Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Neurobiol Dis ; 190: 106363, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37996040

ABSTRACT

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.


Subject(s)
Creutzfeldt-Jakob Syndrome , Prion Diseases , Prions , Mice , Humans , Animals , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/pathology , Prions/genetics , Prions/metabolism , Genome-Wide Association Study , Mice, Transgenic , Brain/metabolism , Prion Diseases/genetics , Prion Diseases/pathology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism
3.
Asian Pac J Cancer Prev ; 24(6): 2003-2010, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37378930

ABSTRACT

OBJECTIVES: The aim of this study is to evaluate the expression of syntaxin 6 (STX6) in epithelial ovarian cancer (EOC) and assess the effects of STX6 on the prognosis of patient. METHODS: Using information from the Kaplan-Meier Plotter database, the effects of STX6 expression on overall survival (OS) and progression-free survival (PFS) in ovarian cancer patients were examined. The clinical information of 147 patients with epithelial ovarian cancer was evaluated, and immunohistochemical staining was used to identify STX6 expression in postoperative tumor specimens, and the affection of STX6 expression on patient prognosis was assessed. In addition, the expression of STX6 in tumor tissue, peritoneal metastases (PM) derived from 13 patients with epithelial ovarian cancer and 6 normal ovarian specimens was detected by PCR and Western blot. In order to investigate how STX6 affects the proliferation of tumor cells, STX6 was also over expressed and knock down in ovarian cancer cell lines. Then colony formation assay was used to explore the effect of STX6 regulating on cell proliferation. RESULTS: Kaplan-Meier Plotter enrollment data analysis revealed that patients with overexpressed STX6 had substantially worse OS and PFS than individuals with low STX6 expression. Retrospective study revealed a significant (P<0.05) correlation between the STX6 expression and tumor classifications, tumor stage, peritoneal carcinomatosis index (PCI), and PFS survival of patients. Western blot and PCR findings for fresh samples showed that STX6 was overexpressed in both primary lesions and PM nodules of OC. SKOV3 cell proliferation was shown to be dramatically reduced by STX6 knockdown and promoted by STX6 overexpression, according to the in vitro experiments. CONCLUSION: STX6 may increase the progression of epithelial OC by encouraging the proliferation of cancer cells, indicating that STX6 was a viable therapeutic target of epithelial OC.


Subject(s)
Ovarian Neoplasms , Female , Humans , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/pathology , Prognosis , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Retrospective Studies
4.
Oncol Lett ; 24(4): 371, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36238841

ABSTRACT

Syntaxin 6 (STX6), a soluble N-ethylmaleimide-sensitive factor-activating receptor protein, has formed an increasing part of cancer research. However, to the best of our knowledge, the role of STX6 in hepatocellular carcinoma (HCC) is still unclear. In the present study, data from multiple bioinformatics databases, including The Cancer Genome Atlas, Gene Expression Omnibus, Kaplan-Meier plotter, Tumor Immune Estimation Resource (TIMER) and Gene Expression Profiling Integrative Analysis (GEPIA2), and immunohistochemistry (IHC) were utilized to assess the role of STX6 in HCC. The results demonstrated that STX6 expression was upregulated in HCC tissues compared with normal tissues. STX6 expression was significantly associated with tumor size, Edmondson grade and α-fetoprotein (AFP) level. Furthermore, survival analysis demonstrated that high STX6 expression was significantly associated with poor prognosis in patients with HCC. Furthermore, assessment of the immune infiltrates demonstrated that CD163 expression was positively correlated with the STX6 level when analyzed using the TIMER and GEPIA2 databases. IHC results further demonstrated this association. Furthermore, compared with the typically used AFP, STX6 could have an improved diagnostic value in the diagnosis of HCC. In conclusion, STX6 expression was not only positively associated with poor prognosis but may also be involved in the immune inflammatory reaction in HCC. STX6 may become a potential therapeutic and diagnosis maker for patients with HCC.

