Visual Abstract

Soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) regulate the vesicle transport machinery in phagocytic cells. Within the secretory pathway, Sec22b is an endoplasmic reticulum–Golgi intermediate compartment (ERGIC)-resident SNARE that controls phagosome maturation and function in macrophages and dendritic cells. The secretory pathway controls the release of cytokines and may also impact the secretion of NO, which is synthesized by the Golgi-active inducible NO synthase (iNOS). Whether ERGIC SNARE Sec22b controls NO and cytokine secretion is unknown. Using murine bone marrow-derived dendritic cells, we demonstrated that inducible NO synthase colocalizes with ERGIC/Golgi markers, notably Sec22b and its partner syntaxin 5, in the cytoplasm and at the phagosome. Pharmacological blockade of the secretory pathway hindered NO and cytokine release, and inhibited NF-κB translocation to the nucleus. Importantly, RNA interference–mediated silencing of Sec22b revealed that NO and cytokine production were abrogated at the protein and mRNA levels. This correlated with reduced nuclear translocation of NF-κB. We also found that Sec22b co-occurs with NF-κB in both the cytoplasm and nucleus, pointing to a role for this SNARE in the shuttling of NF-κB. Collectively, our data unveiled a novel function for the ERGIC/Golgi, and its resident SNARE Sec22b, in the production and release of inflammatory mediators.

The innate immune function of professional phagocytes is channeled through their capacity to ingest and degrade foreign particles and microbes (1). Following particle binding and uptake, phagocytes release a series of small molecules such as NO and cytokines that dictate the nature and intensity of the immune response. Particularly in dendritic cells, the phagosome also serves as a platform for Ag processing and cross-presentation (2). The aforementioned processes exert a membrane demand that is actively supplied by endosomes and the secretory pathway, a dynamic group of organelles through which proteins and lipids are synthesized, processed, and transported (3, 4). Although recycling/early endosomes and the endoplasmic reticulum (ER) supply membrane and protein effectors to nascent phagosomes (5, 6), the latter also serve as cytokine secretion sites (7). Through sequential membrane exchanges with organelles such as the lysosome, maturing phagosomes acidify and become highly degradative.

Interorganellar vesicular transport is regulated by proteins that determine cargo selectivity and vesicle destination (3). Vesicle docking and fusion rely on organelle-specific members of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) family (4, 8). For instance, the syntaxin (Stx) 4/SNAP-23/VAMP3 complex facilitates the passage of TNF from recycling endosomes to the cell membrane (7, 9). On the other hand, trafficking within the ER, ER-Golgi intermediate compartment (ERGIC), Golgi apparatus, and phagosome circuitry is orchestrated by SNARE complexes involving Sec22b, Stx5, and Stx4 (10, 11). Notably, their absence hinders phagosome maturation and Ag cross-presentation (10, 11), which translates into phenotypes such as impaired bacterial and tumor clearance (1214). Moreover, a pan-mouse knockout of Sec22b is embryonically lethal (15), highlighting the key importance of this ERGIC-resident SNARE in immunity and development.

The inducible NO synthase (iNOS) bridges homeostatic regulation and immunity via the production of NO from l-arginine (16). NO modulates inflammation, vasodilation, and neurotransmission. Importantly, this reactive molecule contributes to the generation of highly microbicidal reactive nitrogen species within the phagosome (17, 18) that kill internalized bacteria and Leishmania parasites (19). iNOS expression is mainly regulated at the transcriptional level by NF-κB, and enzymatic activity is regulated at the translational and posttranslational levels. Indeed, iNOS has to assemble into homodimers to transform l-arginine into reactive nitrogen species (20). Although its intracellular trafficking remains uncharacterized, iNOS-positive vesicles have been observed to reside in ER-associated aggresomes and the trans Golgi (2123), as well as the phagosome surface (17, 22). Therefore, the question emerges as to whether secretory pathway–resident SNAREs regulate iNOS trafficking and function. We showed that iNOS is mainly localized to the ERGIC/Golgi both in the cytoplasm and at the phagosome. Notably, Sec22b knockdown hindered NF-κB nuclear translocation, thereby decreasing iNOS and TNF/IL-6 cytokine production. We also observed that Sec22b and NF-κB co-occur in the cytoplasm and the nucleus. These findings highlight a new role for this SNARE in the control of inflammatory responses.

Mouse work was done as stipulated by protocol 1706-07, which was approved by the Comité Institutionnel de Protection des Animaux of the INRS–Centre Armand-Frappier Santé Biotechnologie. This protocol respects procedures on animal practice promulgated by the Canadian Council on Animal Care, described in the Guide to the Care and Use of Experimental Animals.

Rabbit and mouse Abs anti-SAPK/JNK, -p-SAPK/JNK (Thr183/Tyr185), -p38, -p-p38 (Thr180/Tyr182), -p44/42 MAPK, -p-p44/42 MAPK (Thr202/Tyr204), -IκBα, -NF-κB, -iNOS, and -TBP were purchased from Cell Signaling (Cat. Nos. 9252, 9251, 9212, 9211, 9102, 9101, 9242, 8242, 6956, 13120, 8515); rabbit Abs anti-Sec22b and -Stx5 from Synaptic Systems (186003, 110053); rabbit anti-ERGIC53/p58 and mouse anti-β-actin from Sigma (E1031, A1978); rabbit anti-PDI and -CNX from Enzo Life Sciences (ADISPA890, ADISPA865); anti-iNOS, -fibrillarin, and -TOMM20 from Abcam (ab49999, ab186735, ab4566); and rabbit anti-P115 from Proteintech (135091AP). Pharmacological inhibitors brefeldin A (diluted to 0.5 µg/ml), monensin (5 µM), and retro-2 (100 µM) were obtained from Molecular Probes (B7450), Sigma (M5273), and Calbiochem (554715), respectively, and were reconstituted in DMSO from Bioshop (DMS555).

Bone marrow-derived dendritic cells (BMDCs) were differentiated from the bone marrow of 6–8-wk-old female C57BL/6 mice. BMDCs were differentiated for 7 d in RPMI 1640 (Life Technologies) containing 10% v/v heat-inactivated FBS (Wisent), 10 mM HEPES (Bioshop) at pH 7.4, penicillin-streptomycin (Life Technologies), and 10% v/v X63 cell-conditioned medium as a source of GM-CSF (24, 25). Experiments were carried out in complete medium containing 5% X63 cell-conditioned medium. JAWS-II dendritic cell–like lines stably transduced with short hairpin RNA (shRNA) against Sec22b or a scrambled sequence (10) were a gift from D. S. Amigorena (Institut Curie). JAWS-II cells were cultured in RPMI 1640 containing 20% heat-inactivated FBS, 10 mM HEPES at pH 7.4, penicillin-streptomycin, 10% X63 cell-conditioned medium and 40 μg/ml puromycin (Bioshop). The mouse macrophage cell line RAW264.7 clone D3 was cultured in DMEM containing l-glutamine (Life Technologies), 10% heat-inactivated FBS, 20 mM HEPES at pH 7.4, and penicillin-streptomycin. For microscopy experiments, BMDCs and JAWS-II cells were seeded onto poly-l-lysine–coated coverslips (BD Biosciences, 354085) and RAW264.7 cells onto uncoated glass coverslips (Fisher, 1254581). All mammalian cells were kept in a humidified 37°C incubator with 5% CO2.

Leishmania major NIH S (MHOM/SN/74/Seidman) clone A2 (A2WF) promastigotes were cultured in Leishmania medium (M199-1× [Sigma] with 10% heat-inactivated FBS, 10 mM HEPES at pH 7.4, 100 μM hypoxanthine, 5 μM hemin, 3 μM biopterin, 1 μM biotin, and penicillin-streptomycin) in a 26°C incubator. Freshly differentiated promastigotes in late stationary phase (5-d cultures at > 50 × 106 promastigotes/ml) were used for infections.

RAW264.7 macrophages in the second passage were seeded onto 24-well plates and reverse-transfected with Lipofectamine RNAiMAX (Thermo Fisher Scientific, 13778075) as per the manufacturer’s recommendations. Cells were transfected with small interfering RNAs (siRNAs) targeting a scrambled sequence, Sec22b (Thermo Fisher Scientific, D0018101005 and 4390815) or Stx5 (Dharmacon, L063346010005) at a final concentration of 25 nM in a 600-µl volume of complete DMEM without antibiotics for 48 h (26). Prior to experiments, macrophages were replenished with complete medium for 4 h before LPS (diluted to 100 ng/ml; Sigma, L3129) stimulation. Cells were subsequently prepared for microscopy, and their culture supernatants used for NO measurements.

Cells were incubated at 4°C for 5 min with mouse serum-opsonized zymosan (Sigma, Z4250) or L. major promastigotes, followed by a 2-min spin at 298.2 g in a Sorvall RT7 centrifuge (27). Particle internalization was triggered by transferring cells to 37°C, and after 15 min (zymosan) or 2 h (promastigotes), excess particles were washed thrice with warm medium. Cells were then lysed or prepared for confocal microscopy (27).

For phagosome isolation assays, BMDCs were fed with LPS-coated (28) 3-μm magnetic beads (Spherotech, PM-30-10). After a 24-h incubation, cells were washed, resuspended and lysed in filtered purification buffer (2 mM EDTA in PBS at pH 7.2 and 4°C) containing 1× protease inhibitors (Roche) (26). Homogenates were clarified and phagosomes were isolated with a magnet. This was followed by six washes in purification buffer with protease inhibitors, and two washes in filtered PBS (4°C) with no additives. Subsequently, phagosomes were incubated 0.5–1 h (at 4°C) in PBS or in 150 μg/ml proteinase K (Bioshop, PRK403) ± 10% Triton-X100 (27). Reactions were stopped by adding protease inhibitors, 1 μM PMSF and Laemmli sample buffer, followed by immediate boiling at 100°C for 6 min. Proteins were then separated by SDS-PAGE.

Cells were pretreated for 3 h with DMSO or the aforementioned chemical inhibitors and subsequently stimulated (in the presence or absence of inhibitors) with LPS, peptidoglycan (PGN) (40 µg/ml; Sigma, 77140), imiquimod (12.5 µg/ml; Invivogen, tlrl-imq), CpG (4 µg/ml; Invivogen, tlrl-1585), zymosan, or L. major promastigotes. Cell culture supernatants were spun to remove debris, and nitrite concentrations were assayed via the Griess reaction (29). ELISA kits were used to quantify secreted TNF (Ready-SET-Go! Mouse TNFα Kit, eBiosciences, 88732488) and IL-6 (BD OptiEIA, Becton-Dickinson Biosciences, 555240), as per the manufacturers’ instructions.

