The exocyst complex is an essential component of the mammalian constitutive secretory pathway

Secreted proteins fulfill a vast array of functions, including immunity, signaling, and extracellular matrix remodeling. In the trans-Golgi network, proteins destined for constitutive secretion are sorted into post-Golgi carriers which fuse with the plasma membrane. The molecular machinery involved is poorly understood. Here, we have used kinetic trafficking assays and transient CRISPR-KO to study biosynthetic sorting from the Golgi to the plasma membrane. Depletion of all canonical exocyst subunits causes cargo accumulation in post-Golgi carriers. Exocyst subunits are recruited to and co-localize with carriers. Exocyst abrogation followed by kinetic trafficking assays of soluble cargoes results in intracellular cargo accumulation. Unbiased secretomics reveals impairment of soluble protein secretion after exocyst subunit knockout. Importantly, in specialized cell types, the loss of exocyst prevents constitutive secretion of antibodies in lymphocytes and of leptin in adipocytes. These data identify exocyst as the functional tether of secretory post-Golgi carriers at the plasma membrane and an essential component of the mammalian constitutive secretory pathway.

The complex process of membrane trafficking is fundamental to cellular organization. Proteins are transported from their site of synthesis in the ER to the Golgi apparatus, where they are sorted to different subcellular localizations, such as the endolysosomal system or directly to the plasma membrane for secretion (Chen et al., 2017; Stalder and Gershlick, 2020). In higher eukaryotes, ∼12% of all proteins are secreted from the cell (Kanapin et al., 2003; Uhlén et al., 2019; Thul et al., 2017), where they fulfill a vast array of different functions, including cell signaling, the immune response, and extracellular matrix remodeling (Stalder and Gershlick, 2020).

Soluble secreted proteins are synthesized in the ER. After proper folding, they are trafficked to the Golgi apparatus in COPII carriers, where they are glycosylated (Xu and Ng, 2015; Chen et al., 2017; Patterson et al., 2008; Clermont et al., 1995; Keller et al., 2001). At the trans-Golgi apparatus, soluble secreted proteins are sorted into pleomorphic post-Golgi tubular carriers (Stalder and Gershlick, 2020). These carriers are then trafficked directly to the plasma membrane, where they fuse and their contents are delivered to the extracellular milieu (Polishchuk et al., 2003; Stalder and Gershlick, 2020).

The fusion of intracellular carriers is understood to be a two-step process. Molecular tethers, either long coil-coiled tethers or multisubunit tethering complexes, interact with the carrier prior to the subsequent SNARE-mediated fusion (Lupashin and Sztul, 2005; Whyte and Munro, 2001). The initial “capture” with the tether is therefore essential for correct vesicle targeting and fidelity of cargo delivery.

Long coil-coiled tethers tend to be large (>60 kD) and form a coiled-coil domain structure. Examples of long coil-coiled tethering factors include the golgin family of proteins at the Golgi apparatus, and EEA1 on the endosomes (Lupashin and Sztul, 2005; Murray et al., 2016). They interact with the acceptor compartment on one side and the incoming vesicle on the other, “bringing” the vesicle closer to the target membrane (Murray et al., 2016).

The second class of membrane tether are multisubunit tethering complexes, which include Golgi-associated retrograde protein (Pérez-Victoria et al., 2009) complex on the trans-Golgi network, the conserved oligomeric Golgi (Smith and Lupashin, 2008) complex on the medial-Golgi, and the homotypic fusion and protein sorting (Spang, 2016) complex on the lysosomal-endosomal system. These tend to be large multisubunit assemblies and are sometimes, but not exclusively, complexes associated with tethering containing helical rods (CATCHR), which, when assembled, form helical bundles arranged in tandem through a coiled-coil region at the N-terminus (Chou et al., 2016). Multisubunit tethering complexes have also been found to be important for proper SNARE assembly in addition to vesicle catching (Pérez-Victoria et al., 2009).

On the plasma membrane, two molecular tethers have been identified. The long coil-coiled protein ELKS (also: ERC, RAB6IP2, or CAST) localizes to patches on the plasma membrane termed “fusion hotspots” due to the higher frequency of vesicle fusion events at these sites (Deguchi-Tawarada et al., 2004; Monier et al., 2002; Nakata et al., 1999; Wang et al., 2002; Fourriere et al., 2019). ELKS was identified as an interactor and probable effector of all three RAB6 isoforms (RAB6A, A′, and B; Monier et al., 2002). ELKS is implicated in secretion of neuropeptide Y in RAB6, MICAL3, and RAB8 positive carriers (Grigoriev et al., 2007, 2011), and synaptic vesicle tethering to the plasma membrane in a neuronal cell model (Nyitrai et al., 2020).