5.
J Virol ; 96(14): e0081922, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35862696

ABSTRACT

Human cytomegalovirus (HCMV) exhibits a complex host-pathogen interaction with peripheral blood monocytes. We have identified a unique, cell-type specific retrograde-like intracellular trafficking pattern that HCMV utilizes to gain access to the monocyte nucleus and for productive infection. We show that infection of primary human monocytes, epithelial cells, and fibroblasts leads to an increase in the amount of the trafficking protein Syntaxin 6 (Stx6). However, only knockdown (KD) of Stx6 in monocytes inhibited viral trafficking to the trans-Golgi network (TGN), a requisite step for nuclear translocation in monocytes. Conversely, KD of Stx6 in epithelial cells and fibroblasts did not change the kinetics of nuclear translocation and productive infection. Stx6 predominantly functions at the level of the TGN where it facilitates retrograde transport, a trafficking pathway used by only a few cellular proteins and seldom by pathogens. We also newly identify that in monocytes, Stx6 exhibits an irregular vesicular localization rather than being concentrated at the TGN as seen in other cell-types. Lastly, we implicate that viral particles that associate with both Stx6 and EEA1 early in infection are the viral population that successfully traffics to the TGN at later time points and undergo nuclear translocation. Additionally, we show for the first time that HCMV enters the TGN, and that lack of Stx6 prevents viral trafficking to this organelle. We argue that we have identified an essential cell-type specific regulator that controls early steps in efficient productive infection of a cell-type required for viral persistence and disease. IMPORTANCE Human cytomegalovirus (HCMV) infection causes severe and often fatal disease in the immunocompromised. It is one of the leading infectious causes of birth defects and causes severe complications in transplant recipients. By uncovering the unique pathways used by the virus to infect key cells, such as monocytes, responsible for dissemination and persistence, we provide new potential targets for therapeutic intervention.


Subject(s)
Cytomegalovirus , Monocytes , Qa-SNARE Proteins , Cytomegalovirus/pathogenicity , Humans , Monocytes/virology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Signal Transduction , trans-Golgi Network/metabolism
6.
Methods Mol Biol ; 2440: 253-270, 2022.
Article in English | MEDLINE | ID: mdl-35218544

ABSTRACT

Stimulated emission depletion (STED) microscopy is one of the optical superresolution microscopy (SRM) techniques, more recently also referred to as nanoscopy, that have risen to popularity among biologists during the past decade. These techniques keep pushing the physical boundaries of optical resolution toward the molecular scale. Thereby, they enable biologists to image cellular and tissue structures at a level of almost molecular detail that was previously only achievable using electron microscopy. All the while, they retain the advantages of light microscopy, in particular with regards to sample preparation and flexibility of imaging. Commercially available SRM setups have become more and more available and also increasingly sophisticated, both in terms of optical performance and, importantly, ease of use. Institutional microscopy core facilities now offer widespread access to this type of systems. However, the field has grown so rapidly, and keeps growing, that biologists can be easily overwhelmed by the multitude of available techniques and approaches. From this vast array of SRM modalities, STED stands out in one respect: it is essentially an extension to an advanced confocal microscope. Most experienced users of confocal microscopy will find the transition to STED microscopy relatively easy as compared with some other SRM techniques. This also applies to STED sample preparation. Nonetheless, because resolution in STED microscopy does not only depend on the wavelength of the incident light and the numerical aperture of the objective, but crucially also on the square root of the intensity of the depletion laser and, in general, on the photochemical interaction of the fluorophore with the depletion laser, some additional considerations are necessary in STED sample preparation. Here we describe the single color staining of the somatostatin receptor subtype 2A (SSTR2A) and dual color staining of the trans-Golgi-network protein TGN 38 and the t-SNARE syntaxin-6 for STED in the endocrine cell line AtT20 and STED imaging of the samples, providing the protocols in as general a form as possible. The protocols in this chapter are used in this way in an institutional microscopy core facility.