Cells on coverslips were fixed with 2% paraformaldehyde (Thermo Scientific, 28906) for 20 min and blocked and permeabilized for 17 min with a solution of 0.1% Triton X-100, 1% BSA, 6% nonfat milk, 2% goat serum, and 50% FBS. This was followed by a 2-h incubation with primary Abs diluted in PBS. Then, cells were incubated for 35 min in a solution containing suitable combinations of Alexa Fluor–linked secondary Abs (diluted 1:500; Molecular Probes, A11008, A11011, A11031, A11001, A21247) and DAPI (1:40,000; Molecular Probes, D3571). Coverslips were washed three times with PBS after every step. Coverslips were mounted onto glass slides (Fisher) with Fluoromount-G (Southern Biotechnology Associates, 010001) and sealed with nail hardener (Sally Hansen, 45077). Cells were imaged with the 63× objective of an LSM780 confocal microscope (Carl Zeiss Microimaging). Control stainings revealed no cross-reactivity or background (Supplemental Fig. 5). Images were taken in sequential scanning mode via the ZEN 2012 software and mounted with Photoshop (Adobe). Images were analyzed using Icy Software (Institut Pasteur) (30). Colocalization (31) was evaluated using the Pearson R coefficient calculated from regions of interest defined by active contours (32) on the cytoplasm (differential intensity contrast channel) or on the nucleus (DAPI). Nuclear translocation was evaluated by dividing the mean fluorescence intensity of an entire cell by that of its nucleus. Recruitment was evaluated by scoring the presence of a given protein on the phagosome membrane (33, 34).

Imaging flow cytometry experiments were performed as previously described (35). Briefly, cells were washed with cold PBS containing 1% horse serum (Sigma), 0.1% NaN3, and 5 mM EDTA, and fixed, permeabilized, and stained. DNA was stained with DAPI immediately prior to running the samples through an ImageStreamX MKII flow cytometer running IDEAS software (Amnis). Nuclear translocation of NF-κB was evaluated using a score that quantifies the correlation of NF-κB and DAPI pixels on a per cell basis; a score of > 1 indicated nuclear translocation (35).

LPS-treated or control BMDCs were fixed overnight at 4°C in 0.1% glutaraldehyde + 4% paraformaldehyde in a cocodylate buffer at pH 7.2. After washing, cells were treated with 1.3% osmium tetroxide in collidine buffer and dehydrated (27). Pelleted cells were embedded (Spurr, Ted Pella) and placed in BEEM capsules (Pelco Int, 130). After resin polymerization, samples were sectioned using an ultramicrotome (LKB Bromma 2128, Ultratome). Sections were placed on nickel grids, treated with sodium metaperiodate, and blocked with 1% BSA in PBS. Grids were then incubated with primary Abs for 90 min (diluted 1:30 with 0.1% BSA in PBS), washed, and incubated in suitable 10 nm (anti-mouse) or 20 nm (anti-rabbit) gold particle–conjugated secondary Abs (Abcam, ab39619 and ab27237) for 60 min (diluted 1:10 with 0.1% BSA in PBS). Washed samples were treated with uranyl acetate and lead citrate for contrast and imaged via a Hitachi 7100 electron microscope mounted with an AMT XR-111 camera. Control stainings revealed no background or nonspecificity (Supplemental Fig. 5).

Cells were washed with cold PBS containing 1 mM Na3VO4 and lysed in a solution containing 1% NP-40, 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA (pH 8), 1.5 mM EGTA, 1 mM Na3VO4, 50 mM NaF, 10 mM Na4P2O7, and complete protease inhibitors (27). To increase lysis efficiency, whole-cell extracts were passed several times through a 27G needle (for coimmunoprecipitation experiments) or incubated at −70°C. Insoluble material was removed by centrifugation for 15 min at 21,000 g, upon which protein concentrations were determined using the bicinchoninic acid protein assay kit (Pierce). For immunoprecipitation (IP), whole-cell extracts (1 mg) were cleared for 1 h with Sepharose-Protein A beads (Life Technologies, 101041) previously blocked with TBS containing 5% BSA, and then incubated for 2 h with anti-NF-κB (Cell Signaling, 8242) preadsorbed onto BSA-blocked Sepharose-Protein A beads. Immune complexes were collected by brief centrifugation and washed thrice with lysis buffer containing protease and phosphatase inhibitors. Whole-cell extracts and IP samples were boiled (100°C) for 6 min in SDS sample buffer and migrated in SDS-PAGE gels. Proteins were transferred onto Hybond-ECL membranes (Amersham Biosciences, 10600003), blocked for 2 h in TBS-0.1% Tween containing 5% BSA, incubated with primary Abs (diluted in TBS-0.1% Tween containing 5% BSA) overnight at 4°C, and thence with HRP-conjugated anti-rabbit or anti-mouse IgG Abs from GE Healthcare (NA931V and NA934V) for 1 h at room temperature. For IP experiments, HRP-linked Abs specific to the Fc portion (Life Technologies, 31463) or to native rabbit IgG (Cell Signaling, L27A9) were used. Washed membranes were incubated in ECL (GE Healthcare, RPN2106) and immunodetection was achieved via chemiluminescence (34). Densitometric analysis of Western blot bands was done using the AlphaEase FC software (α Innotech) with β-actin used as loading control.

RNA from BMDCs and JAWS-II cells was extracted using TRIzol (Life Technologies, 15596026) as per the manufacturer’s protocol. Purified RNA was reverse-transcribed (36) and cDNA was submitted to quantitative RT-PCR analysis using the iTaq Universal SYBR Green Supermix (Bio-Rad, 1725121). Samples were amplified in duplicate on an MX3000P instrument (Stratagene), and the following primers were used. Inos: 5'-CTGCTGGTGGTGACAAGCACATTT-3' (AD529F) and 5'-ATGTCATGAGCAAAGGCGCAGAAC-3' (AD530R); Tnf: 5'-GACGTGGAAGTGGCAGAAGAG-3' (AD537F) and 5'-TGCCACAAGCAGGAATGAGA-3' (AD538R); Il6: 5'-ACAACCACGGCCTTCCCTACTT-3' (AD539F) and 5'-CACGATTTCCCAGAGAACATGTG-3' (AD540R); and Hprt: 5'-GTTGGATACAGGCCAGACTTTGTTG-3' (AD55F) and 5'-GATTCAACTTGCGCTCATCTTAGGC-3' (AD56R). MxPro-MX2000P software (Stratagene) was used to record cycle threshold (CT) values, which were analyzed by the 2-ΔΔCT method. Data were normalized to Hprt and expressed in terms of fold change differences relative to nonstimulated cells.

JAWS-II cells expressing either shScr or shSec22b were seeded onto 24-well plates in complete JAWS-II medium. After 24 h, the medium was replaced with Opti-MEM I reduced serum medium (Life Technologies) supplemented with 10% heat-inactivated FBS, without antibiotics. Cells were transfected with 750 ng of the NF-κB-Firefly luciferase reporter construct pGL2-NF-κB-Luc (provided by Dr. P. Duplay, INRS) and 30 ng of the Renilla luciferase control vector pRL-TK (Promega). Plasmids were premixed for 10 min at room temperature and then incubated for 30 min at room temperature with TransIT-TKO reagent (MJS BioLynx MIR2152) (37) in Opti-MEM I reduced serum medium, in the absence of serum or antibiotics. Lipid:DNA complexes were subsequently deposited dropwise on different areas of the wells, followed by gentle rocking from side to side. At 24 h posttransfection, complexes were removed, and cells were stimulated with 100 ng/ml LPS for 6 h, collected in microtubes, washed with PBS, and lysed in Passive Lysis Buffer (Promega, E1941) for 15 min. Finally, Firefly and Renilla luciferase activities were measured by analyzing 20 μl of extracts with the Dual-Luciferase Reporter Assay System (Promega, E1910), according to the manufacturer’s instructions, using a Lumat LB 9507 luminometer (EG & G Berthold).

Statistical differences between two or multiple groups were assessed via the Mann–Whitney U test or one/two-way ANOVA followed by Bonferroni post hoc tests, respectively. Two-way ANOVA was used to evaluate time-dependent differences in inflammatory mediator release. Differences were considered significant when p < 0.05, and graphs were constructed using GraphPad Prism 5.0 (GraphPad Software).

All data are contained within the article and its supporting information.

In BMDCs, the release of NO is induced by both soluble and particulate stimuli (Fig. 1A). To elucidate the underlying mechanisms and pathways, we first assessed the localization of iNOS in LPS-stimulated BMDCs, using immunofluorescence confocal microscopy. (Fig. 1B shows that iNOS was present in vesicular structures situated in the cytoplasm and plasmalemma, consistent with its role in NO secretion to the extracellular milieu (16, 38). To further determine the subcellular localization of iNOS in BMDCs, we performed colocalization analyses by immunofluorescence confocal microscopy using markers of the ER (CNX, PDI), the ERGIC (ERGIC53, Sec22b, Stx5), the Golgi apparatus (P115), the nucleolus (fibrillarin), and mitochondria (TOMM20) (Fig. 1C, 1D and Supplemental Fig. 1A). These analyses revealed that iNOS was widely present in the ERGIC and the Golgi, and absent from nucleoli and mitochondria (Fig. 1C, 1D and Supplemental Fig. 1A). Moreover, following phagocytosis of zymosan particles, we observed that iNOS colocalized with Sec22b, Stx5, and LAMP1 on phagosomes (Fig. 2A, 2B and Supplemental Fig. 1B). These findings suggested a role for the secretory pathway in NO secretion. Unlike endothelial NO synthase (39), iNOS lacks a transmembrane domain but binds to membranes (40) via interactions between its C-terminal domain and ezrin-binding adaptor protein EBP50 (38). With this in mind, we asked whether iNOS was present on the phagosome membrane. To this end, we isolated phagosomes from BMDCs that were fed with LPS-coated magnetic beads. (Fig. 2C). Western blot analysis of the phagosomes confirmed the presence of iNOS. To test whether iNOS was in the cytosolic or luminal side of the phagosome, we incubated intact purified phagosomes with proteinase K. Because iNOS was digested (Fig. 2C), we concluded that it faces the cytoplasm. Collectively, these data demonstrate that iNOS is on the cytoplasmic surface of the organelles that it colocalized with.