The second tether associated with the plasma membrane is the CATCHR protein complex exocyst (Wu and Guo, 2015). Exocyst is an octamer composed of EXOC1-8 and was originally identified in yeast as important for secretion based on its localization to the plasma membrane and the Sec phenotype (Stalder and Gershlick, 2020). In mammalian cells, exocyst components localize to the Golgi and plasma membrane as well as at vesicle fusion points (Yeaman et al., 2001; Ahmed et al., 2018; Heider and Munson, 2012). Although exocyst is essential for endosomal recycling to the plasma membrane (Grindstaff et al., 1998; Lipschutz et al., 2000; Langevin et al., 2005; Yeaman et al., 2004; Andersen and Yeaman, 2010; Wu and Guo, 2015; Heider and Munson, 2012), ciliogenesis (Rogers et al., 2004; Zuo et al., 2009; Feng et al., 2012), autophagy (Bodemann et al., 2011; Sáez et al., 2019), innate immunity (Chien et al., 2006; Sáez et al., 2019), and cytokenesis (Chen et al., 2006; Fielding et al., 2005; Neto et al., 2013), the role of exocyst in constitutive protein secretion remains unclear. Inhibition of exocyst with antibodies does not affect delivery of tsVSV-G, a marker of the secretory pathway, to the plasma membrane (Yeaman et al., 2001; Grigoriev et al., 2007); however, this could be due to ineffective inhibition by antibodies as the epitope may not be exposed under certain exocyst structural conformations (Inamdar et al., 2016). Conversely, some evidence indicates that exocyst is important for biosynthetic sorting to the plasma membrane and depletion of EXOC7 decreases tsVSV-G delivery to the plasma membrane (Liu et al., 2007). It is therefore of interest to examine whether exocyst has a direct role in biosynthetic membrane protein sorting in mammalian cells. Moreover, it is not known if exocyst is necessary or important for soluble protein secretion in mammalian cells (Stalder and Gershlick, 2020; Wu and Guo, 2015).

To investigate the functional machinery in protein secretion, we developed a quantitative trafficking assay to study cell-surface delivery from the Golgi apparatus using the retention using selective hooks (RUSH) system. By designing a synthetic type-1 membrane protein based on LAMP1, we can directly observe post-Golgi carriers that co-localize with previously characterized markers and fuse with the plasma membrane. We have used a transient CRISPR-knockout (KO) system to determine that the exocyst complex is essential for the arrival of these carriers to the plasma membrane. We observe exocyst subunits localizing to the post-Golgi carriers on fusion hotspots on the plasma membrane. Kinetic trafficking assays on a set of soluble secreted proteins reveals a broad dependence on exocyst for protein secretion. We performed unbiased proteomics in an endogenous context to exocyst-KO cells and have demonstrated that the exocyst complex is responsible for the majority if not all soluble protein secretion. In addition, we show that important specialized secretory cells require exocyst for the efficient secretion of both antibodies and hormones. We, therefore, define exocyst as the molecular tether for constitutive protein secretion of soluble proteins and as an essential component of the mammalian secretory pathway.

In order to study post-Golgi carriers, we have generated a synthetic protein that allows monitoring of protein delivery to the plasma membrane with proper spatiotemporal kinetics. The single-pass type-1 integral membrane protein LAMP1 is localized to the lysosome at the steady-state level. After synthesis in the ER, LAMP1 traffics via the Golgi apparatus directly to the plasma membrane (Chen et al., 2017) where it is endocytosed, in clathrin-coated vesicles, to be delivered to the endolysosomal system and finally to the lysosome (Chen et al., 2017). Mutations in, or deletion of, the endocytic trafficking motif causes LAMP1 to accumulate on the plasma membrane after exit from the Golgi apparatus in post-Golgi tubular carriers (Chen et al., 2017). To monitor the kinetics of trafficking, we used the RUSH system (Boncompain et al., 2012). LAMP1ΔYQTI was genetically fused to a streptavidin-binding peptide (SBP) and a fluorescent protein (GFP) and coexpressed with streptavidin fused to the ER-retrieval signal KDEL (Munro and Pelham, 1987) in a stable cell line (Fig. 1 A).

