcA is critical for its association with the LCV membrane. Rab7 ubiquitination could impair its binding with the downstream effector Rab-interacting lysosomal protein (RILP), which partially explains why LCVs avoid fusion with lysosomes despite the acquisition of Rab7. Taken together, our study reveals the biological mechanisms employed by SidC/SdcA to promote the maturation of the LCVs.


Sujet(s)
Legionella pneumophila , Phagosomes , Protéines SNARE , Ubiquitination , Protéines G rab , Legionella pneumophila/métabolisme , Humains , Phagosomes/métabolisme , Phagosomes/microbiologie , Protéines SNARE/métabolisme , Protéines G rab/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Animaux , Protéines Qa-SNARE/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Vacuoles/métabolisme , Vacuoles/microbiologie , Cellules HEK293 , Souris , Protéines Rab7 liant le GTP/métabolisme , Protéines G monomériques/métabolisme , Réticulum endoplasmique/métabolisme
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Autophagy ; 20(8): 1895-1896, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38477940

RÉSUMÉ

ATG14 is a core subunit of the class III phosphatidylinositol 3-kinase complex I (PtdIns3K-C1) for macroautophagy/autophagy initiation and also binds to the STX17 to promote autophagosome-lysosome fusion. Our recent work found that ATG14 also targets lipid droplets (LDs) and interacts with mammalian Atg8-family proteins (ATG8s) to mediate lipophagy (selective autophagic degradation of lipid droplets). We also demonstrated that STX18 (syntaxin 18) acts as a negative regulator that disrupts the interactions of ATG14-ATG8s and the formation of the PtdIns3K-C1 through binding to ATG14. Furthermore, we found that knockdown of STX18 induces LD-associated anti-viral protein RSAD2/Viperin degradation dependent on ATG14-mediated lipophagy. Additionally, coronavirus M protein hijacks STX18 to induce lipophagy and degrade RSAD2, facilitating virus production. In summary, our findings reveal new roles of ATG14 in lipid metabolism and viral replication as an autophagic receptor.


Sujet(s)
Protéines associées à l'autophagie , Protéines Qa-SNARE , Humains , Protéines Qa-SNARE/métabolisme , Protéines associées à l'autophagie/métabolisme , Autophagie/physiologie , Animaux , Réplication virale , Gouttelettes lipidiques/métabolisme , Macroautophagie , COVID-19/métabolisme , COVID-19/virologie , Autophagosomes/métabolisme , SARS-CoV-2/métabolisme , Protéines adaptatrices du transport vésiculaire
14.
J Cell Biol ; 223(6)2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38478017

RÉSUMÉ

SM proteins including Sly1 are essential cofactors of SNARE-mediated membrane fusion. Using SNARE and Sly1 mutants and chemically defined in vitro assays, we separate and assess proposed mechanisms through which Sly1 augments fusion: (i) opening the closed conformation of the Qa-SNARE Sed5; (ii) close-range tethering of vesicles to target organelles, mediated by the Sly1-specific regulatory loop; and (iii) nucleation of productive trans-SNARE complexes. We show that all three mechanisms are important and operate in parallel, and that close-range tethering promotes trans-complex assembly when cis-SNARE assembly is a competing process. Further, we demonstrate that the autoinhibitory N-terminal Habc domain of Sed5 has at least two positive activities: it is needed for correct Sed5 localization, and it directly promotes Sly1-dependent fusion. "Split Sed5," with Habc presented solely as a soluble fragment, can function both in vitro and in vivo. Habc appears to facilitate events leading to lipid mixing rather than promoting opening or stability of the fusion pore.


Sujet(s)
Fusion membranaire , Protéines Munc18 , Protéines SNARE , Protéines de Saccharomyces cerevisiae , Protéines Munc18/métabolisme , Liaison aux protéines , Protéines Qa-SNARE/métabolisme , Saccharomyces cerevisiae/cytologie , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines SNARE/génétique , Protéines SNARE/métabolisme , Protéines du transport vésiculaire/métabolisme
15.
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
16.
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
17.
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
18.
Autophagy ; 20(7): 1639-1650, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38411137

RÉSUMÉ

The autophagosomal SNARE STX17 (syntaxin 17) promotes lysosomal fusion and degradation, but its autophagosomal recruitment is incompletely understood. Notably, PtdIns4P is generated on autophagosomes and promotes fusion through an unknown mechanism. Here we show that soluble recombinant STX17 is spontaneously recruited to negatively charged liposomes and adding PtdIns4P to liposomes containing neutral lipids is sufficient for its recruitment. Consistently, STX17 colocalizes with PtdIns4P-positive autophagosomes in cells, and specific inhibition of PtdIns4P synthesis on autophagosomes prevents its loading. Molecular dynamics simulations indicate that C-terminal positively charged amino acids establish contact with membrane bilayers containing negatively charged PtdIns4P. Accordingly, Ala substitution of Lys and Arg residues in the C terminus of STX17 abolishes membrane binding and impairs its autophagosomal recruitment. Finally, only wild type but not Ala substituted STX17 expression rescues the autophagosome-lysosome fusion defect of STX17 loss-of-function cells. We thus identify a key step of autophagosome maturation that promotes lysosomal fusion.Abbreviations: Cardiolipin: 1',3'-bis[1-palmitoyl-2-oleoyl-sn-glycero-3-phospho]-glycerol; DMSO: dimethyl sulfoxide; GST: glutathione S-transferase; GUV: giant unilamellar vesicles; LAMP1: lysosomal associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PA: 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphate; PC/POPC: 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine; PG: 1-palmitoyl-2-linoleoyl-sn-glycero-3-phospho-(1'-rac-glycerol); PI: L-α-phosphatidylinositol; PI4K2A: phosphatidylinositol 4-kinase type 2 alpha; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; POPE/PE: 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine; PS: 1-stearoyl-2-linoleoyl-sn-glycero-3-phospho-L-serine; PtdIns(3,5)P2: 1,2-dioleoyl-sn-glycero-3-phospho-(1"-myo-inositol-3',5'-bisphosphate); PtdIns3P: 1,2- dioleoyl-sn-glycero-3-phospho-(1'-myo-inositol-3'-phosphate); PtdIns4P: 1,2-dioleoyl-sn-glycero-3-phospho-(1"-myo-inositol-4'-phosphate); SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; STX17: syntaxin 17.