Subject(s)
Fluorescent Dyes , Lasers , Microscopy, Confocal , Microscopy, Fluorescence/methods
7.
Cancers (Basel) ; 15(1)2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36612024

ABSTRACT

Syntaxin-6 (STX6), a vesicular transport protein, is a direct target of the tumor suppressor gene P53, supporting cancer growth dependent on P53. However, STX6's function in the tumor microenvironment has yet to be reported. In this research, we comprehensively explored the role of the oncogene STX6 in pan-cancer by combining data from several databases, including the Cancer Genome Atlas, CPTAC, cBioPortal, and TIMER. Then, we verified the carcinogenic effect of STX6 in hepatocellular carcinoma (HCC) and colorectal cancer (CRC) through a series of experiments in vitro and in vivo. Bioinformatics analysis demonstrated that STX6 is an oncogene for several cancers and is mainly involved in the cell cycle, epithelial-mesenchymal transition, oxidative phosphorylation, and tumor immune modulation, especially for tumor-associated fibroblasts (CAFs) and NKT cells. Additionally, a high level of STX6 could indicate patients' resistance to immunotherapy. Our own data indicated that the STX6 level was upregulated in HCC and CRC. Knockdown of the STX6 levels could arrest the cell cycle and restrain cell proliferation, migration, and invasion. RNA-seq indicated that STX6 was significantly involved in pathways for cancer, such as the MAPK signal pathway. In a mouse model, knockdown of STX6 inhibited tumor growth and potentiated anti-PD-1 efficacy. In light of the essential roles STX6 plays in carcinogenesis and cancer immunology, it has the potential to be a predictive biomarker and a target for cancer immunotherapy.

8.
ACS Nano ; 15(5): 8267-8282, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33915044

ABSTRACT

Cytosolic delivery of small interfering RNA (siRNA) remains challenging, and a profound understanding of the cellular uptake and intracellular processing of siRNA delivery systems could greatly improve the development of siRNA-based therapeutics. Here, we show that caveolae-mediated endocytosis (CvME) accounts for the robust siRNA delivery of mannose-modified trimethyl chitosan-cysteine/tripolyphosphate nanoparticles (MTC/TPP NPs) to macrophages by circumventing lysosomes. We show that the Golgi complex and ER are key organelles required for the efficient delivery of siRNA to macrophages in which the siRNA accumulation positively correlates with its silencing efficiency (r = 0.94). We also identify syntaxin6 and Niemann-Pick type C1 (NPC1) as indispensable regulators for MTC/TPP NPs-delivered siRNA into macrophages both in vitro and in vivo. Syntaxin6 and NPC1 knockout substantially decrease the cellular uptake and gene silencing of the siRNA delivered in MTC/TPP NPs in macrophages, which result in poor therapeutic outcomes for mice bearing acute hepatic injury. Our results suggest that highly efficient siRNA delivery can be achieved via CvME, which would give ideas for designing optimal delivery vectors to facilitate the clinical translation of siRNA drugs.


Subject(s)
Caveolae , Nanoparticles , Animals , Endocytosis , Macrophages/metabolism , Mice , RNA Interference , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Mol Carcinog ; 59(1): 62-72, 2020 01.
Article in English | MEDLINE | ID: mdl-31674708

ABSTRACT

Prostate cancer (PCa) deaths are typically the result of metastatic castration-resistant PCa (mCRPC). Recently, enzalutamide (Enz), an oral androgen receptor inhibitor, was approved for treating patients with mCRPC. Invariably, all PCa patients eventually develop resistance against Enz. Therefore, novel strategies aimed at overcoming Enz resistance are needed to improve the survival of PCa patients. The role of exosomes in drug resistance has not been fully elucidated in PCa. Therefore, we set out to better understand the exosome's role in the mechanism underlying Enz-resistant PCa. Results showed that Enz-resistant PCa cells (C4-2B, CWR-R1, and LNCaP) secreted significantly higher amounts of exosomes (2-4 folds) compared to Enz-sensitive counterparts. Inhibition of exosome biogenesis in resistant cells by GW4869 and dimethyl amiloride strongly decreased their cell viability. Mechanistic studies revealed upregulation of syntaxin 6 as well as its increased colocalization with CD63 in Enz-resistant PCa cells compared to Enz-sensitive cells. Syntaxin 6 knockdown by specific small interfering RNAs in Enz-resistant PCa cells (C4-2B and CWR-R1) resulted in reduced cell number and increased cell death in the presence of Enz. Furthermore, syntaxin 6 knockdown significantly reduced the exosome secretion in both Enz-resistant C4-2B and CWR-R1 cells. The Cancer Genome Atlas analysis showed increased syntaxin 6 expressions associated with higher Gleason score and decreased progression-free survival in PCa patients. Importantly, IHC analysis showed higher syntaxin 6 expression in cancer tissues from Enz-treated patients compared to Enz naïve patients. Overall, syntaxin 6 plays an important role in the secretion of exosomes and increased survival of Enz-resistant PCa cells.