After biotin addition, the LAMP1ΔYQTI-RUSH (referred to from here as LAMP1Δ-RUSH) can be observed trafficking with appropriate kinetics (Fig. 1 B and Video 1) as previously observed (Chen et al., 2017). Using lattice-SIM (Structured Illumination Microscopy) live-cell imaging, carriers can be observed budding from the Golgi apparatus (Fig. 1 C and Video 2), trafficking along microtubules (Fig. 1 D and Video 3), and using Total Internal Reflection Fluorescence (TIRF) microscopy, we can observe them fuse with the plasma membrane (Fig. 1 E and Video 4). The LAMP1Δ-RUSH is progressively glycosylated after the addition of biotin (Fig. S1), indicating that the protein trafficks through the secretory pathway and is processed in the same manner as an endogenous cargo.

To quantitatively study this secretory route, we have developed a flow cytometry–based quantitative cell-surface protein delivery assay (Fig. 1 F). Cells stably expressing LAMP1Δ-RUSH were incubated with biotin across a time series of 1 h, the previously established time to achieve a steady-state level (Chen et al., 2017). By labeling intact cells with anti-GFP nanobody fused to a fluorescent mCherry (Buser et al., 2018) and quantitatively measuring both mCherry and GFP levels via flow cytometry, we can detect and quantify LAMP1Δ-RUSH delivery to the plasma membrane. The ratio of the cell surface (mCherry) to total cargo available (GFP) then allows for per-cell quantification of protein cargo arrival at the plasma membrane (Fig. 1 G). In line with previous data, the cargo reaches a steady state ∼1 h after release from the ER with biotin. At 35 min, the assay has a high signal-to-noise but is also sensitive to kinetic changes in trafficking, and this time point was selected for future assays. In summary, the use of the RUSH system with a LAMP1ΔYQTI cargo allows for observation of post-Golgi tubular carriers and their fusion with the plasma membrane with appropriate trafficking kinetics and a quantitative readout.

To validate that the post-Golgi carriers observed using LAMP1Δ-RUSH are representative of secretory carriers, we tested if known markers of these carriers co-localize with LAMP1Δ-RUSH. The small G protein RAB6A/A′ has been observed associated with secretory vesicles that fuse directly with the plasma membrane and has an important role in the fission of the vesicles at the Golgi apparatus and their transport toward the cell surface (Grigoriev et al., 2007; Miserey-Lenkei et al., 2010). Lattice-SIM live-cell imaging showed that overexpressed HALO-RAB6A is associated with tubular carriers emerging from the Golgi apparatus, detaching, and traveling toward the plasma membrane (Fig. 2 A and Video 5). Further, overexpressed HALO-RAB6A co-localized with LAMP1Δ-RUSH at the Golgi apparatus and on these carriers (Fig. 2 B and Video 6), with quantification of carrier co-localization revealing more than 80% of post-Golgi LAMP1 positive carriers have detectable HALO-RAB6A (Fig. 2 C). Another small G protein, RAB8A, associates with exocytotic vesicles in a RAB6-dependent manner through the recruitment of the exchange factor ARHGEF10 (Grigoriev et al., 2011; Shibata et al., 2016). Live-cell imaging demonstrated that both HALO-ARHGEF10 and HALO-RAB8A co-localize with LAMP1Δ-RUSH (Fig. 2, D and F; and Video 7 and Video 8), with significant detectable accumulation on post-Golgi carriers (Fig. 2, E and G) recapitulating previous evidence of a Rab cascade. In conclusion, the post-Golgi carriers observable using LAMP1Δ-RUSH co-localize with a number of markers for secretory carriers.

To demonstrate a functional relationship between RAB6 and LAMP1Δ-RUSH carriers, we transiently abrogated RAB6A using CRISPR-Cas9. Loss of RAB6A drastically and significantly decreased LAMP1Δ-RUSH trafficking to the plasma membrane (Fig. 2 H). Unlike the inactive form of RAB6A (HALO-RAB6A [TN]), overexpression of Rab6A WT (HALO-RAB6A) as well as the dominant active form (HALO-RAB6A [QL]) rescued the defect in cell-surface delivery of LAMP1Δ-RUSH (Fig. 2 H). RAB6A-KO caused accumulation of LAMP1Δ-RUSH in the Golgi apparatus, which was rescued by overexpressing HALO-RAB6A (Fig. 2, I and J), showing that RAB6A is required for the generation of LAMP1Δ-RUSH post-Golgi carriers. Together, these results show that markers of secretory carriers co-localize with LAMP1Δ-RUSH carriers and are necessary for their trafficking from the Golgi, thus demonstrating the LAMP1Δ-RUSH carriers represent the secretory pathway to the plasma membrane, providing a tractable experimental system to study the secretory pathway.