FIGURE 1.

iNOS colocalizes with ERGIC and Golgi markers. (A) BMDCs were incubated with LPS, PGN, and zymosan and NO release was assessed by measuring NO2 over a period of 2–24 h. Data are presented as mean concentration ± SEM of three independent experiments done in triplicate wells. (B) The intracellular expression of iNOS (green) in nonstimulated (NS) or LPS-stimulated BMDCs was visualized via confocal immunofluorescence microscopy. DNA is in blue; scale bar, 5 μm. (C) BMDCs were stimulated with LPS for 24 h and the colocalization (white pixels, rightmost panels) of iNOS (green) with ER/ERGIC-associated proteins Sec22b and ERGIC53, Golgi marker P115, and nucleolar marker fibrillarin (all in red) was assessed by immunofluorescence. DNA is in blue; scale bar, 5 μm. Images are representative of three independent experiments. (D) iNOS colocalization was quantified using the Pearson method (see also Supplemental Fig. 1). Data are presented as mean Pearson R coefficient ± SEM of three independent experiments (≥ 10 cells per experiment); each point represents the coefficient of a single cell. ***p < 0.001; ns, not significant.

FIGURE 1.

iNOS colocalizes with ERGIC and Golgi markers. (A) BMDCs were incubated with LPS, PGN, and zymosan and NO release was assessed by measuring NO2 over a period of 2–24 h. Data are presented as mean concentration ± SEM of three independent experiments done in triplicate wells. (B) The intracellular expression of iNOS (green) in nonstimulated (NS) or LPS-stimulated BMDCs was visualized via confocal immunofluorescence microscopy. DNA is in blue; scale bar, 5 μm. (C) BMDCs were stimulated with LPS for 24 h and the colocalization (white pixels, rightmost panels) of iNOS (green) with ER/ERGIC-associated proteins Sec22b and ERGIC53, Golgi marker P115, and nucleolar marker fibrillarin (all in red) was assessed by immunofluorescence. DNA is in blue; scale bar, 5 μm. Images are representative of three independent experiments. (D) iNOS colocalization was quantified using the Pearson method (see also Supplemental Fig. 1). Data are presented as mean Pearson R coefficient ± SEM of three independent experiments (≥ 10 cells per experiment); each point represents the coefficient of a single cell. ***p < 0.001; ns, not significant.

Close modal
FIGURE 2.

iNOS and Sec22b are recruited to the phagosome and face its outer leaflet. (A) BMDCs were fed with opsonized zymosan and recruitment of iNOS (green), Sec22b, LAMP1, and fibrillarin (all in red) was visualized via immunofluorescence 24 h postinternalization. Cytoplasmic and phagosomal (3×-enlarged insets) colocalization can be observed in the rightmost panels (white pixels). DNA is in blue; scale bar, 5 μm; magenta asterisks indicate phagosome location. Images are representative of three independent experiments. (B) Phagosomal recruitment quantification of the aforementioned proteins at 1 h and 24 h postphagocytosis. LAMP1 (lysosomal marker) and fibrillarin (nucleolus)/TOMM20 (mitochondria) were used as positive and negative controls, respectively (see also Supplemental Fig. 1). Data are presented as mean percentage positive phagosomes ± SEM of three independent experiments done in triplicate coverslips; each point represents percentage recruitment in 100 phagosomes per coverslip. *p < 0.05; ***p < 0.001; ns, not significant. (C) To test whether phagosomal iNOS was protected from protease digestion, isolated phagosomes were mock-treated or incubated with proteinase K ± Triton X-100. After reaction inactivation, 2.7% of homogenate (total cell lysate) or 5.1% of phagosome preparation was loaded onto SDS-PAGE. Blots are representative of two independent experiments. Prot K, proteinase K; TCL, total cell lysate.

FIGURE 2.

iNOS and Sec22b are recruited to the phagosome and face its outer leaflet. (A) BMDCs were fed with opsonized zymosan and recruitment of iNOS (green), Sec22b, LAMP1, and fibrillarin (all in red) was visualized via immunofluorescence 24 h postinternalization. Cytoplasmic and phagosomal (3×-enlarged insets) colocalization can be observed in the rightmost panels (white pixels). DNA is in blue; scale bar, 5 μm; magenta asterisks indicate phagosome location. Images are representative of three independent experiments. (B) Phagosomal recruitment quantification of the aforementioned proteins at 1 h and 24 h postphagocytosis. LAMP1 (lysosomal marker) and fibrillarin (nucleolus)/TOMM20 (mitochondria) were used as positive and negative controls, respectively (see also Supplemental Fig. 1). Data are presented as mean percentage positive phagosomes ± SEM of three independent experiments done in triplicate coverslips; each point represents percentage recruitment in 100 phagosomes per coverslip. *p < 0.05; ***p < 0.001; ns, not significant. (C) To test whether phagosomal iNOS was protected from protease digestion, isolated phagosomes were mock-treated or incubated with proteinase K ± Triton X-100. After reaction inactivation, 2.7% of homogenate (total cell lysate) or 5.1% of phagosome preparation was loaded onto SDS-PAGE. Blots are representative of two independent experiments. Prot K, proteinase K; TCL, total cell lysate.

Close modal

The ER-ERGIC-Golgi circuitry is critical for the biogenesis and secretion of proinflammatory cytokines such as TNF and IL-6 (4). Here, we assessed the potential role of ER-Golgi trafficking in the control of iNOS activity and NO release by BMDCs, and we included TNF and IL-6 in our analysis because their release is mediated by the secretory pathway (4). To this end, we treated BMDCs with DMSO or with chemical inhibitors that affect the secretory pathway at different steps (brefeldin A, monensin, and retro-2), prior to stimulation with LPS. Brefeldin A targets GBF1 thereby preventing COP-I recruitment (41). This induces movement of proteins from the Golgi back to the ER, thereby disrupting Golgi structure and blocking protein secretion (42). This can be observed by the dispersal of Golgi protein P115 (Supplemental Fig. 2) (43). On the other hand, monensin is a Na+/H+ ionophore that blocks endosomal acidification (44), intra-Golgi trafficking from the medial to the trans Golgi complex (45, 46). Monensin treatment also leads to Golgi dispersal (47), as verified by P115 staining in Supplemental Fig. 2. Retro-2 has a more selective effect: it blocks retrograde transport to the ER (48) and did not induce Golgi dispersal (Supplemental Fig. 2).

Although monensin and brefeldin A inhibited NO, TNF, and IL-6 release, retro-2 only inhibited TNF release, highlighting individual differences in the intracellular trafficking of those molecules (Fig. 3A–C). Of note, both monensin and brefeldin A inhibited iNOS protein expression (Fig. 3D, 3E), suggesting that ER-Golgi trafficking is part of the pathway leading to iNOS expression. Inhibition of LPS-induced iNOS protein expression in monensin-treated cells was the consequence of reduced iNOS mRNA levels, as determined by quantitative RT-PCR (Fig. 3F), suggesting that the secretory pathway plays a role in the function of LPS-mediated signaling cascades leading to iNOS gene expression. To understand how perturbation of the secretory pathway lead to the inhibition of NO and cytokine release, we first assessed the impact of monensin on the integrity of LPS-induced signal cascades associated with the expression of iNOS, TNF, and IL-6 (49, 50). As shown in (Fig. 4A and Supplemental Fig. 3A, pretreatment with monensin altered the phosphorylation kinetics of JNK (p54) but had no effect on LPS-induced phosphorylation of p44/p42 MAPK (ERK1/2). Monensin-treated BMDCs also exhibited lower IκBα degradation. One of the key downstream events of inducible IκBα degradation is the nuclear translocation of the transcription factor NF-κB, which mediates the expression of a large number of LPS-inducible genes including iNOS, TNF, and IL-6 (49, 50). As shown in (Fig. 4B and 4C, pretreatment of BMDCs with monensin impaired LPS-induced nuclear translocation of NF-κB. Collectively, these findings are consistent with the notion that the integrity of the secretory pathway is necessary for optimal NF-κB–mediated LPS responses in BMDCs.

FIGURE 3.

Inhibition of ER-Golgi trafficking blocks the production of inflammatory mediators. BMDCs were incubated with DMSO or the indicated pharmacological inhibitors for 2 h prior to LPS stimulation for 24 h. Cell culture supernatants were then assayed for NO (A), TNF (B), and IL-6 (C) release. Data are presented as mean concentration ± SEM of three independent experiments done in triplicate wells; each point represents the concentration of a well. Immunoblotting was used to assay iNOS levels in nonstimulated (NS) or LPS-stimulated cells treated with monensin (D) or brefeldin A (BrefA) (E). iNOS densitometries were normalized with those of the β-actin loading control and expressed relative to nonstimulated DMSO-treated cells. Images are representative of at least two experiments. (F) Quantitative RT-PCR was used to measure Inos mRNA levels in NS or LPS-stimulated cells treated with DMSO or monensin. Data are presented as mean relative expression ± SEM of two independent experiments done in duplicate wells; points represent relative expression in each well. **p < 0.01; ***p < 0.001; ns, not significant.

FIGURE 3.

Inhibition of ER-Golgi trafficking blocks the production of inflammatory mediators. BMDCs were incubated with DMSO or the indicated pharmacological inhibitors for 2 h prior to LPS stimulation for 24 h. Cell culture supernatants were then assayed for NO (A), TNF (B), and IL-6 (C) release. Data are presented as mean concentration ± SEM of three independent experiments done in triplicate wells; each point represents the concentration of a well. Immunoblotting was used to assay iNOS levels in nonstimulated (NS) or LPS-stimulated cells treated with monensin (D) or brefeldin A (BrefA) (E). iNOS densitometries were normalized with those of the β-actin loading control and expressed relative to nonstimulated DMSO-treated cells. Images are representative of at least two experiments. (F) Quantitative RT-PCR was used to measure Inos mRNA levels in NS or LPS-stimulated cells treated with DMSO or monensin. Data are presented as mean relative expression ± SEM of two independent experiments done in duplicate wells; points represent relative expression in each well. **p < 0.01; ***p < 0.001; ns, not significant.