Subject(s)
Antineoplastic Agents/pharmacology , Exosomes/metabolism , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Qa-SNARE Proteins/metabolism , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Exosomes/drug effects , Humans , Male , Nitriles , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/metabolism
10.
Int J Biol Sci ; 15(1): 114-126, 2019.
Article in English | MEDLINE | ID: mdl-30662352

ABSTRACT

As a non-ligand-dependent activation protein, EGFRvIII is the most common mutant of EGFR, and its existence or especially its nuclear translocation in tumors can exacerbate the malignancy. Compared with the nuclear translocation of EGFR, which has been studied extensively, the specific mechanism by which EGFRvIII undergoes nuclear translocation has not yet been reported. Here, we found that EGFRvIII eventually reached the nucleus with the involvement of the Golgi and endoplasmic reticulum (ER) in glioma cells. In this process, syntaxin-6 was responsible for the identification and transport of EGFRvIII on Golgi. We also demonstrated that COPI mediated the reverse transport of EGFRvIII from the Golgi to ER, which process was also important for EGFRvIII's nuclear accumulation. EGFRvIII's nuclear translocation can significantly promote STAT3 phosphorylation and PKM2 nuclear localization. Finally, we showed that EGFRvIII's nuclear translocation obviously induced the growth of gliomas in an intracranial xenotransplantation experiment. These data suggested that searching methods that inhibit EGFRvIII entry into the nucleus will be effective glioma treatments.


Subject(s)
Cell Nucleus/metabolism , ErbB Receptors/metabolism , Glioblastoma/metabolism , Glioma/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Protein Transport , Rats
11.
J Histochem Cytochem ; 65(11): 637-653, 2017 11.
Article in English | MEDLINE | ID: mdl-28914590

ABSTRACT

The comparative structure and expression of salivary components and vesicular transport proteins in the canine major salivary glands were investigated. Histochemical analysis revealed that the morphology of the five major salivary glands-parotid, submandibular, polystomatic sublingual, monostomatic sublingual, and zygomatic glands-was greatly diverse. Immunoblot analysis revealed that expression levels of α-amylase and antimicrobial proteins, such as lysozyme, lactoperoxidase, and lactoferrin, differed among the different glands. Similarly, Rab proteins (Rab3d, Rab11a, Rab11b, Rab27a, and Rab27b) and soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor (SNARE) proteins VAMP4, VAMP8, syntaxin-2, syntaxin-3, syntaxin-4, and syntaxin-6 were expressed at various levels in individual glands. mmunohistochemistry of Rab3d, Rab11b, Rab27b, VAMP4, VAMP8, syntaxin-4, and syntaxin-6 revealed their predominant expression in serous acinar cells, demilunes, and ductal cells. The VAMP4/syntaxin-6 SNARE complex, which is thought to be involved in the maturation of secretory granules in the Golgi field, was found more predominantly in the monostomatic sublingual gland than in the parotid gland. These results suggest that protein expression profiles in canine salivary glands differ among individual glands and reflect the properties of their specialized functions.