Having established a quantitative system for studying the secretory pathway, we utilized it to study the role of plasma membrane tethers in secretory carrier fusion to the cell surface. The long coil-coiled membrane tether ELKS has been previously associated with the fusion of carriers at the plasma membrane (Fourriere et al., 2019). Consistent with these studies, ELKS co-localizes on post-Golgi carrier fusion hotspots by TIRF microscopy (Fig. S2 A and Video 9). Transient or stable KO of ELKS or double KO of ELKS and its characterized cofactors or homologs by CRISPR-Cas9 was performed and validated by immunoblot or quantitative PCR (qPCR; Fig. S2 C and Fig. S3 F). No significant decrease in cell-surface delivery of LAMP1Δ-RUSH to the cell surface could be detected (Fig. S2, B–E).

Based on previous evidence in yeast model systems we hypothesized that the octameric protein complex exocyst is essential for the fusion of post-Golgi carriers with the plasma membrane (Bowser and Novick, 1991; Bowser et al., 1992; TerBush and Novick, 1995). The exocyst is considered an essential component of eukaryotic cells (Novick et al., 1980; Wang et al., 2015), so to avoid lethality and clonal selection artifacts we performed independent transient CRISPR-Cas9 KO of all canonical exocyst components. Using this approach, loss of any canonical significantly decreased cell-surface delivery of LAMP1Δ-RUSH (Fig. 3 A). Rescue with overexpression of EXO-HALO fusions demonstrated that the defect in cell-surface delivery of LAMP1Δ-RUSH is not due to off-target effects or generalized cell lethality (Fig. 3 A). Interestingly, EXOC6-HALO demonstrated poor recovery (Fig. S3 D) in comparison to N-terminally tagged EXOC6 (Fig. 3 A), and overexpression resulted in a moderate but significant decrease in cell-surface arrival of LAMP1Δ-RUSH (Fig. S3 E), compared to N-terminally tagged HALO-EXOC6, consistent with previous reports that C-terminally tagged EXOC6 is experimentally challenging (Ahmed et al., 2018).

Imaging exocyst-KO from all subunits of the canonical octameric complex demonstrated an accumulation of post-Golgi carriers at the cell tips (Fig. 3 B), indicating that the point of exocyst involvement in the secretory pathway is after carriers have budded from the Golgi apparatus and prior to fusion with the plasma membrane. Interestingly, double KO of ELKS and EXOC1 resulted in a significant increase in the phenotype (Fig. S2 F), highlighting that ELKS contributes to this pathway. Together these data demonstrate that the exocyst complex is of fundamental importance for cell-surface delivery of LAMP1Δ-RUSH carriers.

To ensure that the effect of exocyst-KO on post-Golgi carrier fusion is direct, we tested localization of exocyst components. By tagging EXOC3 with a HALO Tag and expressing it at low levels in our LAMP1Δ-RUSH cell line under EXOC3-KO conditions, we observed EXOC3 at fusion punctae co-localizing with LAMP1Δ-RUSH (Fig. 4, A and B, and Video 10). We also observed EXOC1 and EXOC6 co-localizing with post-Golgi carriers prior to fusion with the plasma membrane (Fig. 4, C and D; Fig. S3, A and B; and Video 11 and Video 12). To test the localization of the exocyst subunits to the carriers, we developed an unbiased biochemical approach to localize proteins of interest to post-Golgi carriers. We generated a second LAMP1Δ-RUSH system with the GFP on the cytosolic side of the membrane (Fig. 4 E). This C-terminal fusion had comparable kinetics to the lumenal/extracellular tagged variant (Fig. S4). 35 min after the addition of biotin, there is an accumulation of post-Golgi carriers in the cytosol. We mechanically lysed the cells and immuno-isolated the carriers using GFPtrap beads, an approach we term “carrierIP.” Expression of the control HALO resulted in no enrichment after carrierIP, and all core exocyst components tested (EXOC1, EXOC2, EXOC3, EXOC4, EXOC5, EXOC6, EXOC7, and EXOC8) as well as the positive control RAB6A were enriched on post-Golgi carriers (Fig. 4 F). These data demonstrate that exocyst is directly recruited to the post-Golgi carriers.