Close modal
FIGURE 4.

Inhibition of ER-Golgi trafficking inhibits MAPK phosphorylation and the translocation of NF-κB to the nucleus. (A) BMDCs were incubated with DMSO or monensin for 2 h prior to LPS stimulation for 0.25–2 h. Western blots show levels of total and phosphorylated MAPK, as well as IκBα and NF-κB. β-actin was used as loading control and images are representative of two experiments (see also Supplemental Fig. 3). (B) DMSO- or monensin-treated BMDCs were incubated with LPS for 2 h, and nuclear translocation of NF-κB (green) was visualized and quantified (C) via immunofluorescence. DNA is in blue; NS, nonstimulated; scale bar, 5 μm. Data are presented as average mean fluorescence intensity (MFI) ratios (NF-κB MFI of nucleus/entire cell) ± SEM of four independent experiments (≥ 20 cells per experiment); each point represents the ratio of a single cell. **p < 0.01; ***p < 0.001; ns, not significant.

FIGURE 4.

Inhibition of ER-Golgi trafficking inhibits MAPK phosphorylation and the translocation of NF-κB to the nucleus. (A) BMDCs were incubated with DMSO or monensin for 2 h prior to LPS stimulation for 0.25–2 h. Western blots show levels of total and phosphorylated MAPK, as well as IκBα and NF-κB. β-actin was used as loading control and images are representative of two experiments (see also Supplemental Fig. 3). (B) DMSO- or monensin-treated BMDCs were incubated with LPS for 2 h, and nuclear translocation of NF-κB (green) was visualized and quantified (C) via immunofluorescence. DNA is in blue; NS, nonstimulated; scale bar, 5 μm. Data are presented as average mean fluorescence intensity (MFI) ratios (NF-κB MFI of nucleus/entire cell) ± SEM of four independent experiments (≥ 20 cells per experiment); each point represents the ratio of a single cell. **p < 0.01; ***p < 0.001; ns, not significant.

Close modal

The SNARE Sec22b plays a role in ER-Golgi trafficking (51) and in the unconventional secretion of IL-1β in cells treated with autophagy inducers, a process known as secretory autophagy (52). To assess the potential role of Sec22b in the release of NO, we stimulated with LPS dendritic cell–like JAWS-II cells transduced with an shRNA to Sec22b (shSec22b) or a scrambled (shScr) sequence (10). As shown in (Fig. 5A–C, Sec22b knockdown diminished the release of NO, as well as TNF and IL-6. To confirm the role of Sec22b in NO secretion and to test whether this mechanism operates in macrophages, we stimulated with LPS RAW264.7 macrophages pretreated with siRNAs to Sec22b, its binding partner Stx5, or both. In that regard, knockdown of one or both SNAREs resulted in the inhibition of NO release (Fig. 5D). Akin to the effect of pharmacological inhibitors on iNOS expression, Sec22b knockdown diminished iNOS protein levels (Fig. 5E), as well as Inos, Tnf, and Il6 expression, as assessed by quantitative RT-PCR (Fig. 5F).

FIGURE 5.

Sec22b is a positive regulator of inflammatory mediator release. To evaluate whether Sec22b modulates inflammatory effector secretion, LPS was used to stimulate JAWS-II cells transduced with an shRNA targeting Sec22b (shSec22b) or a scrambled sequence (shScr). Over a period of 24 h, the release of NO (A), TNF (B), and IL-6 (C) was quantified in cell culture supernatants. Data are presented as mean concentration ± SEM of three independent experiments done in triplicate wells. (D) NO secretion in control or LPS-treated RAW264.7 cells transfected with siRNAs to a scrambled sequence, Sec22b or Stx5. Data are presented as mean concentration ± SEM of at least two independent experiments done in triplicate wells; each point represents the concentration of a well. (E) iNOS protein levels in nonstimulated (NS) or LPS-stimulated Sec22b knockdown JAWS-II cells preincubated with DMSO or brefeldin A (BrefA) were assayed via Western blot. iNOS densitometries were normalized with those of the β-actin loading control and expressed relative to NS DMSO-treated cells. Images are representative of two experiments. (F) Inos, Tnf, and Il6 mRNA levels in LPS-stimulated Sec22b knockdown JAWS-II cells were assayed via quantitative RT-PCR. Data are presented as mean relative expression ± SEM of two independent experiments done in duplicate wells; points represent relative expression in each well. *p < 0.05; **p < 0.01; ***p < 0.001.

FIGURE 5.

Sec22b is a positive regulator of inflammatory mediator release. To evaluate whether Sec22b modulates inflammatory effector secretion, LPS was used to stimulate JAWS-II cells transduced with an shRNA targeting Sec22b (shSec22b) or a scrambled sequence (shScr). Over a period of 24 h, the release of NO (A), TNF (B), and IL-6 (C) was quantified in cell culture supernatants. Data are presented as mean concentration ± SEM of three independent experiments done in triplicate wells. (D) NO secretion in control or LPS-treated RAW264.7 cells transfected with siRNAs to a scrambled sequence, Sec22b or Stx5. Data are presented as mean concentration ± SEM of at least two independent experiments done in triplicate wells; each point represents the concentration of a well. (E) iNOS protein levels in nonstimulated (NS) or LPS-stimulated Sec22b knockdown JAWS-II cells preincubated with DMSO or brefeldin A (BrefA) were assayed via Western blot. iNOS densitometries were normalized with those of the β-actin loading control and expressed relative to NS DMSO-treated cells. Images are representative of two experiments. (F) Inos, Tnf, and Il6 mRNA levels in LPS-stimulated Sec22b knockdown JAWS-II cells were assayed via quantitative RT-PCR. Data are presented as mean relative expression ± SEM of two independent experiments done in duplicate wells; points represent relative expression in each well. *p < 0.05; **p < 0.01; ***p < 0.001.

Close modal

To ensure that the function of Sec22b on inflammatory mediator production is not contingent on LPS stimulation, we treated Sec22b knockdown cells with agonists to TLR2 (PGN), dectin-1/TLR2 (zymosan), TLR2 (L. major promastigotes), TLR7 (imiquimod), or TLR9 (CpG). Twenty-four hours poststimulation, silencing Sec22b abrogated the induction of NO, TNF, and IL-6 by all the tested ligands (Fig. 6). Hence, the phenotype is not exclusively dependent on TLR4.

FIGURE 6.

The role of Sec22b is not specific to LPS stimulation. To evaluate whether Sec22b modulates inflammatory effector secretion in an exclusively LPS-dependent manner, shScr- or shSec22b-transduced JAWS-II cells were stimulated with TLR ligands LPS, PGN, zymosan, imiquimod, CpG, and L. major promastigotes for 24 h. Cell culture supernatants were then probed for the release of NO (A), TNF (B), and IL-6 (C). Data are presented as mean concentration ± SEM of at least two independent experiments done in triplicate wells; each point represents the concentration of a well. **p < 0.01; ***p < 0.001; ns, not significant.

FIGURE 6.

The role of Sec22b is not specific to LPS stimulation. To evaluate whether Sec22b modulates inflammatory effector secretion in an exclusively LPS-dependent manner, shScr- or shSec22b-transduced JAWS-II cells were stimulated with TLR ligands LPS, PGN, zymosan, imiquimod, CpG, and L. major promastigotes for 24 h. Cell culture supernatants were then probed for the release of NO (A), TNF (B), and IL-6 (C). Data are presented as mean concentration ± SEM of at least two independent experiments done in triplicate wells; each point represents the concentration of a well. **p < 0.01; ***p < 0.001; ns, not significant.

Close modal

To understand how Sec22b regulates LPS-induced responses, we evaluated the impact of Sec22b knockdown on LPS-induced signal transduction cascades leading to the expression of iNOS. Apart from an early but temporary impairment of JNK (p54) phosphorylation, and IκBα degradation, silencing of Sec22b in JAWS-II dendritic cells had no significant effect on LPS-induced signaling events and on NF-κB expression (Fig. 7A and Supplemental Fig. 3B). To determine whether Sec22b is involved in the nuclear translocation of NF-κB, we stimulated JAWS-II cells expressing either an shScr or an shSec22b with LPS and quantified NF-κB nuclear translocation using imaging flow cytometry (Fig. 7B, 7C) and immunofluorescence confocal microscopy (Fig. 7D, 7E). Our results show that knockdown of Sec22b hampered LPS-induced NF-κB nuclear translocation. We made a similar observation in LPS-stimulated RAW264.7 macrophages treated with siRNAs to Sec22b (Fig. 7F, 7G). To further characterize the impact of Sec22b knockdown on the inhibition of NF-κB translocation, we transfected JAWS-II cells expressing either shScr or shSec22b with an NF-κB luciferase reporter construct prior to stimulation with LPS. As shown in (Fig. 7H, activation of the NF-κB reporter was significantly impaired in Sec22b knockdown cells. These data indicate that the secretory pathway–resident SNARE Sec22b modulates proinflammatory effector production by participating in the nuclear translocation of NF-κB.

FIGURE 7.

Sec22b modulates MAPK phosphorylation and the translocation of NF-κB to the nucleus. (A) ShScr- or shSec22b-transduced JAWS-II cells were stimulated with LPS for 0.25–4 h. Western blots show levels of total and phosphorylated MAPK, as well as IκBα and NF-κB. β-actin was used as loading control and images are representative of two experiments (see also Supplemental Fig. 3). Nuclear translocation of NF-κB (green) was visualized via imaging flow cytometry (B) and quantified (C). DNA is in magenta; scale bar, 10 μm. Data are presented as average NF-κB translocation ± SEM relative to nonstimulated (NS) cells of at least five independent experiments; each point represents one experiment. NF-κB translocation was also visualized and quantified via confocal microscopy in control and LPS-stimulated JAWS-II cells (D and E) or RAW264.7 macrophages (F and G). DNA is in blue; scale bar, 5 μm. Data are presented as average mean fluorescence intensity (MFI) ratios (NF-κB MFI of nucleus/entire cell) ± SEM of four independent experiments (≥ 10 cells per experiment); each point represents the ratio of a single cell. (H) ShScr- or shSec22b-transduced JAWS-II cells were transiently cotransfected with pGL2-NF-κB and pRL-TK for 24 h and then treated with LPS. After 6 h, cells were harvested to analyze luciferase activities, measured as relative light units (RLUs). Data are expressed as a ratio of Firefly to Renilla RLU values. The means ± SEM of three independent experiments done in duplicate or triplicate wells are shown. **p < 0.01; ***p < 0.001; ns, not significant.