Subject(s)
RNA-Binding Proteins/metabolism , SNARE Proteins/metabolism , Salivary Glands/metabolism , Animals , Blotting, Western , Dogs , Immunohistochemistry , Immunoprecipitation , Male , Protein Binding , Salivary Proteins and Peptides/metabolism
12.
Mol Neurobiol ; 54(8): 6085-6096, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27699600

ABSTRACT

Three early signals of asymmetry have been described to occur in a single neurite of neurons at stage 2 of differentiation (before polarization) and shown to be essential for neuronal polarization: (i) accumulation of stable microtubules, (ii) enrichment of the plasma membrane with activatable IGF-1r, and (iii) polarized transport of the microtubular motor KIF5C. Here, we studied the possible relationship between these three phenomena. Our results show that the activatable (membrane-inserted) IGF-1r and stable microtubules accumulate in the same neurite of cells at stage 2. The polarized insertion of IGF-1r depends on microtubule dynamics as shown using drugs which modify microtubule stability. Silencing of KIF5C expression prevents the polarized insertion of IGF-1r into the neuronal plasmalemma and neuronal polarization. Syntaxin 6 and VAMP4, necessary for the polarized insertion of the IGF-1r, are associated to vesicles carried by the microtubular motor KIF5C and is transported preferentially to the neurite where KIF5C accumulates. We conclude that the enrichment of stable microtubules in the future axon enhances KIF5C-mediated vesicular transport of syntaxin 6 and VAMP4, which in turn mediates the polarized insertion of IGF-1r in the plasmalemma, a key step for neuronal polarization. We herewith establish a mechanistic link between three early polarity events necessary for the establishment of neuronal polarity.


Subject(s)
Cell Polarity/physiology , Kinesins/metabolism , Microtubules/metabolism , Neurons/metabolism , Receptor, IGF Type 1/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Polarity/drug effects , Cells, Cultured , Cytochalasin D/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Microtubules/drug effects , Neurites/drug effects , Neurites/metabolism , Neurons/cytology , Neurons/drug effects , Nocodazole/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Paclitaxel/pharmacology , Qa-SNARE Proteins/metabolism , R-SNARE Proteins/metabolism , Rats , Tubulin Modulators/pharmacology
13.
Autophagy ; 13(1): 57-69, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27791468

ABSTRACT

Macroautophagy/autophagy plays a critical role in immunity by directly degrading invading pathogens such as Group A Streptococcus (GAS), through a process that has been named xenophagy. We previously demonstrated that autophagic vacuoles directed against GAS, termed GAS-containing autophagosome-like vacuoles (GcAVs), use recycling endosomes (REs) as a membrane source. However, the precise molecular mechanism that facilitates the fusion between GcAVs and REs remains unclear. Here, we demonstrate that STX6 (syntaxin 6) is recruited to GcAVs and forms a complex with VTI1B and VAMP3 to regulate the GcAV-RE fusion that is required for xenophagy. STX6 targets the GcAV membrane through its tyrosine-based sorting motif and transmembrane domain, and localizes to TFRC (transferrin receptor)-positive punctate structures on GcAVs through its H2 SNARE domain. Knockdown and knockout experiments revealed that STX6 is required for the fusion between GcAVs and REs to promote clearance of intracellular GAS by autophagy. Moreover, VAMP3 and VTI1B interact with STX6 and localize on the TFRC-positive puncta on GcAVs, and are also involved in the RE-GcAV fusion. Furthermore, knockout of RABGEF1 impairs the RE-GcAV fusion and STX6-VAMP3 interaction. These findings demonstrate that RABGEF1 mediates RE fusion with GcAVs through the STX6-VAMP3-VTI1B complex, and reveal the SNARE dynamics involved in autophagosome formation in response to bacterial infection.