Biochemical studies have identified that the phosphatidylinositol-5 kinase (PIP5K) is important for the recruitment of exocyst to the membrane by modifying phosphatidylinositol to phosphatidylinositol-5 phosphate (Maib and Murray, 2021 Preprint). There are three PIP5K1 homologs in mammalian cells, PIP5K1A, PIP5K1B, and PIP5K1C. KO of either isoform individually had no detectable effect on cell-surface delivery of LAMP1Δ-RUSH (Fig. 4 G). Triple-transient KO of all three isoforms, however, lead to a decrease of around 60% (Fig. 4 G), comparable to the loss of exocyst (Fig. 3, B–D). Imaging the accumulation of intracellular LAMP1Δ-RUSH in PIP5K1A/B/C-KO cells reveals cargo accumulating in post-Golgi carriers prior to fusion at the cell surface consistent with previous observations on exocyst recruitment (Fig. 4 H). Together these data directly tie the machinery known to be important for exocyst recruitment to post-Golgi carriers to the plasma membrane.

In this study, we have identified the exocyst complex as an essential component of the mammalian secretory pathway. Exocyst localizes to post-Golgi carriers and loss of exocyst prevents delivery of cell-surface carriers, causing them to accumulate in cell tips resulting in a global loss of secretion. In addition, in professional secretory cells such as adipocytes and lymphoma cells, secretion of key proteins such as adipokines and antibodies is heavily reduced upon loss of exocyst.

The role of exocyst as the secretory complex in Saccharomyces cerevisiae is well established, due to its original identification in the Sec genetic screen (Novick et al., 1980). Exocyst has been previously implicated in biosynthetic sorting in mammalian cells using tsVSV-G (Yeaman et al., 2001) which co-localizes with exocyst in the Golgi stacks; however, exocyst inhibition with antibodies did not affect VSV-G delivery (Yeaman et al., 2001; Grigoriev et al., 2007). siRNA depletion of EXOC7 decreased the efficiency of tsVSV-G delivery to the plasma membrane (Liu et al., 2007). Studies into post-Golgi carriers and thus secretion have been hampered by the lack of model systems to study the kinetic process. The use of quantitative RUSH assays in this study allows this route to be studied with kinetics that better resemble endogenous trafficking. In addition, loss of the exocyst complex is lethal in cultured cells (Wang et al., 2015). To study KO of exocyst, we have used transient CRISPR-Cas9. This has two key advantages: It allows abrogation of the protein-of-interest to be studied in the appropriate phenotypic window and it avoids artefacts introduced by clonal selection after CRISPR-Cas9 gene editing. The combination of kinetic trafficking assays and transient CRISPR-Cas9 thus provides new insights into the fundamental process of secretion.

We chose LAMP1ΔYQTI as a probe as it has been experimentally demonstrated to traffic directly to the cell surface through the biosynthetic secretory pathway (Chen et al., 2017), and thus can act as an orthologous validated marker of this pathway, as demonstrated in Figs. 1 and 2. Additionally, LAMP1 can tolerate a tag on the N and C termini without affecting the trafficking or kinetics (Chen et al., 2017; Fig. S4). Some studies suggest that LAMP1 traffics through the endolysosomal system to the lysosome (Cook et al., 2004; the so-called “direct pathway”), and we cannot rule out a non-detectable subset of the RUSH cargo taking this pathway. Nevertheless, with the LAMP1ΔYQTI-RUSH, we observed a significant amount of trafficking directly to the cell surface (Fig. 1) in line with other studies (Chen et al., 2017).

Using the RUSH system coupled with kinetic trafficking assays, super-resolution imaging, and TIRF microscopy, we are able to map the machinery of the secretory pathway from the Golgi apparatus to the plasma membrane. RAB6A is essential for budding of the carriers from the Golgi (Fig. 2), where they traffic along microtubules to the cell tips. After budding, we see the tubules acquiring ARHGEF10 and RAB8 through imaging (Fig. 2), which is suggestive of a Rab cascade as previously described (Grigoriev et al., 2011; Shibata et al., 2016). A RAB6A to RAB8A transition has also been described in the tethering and fusion of carriers through ELKS (Grigoriev et al., 2011). Across different species, both RAB8 and RAB11 have been described as EXOC6 interactors (Zhang et al., 2004; Guo et al., 1999; Wu et al., 2005; Luo et al., 2014). It has also been suggested that yeast homologues of RAB8 and RAB11 together with the exocyst complex can promote vesicle transport along the cytoskeleton through EXOC6 interaction with myosin type V (Lipatova et al., 2008; Jin et al., 2011). EXOC5 has been shown to bind GTP-ARF6 (Prigent et al., 2003), which has been recently proposed to mediate the recruitment of a PIP5 kinase (Maib and Murray, 2021 Preprint). In fact, three PIP5 kinases appear to act redundantly to convert the post-Golgi phospholipids (Fig. 4) and allow the recruitment of the exocyst complex, which tethers the carrier to the plasma membrane for the final fusion event. In mammalian cells, the PIP5 kinases are likely recruited to post-Golgi membranes by several GTPases that act redundantly with ARF6. Indeed, this study has started to uncover a complex cargo delivery system, where redundancy most likely marks every step.

Major work has been undertaken on the exact molecular timings of recruitment of exocyst prior to cell-surface fusion which show an exquisite order of complex assembly on carriers, upstream of SNARE complex activity, and prior to cell-surface fusion (Rivera-Molina and Toomre, 2013; Ahmed et al., 2018). Our work is consistent with the observation that the exocyst complex is recruited to secretory carriers near the plasma membrane, prior to their fusion with the plasma membrane (Ahmed et al., 2018).

ELKS has previously been identified as the molecular tether for secretory carriers (Fourriere et al., 2019). Although we observed ELKS localizing to hotspots on the plasma membrane, we did not see a phenotype of ELKS-KO on cell-surface delivery of LAMP1Δ-RUSH (Fig. S2). This does not rule out the role of ELKS in this process and it is likely that for certain cargoes or cell types ELKS has an essential role. Accordingly, we observe an increase in the phenotype when combining ELKS and exocyst KO (Fig. S2 F), indicating a potential functional redundancy, and notably see both ELKS and exocyst on the same hotspots where cell-surface fusion occurs. Indeed, in neurons, studies demonstrate that ELKS acts as a redundant scaffold protein at the active zone site and that when this structure is disrupted, synaptic vesicle fusion is impaired (Wang et al., 2016). Additionally, one of the cargos that ELKS has been shown to have a role in the secretion of is NPY, a soluble secreted cargo that takes the regulated secretory pathway in specialized cell types (Fourriere et al., 2019). Besides a structural role, ELKS has been further shown to capture RAB6 positive cargoes in golgin-like manner contributing to the establishment of a ready to fuse pool of synaptic vesicles at the active zone (Nyitrai et al., 2020). To fully understand the specific balance of roles between exocyst and ELKS will require further studies of other cargoes in specific cell types.

A number of exocyst subunits have been implicated in rare human genetic disorders. To date and to our knowledge, there are five disease-associated subunits, EXOC8 (Coulter et al., 2020; Online Mendelian Inheritance in Man [OMIM]: 615283), EXOC7 (Coulter et al., 2020; OMIM: 608163), EXOC6B (Girisha et al., 2016; Simsek-Kiper et al., 2022; OMIM: 607880), and EXOC4 (Nihalani et al., 2019; OMIM: 608185) and EXOC2 (Van Bergen et al., 2020; OMIM: 615329). Mutations in EXOC8 and EXOC2 are associated with neurodevelopmental disorders, and mutations in EXOC6B with skeletal abnormalities and mutations in EXOC4 have been associated with nephrotic syndrome. In cell models, exocyst has shown to be essential, with single-cell lethality associated with stable KOs (Wang et al., 2015). The severity and rarity of the disorders associated with loss of exocyst subunits are likely linked to both the variety of cellular functions associated with exocyst as well as the fundamental nature of these processes, including secretion.

Exocyst was initially associated with basolateral vesicle trafficking to cell–cell contacts in polarized cells (Grindstaff et al., 1998; Lipschutz et al., 2000; Langevin et al., 2005; Yeaman et al., 2004; Andersen and Yeaman, 2010; Xiong et al., 2012; Blankenship et al., 2007; Oztan et al., 2007; Bryant et al., 2010), and has since been implicated in a variety of processes including cytokinesis (Chen et al., 2006; Fielding et al., 2005; Neto et al., 2013), cell migration and tumor invasion (Rossé et al., 2006; Rosse et al., 2009; Spiczka and Yeaman, 2008; Assaker et al., 2010; Thapa et al., 2012; Lalli, 2009; Das et al., 2014), autophagy (Bodemann et al., 2011), lysosome secretion (Sáez et al., 2019), innate immune response following viral infection (Chien et al., 2006) and primary ciliogenesis (Rogers et al., 2004; Zuo et al., 2009; Feng et al., 2012), though in most cases, exocyst’s role is directly linked to its exocytic function (Wu and Guo, 2015).

Here, we demonstrate that in addition to these roles, exocyst is essential for the fusion of constitutive secretory carriers. The regulation of exocyst and its recruitment to carriers is not fully understood.

There are a plethora of associated proteins that potentially allow for differential recruitment of exocyst to various carriers. These include the RALs (Maib and Murray, 2021,Preprint), ARF6 (Maib and Murray, 2021,Preprint), phospholipids (Maib and Murray, 2021,Preprint; Fig. 4, G and H), CDC42 (Zhang et al., 2001), RAB10 (Babbey et al., 2010), RAB11 (Takahashi et al., 2012), and RAB8A (Mei and Guo, 2018). In addition, EXOC3 has three homologues, EXOC3L1, EXOC3L2, and EXOC3L4; and EXOC6 has EXOC6B. For example, in this study, EXOC6 acts redundantly with EXOC6B for constitutive cargo delivery; however, the absence of EXOC6B alone is sufficient to cause a skeletal disorder in humans (Girisha et al., 2016; Simsek-Kiper et al., 2022). In addition, exocyst subunits have a differential tissue expression in various metazoa (Thisse et al., 2004; Mehta et al., 2005), and there are a number of functional sub-complexes ascribed to specific cellular functions, including the existence of sub-complex 1 (EXOC1–4) and 2 (EXOC5–8; Ahmed et al., 2018), as well as specialized sub-complexes including an EXOC8-depedent sub-complex (Bodemann et al., 2011), an EXOC2-EXOC8 containing sub-complex (Moskalenko et al., 2003; Jin et al., 2005), and a specialized role for EXOC7 (Zhao et al., 2013) and EXOC5 (Lipschutz et al., 2000). Which combination of these sub-complexes, homologues, or associated proteins provide specificity, redundancy, or regulation of exocyst is not fully understood, but could potentially explain the widespread function of the complex with discrete specificities.

The following primary antibodies were used for Western blot in this study: mouse anti-GFP HRP conjugate antibody (GG4-2C2.12.10; 1:5,000; 130-091-833; Miltenyi Biotec), rabbit anti-EXOC1 antibody (1:5,000; ab118798; Abcam), rabbit anti-EXOC3 antibody (EPR10812; 1:5,000; ab156568; Abcam), rabbit anti-ELKS antibody (EPR13777; 1:5,000; ab180507; Abcam), mouse anti-LAMP1 antibody (H4A3; 1:10,000; ab25630; Abcam), rabbit anti-TIMP2 antibody (D18B7; 1:1,000; 5738; Cell Signaling Technology), rabbit anti-Cystatin C antibody (EPR4413; 1:5,000; ab109508; Abcam), rabbit anti-PSAP antibody (1:1,000; HPA004426; Atlas), mouse anti-GM130 antibody (1:1,000; 610822; BD Transduction Laboratories), and rabbit anti-GAPDH HRP conjugate antibody (D16H11; 1:1,000; 8884; Cell Signaling Technology). Goat HRP-conjugated secondary antibodies (1:5,000) were purchased from Abcam (anti-mouse: ab205719; anti-rabbit: ab205718), and goat-anti-human IgG secondary antibody IRDye 800CW (1:15,000; 926-32232) from LI-COR. The prokaryote expression vector encoding an anti-GFP mCherry nanobody (a gift from Martin Spiess, #109421; Addgene plasmid) and pOPINE GFP nanobody:HALO:His6, encoding anti-GFP HALO nanobody (a gift from Lennart Wirthmueller, #111090; Addgene plasmid) were expressed in bacteria and respectively GST- and His-purified in-house. The following cell-permeable dyes were obtained from these vendors: 646 HALO Dye (GA112A; Promega) and DAPI (D21490; Invitrogen).

The following antibiotics were used to select the newly generated stable cell lines: geneticin (875 mg/ml; 10131035; Life Technologies), hygromycin B (250 mg/ml; 10687010; Invitrogen), puromycin (1 μg/ml; A1113803; Gibco), and blasticidin (150 mg/ml; A1113903; Gibco).

The following chemicals were used in this work: dexamethasone (D4902; Sigma-Aldrich), biotin (B4501; Sigma-Aldrich), insulin (I5500; Sigma-Aldrich), 3-Isobutyl-1-methylxanthine (IBMX; I5879; Sigma-Aldrich), polybrene (TR-1003; Sigma-Aldrich).

SBP-GFP-LAMP1ΔYQTI was PCR amplified from the original backbone SBP-GFP-LAMP1 (Chen et al., 2017; a generous gift from Juan Bonifacino, National Institute of Child Health and Human Development, National Institutes of Health) and Gibson assembled (E2621L; New England Biolabs) to the pEGFP-C1 (Clontech) vector backbone in between the AgeI/HindIII restriction sites. The SBP-LAMP1ΔYQTI-GFP was then obtained by a two-step assembly that first removed GFP and then re-cloned its PCR product downstream of LAMP1ΔYQTI.

Strep-KDEL was PCR amplified from Strep-KDEL_SBP-mCherry-GPI (#65295; Addgene plasmid) and assembled to a BamHI/PsrI digested TtTMPV-Neo viral backbone (#27993; Addgene plasmid). The neomycin resistance gene was then replaced with a hygromycin B encoding sequence.

pHALO-C1 and pHALO-N1 were generated by replacing the eGFP in Clontech vectors with HALO using Gibson assembly (E2621L; New England Biolabs). HALO-ARHGEF10, EXOC1-HALO, EXOC2-HALO, EXOC3-HALO, EXOC4-HALO, EXOC5-HALO, EXOC6-HALO, HALO-EXOC6, EXOC7-HALO, EXOC8-HALO, HALO-ELKS, and HALO-EXOC6 were cloned by Gibson assembly using a synthetic, codon optimized version of each gene of interest (Integrated DNA Technologies), cloned in-frame upstream or downstream of HALOTag in pHALO-C1 or pHALO-N1. HALO-RAB6A and HALO-RAB8A were cloned in similar manner, except the gene sequences were PCR amplified from pEGFP-RAB6A (a kind gift from Juan Bonifacino, National Institute of Child Health and Human Development, National Institutes of Health) and pEGFP-RAB8A (Matsui et al., 2011; a gift from Mitsunori Fukuda, Tohoku University, Sendai, Japan), respectively. Point mutations were introduced through Q5 Site-Directed Mutagenesis Kit (M0554S; New England Biolabs).

Microtubules were visualized by expressing a plasmid containing β-tubulin-mCherry (#175829; Addgene plasmid). To generate a stable KO cell line, guide RNAs targeting ELKS (ERC1) were cloned into pSpCas9 (BB)V2.0 (#62988; Addgene plasmid; Ran et al., 2013), using the BbsI restriction sites.

For transient KO cells, guide RNAs targeting a gene of interest (Table S1) were cloned into pKLV-U6gRNA (BbsI)-PGKpuro2ABFP (#50946; Addgene plasmid), using the BbsI restriction sites, as described above. The IDT Alt-R CRISPR-Cas9 guide RNA tool was used to custom design two guide sequences per gene of interest. Cas9 viral expression backbone was a kind gift from Paul Lehner as well as the packaging vectors pMD.G and pCMVR8.91.

To generate the piggybac-RUSH constructs, we first created a piggybac-CMV-StrepKDEL-IRES-SBP-HALO. To achieve this, we Gibson assembled a PCR product containing CMV-StrepKDEL-IRES-SBP (#65295; amplified from Addgene plasmid) to the SalI/MluI digested piggybac backbone (a generous gift from Jonathon Nixon-Abell, Cambridge Institute for Medical Research [CIMR], University of Cambridge, Cambridge, UK) and then digested the result with BamHI to assemble in a HALOTag flanked by BamHI and XbaI restriction sites. Gibson assembly was used to further clone other cDNAs of interest into this piggybac-RUSH-HALO vector. A PCR product of ColX (#110726; from Addgene) and a synthetic gene containing PAUF (Integrated DNA Technologies) were cloned into the XbaI site downstream of HALO. PCR products containing CAB45 and NUCB1 (generous gifts from Liz Miller), and TNFa (#166901; from Addgene) were cloned into the BsrGI/BamHI sites upstream of HALO. Plasmids and primers used in this work are available upon request. All constructs were sequenced to verify their integrity.

HeLa cells were already available in the lab; 3T3-L1 fibroblasts, originally from Howard Green (Harvard Medical School, Boston, MA), were a gift from David James (University of Sydney, Australia); JK6L myeloma cells were a kind gift from Jonathan Keats (Translational Genomics Research Institute, USA). and Lenti-X 293T cells were obtained from Takara Bio (632180). JK6L cel

留言 (0)

沒有登入
gif