FIGURE 7.

Sec22b modulates MAPK phosphorylation and the translocation of NF-κB to the nucleus. (A) ShScr- or shSec22b-transduced JAWS-II cells were stimulated with LPS for 0.25–4 h. Western blots show levels of total and phosphorylated MAPK, as well as IκBα and NF-κB. β-actin was used as loading control and images are representative of two experiments (see also Supplemental Fig. 3). Nuclear translocation of NF-κB (green) was visualized via imaging flow cytometry (B) and quantified (C). DNA is in magenta; scale bar, 10 μm. Data are presented as average NF-κB translocation ± SEM relative to nonstimulated (NS) cells of at least five independent experiments; each point represents one experiment. NF-κB translocation was also visualized and quantified via confocal microscopy in control and LPS-stimulated JAWS-II cells (D and E) or RAW264.7 macrophages (F and G). DNA is in blue; scale bar, 5 μm. Data are presented as average mean fluorescence intensity (MFI) ratios (NF-κB MFI of nucleus/entire cell) ± SEM of four independent experiments (≥ 10 cells per experiment); each point represents the ratio of a single cell. (H) ShScr- or shSec22b-transduced JAWS-II cells were transiently cotransfected with pGL2-NF-κB and pRL-TK for 24 h and then treated with LPS. After 6 h, cells were harvested to analyze luciferase activities, measured as relative light units (RLUs). Data are expressed as a ratio of Firefly to Renilla RLU values. The means ± SEM of three independent experiments done in duplicate or triplicate wells are shown. **p < 0.01; ***p < 0.001; ns, not significant.

Close modal

Because Sec22b is present in the ER-ERGIC-Golgi circuit (8, 53), we postulated that NF-κB and Sec22b are found in the same compartments. To test this, we used immunofluorescence to examine the colocalization of NF-κB with Sec22b, as well as other ERGIC and Golgi proteins. In control and LPS-stimulated BMDCs, we found partial colocalization under steady-state conditions, which disappeared following LPS-induced migration of NF-κB to the nucleus (Fig. 8A, 8B, Supplemental Figs. 4 and 5). We also observed that Sec22b was present in the nucleus; LPS slightly, but significantly, increased the proportion of Sec22b in the nucleus compared with the entire cell (Fig. 8C). Similarly, immunogold electron microscopy revealed that NF-κB and Sec22b co-occur in membranous structures in the cytoplasm, and increasingly in the nucleus after LPS stimulation (Fig. 8D and Supplemental Fig. 5). Both NF-κB and Sec22b were found in nuclear regions that do not costain with DAPI. Because nucleoli also exhibit this characteristic (54), we asked whether Sec22b colocalizes with nucleolar marker fibrillarin (55), and found that Sec22b was not found in these structures (Fig. 8E, 8F and Supplemental Videos 1 and 2). Our observation that Sec22b and NF-κB partially colocalize led us to determine whether these proteins physically interact. To this end, we performed coimmunoprecipitation experiments in BMDCs under steady-state conditions. As shown in (Fig. 8G, NF-κB and Sec22b did not coimmunoprecipitate, in contrast to NF-κB and its inhibitor IκBα that are known to exist as a complex in steady-state cells. This observation indicates that NF-κB and Sec22b are present in the same compartments but do not form complexes. Collectively, these findings revealed that NF-κB is an ER-Golgi–associated protein that necessitates Sec22b to translocate to the nucleus.

FIGURE 8.

NF-κB and Sec22b comigrate to the nucleus. (A) BMDCs were stimulated with LPS for 1 h and the colocalization (white pixels, rightmost panels) of NF-κB (green) with SNARE Sec22b (red) was assessed by immunofluorescence. DNA is in blue; 3.5×-enlarged insets are shown. (B) NF-κB colocalization was quantified using the Pearson method (see also Supplemental Fig. 4). (C) Quantification of Sec22b mean fluorescence intensity ratio in nucleus relative to the entire cell. (D) Immunoelectron microscopy assessment of NF-κB (10 nm nanoparticles) and Sec22b (20 nm nanoparticles) expression in nonstimulated (NS) and LPS-treated BMDCs. Red arrowheads indicate regions where both NF-κB and Sec22b nanoparticles were at a ≤ 100 nm distance (see also Supplemental Fig. 5). (E) Assessment of Sec22b (green) colocalization with nucleolar marker fibrillarin (red). Rightmost panels show orthogonal views of the nuclear region (see also Supplemental Videos 1 and 2). (F) Quantification of Sec22b and fibrillarin colocalization in the nucleus. Image panels are representative of three independent experiments. (G) NF-κB was immunoprecipitated from whole-cell extracts of unstimulated BMDCs and the resulting immune complexes were subjected to blotting using Abs against NF-κB, IκBα, and Sec22b, along with 4% of the initial lysates (input). Images are representative of two experiments. In (A), (D), and (E), scale bars represent 5 μm (white), 500 nm (black), and 100 nm (red). In (B), (C) and (F), data are presented as mean ± SEM of three independent experiments (≥ 10 cells per experiment); each point represents the measurement of a single cell. **p < 0.01; ***p < 0.001; ns, not significant.

FIGURE 8.

NF-κB and Sec22b comigrate to the nucleus. (A) BMDCs were stimulated with LPS for 1 h and the colocalization (white pixels, rightmost panels) of NF-κB (green) with SNARE Sec22b (red) was assessed by immunofluorescence. DNA is in blue; 3.5×-enlarged insets are shown. (B) NF-κB colocalization was quantified using the Pearson method (see also Supplemental Fig. 4). (C) Quantification of Sec22b mean fluorescence intensity ratio in nucleus relative to the entire cell. (D) Immunoelectron microscopy assessment of NF-κB (10 nm nanoparticles) and Sec22b (20 nm nanoparticles) expression in nonstimulated (NS) and LPS-treated BMDCs. Red arrowheads indicate regions where both NF-κB and Sec22b nanoparticles were at a ≤ 100 nm distance (see also Supplemental Fig. 5). (E) Assessment of Sec22b (green) colocalization with nucleolar marker fibrillarin (red). Rightmost panels show orthogonal views of the nuclear region (see also Supplemental Videos 1 and 2). (F) Quantification of Sec22b and fibrillarin colocalization in the nucleus. Image panels are representative of three independent experiments. (G) NF-κB was immunoprecipitated from whole-cell extracts of unstimulated BMDCs and the resulting immune complexes were subjected to blotting using Abs against NF-κB, IκBα, and Sec22b, along with 4% of the initial lysates (input). Images are representative of two experiments. In (A), (D), and (E), scale bars represent 5 μm (white), 500 nm (black), and 100 nm (red). In (B), (C) and (F), data are presented as mean ± SEM of three independent experiments (≥ 10 cells per experiment); each point represents the measurement of a single cell. **p < 0.01; ***p < 0.001; ns, not significant.

Close modal

In dendritic cells, microbial-derived molecules induce the expression of genes encoding proinflammatory mediators and anti-microbial molecules through complex signaling cascades that culminate in the activation of specific transcription factors (56). In the current study, we aimed at further elucidating the mechanisms and pathways associated with the production of NO and inflammatory mediators in BMDCs exposed to LPS. Our main finding is that the secretory pathway, through the action of the SNARE Sec22b, plays a central role in LPS-induced responses by regulating the nuclear translocation of the transcription factor NF-κB (see Visual Abstract).

Whereas the signaling pathways leading to the expression of iNOS and the roles of NO have been explored in great detail in various immune cell types (16, 18), much less is known on the subcellular localization of iNOS. Using electron microscopy and biochemical approaches, Vodovotz and colleagues (22) reported that over 40% of the iNOS activity in whole sonicates was particulate in LPS-activated primary macrophages. They also observed that iNOS was associated with 50–80 nm vesicles of undefined nature, which did not correspond to endosomes, lysosomes, or peroxisomes. Interestingly, the most intense iNOS labeling observed by immunoelectron microscopy was perinuclear, on the trans side of the trans Golgi network (22). Upon phagocytosis, iNOS was found to associate to phagosomes through an actin-dependent mechanism (17). Our results demonstrated that iNOS co-occurs, to varying extents, with proteins found in the ER, ERGIC, and Golgi apparatus. Indeed, iNOS colocalized with ER-Golgi SNAREs Sec22b and Stx5, raising the possibility that the previously observed iNOS-positive vesicular structures originate from these organelles. Because iNOS does not have a signal sequence directing it to the ER lumen (http://www.cbs.dtu.dk/services/TargetP (57)), iNOS is likely to be found on the cytoplasmic side of these secretory pathway organelles via interactions with other proteins. We showed that this topology is conserved in phagosomes, because iNOS was degraded when phagosomes were treated with proteinase K.

Given the large number of genes controlled by NF-κB, this ubiquitous transcription factor plays a central role in the regulation of inflammation, and innate and adaptive immune responses, including LPS-induced responses in dendritic cells (56, 58). In resting cells, NF-κB dimers are sequestrated in an inactive state in the cytoplasm by a member of the inhibitory proteins known as IκBα (59), preventing NF-κB from being recognized by the nuclear import machinery (60). Cell stimulation triggers signaling cascades leading to the phosphorylation of IκB, its ubiquitination, and degradation by the proteasome (61, 62), enabling the nuclear import of NF-κB dimers, their binding to consensus DNA sequences, and the activation of target gene expression (49). Nucleocytoplasmic trafficking of macromolecules such as transcription factors is tightly controlled by importins and exportins, through the recognition of nuclear localization signals and nuclear export signals (63). Using pharmacological inhibitors, we obtained evidence that the ER-Golgi circuitry contributes to optimal LPS-induced responses in BMDCs, through the regulation of MAPK phosphorylation and NF-κB nuclear translocation. The fact that ERK1/2 and p38 are involved in LPS-induced NO release (64) led us to the finding that secretory pathway blockade deregulated JNK (p54) phosphorylation and precluded IκBα degradation. This may explain in part why NO secretion decreases. Consistent with our findings, previous studies with RAW264.7 macrophages and with primary mouse astrocytes revealed that ER stress induced by either tunicamycin or brefeldin A inhibited LPS-induced iNOS expression (65, 66). These findings highlight a previously unappreciated role for the ER-Golgi secretory pathway in the translocation of NF-κB to the nucleus.

Through their role in membrane fusion, the ER/ERGIC-resident SNAREs Sec22b and Stx5 participate in various cellular processes including secretory autophagy (52), embryonic development (15), and plasma membrane expansion (67). These two SNAREs also play a key role in phagosome maturation and function by regulating the delivery of ER- and ERGIC-resident proteins to phagosomes in macrophages and dendritic cells. Remarkably, we found that Sec22b was needed for the expression, at the mRNA and protein levels, of iNOS, TNF, and IL-6. In immune cells, LPS-induced responses are largely dependent on NF-κB, as it regulates the expression of several genes including iNOS, IL-6, and TNF (49). Sec22b knockdown hindered MAPK phosphorylation and IκBα degradation, which implicates this SNARE in the MAPK-mediated regulation of NO release (64). Aside from the effect of Sec22b on IκBα degradation and NF-κB translocation, we do not exclude the possibility that Sec22b knockdown alters ER-Golgi physiology to the extent that it may alter MAPK phosphorylation via an indirect mechanism. Because of their amenability to genetic manipulation (10, 37), we used the immortalized dendritic cell line JAWS-II to study the impact of Sec22b knockdown on NF-κB dynamics. Although different (68), JAWS-II cells are derived from BMDCs of C57BL/6 mice and behave very similarly in a variety of contexts ranging from infection to Ag presentation (10, 6971). Our findings were also corroborated with the RAW264.7 cell line. Whether Sec22b controls NF-κB translocation in other leukocytes remains to be determined.

The role of Sec22b on NF-κB translocation was surprising but not without precedent. For one, ER- and Golgi-resident proteins such as HSP90 and golgin-97 have been implicated in the translocation of NF-κB to the nucleus (72, 73). Because we observed that NF-κB was present in the ERGIC and Golgi, it is reasonable to postulate that Sec22b plays an active role in the cytoplasmic–nuclear shuttling of NF-κB. Second, SNAREs and their chaperones are required for nuclear pore assembly and sealing (75). Our finding that Sec22b and NF-κB co-occurred in the cytoplasm and nucleus further supports the notion that Sec22b is part of the circuitry that, with help from importins (60, 63), shepherds NF-κB translocation.

There is growing evidence that SNAREs and alternatively spliced isoforms access the nucleus where they perform unsuspected functions (7479). In a study aimed at determining the subcellular localization of Stx17, it was found that this SNARE is present in both the cytoplasm and nucleus of several cell types (79). The absence of homology with known nuclear localization signal consensus sequences led the authors to speculate about the existence of a yet uncharacterized nuclear transport mechanism for Stx17. The meaning of the presence of Stx17 in the nucleus remains to be elucidated. Similarly, the SNARE SNAP-47 was found in the ER, ERGIC, and nucleus of HeLa cells (77). Further analyses revealed the presence of several putative nuclear localization signals and two nuclear export signals in SNAP-47. Treatment of HeLa cells with leptomycin B, which inhibits CRM-1/exportin1-dependent nuclear export led to nuclear accumulation of SNAP-47, suggesting a functional nuclear export signal in SNAP-47 (77). SNAP-47 was previously shown to interact with Stx3 (76). Interestingly, a splice variant of Stx3 was found to undergo nuclear translocation, to bind to several transcription factors, and to act as a transcriptional activator (74). An isoform of Stx1, lacking a transmembrane domain, was also found to localize in the nucleus (78). The observation that Sec22b was present in nonnucleolar compartments of the nucleus raises the interesting possibility that Sec22b partakes in nuclear dynamics and gene regulation. Because the a, b, and c isoforms of Sec22 possess transmembrane domains (53), we do not exclude the possibility that Sec22b undergoes cleavage prior to its nuclear translocation. Hence, how Sec22b migrates to the nucleus is the subject of future research.

In sum, our findings demonstrated that the SNARE Sec22b controls inflammatory effector release by facilitating the translocation of NF-κB from the secretory pathway to the nucleus.

We are grateful to D. S. Amigorena for the kind gift of shRNA-transduced JAWS-II cells, J. Tremblay for assistance in immunofluorescence experiments, and A. Nakamura for assistance in electron microscopy. The visual abstract was created with BioRender.com.

This work was supported by Natural Science and Engineering Research Council of Canada Grant 1055043 (to A.D.) and by Canadian Institutes of Health Research (CIHR) Grant PJT-159647 (to S.S.). A.D. is the holder of the Canada Research Chair on the Biology of intracellular parasitism. G.A.D. was partially supported by a CIHR Frederick Banting and Charles Best Doctoral Award and by bridging funds from the Centre for Host-Parasite Interactions.

G.A.D., R.D., C.M., and A.D. conceived and designed the experiments. G.A.D., R.D., C.M., J.D., and A.F. performed the experiments. G.A.D., R.D., C.M., J.D., A.F., S.S., and A.D. analyzed the data. S.S. contributed reagents/materials/analysis tools. G.A.D., C.M., and A.D. wrote the paper. All authors discussed the findings and commented on the manuscript.

The online version of this article contains supplemental material.

Abbreviations used in this article

BMDC

bone marrow-derived dendritic cell

ER

endoplasmic reticulum

ERGIC

ER-Golgi intermediate compartment

iNOS

inducible NO synthase

IP

immunoprecipitation

PGN

peptidoglycan

shRNA

short hairpin RNA

siRNA

small interfering RNA

SNARE

soluble N-ethylmaleimide-sensitive factor attachment protein receptor

Stx

syntaxin

1.
Gordon
S.
2016
.
Phagocytosis: an immunobiologic process.
Immunity
44
:
463
475
.
2.
Guermonprez
P.
,
L.
Saveanu
,
M.
Kleijmeer
,
J.
Davoust
,
P.
Van Endert
,
S.
Amigorena
.
2003
.
ER-phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells.
Nature
425
:
397
402
.
3.
Bonifacino
J. S.
,
B. S.
Glick
.
2004
.
The mechanisms of vesicle budding and fusion.
Cell
116
:
153
166
.
4.
Murray
R. Z.
,
J. L.
Stow
.
2014
.
Cytokine secretion in macrophages: SNAREs, Rabs, and membrane trafficking.
Front. Immunol.
DOI: 10.3389/fimmu.2014.00538.
5.
Levin
R.
,
S.
Grinstein
,
J.
Canton
.
2016
.
The life cycle of phagosomes: formation, maturation, and resolution.
Immunol. Rev.
273
:
156
179
.
6.
Gagnon
E.
,
S.
Duclos
,
C.
Rondeau
,
E.
Chevet
,
P. H.
Cameron
,
O.
Steele-Mortimer
,
J.
Paiement
,
J. J.
Bergeron
,
M.
Desjardins
.
2002
.
Endoplasmic reticulum-mediated phagocytosis is a mechanism of entry into macrophages.
Cell
110
:
119
131
.
7.
Murray
R. Z.
,
J. G.
Kay
,
D. G.
Sangermani
,
J. L.
Stow
.
2005
.
A role for the phagosome in cytokine secretion.
Science
310
:
1492
1495
.
8.
Dingjan
I.
,
P. T. A.
Linders
,
D. R. J.
Verboogen
,
N. H.
Revelo
,
M.
Ter Beest
,
G.
van den Bogaart
.
2018
.
Endosomal and phagosomal SNAREs.
Physiol. Rev.
98
:
1465
1492
.
9.
Pagan
J. K.
,
F. G.
Wylie
,
S.
Joseph
,
C.
Widberg
,
N. J.
Bryant
,
D. E.
James
,
J. L.
Stow
.
2003
.
The t-SNARE syntaxin 4 is regulated during macrophage activation to function in membrane traffic and cytokine secretion.
Curr. Biol.
13
:
156
160
.
10.
Cebrian
I.
,
G.
Visentin
,
N.
Blanchard
,
M.
Jouve
,
A.
Bobard
,
C.
Moita
,
J.
Enninga
,
L. F.
Moita
,
S.
Amigorena
,
A.
Savina
.
2011
.
Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells.
Cell
147
:
1355
1368
.
11.
Nair-Gupta
P.
,
A.
Baccarini
,
N.
Tung
,
F.
Seyffer
,
O.
Florey
,
Y.
Huang
,
M.
Banerjee
,
M.
Overholtzer
,
P. A.
Roche
,
R.
Tampé
, et al
2014
.
TLR signals induce phagosomal MHC-I delivery from the endosomal recycling compartment to allow cross-presentation.
Cell
158
:
506
521
.
12.
Abuaita
B. H.
,
K. M.
Burkholder
,
B. R.
Boles
,
M. X.
O’Riordan
.
2015
.
The endoplasmic reticulum stress sensor inositol-requiring enzyme 1α augments bacterial killing through sustained oxidant production.
MBio
6
:
e00705-15
.
13.
Alloatti
A.
,
D. C.
Rookhuizen
,
L.
Joannas
,
J. M.
Carpier
,
S.
Iborra
,
J. G.
Magalhaes
,
N.
Yatim
,
P.
Kozik
,
D.
Sancho
,
M. L.
Albert
,
S.
Amigorena
.
2017
.
Critical role for Sec22b-dependent antigen cross-presentation in antitumor immunity. [Published erratum appears in 2018 J. Exp. Med. 215: 1001.]
J. Exp. Med.
214
:
2231
2241
.
14.
Kagan
J. C.
,
M.-P.
Stein
,
M.
Pypaert
,
C. R.
Roy
.
2004
.
Legionella subvert the functions of Rab1 and Sec22b to create a replicative organelle.
J. Exp. Med.
199
:
1201
1211
.
15.
Wu
S. J.
,
R.
Khoriaty
,
S. H.
Kim
,
K. S.
O’Shea
,
G.
Zhu
,
M.
Hoenerhoff
,
C.
Zajac
,
K.
Oravecz-Wilson
,
T.
Toubai
,
Y.
Sun
, et al
2019
.
SNARE protein SEC22B regulates early embryonic development.
Sci. Rep.
DOI: 10.1038/s41598-019-46536-7.
16.
Xue
Q.
,
Y.
Yan
,
R.
Zhang
,
H.
Xiong
.
2018
.
Regulation of iNOS on immune cells and its role in diseases.
Int. J. Mol. Sci.
DOI: 10.3390/ijms19123805.
17.
Miller
B. H.
,
R. A.
Fratti
,
J. F.
Poschet
,
G. S.
Timmins
,
S. S.
Master
,
M.
Burgos
,
M. A.
Marletta
,
V.
Deretic
.
2004
.
Mycobacteria inhibit nitric oxide synthase recruitment to phagosomes during macrophage infection.
Infect. Immun.
72
:
2872
2878
.
18.
Bogdan
C.
2015
.
Nitric oxide synthase in innate and adaptive immunity: an update.
Trends Immunol.
36
:
161
178
.
19.
Qadoumi
M.
,
I.
Becker
,
N.
Donhauser
,
M.
Röllinghoff
,
C.
Bogdan
.
2002
.
Expression of inducible nitric oxide synthase in skin lesions of patients with American cutaneous leishmaniasis.
Infect. Immun.
70
:
4638
4642
.
20.
Tzeng
E.
,
T. R.
Billiar
,
P. D.
Robbins
,
M.
Loftus
,
D. J.
Stuehr
.
1995
.
Expression of human inducible nitric oxide synthase in a tetrahydrobiopterin (H4B)-deficient cell line: H4B promotes assembly of enzyme subunits into an active dimer.
Proc. Natl. Acad. Sci. USA
92
:
11771
11775
.
21.
Webb
J. L.
,
M. W.
Harvey
,
D. W.
Holden
,
T. J.
Evans
.
2001
.
Macrophage nitric oxide synthase associates with cortical actin but is not recruited to phagosomes.
Infect. Immun.
69
:
6391
6400
.
22.
Vodovotz
Y.
,
D.
Russell
,
Q. W.
Xie
,
C.
Bogdan
,
C.
Nathan
.
1995
.
Vesicle membrane association of nitric oxide synthase in primary mouse macrophages.
J. Immunol.
154
:
2914
2925
.
23.
Nakano
T.
,
Y.
Hozumi
,
K.
Goto
,
I.
Wakabayashi
.
2012
.
Involvement of diacylglycerol kinase γ in modulation of iNOS synthesis in Golgi apparatus of vascular endothelial cells.
Naunyn Schmiedebergs Arch. Pharmacol.
385
:
787
795
.
24.
Ranatunga
D.
,
C. M.
Hedrich
,
F.
Wang
,
D. W.
McVicar
,
N.
Nowak
,
T.
Joshi
,
L.
Feigenbaum
,
L. R.
Grant
,
S.
Stäger
,
J. H.
Bream
.
2009
.
A human IL10 BAC transgene reveals tissue-specific control of IL-10 expression and alters disease outcome.
Proc. Natl. Acad. Sci. USA
106
:
17123
17128
.
25.
Hammami
A.
,
T.
Charpentier
,
M.
Smans
,
S.
Stäger
.
2015
.
IRF-5-mediated inflammation limits CD8+ T cell expansion by inducing HIF-1α and impairing dendritic cell functions during Leishmania infection.
PLoS Pathog.
DOI: 10.1371/journal.ppat.1004938.
26.
Arango Duque
G.
,
M.
Fukuda
,
A.
Descoteaux
.
2013
.
Synaptotagmin XI regulates phagocytosis and cytokine secretion in macrophages.
J. Immunol.
190
:
1737
1745
.
27.
Arango Duque
G.
,
A.
Jardim
,
É.
Gagnon
,
M.
Fukuda
,
A.
Descoteaux
.
2019
.
The host cell secretory pathway mediates the export of Leishmania virulence factors out of the parasitophorous vacuole.
PLoS Pathog.
DOI: 10.1371/journal.ppat.1007982.
28.
Wei
M.-T.
,
K.-F.
Hua
,
J.
Hsu
,
A.
Karmenyan
,
K.-Y.
Tseng
,
C.-H.
Wong
,
H.-Y.
Hsu
,
A.
Chiou
.
2007
.
The interaction of lipopolysaccharide with membrane receptors on macrophages pretreated with extract of Reishi polysaccharides measured by optical tweezers.
Opt. Express
15
:
11020
11032
.
29.
Classen
A.
,
J.
Lloberas
,
A.
Celada
.
2009
.
Macrophage activation: classical vs. alternative.
In
Macrophages and Dendritic Cells: Methods and Protocols.
N. E.
Reiner
.
Humana Press
,
Totowa, NJ
, p.
29
43
.
30.
de Chaumont
F.
,
S.
Dallongeville
,
N.
Chenouard
,
N.
Hervé
,
S.
Pop
,
T.
Provoost
,
V.
Meas-Yedid
,
P.
Pankajakshan
,
T.
Lecomte
,
Y.
Le Montagner
, et al
2012
.
Icy: an open bioimage informatics platform for extended reproducible research.
Nat. Methods
9
:
690
696
.
31.
Lagache
T.
,
N.
Sauvonnet
,
L.
Danglot
,
J.-C.
Olivo-Marin
.
2015
.
Statistical analysis of molecule colocalization in bioimaging.
Cytometry A
87
:
568
579
.
32.
Dufour
A.
,
R.
Thibeaux
,
E.
Labruyère
,
N.
Guillén
,
J. C.
Olivo-Marin
.
2011
.
3-D active meshes: fast discrete deformable models for cell tracking in 3-D time-lapse microscopy.
IEEE Trans. Image Process.
20
:
1925
1937
.
33.
Vinet
A. F.
,
M.
Fukuda
,
S. J.
Turco
,
A.
Descoteaux
.
2009
.
The Leishmania donovani lipophosphoglycan excludes the vesicular proton-ATPase from phagosomes by impairing the recruitment of synaptotagmin V.
PLoS Pathog.
DOI: 10.1371/journal.ppat.1000628.
34.
Arango Duque
G.
,
M.
Fukuda
,
S. J.
Turco
,
S.
Stäger
,
A.
Descoteaux
.
2014
.
Leishmania promastigotes induce cytokine secretion in macrophages through the degradation of synaptotagmin XI.
J. Immunol.
193
:
2363
2372
.
35.
Fabié
A.
,
L. T.
Mai
,
X.
Dagenais-Lussier
,
A.
Hammami
,
J.
van Grevenynghe
,
S.
Stäger
.
2018
.
IRF-5 promotes cell death in CD4 T cells during chronic infection.
Cell Rep.
24
:
1163
1175
.
36.
Matte
C.
,
A.
Descoteaux
.
2010
.
Leishmania donovani amastigotes impair gamma interferon-induced STAT1α nuclear translocation by blocking the interaction between STAT1α and importin-α5.
Infect. Immun.
78
:
3736
3743
.
37.
Awasthi
S.
,
R. A.
Cox
.
2003
.
Transfection of murine dendritic cell line (JAWS II) by a nonviral transfection reagent.
Biotechniques
35
:
600
602
,
604
.
38.
Glynne
P. A.
,
K. E. A.
Darling
,
J.
Picot
,
T. J.
Evans
.
2002
.
Epithelial inducible nitric-oxide synthase is an apical EBP50-binding protein that directs vectorial nitric oxide output.
J. Biol. Chem.
277
:
33132
33138
.
39.
Andrew
P. J.
,
B.
Mayer
.
1999
.
Enzymatic function of nitric oxide synthases.
Cardiovasc. Res.
43
:
521
531
.
40.
Loughran
P. A.
,
D. B.
Stolz
,
Y.
Vodovotz
,
S. C.
Watkins
,
R. L.
Simmons
,
T. R.
Billiar
.
2005
.
Monomeric inducible nitric oxide synthase localizes to peroxisomes in hepatocytes.
Proc. Natl. Acad. Sci. USA
102
:
13837
13842
.
41.
Helms
J. B.
,
J. E.
Rothman
.
1992
.
Inhibition by brefeldin A of a Golgi membrane enzyme that catalyses exchange of guanine nucleotide bound to ARF.
Nature
360
:
352
354
.
42.
Misumi
Y.
,
Y.
Misumi
,
K.
Miki
,
A.
Takatsuki
,
G.
Tamura
,
Y.
Ikehara
.
1986
.
Novel blockade by brefeldin A of intracellular transport of secretory proteins in cultured rat hepatocytes.
J. Biol. Chem.
261
:
11398
11403
.
43.
Sohda
M.
,
Y.
Misumi
,
S.
Yoshimura
,
N.
Nakamura
,
T.
Fusano
,
S.
Ogata
,
S.
Sakisaka
,
Y.
Ikehara
.
2007
.
The interaction of two tethering factors, p115 and COG complex, is required for Golgi integrity.
Traffic
8
:
270
284
.
44.
Misinzo
G.
,
P. L.
Delputte
,
H. J.
Nauwynck
.
2008
.
Inhibition of endosome-lysosome system acidification enhances porcine circovirus 2 infection of porcine epithelial cells.
J. Virol.
82
:
1128
1135
.
45.
Mollenhauer
H. H.
,
D. J.
Morré
,
L. D.
Rowe
.
1990
.
Alteration of intracellular traffic by monensin; mechanism, specificity and relationship to toxicity.
Biochim. Biophys. Acta
1031
:
225
246
.
46.
Griffiths
G.
,
P.
Quinn
,
G.
Warren
.
1983
.
Dissection of the Golgi complex. I. Monensin inhibits the transport of viral membrane proteins from medial to trans Golgi cisternae in baby hamster kidney cells infected with Semliki Forest virus.
J. Cell Biol.
96
:
835
850
.
47.
Eisenberg-Lerner
A.
,
R.
Benyair
,
N.
Hizkiahou
,
N.
Nudel
,
R.
Maor
,
M. P.
Kramer
,
M. D.
Shmueli
,
I.
Zigdon
,
M.
Cherniavsky Lev
,
A.
Ulman
, et al
2020
.
Golgi organization is regulated by proteasomal degradation.
Nat. Commun.
DOI: 10.1038/s41467-019-14038-9.
48.
Stechmann
B.
,
S.-K.
Bai
,
E.
Gobbo
,
R.
Lopez
,
G.
Merer
,
S.
Pinchard
,
L.
Panigai
,
D.
Tenza
,
G.
Raposo
,
B.
Beaumelle
, et al
2010
.
Inhibition of retrograde transport protects mice from lethal ricin challenge.
Cell
141
:
231
242
.
49.
Liu
T.
,
L.
Zhang
,
D.
Joo
,
S.-C.
Sun
.
2017
.
NF-κB signaling in inflammation.
Signal Transduct. Target. Ther.
DOI: 10.1038/sigtrans.2017.23.
50.
Aktan
F.
2004
.
iNOS-mediated nitric oxide production and its regulation.
Life Sci.
75
:
639
653
.
51.
Jahn
R.
,
R. H.
Scheller
.
2006
.
SNAREs--engines for membrane fusion.
Nat. Rev. Mol. Cell Biol.
7
:
631
643
.
52.
Kimura
T.
,
J.
Jia
,
S.
Kumar
,
S. W.
Choi
,
Y.
Gu
,
M.
Mudd
,
N.
Dupont
,
S.
Jiang
,
R.
Peters
,
F.
Farzam
, et al
2017
.
Dedicated SNAREs and specialized TRIM cargo receptors mediate secretory autophagy.
EMBO J.
36
:
42
60
.
53.
Sun
W.
,
B.-x.
Tian
,
S.-h.
Wang
,
P.-j.
Liu
,
Y.-c.
Wang
.
2020
.
The function of SEC22B and its role in human diseases.
Cytoskeleton
77
:
303
312
.
54.
Kodiha
M.
,
P.
Bański
,
U.
Stochaj
.
2011
.
Computer-based fluorescence quantification: a novel approach to study nucleolar biology.
BMC Cell Biol.
DOI: 10.1186/1471-2121-12-25.
55.
Ochs
R. L.
,
M. A.
Lischwe
,
W. H.
Spohn
,
H.
Busch
.
1985
.
Fibrillarin: a new protein of the nucleolus identified by autoimmune sera.
Biol. Cell
54
:
123
133
.
56.
Qian
C.
,
X.
Cao
.
2018
.
Dendritic cells in the regulation of immunity and inflammation.
Semin. Immunol.
35
:
3
11
.
57.
Almagro Armenteros
J. J.
,
M.
Salvatore
,
O.
Emanuelsson
,
O.
Winther
,
G.
von Heijne
,
A.
Elofsson
,
H.
Nielsen
.
2019
.
Detecting sequence signals in targeting peptides using deep learning.
Life Sci. Alliance.
DOI: 10.26508/lsa.201900429.
58.
Hofer
S.
,
M.
Rescigno
,
F.
Granucci
,
S.
Citterio
,
M.
Francolini
,
P.
Ricciardi-Castagnoli
.
2001
.
Differential activation of NF-κ B subunits in dendritic cells in response to Gram-negative bacteria and to lipopolysaccharide.
Microbes Infect.
3
:
259
265
.
59.
Ghosh
S.
,
D.
Baltimore
.
1990
.
Activation in vitro of NF-κ B by phosphorylation of its inhibitor I κ B.
Nature
344
:
678
682
.
60.
Turpin
P.
,
R. T.
Hay
,
C.
Dargemont
.
1999
.
Characterization of IkappaBalpha nuclear import pathway.
J. Biol. Chem.
274
:
6804
6812
.
61.
Henkel
T.
,
T.
Machleidt
,
I.
Alkalay
,
M.
Krönke
,
Y.
Ben-Neriah
,
P. A.
Baeuerle
.
1993
.
Rapid proteolysis of I κ B-α is necessary for activation of transcription factor NF-κ B.
Nature
365
:
182
185
.
62.
Kanarek
N.
,
Y.
Ben-Neriah
.
2012
.
Regulation of NF-κB by ubiquitination and degradation of the IκBs.
Immunol. Rev.
246
:
77
94
.
63.
Aggarwal
A.
,
D. K.
Agrawal
.
2014
.
Importins and exportins regulating allergic immune responses.
Mediators Inflamm.
DOI: 10.1155/2014/476357.
64.
Feng
G.-J.
,
H. S.
Goodridge
,
M. M.
Harnett
,
X.-Q.
Wei
,
A. V.
Nikolaev
,
A. P.
Higson
,
F.-Y.
Liew
.
1999
.
Extracellular signal-related kinase (ERK) and p38 mitogen-activated protein (MAP) kinases differentially regulate the lipopolysaccharide-mediated induction of inducible nitric oxide synthase and IL-12 in macrophages: Leishmania phosphoglycans subvert macrophage IL-12 production by targeting ERK MAP kinase.
J. Immunol.
163
:
6403
6412
.
65.
Hosoi
T.
,
J.
Noguchi
,
M.
Takakuwa
,
M.
Honda
,
Y.
Okuma
,
Y.
Nomura
,
K.
Ozawa
.
2014
.
Inhibition of inducible nitric oxide synthase and interleukin-1β expression by tunicamycin in cultured glial cells exposed to lipopolysaccharide.
Brain Res.
1558
:
11
17
.
66.
Ho
H.-J.
,
D.-Y.
Huang
,
F.-M.
Ho
,
L.-T.
Lee
,
W.-W.
Lin
.
2012
.
Inhibition of lipopolysaccharide-induced inducible nitric oxide synthase expression by endoplasmic reticulum stress.
Cell. Signal.
24
:
2166
2178
.
67.
Petkovic
M.
,
A.
Jemaiel
,
F.
Daste
,
C. G.
Specht
,
I.
Izeddin
,
D.
Vorkel
,
J.-M.
Verbavatz
,
X.
Darzacq
,
A.
Triller
,
K. H.
Pfenninger
, et al
2014
.
The SNARE Sec22b has a non-fusogenic function in plasma membrane expansion.
Nat. Cell Biol.
16
:
434
444
.
68.
Jørgensen
T. N.
,
C.
Haase
,
B. K.
Michelsen
.
2002
.
Treatment of an immortalized APC cell line with both cytokines and LPS ensures effective T-cell activation in vitro.
Scand. J. Immunol.
56
:
492
503
.
69.
Hume
P. S.
,
J.
He
,
K.
Haskins
,
K. S.
Anseth
.
2012
.
Strategies to reduce dendritic cell activation through functional biomaterial design.
Biomaterials
33
:
3615
3625
.
70.
Jiang
X.
,
C.
Shen
,
J.
Rey-Ladino
,
H.
Yu
,
R. C.
Brunham
.
2008
.
Characterization of murine dendritic cell line JAWS II and primary bone marrow-derived dendritic cells in Chlamydia muridarum antigen presentation and induction of protective immunity.
Infect. Immun.
76
:
2392
2401
.
71.
Otsu
S.
,
K.
Gotoh
,
T.
Yamashiro
,
J.
Yamagata
,
K.
Shin
,
T.
Fujioka
,
A.
Nishizono
.
2006
.
Transfer of antigen-pulsed dendritic cells induces specific T-Cell proliferation and a therapeutic effect against long-term Helicobacter pylori infection in mice.
Infect. Immun.
74
:
984
993
.
72.
Feldman
I.
,
G. M.
Feldman
,
C.
Mobarak
,
J. C.
Dunkelberg
,
K. K.
Leslie
.
2007
.
Identification of proteins within the nuclear factor-κ B transcriptional complex including estrogen receptor-α.
Am. J. Obstet. Gynecol.
196
:
394.e1
394.e13
.
73.
Hsu
R.-M.
,
C.-Y.
Zhong
,
C.-L.
Wang
,
W.-C.
Liao
,
C.
Yang
,
S.-Y.
Lin
,
J.-W.
Lin
,
H.-Y.
Cheng
,
P.-Y.
Li
,
C.-J.
Yu
.
2018
.
Golgi tethering factor golgin-97 suppresses breast cancer cell invasiveness by modulating NF-κB activity.
Cell Commun. Signal.
DOI: 10.1186/s12964-018-0230-5.
74.
Baur
T.
,
K.
Ramadan
,
A.
Schlundt
,
J.
Kartenbeck
,
H. H.
Meyer
.
2007
.
NSF- and SNARE-mediated membrane fusion is required for nuclear envelope formation and completion of nuclear pore complex assembly in Xenopus laevis egg extracts.
J. Cell Sci.
120
:
2895
2903
.
75.
Giovannone
A. J.
,
C.
Winterstein
,
P.
Bhattaram
,
E.
Reales
,
S. H.
Low
,
J. E.
Baggs
,
M.
Xu
,
M. A.
Lalli
,
J. B.
Hogenesch
,
T.
Weimbs
.
2018
.
Soluble syntaxin 3 functions as a transcriptional regulator.
J. Biol. Chem.
293
:
5478
5491
.
76.
Jurado
S.
,
D.
Goswami
,
Y.
Zhang
,
A. J.
Miñano Molina
,
T. C.
Südhof
,
R. C.
Malenka
.
2013
.
LTP requires a unique postsynaptic SNARE fusion machinery.
Neuron
77
:
542
558
.
77.
Kuster
A.
,
S.
Nola
,
F.
Dingli
,
B.
Vacca
,
C.
Gauchy
,
J.-C.
Beaujouan
,
M.
Nunez
,
T.
Moncion
,
D.
Loew
,
E.
Formstecher
,
T.
Galli
,
V.
Proux-Gillardeaux
.
2015
.
The Q-soluble-N-ethylmaleimide-sensitive factor attachment protein receptor (Q-SNARE) SNAP-47 regulates trafficking of selected vesicle-associated membrane proteins (VAMPs).
J. Biol. Chem.
290
:
28057
28069
.
78.
Pereira
S.
,
A.
Massacrier
,
P.
Roll
,
A.
Vérine
,
M.-C.
Etienne-Grimaldi
,
Y.
Poitelon
,
A.
Robaglia-Schlupp
,
S.
Jamali
,
N.
Roeckel-Trevisiol
,
B.
Royer
, et al
2008
.
Nuclear localization of a novel human syntaxin 1B isoform.
Gene
423
:
160
171
.
79.
Zhang
Q.
,
J.
Li
,
M.
Deavers
,
J. L.
Abbruzzese
,
L.
Ho
.
2005
.
The subcellular localization of syntaxin 17 varies among different cell types and is altered in some malignant cells.
J. Histochem. Cytochem.
53
:
1371
1382
.

The authors have no financial conflicts of interest.