Subject(s)
Autophagosomes/metabolism , Endosomes/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , Autophagy , CRISPR-Cas Systems , DNA, Complementary/metabolism , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , Phagocytosis , Phagosomes/metabolism , Protein Domains , Protein Transport , Recombinant Fusion Proteins/metabolism , Transgenes , Vacuoles/metabolism , rab GTP-Binding Proteins/metabolism
14.
J Cell Sci ; 129(22): 4238-4251, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27802163

ABSTRACT

Receptor tyrosine kinases (RTKs) are embedded in the lipid bilayer of the plasma membrane, but the specific roles of various lipids in cell signaling remain largely uncharacterized. We have previously found that acid sphingomyelinase (ASM; also known as SMPD1) regulates the conserved DAF-2 (the ortholog IGF-1R in mammals) RTK signaling pathway in Caenorhabditis elegans How ASM and its catalytic products, ceramides, control RTK signaling pathways remain unclear. Here, we report that ASM regulates the homeostasis of Met, an RTK that is frequently overexpressed in various cancers. Inactivation of ASM led to a rapid loss of Met from the plasma membrane, reduced Met phosphorylation and activation, and induced Met accumulation in the trans-Golgi network (TGN). However, trafficking of integrin ß3 and vesicular stomatitis virus glycoprotein (VSVG) was largely unaffected. Knockdown of syntaxin 6 (STX6) also blocked the Golgi exit of Met. Depletion of either ASM or STX6 led to aberrant trafficking of Met to lysosomes, promoting its degradation. Our studies reveal that ASM and ceramides, together with STX6 and cholesterol, constitute a new regulatory mechanism for the exit of Met from the Golgi during its biosynthetic route, which is used to rapidly replenish and regulate the plasma membrane levels of Met in various cancer cells.


Subject(s)
Cell Membrane/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-met/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cholesterol/metabolism , Enzyme Activation , Female , Humans , Integrin beta3/metabolism , Ligands , Melanoma/metabolism , Melanoma/pathology , Membrane Glycoproteins/metabolism , Neoplasms/pathology , Phosphotyrosine/metabolism , Qa-SNARE Proteins/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Viral Envelope Proteins/metabolism , trans-Golgi Network/metabolism
15.
J Biol Chem ; 291(3): 1320-35, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26578516

ABSTRACT

Annexins are a family of proteins that bind to phospholipids in a calcium-dependent manner. Earlier studies implicated annexin A6 (AnxA6) to inhibit secretion and participate in the organization of the extracellular matrix. We recently showed that elevated AnxA6 levels significantly reduced secretion of the extracellular matrix protein fibronectin (FN). Because FN is directly linked to the ability of cells to migrate, this prompted us to investigate the role of AnxA6 in cell migration. Up-regulation of AnxA6 in several cell models was associated with reduced cell migration in wound healing, individual cell tracking and three-dimensional migration/invasion assays. The reduced ability of AnxA6-expressing cells to migrate was associated with decreased cell surface expression of αVß3 and α5ß1 integrins, both FN receptors. Mechanistically, we found that elevated AnxA6 levels interfered with syntaxin-6 (Stx6)-dependent recycling of integrins to the cell surface. AnxA6 overexpression caused mislocalization and accumulation of Stx6 and integrins in recycling endosomes, whereas siRNA-mediated AnxA6 knockdown did not modify the trafficking of integrins. Given our recent findings that inhibition of cholesterol export from late endosomes (LEs) inhibits Stx6-dependent integrin recycling and that elevated AnxA6 levels cause LE cholesterol accumulation, we propose that AnxA6 and blockage of LE cholesterol transport are critical for endosomal function required for Stx6-mediated recycling of integrins in cell migration.


Subject(s)
Annexin A6/metabolism , Cholesterol/metabolism , Endosomes/metabolism , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Qa-SNARE Proteins/metabolism , Animals , Annexin A6/antagonists & inhibitors , Annexin A6/genetics , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Movement , Cells, Cultured , Cricetulus , Endosomes/ultrastructure , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Integrin alpha5beta1/antagonists & inhibitors , Integrin alphaVbeta3/antagonists & inhibitors , Mice , Microscopy, Confocal , Microscopy, Video , Qa-SNARE Proteins/antagonists & inhibitors , Qa-SNARE Proteins/genetics , RNA Interference , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Time-Lapse Imaging
16.
Article in English | MEDLINE | ID: mdl-25309881

ABSTRACT

Understanding how host proteins are targeted to pathogen-specified organelles, like the chlamydial inclusion, is fundamentally important to understanding the biogenesis of these unique subcellular compartments and how they maintain autonomy within the cell. Syntaxin 6, which localizes to the chlamydial inclusion, contains an YGRL signal sequence. The YGRL functions to return syntaxin 6 to the trans-Golgi from the plasma membrane, and deletion of the YGRL signal sequence from syntaxin 6 also prevents the protein from localizing to the chlamydial inclusion. YGRL is one of three YXXL (YGRL, YQRL, and YKGL) signal sequences which target proteins to the trans-Golgi. We designed various constructs of eukaryotic proteins to test the specificity and propensity of YXXL sequences to target the inclusion. The YGRL signal sequence redirects proteins (e.g., Tgn38, furin, syntaxin 4) that normally do not localize to the chlamydial inclusion. Further, the requirement of the YGRL signal sequence for syntaxin 6 localization to inclusions formed by different species of Chlamydia is conserved. These data indicate that there is an inherent property of the chlamydial inclusion, which allows it to recognize the YGRL signal sequence. To examine whether this "inherent property" was protein or lipid in nature, we asked if deletion of the YGRL signal sequence from syntaxin 6 altered the ability of the protein to interact with proteins or lipids. Deletion or alteration of the YGRL from syntaxin 6 does not appreciably impact syntaxin 6-protein interactions, but does decrease syntaxin 6-lipid interactions. Intriguingly, data also demonstrate that YKGL or YQRL can successfully substitute for YGRL in localization of syntaxin 6 to the chlamydial inclusion. Importantly and for the first time, we are establishing that a eukaryotic signal sequence targets the chlamydial inclusion.


Subject(s)
Chlamydia/physiology , Host-Pathogen Interactions , Inclusion Bodies/metabolism , Inclusion Bodies/microbiology , Amino Acid Motifs , Cell Line , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Humans , Lipid Metabolism , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Sequence Deletion
17.
Biol Open ; 3(5): 314-25, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24705014

ABSTRACT

GLUT4 constitutively recycles between the plasma membrane and intracellular depots. Insulin shifts this dynamic equilibrium towards the plasma membrane by recruiting GLUT4 to the plasma membrane from insulin-responsive vesicles. Muscle is the primary site for dietary glucose deposition; however, how GLUT4 sorts into insulin-responsive vesicles, and if and how insulin resistance affects this process, is unknown. In L6 myoblasts stably expressing myc-tagged GLUT4, we analyzed the intracellular itinerary of GLUT4 as it internalizes from the cell surface and examined if such sorting is perturbed by C2-ceramide, a lipid metabolite causing insulin resistance. Surface-labeled GLUT4myc that internalized for 30 min accumulated in a Syntaxin-6 (Stx6)- and Stx16-positive perinuclear sub-compartment devoid of furin or internalized transferrin, and displayed insulin-responsive re-exocytosis. C2-ceramide dispersed the Stx6-positive sub-compartment and prevented insulin-responsive re-exocytosis of internalized GLUT4myc, even under conditions not affecting insulin-stimulated signaling towards Akt. Microtubule disruption with nocodazole prevented pre-internalized GLUT4myc from reaching the Stx6-positive perinuclear sub-compartment and from undergoing insulin-responsive exocytosis. Removing nocodazole allowed both parameters to recover, suggesting that the Stx6-positive perinuclear sub-compartment was required for GLUT4 insulin-responsiveness. Accordingly, Stx6 knockdown inhibited by ∼50% the ability of internalized GLUT4myc to undergo insulin-responsive re-exocytosis without altering its overall perinuclear accumulation. We propose that Stx6 defines the insulin-responsive compartment in muscle cells. Our data are consistent with a model where ceramide could cause insulin resistance by altering intracellular GLUT4 sorting.

SELECTION OF CITATIONS
SEARCH DETAIL