Sujet(s)
Autophagosomes , Lysosomes , Fusion membranaire , Phosphates phosphatidylinositol , Protéines Qa-SNARE , Lysosomes/métabolisme , Humains , Autophagosomes/métabolisme , Fusion membranaire/effets des médicaments et des substances chimiques , Protéines Qa-SNARE/métabolisme , Phosphates phosphatidylinositol/métabolisme , Autophagie/physiologie , Autophagie/effets des médicaments et des substances chimiques , Liposomes/métabolisme , Simulation de dynamique moléculaire , Cellules HeLa
19.
Sci Rep ; 14(1): 3200, 2024 02 08.
Article de Anglais | MEDLINE | ID: mdl-38331993

RÉSUMÉ

In the Drosophila larval salivary gland, developmentally programmed fusions between lysosomes and secretory granules (SGs) and their subsequent acidification promote the maturation of SGs that are secreted shortly before puparium formation. Subsequently, ongoing fusions between non-secreted SGs and lysosomes give rise to degradative crinosomes, where the superfluous secretory material is degraded. Lysosomal fusions control both the quality and quantity of SGs, however, its molecular mechanism is incompletely characterized. Here we identify the R-SNARE Ykt6 as a novel regulator of crinosome formation, but not the acidification of maturing SGs. We show that Ykt6 localizes to Lamp1+ carrier vesicles, and forms a SNARE complex with Syntaxin 13 and Snap29 to mediate fusion with SGs. These Lamp1 carriers represent a distinct vesicle population that are functionally different from canonical Arl8+, Cathepsin L+ lysosomes, which also fuse with maturing SGs but are controlled by another SNARE complex composed of Syntaxin 13, Snap29 and Vamp7. Ykt6- and Vamp7-mediated vesicle fusions also determine the fate of SGs, as loss of either of these SNAREs prevents crinosomes from acquiring endosomal PI3P. Our results highlight that fusion events between SGs and different lysosome-related vesicle populations are critical for fine regulation of the maturation and crinophagic degradation of SGs.


Sujet(s)
Protéines SNARE , Vésicules de sécrétion , Protéines SNARE/génétique , Protéines SNARE/métabolisme , Protéines R-SNARE/génétique , Protéines R-SNARE/métabolisme , Protéines Qa-SNARE/métabolisme , Vésicules de sécrétion/métabolisme , Fusion membranaire/physiologie , Lysosomes/métabolisme
20.
Int J Mol Sci ; 25(3)2024 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-38339183

RÉSUMÉ

The main characteristic of polycystic kidney disease is the development of multiple fluid-filled renal cysts. The discovery of mislocalized sodium-potassium pump (Na,K-ATPase) in the apical membrane of cyst-lining epithelia alluded to reversal of polarity as a possible explanation for the fluid secretion. The topic of apical Na,K-ATPase in cysts remains controversial. We investigated the localization of the Na,K-ATPase and assessed the apical-basolateral polarization of cyst-lining epithelia by means of immunohistochemistry in kidney tissue from six polycystic kidney disease patients undergoing nephrectomy. The Na,K-ATPase α1 subunit was conventionally situated in the basolateral membrane of all immunoreactive cysts. Proteins of the Crumbs and partitioning defective (Par) complexes were localized to the apical membrane domain in cyst epithelial cells. The apical targeting protein Syntaxin-3 also immunolocalized to the apical domain of cyst-lining epithelial cells. Proteins of the basolateral Scribble complex immunolocalized to the basolateral domain of cysts. Thus, no deviations from the typical epithelial distribution of basic cell polarity proteins were observed in the cysts from the six patients. Furthermore, we confirmed that cysts can originate from virtually any tubular segment with preserved polarity. In conclusion, we find no evidence of a reversal in apical-basolateral polarity in cyst-lining epithelia in polycystic kidney disease.


Sujet(s)
Kystes , Polykystoses rénales , Humains , Sodium-Potassium-Exchanging ATPase/métabolisme , Polarité de la cellule , Polykystoses rénales/métabolisme , Épithélium/métabolisme , Membrane cellulaire/métabolisme , Protéines Qa-SNARE/métabolisme , Kystes/métabolisme , Rein/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE