Skip to main content
eLife logoLink to eLife
. 2019 Oct 10;8:e49796. doi: 10.7554/eLife.49796

Selective clearance of the inner nuclear membrane protein emerin by vesicular transport during ER stress

Abigail Buchwalter 1,2,3,, Roberta Schulte 4, Hsiao Tsai 4, Juliana Capitanio 4, Martin Hetzer 4,
Editors: Elizabeth A Miller5, David Ron6
PMCID: PMC6802967  PMID: 31599721

Abstract

The inner nuclear membrane (INM) is a subdomain of the endoplasmic reticulum (ER) that is gated by the nuclear pore complex. It is unknown whether proteins of the INM and ER are degraded through shared or distinct pathways in mammalian cells. We applied dynamic proteomics to profile protein half-lives and report that INM and ER residents turn over at similar rates, indicating that the INM’s unique topology is not a barrier to turnover. Using a microscopy approach, we observed that the proteasome can degrade INM proteins in situ. However, we also uncovered evidence for selective, vesicular transport-mediated turnover of a single INM protein, emerin, that is potentiated by ER stress. Emerin is rapidly cleared from the INM by a mechanism that requires emerin’s LEM domain to mediate vesicular trafficking to lysosomes. This work demonstrates that the INM can be dynamically remodeled in response to environmental inputs.

Research organism: Mouse

Introduction

The biogenesis of roughly one-third of the cell’s proteome takes place within the endoplasmic reticulum (ER) network. The ER is contiguous with the nuclear envelope (NE) membrane, a double bilayer membrane that defines the boundary of the nucleus. The NE is punctuated by nuclear pore complexes (NPCs) which control transport between the nuclear and cytoplasmic compartments. The outer nuclear membrane (ONM) and the bulk ER membrane network have a similar protein composition, including ribosomes that can be seen associated with the ONM. The inner nuclear membrane (INM), in contrast, is cloistered away from the bulk ER network by the NPC (Figure 1A). Proteomic analyses suggest that hundreds of proteins are selectively concentrated at the INM (Schirmer et al., 2003), and mutations to these proteins cause a broad array of rare pathologies (Schreiber and Kennedy, 2013).

Figure 1. Dynamic proteomic analysis of inner nuclear membrane protein turnover.

(A) Diagram of the ER with associated ribosomes, the NE composed of the ONM and INM, the NPCs, and the underlying nuclear lamina. INM proteins are synthesized in the ER, pass through the NPC, and enrich at the INM. (B) Overview of dynamic SILAC labeling experimental design. C2C12 mouse myoblasts were cultured for five population doublings in medium containing 13C6-lysine and 13C6, 15N4-arginine to completely label the proteome. After 3 days of culturing under differentiating conditions to generate non-dividing myotubes, cultures were switched to chase medium containing 12C-lysine and 12C, 14N-arginine for 1 to 3 days. Nuclear extracts were prepared at day 0, day 1, day 2, and day three for proteomic identification. (C,D) Representative peptide scans for a slowly degraded protein (Nup160) and (D) for a rapidly degraded protein (Topo2α) at the starting and ending points of the experiment outlined in (B). (E) Histogram of calculated half-lives for 1677 proteins with a median half-life of 2.4 days. (F) Features of nuclear proteome turnover. Median turnover behavior of 1677 proteins detected in at least three timepoints with at least one peptide (black line) with one standard deviation (gray); compared to turnover of the slowly exchanged protein Nup160 (black) and the rapidly exchanged protein Topo2α (blue). Error bars indicate SEM. (G) Calculated half-lives of 10 bona fide INM proteins, ranging from slowly degraded (nurim, purple) to rapidly degraded (emerin, green); 12 nuclear envelope transmembrane proteins (NETs) identified as NE residents by subtractive proteomics (see Schirmer et al., 2003); and 112 ER membrane proteins. ns indicates lack of statistical significance by Mann-Whitney test. Error bars indicate SEM. (H) There is no significant correlation between extraluminal domain size of INM proteins and their half-lives. See also Source Data 1–2, Supplementary files 13, and Figure 1—figure supplement 1.

Figure 1.

Figure 1—figure supplement 1. Example half life fits.

Figure 1—figure supplement 1.

Examples of half-life fits for proteins with predicted half-lives of 0.5 days (A), 1 day (B), 2 days (C), 4 days (D), 8 days (E), and 17 days (F).

As the INM is devoid of ribosomes and translocation machinery, INM proteins must be synthesized in the ONM/ER and transported into the INM. Proteins concentrate at the INM by mechanisms including diffusion followed by stable binding to a nuclear structure, such as chromatin or the nuclear lamina, or signal-mediated import through the NPC (Katta et al., 2014). Transport across the NPC is a major kinetic barrier to accumulation of proteins at the INM (Boni et al., 2015; Ungricht et al., 2015). While mechanisms of INM targeting have been extensively studied, it is less clear how INM proteins are targeted for degradation if misfolded, damaged, or mistargeted.

Protein folding is inefficient, and newly synthesized proteins often become terminally misfolded and require degradation (Hegde and Zavodszky, 2019). Mature proteins also become damaged or misfolded over time and require selective degradation and replacement. Within the ER membrane network, the major degradation pathway is ER-associated degradation, or ERAD. ERAD is initiated by poly-ubiquitination of a target protein by an E3 ubiquitin ligase, followed by extraction from the membrane and proteolysis by proteasomes in the cytosol (Hegde and Zavodszky, 2019). Flux through ERAD helps to maintain organelle homeostasis and cell function by clearing damaged, misfolded, or mislocalized proteins.

Recent work in S. cerevisiae has identified a small number of ubiquitin ligases that target INM-localized proteins for degradation by ERAD, but the mammalian homologs remain elusive, perhaps because of the massive expansion of the E3 ubiquitin ligase family in recent evolution (Deshaies and Joazeiro, 2009). Degradation of mammalian INM proteins also appears to rely on activity of the proteasome and on the ERAD ATPase p97 (Tsai et al., 2016), suggesting that mammalian INM proteins may be subject to ERAD. However, we lack a broad understanding of the lifetimes of INM proteins in this compartment and the pathways used for their degradation within mammalian cells.

We sought to understand features of INM protein turnover in mammalian cells, and applied both proteome-wide and targeted candidate approaches to address this question. Here we show that the LEM domain protein emerin (EMD) is a rapidly degraded constituent of the INM. We use EMD as a model for dissecting INM protein turnover pathways and demonstrate that EMD is subject to both proteasome-dependent and lysosome-dependent modes of degradation. We report that both misfolded and normally folded variants of EMD are selectively exported from the INM and ER during acute ER stress by vesicular transport through the secretory pathway and delivery to the lysosome. These findings indicate that the INM sub-compartment senses and responds to ER stress.

Results

Trends in protein turnover across the NE/ER membrane network

We first used a dynamic proteomic approach to understand trends in protein turnover across ER sub-compartments. Since the nucleus is entirely disassembled during mitosis in mammalian cells, we chose a system that would allow us to profile protein turnover in the absence of cell division. We made use of the C2C12 myoblast culture system, which can be induced to irreversibly differentiate into myotubes by serum withdrawal (D'Angelo et al., 2009). We subjected these non-dividing mouse muscle myotubes to a pulse-chase timecourse using stable isotope labeling in cell culture (SILAC) (Ong and Mann, 2006) for timepoints ranging from 1 to 3 days (Figure 1B, see Materials and methods). Crude nuclear extracts were prepared and analyzed by mass spectrometry, and the ratio of ‘old’ (13C6-Lys, 15N4 + 13C6-Arg-labeled) to ‘new’ (12C6-Lys, 14N4 + 12C6-Arg-labeled) protein was quantified at the peptide level over time; peptides that passed stringent quality control filters were retained for estimation of half-lives by a linear regression fitting method (Dörrbaum et al., 2018)(see Materials and methods). We evaluated 1677 proteins and predicted half-lives over a wide range, from less than a day to greater than 15 days (Figure 1E, Table S3). Linear regression performs well when a line can be fitted with high fidelity and a non-zero slope is detectable; these conditions were generally met for proteins with predicted half-lives ranging from 1 to 8 days. We observed more frequent deviations in linearity at the low extreme (predicted t1/2 <1 day) and slopes approaching zero at the high extreme (predicted t1/2 > 8 days) (Figure 1—figure supplement 1). We expect that these factors limit the precision of half-life determination below 1 day and above 8 days from our 3 day timecourse. The median turnover rate that we observed (2.4 days) corresponds well with previous analyses in non-dividing mammalian cell cultures (Cambridge et al., 2011).

We observed some extremes in protein stability that are consistent with previous reports; for example, the long-lived nuclear pore complex component Nup160 (Toyama et al., 2013) was in the top 10% of predicted half-lives, with a calculated half-life of approximately 18 days (Figure 1C,F, Table S1). Near the other extreme, we observed that the enzyme topoisomerase 2α (Topo2α) had a predicted half-life in the bottom 10%, of less than 1 day (Figure 1D,F). This is consistent with this enzyme’s known regulation by ubiquitination and proteolysis (Gao et al., 2014).

Having established this framework, we then quantified the turnover kinetics of known inner nuclear membrane (INM) proteins. For this analysis, we focused on proteins whose preferential enrichment in this membrane compartment had been experimentally verified; we identified ten such proteins in our dataset (see Table S1) and determined their half-lives (see Materials and methods). We observed half-lives for these proteins ranging from 8.8 days (nurim) to 1.5 days (emerin) (Figure 1G). While very little is known about nurim’s function, its intrinsic biophysical properties may contribute to its long half-life: nurim contains six transmembrane domains, is extremely insoluble (Hofemeister and O'Hare, 2005), and diffuses very slowly within the INM (Rolls et al., 1999). Emerin (EMD) is a founding member of the LEM domain family of INM proteins with essential functions in muscle development (Brachner and Foisner, 2011). Unlike nurim, EMD is a small (~25 kDa), single-pass, tail-anchored transmembrane protein that diffuses freely through the NPC and enriches at the INM by virtue of its affinity for lamin A (Vaughan et al., 2001).

Given the INM’s status as a restricted sub-compartment of the ER, we reasoned that it might be possible that INM proteins would be generally less accessible to protein turnover than ER membrane proteins. Alternatively, similarly effective turnover in both compartments might support the possibility that turnover can occur in situ at the INM. ER membrane proteins were well represented in our dataset, as a significant proportion of ER membranes remain attached to and co-purify with nuclei (Schirmer et al., 2003). We could thus query whether INM proteins exhibited distinct turnover kinetics from membrane proteins of the bulk ER by comparing INM and ER transmembrane protein half-lives. We also compared bona fide INM proteins to proteins that had been identified as preferentially associated with either the inner or outer bilayer of the nuclear envelope (NE) membrane by comparative proteomics (Schirmer et al., 2003), termed NE transmembrane proteins (NETs). Altogether, these analyses indicate that INM proteins do not exhibit unique turnover kinetics as a protein class, compared to ER membrane proteins in general or to the overlapping designation of NETs (Figure 1G).

The size of INM proteins determines whether a protein must rely on signal-mediated transport through the NPC (Katta et al., 2014), and live imaging assays indicate that INM proteins with larger nucleoplasm-facing domains move more slowly across the NPC barrier (Boni et al., 2015; Ungricht et al., 2015). If transit across the NPC and out of the INM were a prerequisite for turnover in the bulk ER, we reasoned that turnover efficiency would also exhibit some dependence on protein size, because of the relationship between protein size and transport efficiency between the two compartments. The short half-life and small size of EMD is in line with this possibility. Our dataset of INM proteins included four single-pass INM and six multi-pass INM proteins, with total size of extraluminal domains ranging from 40 amino acids to 733 amino acids (Table S5). If export out of the NPC were a prerequisite for INM protein turnover, we reasoned that half-life should increase as the bulk of nucleoplasm-facing domains increases. We found no evidence for such a correlation (Figure 1H). We infer from this analysis that other factors distinct from monomeric protein size regulate protein turnover rate. This indicates that for INM proteins, export out of the INM is not a rate-limiting step for protein turnover. Rather, this is consistent with evidence in S. cerevisiae (Foresti et al., 2013; Khmelinskii et al., 2014) and in mammalian cells (Tsai et al., 2016) that turnover of INM proteins can take place in situ at the INM.

Recombination-induced tag exchange confirms INM protein lifetimes

We observed a wide range of half-lives for INM proteins in our proteomic analyses (Figure 1G), with the polytopic INM protein nurim turning over most slowly and the single-pass INM protein EMD turning over most rapidly. To directly visualize these relative differences in protein stability, we used recombination-induced tag exchange (RITE) (Toyama et al., 2019; Verzijlbergen et al., 2010) (Figure 2A) to perform a microscopy-based pulse-chase experiment. We expressed either nurim or EMD in a cassette encoding two C-terminal epitope tags separated by LoxP sites and by a stop codon, such that the resulting transcript will encode a protein that will be C-terminally tagged with the first tag. Upon adenoviral introduction of Cre recombinase, the RITE cassette is recombined to remove the first tag and position the second tag downstream of the open reading frame, so that all newly synthesized mRNA encodes a protein marked with the second tag. This enables simultaneous tracking of older and newer pools of protein that were synthesized before and after Cre addition, respectively (Toyama et al., 2019; Verzijlbergen et al., 2010). Using this approach, we visualized the rate of decline in the fluorescence intensity of ‘old’ myc-tagged nurim or EMD over several days in quiescent C2C12 cells. Consistent with our proteomic observations, we observed that RITE-tagged nurim decayed significantly more slowly than RITE-tagged EMD at the NE (Figure 2B–C,H).

Figure 2. RITE analysis of INM proteins enables visualization of proteasome-dependent turnover.

RITE analysis of INM proteins corroborates protein turnover determined by proteomics. (A) Schematic of recombination-induced tag exchange (RITE) expression cassette for visualizing protein turnover using Cre recombinase-mediated tag switching. (B-C) RITE timecourses of nurim-RITE (B) and EMD-RITE (C) in quiescent C2C12 cells. Maximum intensity projections of confocal z-series shown. (D) Diagram of emerin domain organization and position of EDMD-linked deletion mutant (EMDΔ95-99) within the lamin-binding domain. (E-G) RITE timecourses of nurim-RITE (E), EMD-RITE (F), and EMDΔ-RITE (G) with or without 1 day of cotreatment with the proteasome inhibitor MG132 (right panels). Single confocal z-slices shown. (H) Quantification of normalized intensity of old NE-localized RITE-tagged protein in maximum intensity projections of confocal z series acquired across the conditions shown in (E-G). Bars indicate average values with error bars indicating SEM for N > 42 cells per condition from 2 independent experiments. **** indicates p-value < 0.0001 (by t-test) for comparison between untreated and treated conditions. Scale bar, 10 mm. See also Figure 2—figure supplement 1.

Figure 2.

Figure 2—figure supplement 1. Identification of a disease-linked emerin mutant with normal NE localization.

Figure 2—figure supplement 1.

(A) EMDΔ95–99-GFP (EMDΔ-GFP) has normal localization at the NE (B) and normal residence time at the NE, based on fluorescence recovery after photobleaching timecourse at the NE (C,D).

The RITE system allows unambiguous dissection of the fates of maturely folded protein as well as nascent, newly synthesized protein. Recent work in yeast (Foresti et al., 2013; Khmelinskii et al., 2014) and in mammalian cells (Tsai et al., 2016) strongly suggests that INM proteins are subject to proteasome-mediated degradation via the ERAD pathway. The RITE system provides a means to distinguish the effects of proteasome inhibition on maturely folded proteins by inhibiting the proteasome after RITE tag switching, and monitoring the effects on maturely folded proteins. Mature nurim-RITE decreases only modestly within 2 days of tag switching, but co-incubation with the proteasome inhibitor MG132 for 1 day causes accumulation of nurim-RITE through the NE and ER (Figure 2E,H). Maturely folded EMD-RITE diminishes significantly at the NE within 2 days of tag switching but is partially stabilized at the NE in the presence of MG132 (Figure 2F,H). This indicates that mature, INM-localized proteins can be degraded in a proteasome-dependent pathway in situ at the INM. Notably, abundant proteasomes have been observed along the INM in cryo-EM studies and could possibly engage with substrate there (Albert et al., 2017). This is also consistent with a recent report that an unstable INM protein mutant accumulates within the nucleus of mammalian cells when the proteasome is inhibited (Tsai et al., 2016).

Identification of a model substrate for dissecting INM protein turnover

In order to gain more insight into the pathways that control INM protein turnover in mammalian cells, we chose to focus on EMD for its relatively fast turnover rate (Figure 1G, Table S1) and for the variety of disease-linked mutations to EMD that appear to influence protein stability (Fairley et al., 1999). Loss-of-function mutations to EMD cause Emery-Dreifuss muscular dystrophy (EDMD) (Bonne and Quijano-Roy, 2013). In some cases, EDMD-linked mutations cause loss of detectable EMD protein without affecting mRNA levels, suggesting that these mutations might cause misfolding and degradation of EMD (Fairley et al., 1999). We sought to identify such an EDMD-linked EMD variant for use as a model substrate for dissecting INM protein turnover. We selected a small in-frame deletion (Δ95–99) within EMD’s lamin-binding domain (Figure 2D) that had been previously shown to localize to the NE when ectopically expressed (Fairley et al., 1999). Consistently, when we expressed either EMD-GFP or EMDΔ95–99-GFP in C2C12 cells, we observed similar enrichment at the NE (Figure 2—figure supplement 1). Further, both protein variants exhibited identical residence times at the NE as assayed by fluorescence recovery after photobleaching (FRAP) analysis (Figure 2—figure supplement 1). Directly monitoring the stability of EMDΔ95–99 by RITE tagging indicates that it disappears from the NE faster than wild type EMD (Figure 2G), but is also stabilized at the NE by proteasome inhibition (Figure 2G–H). These observations indicate that EMDΔ95–99 is an unstable EMD variant that resides within the INM. We next moved to dissect that pathway(s) involved in EMDΔ95–99 degradation.

Proteasome-dependent and proteasome-independent modes of emerin clearance

Misfolded ER resident proteins are cleared by the ER-associated degradation (ERAD) pathway. ERAD clients are marked for degradation by ubiquitination, extracted from the ER membrane by the ATPase enzyme p97, and degraded by the proteasome in the cytosol (Ruggiano et al., 2014). INM proteins may also be targeted to an arm of the ERAD pathway in mammalian cells (Tsai et al., 2016), and our data indicate that multiple INM proteins are stabilized in situ by proteasome inhibition. However, ubiquitin ligase(s) that recognize INM-localized substrates in mammalian cells have not been identified.

To sensitively probe factors that influence INM protein stability, we tracked the stability of GFP-tagged EMD variants. When de novo protein synthesis was blocked by cycloheximide (CHX), we observed rapid loss of EMDΔ95–99-GFP within 4–8 hr (Figure 3A–B) while wild type EMD remained stable (Figure 3—figure supplement 1). This loss is blunted by co-treatment with the proteasome inhibitor MG132 (Figure 3A, third panel), consistent with our observations that INM protein turnover is slowed by proteasome inhibition using the RITE system (Figure 2E–G). If EMDΔ95–99-GFP is directed to the proteasome through ERAD, inhibition of earlier steps in this pathway should similarly cause accumulation of EMDΔ95–99-GFP. Indeed, pharmacological inhibition of p97 with the drug eeyarestatin I (Wang et al., 2008) causes modest accumulation of EMDΔ95–99 and of higher molecular weight species, a similar effect to proteasome inhibition itself (Figure 3B). In contrast, the drug kifunensine, which prevents ERAD targeting of misfolded glycosylated proteins (Fagioli and Sitia, 2001), has no effect on EMDΔ95–99-GFP levels as would be expected given the lack of glycosylation sites within EMD’s small luminal domain (Figure 3B).

Figure 3. Acute stressors destabilize mutant emerin protein levels.

(A) C2C12 cells stably expressing EMDΔ-GFP and treated with DMSO vehicle control, CHX alone, CHX and MG132, or MG132 alone for 8 hours. All images were acquired using the same laser power and detector gain settings. Single confocal z slices shown. (B) Western blot analysis of protein levels in C2C12 cells stably expressing EMDΔ-GFP and treated with DMSO vehicle, the translation inhibitor CHX, the proteasome inhibitor MG132, the p97 ATPase inhibitor eeyarestatin, or the glycosylation trimming inhibitor kifunensine for the time periods shown. a-tubulin shown as loading control. (C) Western blot analysis of U2OS cells stably expressing EMDΔ-GFP and doxycycline-inducible RNAi targeting the E2 ubiquitin ligases UBE2G1 and UBE2G2 and treated with DMSO vehicle control (-) or with doxycycline (+) for 48 hours. Free GFP indicates RNAi induction. a-tubulin shown as loading control. (D) Western blot detection of EMDΔ-GFP levels in cells treated with DMSO vehicle, or co-treated with CHX and the ER stress inducer THG for the time periods shown. a-tubulin shown as loading control. (E) C2C12 cells stably expressing EMDΔ-GFP and treated with vehicle control or with THG for the time periods shown. Insets show nuclei in the same ~50 µm field of view stained with Hoechst. All images acquired using the same laser power and detector gain settings; single confocal z slices shown. (F) Quantification of total NE-localized GFP fluorescence in maximum intensity projections of confocal z slices acquired across the conditions shown in (E) for N > 410 cells per condition. (G) Diagram of emerin domain organization and the sequence of an inserted C-terminal glycosylation sequence derived from the opsin protein, with glycosylation acceptor site marked (*). (H) Analysis of EMDΔ-GFP* glycosylation state in cells subjected to treatment with DMSO vehicle control or CHX and THG cotreatments for the times indicated. Red arrowhead indicates EndoH-sensitive glycosylated state of EMDΔ-GFP*; orange arrowhead indicates EndoH-resistant states of EMDΔ-GFP*; black arrowhead indicates deglycosylated EMDΔ-GFP*. a-tubulin shown as loading control. Numbers to left of blots indicate molecular weights in kDa. Scale bars in micrographs indicate 10 mm. See also Figure 3 – figure supplement 1, 2, and 3.

Figure 3.

Figure 3—figure supplement 1. Localization and stability of a disease-linked emerin variant.

Figure 3—figure supplement 1.

(A–B) C2C12 cells stably expressing EMDΔ-GFP (A) or EMD-WT-GFP (B) and treated with DMSO vehicle control, CHX alone, CHX and MG132, or MG132 alone for 8 hr. All images were acquired using the same laser power and detector gain settings. Single confocal z slices shown. Scale bar, 10 μm. (C–D) Western blot analysis of EMDΔ-GFP (C) or EMD-WT-GFP (D) protein levels after treatment with CHX for the indicates times.
Figure 3—figure supplement 2. siRNA-mediated E2 or E3 ubiquitin ligase knockdowns do not stabilize EMDΔ-GFP.

Figure 3—figure supplement 2.

(A–B) EMDΔ-GFP protein levels do not change in C2C12 cells stably expressing EMDΔ-GFP and transfected in duplicate with 50 nM RNAi targeting the E3 ubiquitin ligases Rnf26, CGRRF1, MARCH6, or a scrambled control (A), or targeting the E2 ubiquitin conjugating enzymes UBE2G1, UBE2G2, UBE2J1, and UBE2J2, or MARCH6 (B). (C) qPCR quantification of RNAi knockdown efficiency for conditions shown in (A–B). (D) EMDΔ-GFP protein levels either decrease or do not change in U2OS cells stably expressing EMDΔ-GFP and induced to express a miR-E RNAi cassette targeting the E3 ubiquitin ligase MARCH6 or the E2 ubiquitin conjugating enzyme UBE2J1 for 48 hr. Free GFP is also expressed from this cassette and indicates RNAi expression. miR-E RNAis used were from a validated set described in Knott et al. (2014).
Figure 3—figure supplement 3. Glycosylation reporter variants are destabilized by ER stress and recovered by BFA treatment.

Figure 3—figure supplement 3.

Glycosylation reporter variant of EMDΔ-GFP localizes normally to the NE and responds to ER stress induced by THG and secretory pathway disruption caused by BFA (A–B). (C) Pattern of glycosylation modifications in DMSO vehicle control treated cells or cells treated with THG for 2–4 hr. A larger proportion of EMDΔ-GFP becomes Endo H-resistant during acute ER stress.

E3 ubiquitin ligases transfer ubiquitin to ERAD substrates, and each E3 ligase exhibits preference for a small number of substrates. A few E3 ligases have been implicated in ERAD of INM-localized substrates in yeast, including Doa10 and Asi1 (Khmelinskii et al., 2014). MARCH6 is a mammalian ortholog of Doa10 (Zattas et al., 2016). Mammalian orthologs of Asi1 have not been identified. Based on iterative sequence homology analysis through the MetaPhORs database (Pryszcz et al., 2011) we identified two possible Asi1 homologs: Rnf26 and CGRRF1. We depleted MARCH6, Rnf26, and CGRRF1 with short interfering RNA (siRNA), but observed no effect on EMDΔ95–99 protein levels (Figure 3—figure supplement 2), suggesting that these ligases do not catalyze EMD turnover, or alternatively that multiple E3 ligases are redundant in this process. Importantly, the broad group of ERAD-implicated E3 ligases rely on a handful of E2 ubiquitin conjugating enzymes for ubiquitin transfer. These E2 ligases – four in mammals – thus represent a key control point for ERAD (Christianson et al., 2011; Leto et al., 2019). We targeted these four E2 ubiquitin ligases by siRNA transfection (Figure 3—figure supplement 2) and a subset of these by inducible RNAi using a potent microRNA-based system (Fellmann et al., 2013) and analyzed the effects on EMDΔ95–99-GFP levels. To our surprise, knockdown of UBE2G1, UBE2G2, UBE2J1, and UBE2J2 either did not stabilize or instead decreased EMDΔ95–99-GFP levels (Figure 3C; Figure 3—figure supplement 2). This finding suggests that when ERAD is perturbed, EMD variants can be cleared by an alternative pathway.

Emerin is subject to rapid stress-dependent clearance from the ER and NE

Global inhibition of ERAD places profound protein folding stress on the ER membrane network and induces the unfolded protein response (UPR) (Christianson et al., 2011). We considered whether direct induction of ER stress was sufficient to accelerate the turnover of EMDΔ95–99-GFP. We tested the effect of the ER stressor thapsigargin (THG), which disrupts ER homeostasis by causing release of Ca2+ from the ER lumen, on EMDΔ95–99 protein stability. Compared to CHX treatment alone (Figure 3—figure supplement 1), THG co-treatment further destabilized EMDΔ95–99-GFP (Figure 3D). Strikingly, when we tracked EMDΔ95–99 protein localization by time-lapse microscopy (Figure 3E), it became apparent that NE localization of EMDΔ95–99 significantly decreases within 2 hr of THG treatment, concomitant with accumulation in a perinuclear membrane compartment that morphologically resembles the Golgi apparatus. By 8 hr after THG administration, EMDΔ95–99 was undetectable (Figure 3E–F). These data suggest that under conditions of ER stress, EMDΔ95–99 is cleared from the NE/ER membrane network by transport out of the ER and eventual disposal of the protein in a post-ER compartment.

Acute ER stress reroutes emerin through the secretory pathway

We took several approaches to test the possibility that EMDΔ95–99 leaves the NE/ER network during ER stress. Firstly, we made use of the characteristic sugar modifications that occur as cargoes progress through the secretory pathway to determine whether EMDΔ95–99 accesses post-ER compartments. Since the short lumenal domain of EMD lacks a glycosylation consensus site, we engineered the glycosylation site from the opsin protein (Bulbarelli et al., 2002) (SSNKTVD) onto the lumen-facing C terminus of EMDΔ95–99. If EMDΔ95–99 is retained in the ER, all of its N-linked glycans should remain sensitive to the trimming enzyme Endo H. On the other hand, if EMDΔ95–99 exits the ER, its N-linked glycans will be elaborated with further modifications so that Endo H can no longer trim them. These glycosylation states can be detected as progressive increases in molecular weight, and can be completely removed by incubation with the enzyme PNGase F. This engineered variant, EMDΔ95–99-GFP*, localizes normally to the NE and also disappears from the NE upon ER stress induction (Figure 3—figure supplement 3). In unstressed cells, EMDΔ95–99-GFP* is predominantly observed in an Endo H-sensitive glycosylation state (Figure 3H, red arrowhead), with a minor pool of Endo H-resistant protein (Figure 3H, orange arrowhead). In contrast, EMDΔ95–99-GFP* shifts progressively to a higher molecular weight, Endo H-resistant state over 2 to 4 hr of co-treatment with CHX and THG. This indicates that ER stress induction increases the proportion of EMDΔ95–99 that exits the NE/ER and samples post-ER compartments.

Upon ER stress induction, EMDΔ95–99-GFP accumulates in a perinuclear domain that resembles the Golgi apparatus (Figure 3E). We evaluated the extent of colocalization of EMDΔ95–99-GFP with the medial Golgi resident protein giantin in untreated cells and cells that had been treated with THG for 2–4 hr (Figure 4A–C). In untreated cells, EMDΔ95–99-GFP was not detectable in the Golgi, but THG treatment rapidly induced Golgi colocalization as NE-localized EMDΔ95–99 levels decreased (Figure 4A–C). Comparing GFP fluorescence intensity in the Golgi versus the NE over time revealed that ER stress induces significant enrichment of EMDΔ95–99-GFP in the Golgi accompanied by loss from the NE within 2 hr (Figure 4D).

Figure 4. Stress-induced clearance of mutant emerin from the ER and NE involves the secretory pathway.

(A-C) Representative confocal slices of cells stably expressing EMDΔ-GFP, treated with DMSO or THG for the indicated times and costained for giantin to mark the Golgi (magenta). All images were acquired using the same laser power and detector gain settings. (A’-C’) Are contrast-adjusted to show relative levels of EMDΔ-GFP in NE and Golgi. Dotted lines mark positions of linescans in (A’’-C’’). (D) Quantification of GFP fluorescence intensity abundance ratio in Golgi versus NE in single, non-contrast-adjusted z slices over THG treatment timecourse. Columns indicate average with error bars indicating SEM for N > 37 cells from two independent experiments. **** indicates p-value<0.0001 compared to untreated (by t-test). (E-F) Representative confocal slices of cells stably expressing EMDΔ-GFP (F) after 16 hr of treatment with DMSO vehicle control, THG, BFA, or co-treatment with THG and BFA. All images were acquired using the same laser power and detector gain settings. Insets show nuclei in the same ~ 50 µm field of view stained with Hoechst. (E) Quantification of GFP fluorescence intensity at the NE in maximum intensity projections of confocal z series acquired across the conditions represented in (F). Columns indicate average with error bars indicating SEM for N > 386 cells from three independent experiments. **** indicates p-value<0.0001 compared to untreated (by t-test). Scale bars in micrographs indicate 10 μm. See also Figure 4—figure supplement 1.

Figure 4.

Figure 4—figure supplement 1. Colocalization of emerin with the Golgi.

Figure 4—figure supplement 1.

(A–B) Representative confocal slices of cells stably expressing EMD-WT-GFP, treated with DMSO or THG for the indicated times and costained for giantin to mark the Golgi (magenta). All images were acquired using the same laser power and detector gain settings. Dotted lines mark positions of linescans shown in (B). (C) Quantification of GFP fluorescence intensity abundance ratio in Golgi versus NE in single, non-contrast-adjusted z slices over THG treatment timecourse. Columns indicate average with error bars indicating SEM for N > 47 cells from two independent experiments. **** indicates p-value<0.0001. Scale bars in micrographs indicate 10 μm.

EMDΔ95–99 could be delivered to the Golgi by vesicular transport from the ER. Transport between the ER and the Golgi is mediated by packaging of cargoes into COP-coated vesicles (Barlowe and Miller, 2013), a process which can be inhibited by the drug brefeldin A (BFA). BFA acts by disrupting COPI vesicle formation, leading to the collapse of the Golgi into the ER membrane network (Chardin and McCormick, 1999). To test whether clearance of EMDΔ95–99-GFP from the NE/ER requires vesicle-mediated ER-to-Golgi transport, we co-incubated cells expressing EMDΔ95–99-GFP with THG and BFA. Strikingly, co-treatment with BFA nearly quantitatively reversed loss of EMDΔ95–99-GFP from the NE (Figure 4E–F). Taken together with the time-dependent enrichment of EMD variants in the Golgi apparatus (Figure 4A–C), and the time-dependent accumulation of more complex N-glycosylated variants of EMDΔ95–99-GFP* (Figure 3H), this indicates that under ER stress, EMD variants can be cleared from both the NE and ER by vesicular transport through the Golgi.

While vesicle-mediated transport is the major pathway by which proteins move from the ER and onward through the secretory pathway, alternative modes of removing protein from the ER exist, in particular during ER stress. Recent evidence indicates that the ER can undergo autophagy under various conditions, including acute ER stress (Smith et al., 2018). To evaluate the possibility that EMDΔ95–99-GFP could be engulfed and removed from the NE and ER by autophagosomes, we tested the ability of the PI3K inhibitor KU55933 to reverse EMDΔ95–99-GFP loss. PI3K signaling promotes the formation of isolation membranes that engulf autophagic cargo (Farkas et al., 2011; Klionsky et al., 2016). We observed that cotreatment with KU55933 during acute ER stress could not prevent loss of EMDΔ95–99-GFP from the NE/ER (Figure 5A–C), in contrast to the ability of BFA treatment to rescue EMD loss. This indicates that vesicle-mediated transport to the Golgi and not autophagic engulfment mediates EMDΔ95–99-GFP’s exit from the ER during stress.

Figure 5. Mutant emerin trafficking is dependent on lysosomal but not autophagosomal function.

Figure 5.

(A) Representative confocal slices of cells stably expressing EMDΔ-GFP after 8 hr of treatment with DMSO vehicle control, THG, co-treatment with THG and BFA, or co-treatment with THG and KU55933. Insets show nuclei in the same field of view stained with Hoechst. (B) Quantification of NE-localized GFP fluorescence intensity in maximum intensity projections of confocal z series acquired across the conditions shown in (A). Columns indicate average and error bars indicate SEM for N > 56 cells from three independent experiments. **** indicates p-value<0.0001 compared to untreated (by t-test). (C) Diagram of processes perturbed by KU55933, BFA, and Baf A1 treatment. (D-F) Representative confocal slices of C2C12 cells stably expressing EMDΔ-GFP and costained for LAMP1 after treatment with DMSO vehicle control (D), Baf A1 (E), or co-treatment with THG and Baf A1 (F) for the indicated times. (D’-F’) Insets show GFP-LAMP1 colocalization within ~ 15 µm field of view demarcated by dashed rectangles in (D-F). (G) Analysis of EMDΔ-GFP* glycosylation state in cells subjected to treatment with DMSO vehicle control or THG and Baf A1 cotreatments for the times indicated. Red arrowhead indicates EndoH-sensitive glycosylated state of EMDΔ-GFP*; orange arrowhead indicates EndoH-resistant states of EMDΔ-GFP*; black arrowhead indicates deglycosylated EMDΔ-GFP*. α-tubulin shown as loading control. Numbers to left of blots indicate molecular weights in kDa. Scale bars in micrographs indicate 10 μm.

Proteins in post-ER compartments can be degraded by vesicle-mediated traffic to the lysosome (Saftig and Klumperman, 2009). To investigate whether the lysosome was the eventual destination of EMDΔ95–99-GFP after ER export, we co-incubated cells expressing EMDΔ95–99-GFP with THG and bafilomycin A1 (Baf A1), which impairs lysosome acidification and thus slows protein degradative processes within lysosomes.

Under these conditions, we observed complete translocation of EMDΔ95–99-GFP out of the NE and ER and into numerous vesicles that are decorated with the lysosomal marker LAMP1 (Figure 5F). Notably, bafilomycin A1 alone did not trap EMDΔ95–99-GFP in lysosomes (Figure 5E), indicating that ER stress potentiates exit from the NE/ER and lysosomal accumulation. This indicates that the eventual destination of EMDΔ95–99-GFP after export from the NE/ER network is the lysosome.

If EMDΔ95–99 arrives at the lysosome by trafficking through the secretory pathway, this should be accompanied by the accumulation of Endo H-resistant N-glycosylation modifications on our engineered reporter EMDΔ95–99-GFP*. Indeed, we observe that the majority of EMDΔ95–99-GFP* exists as an Endo H-sensitive species in unperturbed cells (Figure 5G, red arrowhead), but shifts progressively to a higher molecular weight, Endo H-resistant state over 2 to 4 hr of co-treatment with THG and Baf A1 (Figure 5G, orange arrowhead). This indicates that EMDΔ95–99 traverses the secretory pathway before being delivered to the lysosome.

Emerin transiently accesses the plasma membrane during ER stress

A possible route from the early secretory pathway to the lysosome could involve anterograde transport following the ‘bulk flow’ of the secretory pathway, through the Golgi and into vesicles destined for the plasma membrane (PM). There, mislocalized proteins may be selectively endocytosed and trafficked to lysosomes for degradation through retrograde transport (Saftig and Klumperman, 2009). To explore this possibility, we performed antibody uptake assays in cells expressing EMDΔ95–99-GFP under homeostatic or stressed conditions. EMD is a tail-anchored protein with its final C-terminal amino acids facing the ER lumen; the C-terminal GFP tag will thus face the extracellular space if EMDΔ95–99-GFP accesses the PM (Figure 6A). We tested whether EMDΔ95–99’s GFP tag is accessible to an anti-GFP antibody applied to the surface of intact cells. In untreated cells, a small amount of EMDΔ95–99-GFP (Figure 6B–D) is accessible to an anti-GFP antibody, but not to an anti-myc antibody, indicating that EMDΔ95–99 is not completely restricted to intracellular membrane compartments under homeostatic conditions. Importantly, the signal from the anti-GFP antibody is specific to cells that express a GFP fusion protein (Figure 6D–E). Upon induction of ER stress by THG, the amount of PM-accessible EMDΔ95–99-GFP rapidly increases within 2–4 hr, and begins to taper off within 6 hr. This implies that ER stress induces the export of EMDΔ95–99-GFP from the NE/ER to the PM as well as its internalization. Importantly, the GFP antibody signal is visible within intracellular puncta, consistent with EMDΔ95–99-GFP:antibody conjugates being rapidly internalized into vesicles after PM delivery. Based on the timescale when the levels of EMDΔ95–99-GFP begin to significantly decrease at the NE (Figure 3E–F), increase in the Golgi (Figure 4A–D), transit through the PM (Figure 6) and accumulate in lysosomes (Figure 5F), we infer that EMDΔ95–99-GFP is transported through the secretory pathway to the PM, then internalized and delivered to lysosomes for degradation.

Figure 6. Mutant emerin traffics through the plasma membrane upon ER stress.

(A) Schematic of antibody uptake assay experimental design. If emerin accesses the plasma membrane (PM), it will be detected by anti-GFP antibody (green), which will bind the surface-exposed GFP tag. (B) Uptake of anti-GFP antibody (magenta) by cells stably expressing EMDΔ-GFP and treated with DMSO vehicle control or THG for 2, 4, or 6 hours. Cells were incubated with anti-GFP antibody for the final hour of these treatment periods before fixation and imaging. (C) Control demonstrating lack of uptake of anti-myc antibody by cells stably expressing EMDΔ-GFP and treated with THG for 2 hours. (D) Quantification of internalized antibody signal in EMDΔ-GFP expressing cells. Columns indicate average and error bars indicate SEM for N > 235 cells from 3 independent experiments. **** indicates p-value < 0.0001 (t-test) compared to untreated. (E) Control demonstrating lack of uptake of anti-GFP antibody by untreated C2C12 cells that do not express a GFP fusion protein. WGA is used to define cell boundaries. All images were acquired using the same laser power and detector gain settings. Scale bars in micrographs indicate 10 µm. See also Figure 6—figure supplement 1.

Figure 6.

Figure 6—figure supplement 1. Access of emerin to the plasma membrane.

Figure 6—figure supplement 1.

(A) Uptake of anti-GFP antibody (magenta) by cells stably expressing EMD-WT-GFP and treated with DMSO vehicle control or THG for the indicated times. Cells were incubated with anti-GFP antibody for the final hour of the treatment period before fixation and imaging. (B) control demonstrating lack of uptake of anti-myc antibody by cells stably expressing EMD-WT-GFP and treated with THG for 2 hr. (D) Quantification of internalized antibody signal in EMD-WT-GFP expressing cells. Columns indicate average and error bars indicate SEM for N > 66 cells from two independent experiments. **** indicates 2-value < 0.0001 (t-test) compared to untreated.

Other INM proteins do not undergo stress-dependent clearance

Our findings indicate that EMDΔ95–99 is subject to proteasome-dependent turnover at the INM (Figure 2G–H; Figure 3A–B), but can also be rapidly removed from the INM and ER membrane network and targeted for degradation during ER stress. This raises the possibility that additional INM proteins are susceptible to stress-dependent degradation. To address this, we tested the response of additional INM proteins to ER stress induction by THG, to ER export blockage by BFA, and to lysosomal maturation blockage by Baf A1. We selected proteins with distinct topologies and half-lives, including the long-lived polytopic INM protein nurim and the less stable single-pass transmembrane protein Sun2 (see Figure 1H, Table S5). Prolonged treatment with THG modestly decreased nurim protein levels and significantly decreased Sun2 protein levels (Figure 7A–B,D), likely as a consequence of translational inhibition caused by ER stress (Harding et al., 1999). Consistent with this interpretation, the sensitivity of these two proteins tracks with the relative differences in their half-lives (Table S5); nurim has a half-life of ~ 9 days, while Sun2 has a half-life of ~ 3 days in non-dividing cells. Importantly, however, co-incubation with THG and BFA had no effect on either the subcellular localization or abundance of nurim or Sun2 (Figure 7A–B,D), indicating that loss of these proteins is not mediated by ER export. Further, neither protein leaves the NE/ER to accumulate in lysosomes when lysosome acidification is blocked by Baf A1 (Figure 7A’, B’).

Figure 7. Emerin, but not other INM proteins, undergoes stress-dependent clearance from the NE and ER and accesses lysosomes.

(A-C) Representative confocal slices of cells stably expressing NRM-GFP (A), Sun2-GFP (B), or EMD-GFP (C) after 16 hr of treatment with DMSO vehicle control, THG, or co-treatment with THG and BFA. Insets show nuclei in the same ~ 50 µm field of view stained with Hoechst. (A’-C’) Representative confocal slices of cells co-treated with THG and Baf A1. All images were acquired using the same laser power and detector gain settings. (D) Quantification of GFP fluorescence intensity at the NE in maximum intensity projections of confocal z series acquired across conditions represented in (A-C). Columns indicate average and error bars indicate SEM for N > 690 cells from three independent experiments. **** indicates p-value<0.0001 compared to untreated (t-test). (E-F) Analysis of EMD-WT-GFP* glycosylation state in cells subjected to treatment with DMSO vehicle control or THG and CHX (E) or THG and Baf A1 (F) cotreatments for the times indicated. Red arrowhead indicates EndoH-sensitive glycosylated state of EMD-WT-GFP*; orange arrowhead indicates EndoH-resistant states of EMD-WT-GFP*; black arrowhead indicates deglycosylated EMD-WT-GFP*. α-tubulin shown as loading control. Numbers to left of blots indicate molecular weights in kDa. Scale bars in micrographs indicate 10 μm. See also Figure 7—figure supplement 1.

Figure 7.

Figure 7—figure supplement 1. Glycosylation reporter variants are destabilized by ER stress and recovered by BFA treatment.

Figure 7—figure supplement 1.

(A–B) Glycosylation reporter variant of EMD-WT-GFP localizes normally to the NE and responds to ER stress induced by THG and secretory pathway disruption caused by BFA.

We next asked whether wild type EMD was also subject to this pathway. Similarly to EMDΔ95–99, NE-localized EMD-GFP decreases when stress is induced by THG, but remains stable when stress induction by THG is accompanied by secretory pathway disruption by BFA (Figure 7C). ER stress also induces EMD-GFP to enrich in the Golgi (Figure 4—figure supplement 1) and access the plasma membrane (Figure 6—figure supplement 1). EMD-GFP also accumulates in perinuclear puncta that are likely lysosomes when cells are co-incubated with THG and Baf A1 (Figure 7C’). We again engineered an opsin glycosylation site onto the C-terminus of EMD-GFP in order to track movement of EMD through membrane compartments. EMD-WT-GFP* localizes to the NE and responds similarly to ER stress and secretory pathway blockage (Figure 7—figure supplement 1). As we observed with EMDΔ95–99, EMD-WT-GFP* exists predominantly in an Endo H-sensitive modification state in unstressed cells (Figure 7E–F, red arrowheads). Higher molecular weight, Endo H-resistant species increase in abundance when cells are coincubated with THG and CHX (Figure 7E, orange arrowhead) or with THG and Baf A1 (Figure 7F, orange arrowhead). We thus conclude that wild type EMD is subject to the same stress-induced lysosomal degradation pathway as EMDΔ95–99. However, when EMD-GFP’s response to THG is tracked over time, it is clear that displacement of wild type EMD from the NE proceeds significantly more slowly than displacement of EMDΔ95–99 (Figure 8B,D). This indicates that stress-dependent trafficking out of the INM is selective to variants of EMD, and that intrinsic features of EMD control its clearance from the NE/ER and targeting into lysosomes.

Figure 8. A signal within emerin’s.

LEM domain is required for stress-dependent clearance from the NE and ER (A) Diagram of emerin domain organization with N-terminal LEM domain deletion indicated (amino acids 1-45). (B-C) Representative confocal slices of C2C12 cells stably expressing EMD-WT-GFP (B) or EMDΔLEM-GFP (C) after treatment with DMSO vehicle control or THG for the indicated times. Insets show nuclei in the same ~50 µm field of view stained with Hoechst. (D) Quantification of EMDΔ95-99-GFP (as also shown in Figure 3F), EMD-WT-GFP, and EMDΔLEM-GFP fluorescence intensity at the NE in maximum intensity projections of confocal z series acquired across the timecourse shown in (B-C). Columns indicate average and error bars indicate SEM for N > 146 cells from 3 independent experiments. **** indicates p-value < 0.0001 compared to untreated (t-test). (E) Western blot of EMDΔLEM-GFP in cells treated with DMSO vehicle control or co-treated with THG and CHX for the indicated times. (F-G) Representative confocal slices of C2C12 cells stably expressing EMDΔLEM-GFP after treatment with (F) DMSO vehicle control, THG, THG + BFA, or (G) THG + Baf A1. (H) Quantification of GFP fluorescence intensity at the NE across the conditions shown in (F). Columns indicate average and error bars indicate SEM for N > 776 cells from 3 independent experiments. **** indicates p-value < 0.0001 compared to untreated (t-test). (I-J) Analysis of EMD-ΔLEM-GFP* glycosylation state in cells subjected to treatment with DMSO vehicle control or THG and CHX (I) or THG and Baf A1 (J) cotreatments for the times indicated. Red arrowhead indicates EndoH-sensitive glycosylated state of EMDΔLEM-GFP*; black arrowhead indicates deglycosylated EMDΔLEM-GFP*. a-tubulin shown as loading control. Numbers to left of blots indicate molecular weights in kDa. Scale bars in micrographs indicate 10 mm. See also Figure 8—figure supplements 1 and 2.

Figure 8.

Figure 8—figure supplement 1. Stability of EMDΔLEM-GFP over time of cycloheximide treatment.

Figure 8—figure supplement 1.

Western blot detection of EMDΔLEM-GFP protein levels in cells treated with DMSO vehicle control or CHX for the indicated times.
Figure 8—figure supplement 2. Emerin is mislocalized to the ER, but not degraded in lmna - /- MEFs.

Figure 8—figure supplement 2.

(A) representative immunostaining of WT MEFs or of lmna - /- MEFs with lamin A antibody (red) and Hoechst (blue). (B) Representative immunofluorescence of EMD-GFP stably expressed in WT MEFs (top panel) or lmna - /- MEFs (bottom panel).

A signal within emerin’s LEM domain is required for stress-dependent export

Why are EMD variants selectively targeted for stress-dependent clearance from the INM and ER? We considered functional domains that might be involved in responding to ER stress. EMD is a tail-anchored protein with a ~ 10 amino acid tail that protrudes into the ER lumen (Figure 3A). This short sequence lacks any known motifs for engaging with proteins within the ER lumen. EMD’s nucleoplasmic domain includes an N-terminal LEM domain and an internal lamin A-binding region (Figure 3A). Emerin relies on lamin A for targeting to the INM (Vaughan et al., 2001) but we noted that in lmna - /- MEFs, EMD-GFP remained stably expressed even while mislocalized to the peripheral ER (Figure 8—figure supplement 2). The small deletion within EMDΔ95–99 falls within the lamin A-binding domain, but does not appear to affect the protein’s affinity for the lamina as judged by FRAP (Figure 2—figure supplement 1), even though this variant responds more potently than wild type EMD to ER stress (Figure 8D). Taken together, these observations indicate that dissociation from the lamina is not sufficient to promote clearance of EMD from the NE/ER membrane system.

We next evaluated whether interactions mediated by the LEM domain could contribute to stress-dependent EMD export. The LEM domain (Figure 3A) is a protein fold that binds with high affinity to the soluble nucleoplasmic protein BAF (Lee et al., 2001). We deleted this domain and queried the effects on EMD localization and trafficking. When expressed within unperturbed cells, EMDΔLEM exhibited normal enrichment in the NE (Figure 8C), consistent with its ability to bind the lamina independently of the LEM domain. However, we observed that this mutant was less responsive than other EMD variants to ER stress induction; NE-localized EMDΔLEM was clearly detectable over several hours of THG treatment and was significantly less sensitive than full-length EMD to ER stress (Figure 8C–E). We surmise that the eventual loss of EMDΔLEM results from translational inhibition resulting from ER stress (Harding et al., 1999) and degradation by alternative pathways. Consistent with the interpretation that the LEM domain mediates post-ER trafficking, co-incubation of EMDΔLEM-expressing cells with THG and BFA or Baf A1 each had no effect on protein levels or localization (Figure 8F–H). These results suggest that without the LEM domain, EMD does not access post-ER compartments. To directly evaluate this, we generated a glycosylation-reporting variant, EMDΔLEM-GFP*, with a glycosylation consensus site at the lumenal C terminus. In contrast to variants of EMD with an intact LEM domain, EMDΔLEM-GFP* accumulates only Endo H-sensitive modifications and remains equivalently Endo H-sensitive during ER stress and lysosome blockage (Figure 8I–J, red arrowheads). Altogether, these data indicate that a signal within emerin’s LEM domain enables selective export from the ER under stress conditions.

Discussion

In this work, we applied a dynamic proteomic strategy to define organelle-wide trends in protein turnover across the NE/ER membrane network in mammalian cells. While the INM is separated from the bulk ER by the selective barrier of the NPC, we observe no difference in global protein turnover rates between the ER and INM compartments or any correlation between INM protein size and turnover kinetics. This, along with specific visualization of mature INM proteins by RITE tagging and microscopy (Figure 2) and previous studies (Tsai et al., 2016), suggests that turnover of INM proteins can be effectively achieved in situ.

Moving forward with the rapidly turned over INM protein EMD as a model substrate for dissecting INM protein turnover, we identified an even less stable, EDMD-linked variant of EMD as an ideal substrate for sensitively probing INM protein turnover pathways. We noted that turnover of maturely folded EMD and EMDΔ95–99 exhibits proteasome dependence at the INM (Figure 2F–H; Figure 3A), while nascent EMDΔ95–99 accumulates in multiple cellular compartments when the proteasome is inhibited (Figure 3A, fourth panel). Taken together, these observations lead us to infer that mature EMD variants and potentially other INM proteins can be turned over in situ at the INM by a pathway that terminates in proteasomal degradation, while immature EMD variants (and potentially other INM proteins) are also subject to co-translational quality control that terminates in proteasomal degradation. As proteasomal inhibition and p97 inhibition each stabilize EMDΔ95–99 (Figure 3B), we expect that EMDΔ95–99 is an ERAD client under some conditions.

Surprisingly, however, we also find that EMD can be selectively shunted to an alternative turnover pathway under conditions of acute ER stress. This pathway is rapidly induced by ERAD blockage or by pharmacological induction of acute ER protein folding stress (Figure 3) and requires ER export (Figures 3 and 4). Notably, changes to EMDΔ95–99 localization and levels are apparent at a timescale shorter than the normal half-life of EMDΔ95–99, within 2–4 hr of ER stress induction. Based on the transient appearance of EMDΔ95–99 at the PM (Figure 6) and its accumulation in lysosomes (Figure 5), we conclude that EMD transits through the secretory pathway and is then internalized into lysosomes. While our data indicate that a significant proportion of EMD leaves the NE/ER during ER stress, we cannot rule out the possibility that ERAD-mediated degradation of some proportion of EMD takes place within the NE/ER network in parallel to the lysosome-mediated pathway that we have identified. Nonetheless, this dynamic and selective removal of an INM protein is quite surprising and is inconsistent with models of the INM as a terminal depot for its resident proteins.

Our findings have some intriguing parallels to the fate of a misfolded variant of the GPI-anchored prion protein, PrP, during ER stress (Satpute-Krishnan et al., 2014). PrP is normally targeted to the PM, but a misfolded variant is retained within the ER by persistent association with protein folding chaperones. Similarly to what we observe for an INM protein, ER stress induces the rapid export of misfolded PrP through the secretory pathway, followed by transit through the PM and internalization and delivery to lysosomes for degradation. This mode of clearance has been referred to as rapid ER stress-induced export, or RESET (Satpute-Krishnan et al., 2014).

There are several notable contrasts between PrP’s export from the peripheral ER and EMD’s export from the INM and ER. For instance, the topologies of PrP and EMD are quite disparate. As a GPI-anchored protein, misfolded PrP faces the lumen of the ER, and an interaction between PrP and Tmp21, a sorting adaptor for luminal proteins, controls RESET (Satpute-Krishnan et al., 2014). Misfolded PrP remains associated with additional luminal ER-derived proteins during its transit through the secretory pathway, and these associations appear to enable recognition of misfolded PrP at the cell surface for internalization (Zavodszky and Hegde, 2019). In contrast, EMD is a tail-anchored protein, and interactions mediated by EMD’s nucleoplasmic-facing LEM domain (Figure 8) control its stress-dependent clearance. We do not yet know whether EMD remains associated with other proteins as it transits through the secretory pathway, or what role those associations might play in targeting EMD for degradation.

PrP and EMD also exhibit distinct subcellular localization when not undergoing RESET. Misfolded PrP is retained in the ER network until RESET is initiated, while EMD is enriched in the INM and associated with the nuclear lamina. Importantly, EMD is small enough (~25 kDa) to diffuse freely across the NPC, meaning that it may release INM-localized binding partners and sample the ER with some frequency. This spatial separation between EMD’s normal site of enrichment and its site of ER export may explain the longer timescale of RESET for EMD (2–4 hr) compared to ER-localized misfolded PrP (~1 hr) (Satpute-Krishnan et al., 2014).

Finally, PrP and EMD variants exhibit clear differences in selectivity for the RESET pathway. Only misfolded, ER-retained mutants of PrP are subject to RESET. On the other hand, both wild type EMD (Figure 7) and a less stable disease-linked variant (EMDΔ95–99) are subject to stress-dependent clearance, although EMDΔ95–99 is more rapidly cleared from the NE and ER. Both EMD variants appear functional until ER stress is induced, as judged by their localization and affinity for the INM (Figure 3, Figure 2—figure supplement 1). This suggests that clearance of EMD from the NE/ER is not strictly contingent on protein misfolding.

We find that selective, stress dependent clearance of EMD depends on its 45-amino acid LEM domain. LEM domains bind dynamically to the small soluble protein BAF, which exists in both nuclear and cytoplasmic pools (Shimi et al., 2004). While glycosylation reporters indicate that EMD variants also exit the NE/ER with some frequency under homeostatic conditions (Figure 3H, Figure 7E–F), this is completely abolished by deletion of the LEM domain (Figure 8). One model that could explain the dichotomy between LEM-mediated BAF binding and LEM-mediated ER export is that BAF and ER export-promoting factor(s) bind competitively to the same surface of EMD’s LEM domain (Figure 9). It could be that acute ER stress is relayed to EMD via a structural reorganization or post-translational modification that disrupts the LEM:BAF interface. It is possible that this system could be used to rapidly remove EMD in response to ER stress and potentially other physiological stressors. This could in turn rapidly inhibit the normal functions of EMD at the INM, including participating in mechanosensitive signaling pathways (Guilluy et al., 2014) and contributing to the scaffolding of heterochromatic domains at the nuclear periphery (Demmerle et al., 2013). Overall, our findings indicate that the INM can be rapidly remodeled in response to environmental stimuli, and that the function of the INM protein EMD may be dynamically controlled by integration of environmental inputs via its LEM domain.

Figure 9. Competition model for emerin sorting via its LEM domain competitively binding to BAF or to the ER export machinery.

Figure 9.

Notably, muscular dystrophy and cardiomyopathy diseases are caused by loss-of-function mutations to EMD, many of which further destabilize the protein (Bonne and Quijano-Roy, 2013; Fairley et al., 1999). We find evidence that a muscular dystrophy-linked EMD variant (EMDΔ95–99) is more rapidly degraded under acute stress conditions, suggesting that an overzealous response to ER stress could contribute to the pathogenesis of EDMD. EMD is broadly expressed (Uhlen et al., 2015), but mutations predominantly affect muscle tissues. Intriguingly, skeletal muscle undergoes significant ER stress both during development and during normal function (Deldicque et al., 2012). We speculate that these features of muscle physiology may make muscle-localized EMD mutants especially vulnerable to ER stress-induced degradation.

Materials and methods

Key resources table.

Reagent
type (species)
or resource
Designation Source or
reference
Identifiers Additional
information
Gene (Mus musculus) emerin NCBI RefSeq NM_007927
Gene (Mus musculus) nurim NCBI RefSeq NM_134122
Gene (Mus musculus) Sun2 NCBI RefSeq NM_001205346
Cell line (Mus musculus) C2C12 ATCC CRL-1772
Cell line (Homo sapiens) U-2-OS ATCC HTB-96
Recombinant DNA reagent (plasmid) pQCXIB vector Campeau et al. (2009) Addgene Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) Myc/FLAG RITE vector Toyama et al. (2019) Lentiviral contruct for stable expression of RITE-tagged protein
Recombinant DNA reagent (plasmid) pQCXIB emerin-GFP This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB emerin-D95-99-GFP This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB emerin-DLEM-GFP This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB emerin-GFP-SSNKTVD This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB emerin-Δ95–99-GFP-SSNKTVD This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB emerin-ΔLEM-GFP-SSNKTVD This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB Sun2-GFP This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) pQCXIB nurim-GFP This paper Retroviral construct for stable expression
Recombinant DNA reagent (plasmid) Emerin-RITE This paper Lentiviral contruct for stable expression of RITE-tagged protein
Recombinant DNA reagent (plasmid) Nurim-RITE This paper Lentiviral contruct for stable expression of RITE-tagged protein
Recombinant DNA reagent (plasmid) Emerin-Δ95–99-RITE This paper Lentiviral contruct for stable expression of RITE-tagged protein
Antibody Rabbit polyclonal anti-emerin Santa Cruz Biotechnology Sc-15378 WB (1:1000)
Antibody GFP Abcam ab290 Ab uptake (1:500); WB (1:1000)
Antibody Mouse monoclonal anti-FLAG Sigma-Aldrich F1804 IF (1:1000)
Antibody Mouse monoclonal anti-Myc Cell Signaling 2233 IF (1:1000); Ab uptake (1:500)
Antibody Mouse monoclonal anti-tubulin Sigma-Aldrich T5168 WB (1:2500)
Antibody giantin BioLegend PRB-114C IF (1:1000)
Antibody LAMP1 Abcam ab24170 IF(1:100)
Other Alexa-647 WGA Life Technologies W32466 IF (5 ug/ml)
Commercial assay or kit PNGase F NEB P0704
Commercial assay or kit Endo H NEB P0702
Chemical compound, drug Thapsigargin Thermo Fisher T7459 Used at 100 nM
Chemical compound, drug MG132 Cayman Chemical 1211877-36-9 Used at10 uM
Chemical compound, drug Bafilomycin A1 BioViotica BVT-0252 Used at100 nM
Chemical compound, drug Brefeldin A Tocris 1231 Used at2.5 uM
Chemical compound, drug Leupeptin Sigma-Aldrich L5793 Used at 125 uM
Chemical compound, drug cycloheximide Sigma-Aldrich C-7698 Used at200 ug/ml
Other 13C6-Lysine Cambridge Isotopes CLM-2247
Other 13C6, 15N4-Arginine Cambridge Isotopes CNLM-539
Other Lysine/arginine free DMEM Thermo Fisher 88364
Other Dialyzed fetal bovine serum Thermo Fisher 26400044
Other Hoechst stain Molecular Probes H1399 Used at 10 ug/ml
Recombinant DNA reagent (plasmid) UBE2G1 miR-E LT3GEPIR Knott et al., 2014 TGCTGTTGACAGTGAGCGAAAGACAGCTGGCAGAACTCAATAGTGAAGCCACAGATGTATTGAGTTCTGCCAGCTGTCTTCTGCCTACTGCCTCGGA
Recombinant DNA reagent (plasmid) UBE2G2 miR-E LT3GEPIR Knott et al., 2014 TGCTGTTGACAGTGAGCGAACCGGGAGCAGTTCTATAAGATAGTGAAGCCACAGATGTATCTTATAGAACTGCTCCCGGTCTGCCTACTGCCTCGGA
Recombinant DNA reagent (plasmid) UBE2J1 miR-E LT3GEPIR Knott et al., 2014 TGCTGTTGACAGTGAGCGAAAGGTTGTCTACTTCACCAGATAGTGAAGCCACAGATGTATCTGGTGAAGTAGACAACCTTCTGCCTACTGCCTCGGA
Recombinant DNA reagent (plasmid) MARCH6 miR-E LT3GEPIR Knott et al., 2014 TGCTGTTGACAGTGAGCGACTGGATCTTCATTCTTATTTATAGTGAAGCCACAGATGTATAAATAAGAATGAAGATCCAGCTGCCTACTGCCTCGGA
Software, algorithm Fiji https://fiji.sc/
Software, algorithm RStudio https://rstudio.com/

SILAC labeling

SILAC labeling was performed as a pulse-chase (Ong and Mann, 2006). Proliferating C2C12 mouse myoblasts were subcultured for > 5 population doublings in culture medium containing stable heavy isotopes of lysine and arginine (13C6-Lysine, 13C6, 15N4-Arginine) to completely label the cellular proteome. Cells were grown in SILAC-formulated DMEM lacking lysine and arginine and supplemented with 20% dialyzed FBS, penicillin/streptomycin, and SILAC amino acids. Complete label incorporation was verified by LC-MS/MS. Myoblasts were then grown to confluency and switched to differentiation medium containing heavy isotopes for 5 days to induce myotube differentiation. Differentiation medium contained SILAC DMEM, 2% dialyzed FBS, penicillin/streptomycin, and SILAC amino acids. Media was refreshed every other day. After differentiation, the mature myotube culture was switched to low serum differentiation medium containing the normal isotopes of lysine and arginine: 12C6-Lysine, 12C6, 14N4-Arginine for 1–3 days.

Crude nuclear extracts were prepared similarly to previous work (Buchwalter and Hetzer, 2017; Schirmer et al., 2003). Cells were harvested by scraping into PBS, then swollen in hypotonic lysis buffer (10 mM potassium acetate, 20 mM Tris acetate pH 7.5, 0.5 mM DTT, 1.5 mM MgCl2, and protease inhibitors), followed by mechanical lysis through a 25-gauge needle and syringe. The nuclei were pelleted and the supernatant (containing cytosol) was decanted. Nuclei were then resuspended in buffer containing 10 mM Tris pH 8.0, 10% sucrose, 1 mM DTT, 0.1 mM MgCl2, 20 ug/ml DNase I, and 1 ug/ml RNase I. After nuclease treatment, nuclei were layered on top of a 30% sucrose cushion and pelleted. Crude nuclei were then extracted in 10 mM Tris pH 8, 1% n-octyl glucoside, 400 mM NaCl, and 1 mM DTT, and extracts and pellets were prepared separately for liquid chromatography-mass spectrometry.

Lc-ms/MS

Samples were denatured in 8M urea/100 mM TEAB, pH 8.5; reduced with TCEP; alkylated with chloroacetamide; and digested overnight with trypsin. Digestion was quenched with 5% formic acid. Detergent was removed from pulse-labeled SILAC samples with SCX tips (EMD Millipore). Samples were run on a Thermo Orbitrap Fusion Tribrid MS/MS with CID fragmentation. The digest was injected directly onto a 30 cm, 75 um ID column packed with BEH 1.7 um C18 resin. Samples were separated at a flow rate of 200 nl/min on a nLC 1000. Buffer A and B were 0.1% formic acid in water and acetonitrile, respectively. A gradient of 1–25% B over 160 min, an increase to 35% B over 60 min, an increase to 90% B over another 10 min and a hold at 90%B for a final 10 min of washing was used for a total run time of 240 min. The column was re-equilibrated with 20 ul of buffer A prior to the injection of sample. Peptides were eluted directly from the tip of the column and nanosprayed into the mass spectrometer by application of 2.5 kV voltage at the back of the column. The Orbitrap Fusion was operated in data dependent mode. Full MS1 scans were collected in the Orbitrap at 120K resolution with a mass range of 400 to 1500 m/z and an AGC target of 4e. The cycle time was set to 3 s, and within this 3 s the most abundant ions per scan were selected for CID MS/MS in the ion trap with an AGC target of 1e and minimum intensity of 5000. Maximum fill times were set to 50 ms and 100 ms for MS and MS/MS scans, respectively. Quadrupole isolation at 1.6 m/z was used, monoisotopic precursor selection was enabled, charge states of 2–7 were selected and dynamic exclusion was used with an exclusion duration of 5 s.

Analysis of proteomic data

Peptide and protein identification, quantification, and analysis were performed with Integrated Proteomics Pipeline (IP2) (Integrated Proteomics Applications; www.integratedproteomics.com). Tandem mass spectra were extracted from raw files using RawConverter (He et al., 2015) and searched with ProLUCID (Xu et al., 2015) against the mouse UniProt database (ID). The search space included all fully tryptic and half-tryptic peptide candidates. Carbamidomethylation on cysteine was allowed as a static modification. Data were searched with 50 ppm precursor ion tolerance and 600 ppm fragment ion tolerance. Data were filtered to 10 ppm precursor ion tolerance post-search. Identified proteins were filtered using DTASelect (Tabb et al., 2002) and utilizing a target-decoy database strategy to control the false discovery rate to 1% at the protein level.

Census2 (Park et al., 2014) was used for quantitative analysis of SILAC-labeled peptides. Peptides were subjected to stringent quality control criteria before inclusion in half-life determination analyses. Firstly, any peptide with a profile score < 0.8 was discarded. Secondly, peptides were filtered based on the extent of correlation between the heavy and light chromatograms, which is quantified as a regression score in Census. Peptides with extreme area ratios (less than 0.111 or greater than 9) were retained only if their regression score was > 0. Peptides with intermediate area ratios (between 0.111 and 9) were retained only if their regression score was > 0.8.

Half-life calculation

For estimation of protein half-lives, we restricted our analysis to peptides that were detected in at least three timepoints. 1685 proteins passed this criterion with at least one peptide; individual peptides and protein-level data are reported in Tables S1 and S2, respectively. Area ratio values were transformed into % old values using the following equation:

%old=100(1/(1+AR))

And individual peptides were fit to a line corresponding to the following equation:

Ln(%old)=kt+a

Individual peptide fits with r2 > 0.8 and values of k < 0 were retained for protein-level estimation of half-life. The slope of the fit for all peptides detected were averaged to produce an average value and standard deviation at the protein level. These average slope values were converted to half-life estimates using the equation below.

T1/2=ln(2)/k

These values are reported in Table S3 for 1677 proteins. While calculated half-lives range from ~0.33 days to ~30 days, we note that half-lives at either extreme should be considered rough estimates of protein stability. For illustration, we have included example fits for proteins in Figure S1 with predicted half-lives of 0.5 day, 1 day, 2 days, 4 days, 8 days, and 16 days. Linear regression predicts half-life well under conditions where a line can be fitted with high fidelity and a non-zero slope is detectable. We note the good performance and clear distinctions in slope for proteins with predicted half-lives ranging from 1 to 8 days, and observed more frequent deviations in linearity at the low extreme (predicted T1/2 < 1 day) and slopes approaching zero at the high extreme (predicted T1/2 > 8 days). We expect that these factors limit the precision of half-life determination below 1 day and above 8 days from a 3 day timecourse. Shorter or longer timecourses would be required to investigate turnover at these timescales.

The TMHMM server (Krogh et al., 2001) was used to define the positions of transmembrane domains in INM proteins and infer extraluminal domain sequences.

Plasmid construction

All emerin constructs are based on the emerin sequence from mouse (Uniprot ID O08579); all nurim constructs based on the nurim sequence from mouse (Uniprot ID Q8VC65); and all Sun2 constructs based on the Sun2 sequence from mouse (Uniprot ID Q8BJS4). Emerin, Sun2, and nurim were each C-terminally tagged with GFP by stitching PCR (Heckman and Pease, 2007), where each open reading frame was amplified with the start codon included and the stop codon omitted, while GFP was amplified with its start codon omitted and stop codon included. Primersets were designed with overhangs including homology between each ORF and GFP, so that a second round of PCR with flanking primers and the first two PCR products used as templates generates an ORF-GFP fusion. EMDΔ95–99 was generated by Quickchange mutagenesis of the emerin-GFP sequence followed by sequence verification. EMDΔLEM-GFP was constructed by stitching PCR of emerin nucleic acid sequence 136–777 corresponding to residues 46–258 of emerin protein with the stop codon omitted. A new ATG start codon was introduced by PCR, and the C-terminal GFP tag introduced by stitching PCR. All ORFs were introduced into the pQCXIB vector (Campeau et al., 2009) for retroviral delivery and constitutive expression under a CMV promoter by Gateway cloning.

INM-RITE tag plasmids were constructed as described in Toyama et al. (2019). ORFs of interest were introduced into the FLAG/myc-RITE or myc/FLAG-RITE plasmid backbones, then the entire ORF-RITE construct was amplified and recombined into a pDONR207 Gateway entry vector, followed by recombination into the pQCXIB retroviral vector for constitutive mammalian expression.

Glycosylation reporter plasmids were constructed by introducing a 21 base-pair sequence encoding the glycosylation acceptor site SSNKTVD within a 3’ PCR primer for amplification of EMD-WT-GFP, EMDΔYEESY-GFP, and EMDΔLEM-GFP. The resulting PCR product was inserted into the pQCXIB vector by Gateway cloning. Sequence verified clones were used for stable cell line generation.

UBE2G1, UBE2G2, UBE2J1, and MARCH6 miR-E inducible RNAi plasmids were constructed as described in Fellmann et al. (2013). Validated shRNA sequences with the highest score for targeting mouse UBE2G1, UBE2G2, UBE2J1, and MARCH6 were selected from the shERWOOD database (www.sherwood.cshl.edu), and ~100 bp oligonucleotides with the corresponding sequence were synthesized. This sequence was amplified by PCR using degenerate primers with XhoI and EcoRI restriction sites. The PCR was digested with XhoI and EcoRI, gel purified, and ligated into the LT3GEPIR lentiviral vector for doxycycline-inducible RNAi expression with GFP reporter fluorescence. The LT3GEPIR vector was the kind gift of Johannes Zuber.

Cell line generation

GFP-tagged cell lines were generated in C2C12 mouse myoblasts. Low-passage C2C12 cells were obtained from ATCC, and identity was validated by a functional assay: cells were grown to confluency, switched to low serum medium for several days, and evaluated for the formation of multinucleated myotubes. Parallel cultures of C2C12 cells were infected with virus encoding GFP fusion proteins. 293 T cells were transfected with delivery vectors and viral packaging vectors for retroviral or lentiviral production. Conditioned media were collected 48–72 hr after transfection and applied to low-passage C2C12 cells in the presence of polybrene. Integrated clones were selected using the relevant antibiotic selections for each vector backbone. Fluorescent cell populations were isolated by FACS. The resulting stable GFP-expressing C2C12 cell lines were tested to verify the absence of mycoplasma contamination. miR-E RNAi cell lines were generated in U2OS cells. U2OS cells were obtained from ATCC and were periodically tested for mycoplasma contamination. EMDΔ95–99-GFP was introduced by retroviral infection and FACS sorted as described above. miR-E RNAi expression vectors were then introduced into these stable cell lines by lentiviral infection.

RITE tag switching

RITE tag switching experiments were performed in quiescent C2C12 cells stably expressing RITE-tagged INM proteins. C2C12s were grown in Ibidi chamber slides and induced to enter quiescence as previously described (Zhang et al., 2010) by growing C2C12 myoblasts to ~ 75% confluence, washing twice in warm PBS, and switching to quiescent medium (DMEM without methionine, 2% FBS, and pen-strep). Cells were maintained in quiescent medium for 3 days with media changes every other day before initiation of RITE timecourses. To induce tag exchange, concentrated adenovirus expressing Cre recombinase was added to the culture medium. Tag switching was initiated at the indicated timepoints such that the entire slide containing all time points could be fixed, stained, imaged, and quantified in parallel. To quantify loss of ‘old’ RITE-tagged protein over time, intensity per unit area of the ‘old’ tag was quantified across all conditions. Background measurements were taken from cell-free regions of the imaging dish and subtracted from all signals as a background correction. All signals were then normalized to the day 0 timepoint (no tag switch).

Antibody uptake assays

For antibody uptake assays, cells were pre-treated with drugs for the indicated times, then incubated in medium containing antibody, drug, and 125 μM leupeptin for 1 hr before fixation in paraformaldehyde and staining. Cells were stained with Alexa Fluor-conjugated secondary antibody to visualize internalized primary antibody:GFP conjugates. Cell surfaces were stained with Alexa Fluor-conjugated WGA; the WGA signal was used as a guide for outlining individual cells and quantifying internalized antibody fluorescence.

Preparation of protein lysates and western blotting

Cells were washed in PBS, then lysed directly in plates in PBS lysis buffer (PBS supplemented with 1% Triton-X-100, 0.1% SDS, and protease inhibitors). Cells were further lysed by passage through a 25-gauge needle. Protein concentrations were quantified by BCA assay, and ~20 ug of total protein was loaded per lane of 4–12% gradient gels (Invitrogen). Blots were incubated with emerin antibody (1:1000) or alpha-tubulin antibody (1:5000) followed by IR Dye-conjugated secondary antibodies (1:5000) for multiplexed detection on the Odyssey imaging system.

Microscopy and image analysis

Cells were grown in Ibidi culture chambers, treated as indicated, and fixed in 4% PFA for 5 min, then permeabilized in IF buffer (PBS, 0.1% Tx100, 0.02% SDS, 10 mg/ml BSA) before staining with Hoechst DNA dye. Wells were rinsed in PBS before imaging on a Zeiss LSM 710 scanning confocal microscope with a 63 × 1.4 NA objective. Images shown are single confocal slices. All image quantification was performed on maximum intensity projections of z-series with ImageJ. To quantify NE-localized protein levels, the DNA stain was used as a mask, and all GFP fluorescence within that mask was quantified.

For lysosomal staining, cells were prepared as described (Castellano et al., 2017) with the following modifications. Following fixation in 4% PFA for 5 min, cells were rinsed in PBS, then permeabilized in freshly prepared 0.1% digitonin in PBS for 10 min at 4C. Cells were rinsed again in PBS, then blocked in 2% goat serum in PBS for 30 min before staining with LAMP1 antibody (1:100 in 2% goat serum) for 1–2 hr at RT. Cells were rinsed again in PBS, then stained with Alexa Fluor-conjugated secondary antibody and Hoechst DNA stain for 1 hr at RT.

Funding Statement

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Contributor Information

Abigail Buchwalter, Email: abigail.buchwalter@ucsf.edu.

Martin Hetzer, Email: hetzer@salk.edu.

Elizabeth A Miller, MRC Laboratory of Molecular Biology, United Kingdom.

David Ron, University of Cambridge, United Kingdom.

Funding Information

This paper was supported by the following grants:

  • NIH Office of the Director NS096786 to Martin Hetzer.

  • National Institute of General Medical Sciences R01GM126829 to Martin Hetzer.

  • National Cancer Institute P30 014195 to Martin Hetzer.

  • Chapman Foundation to Martin Hetzer.

  • Helmsley Charitable Trust to Martin Hetzer.

Additional information

Competing interests

No competing interests declared.

Author contributions

Conceptualization, Data curation, Formal analysis, Validation, Investigation, Visualization, Methodology, Writing—original draft, Project administration, Writing—review and editing.

Data curation, Investigation, Methodology, Project administration.

Investigation, Methodology.

Software, Formal analysis, Methodology.

Resources, Supervision, Funding acquisition, Writing—review and editing.

Additional files

Source data 1. Filtered peptide data for half life calculations.

Peptide turnover data for all peptides passing quality control filters. See R script and Materials and methods for details.

elife-49796-data1.xlsx (2.2MB, xlsx)
DOI: 10.7554/eLife.49796.021
Source data 2. Filtered protein data for half life calculations.

Filtered and averaged protein turnover data. See R script and Materials and methods for details.

elife-49796-data2.xlsx (459.4KB, xlsx)
DOI: 10.7554/eLife.49796.022
Supplementary file 1. Results of half life fits passing quality filters.
elife-49796-supp1.xlsx (131KB, xlsx)
DOI: 10.7554/eLife.49796.023
Supplementary file 2. Complete list of half life fits.
elife-49796-supp2.xlsx (386.6KB, xlsx)
DOI: 10.7554/eLife.49796.024
Supplementary file 3. Half lives and protein topology data.

Selected data related to Figure 1G-H.

elife-49796-supp3.xlsx (24.7KB, xlsx)
DOI: 10.7554/eLife.49796.025
Transparent reporting form
DOI: 10.7554/eLife.49796.026

Data availability

Raw and analyzed mass spectrometric data and associated scripts and tables have been deposited in Dryad. Analyzed data are also included with the manuscript as supplementary tables.

The following dataset was generated:

Buchwalter A, Schulte R, Tsai H, Capitanio J, Hetzer MW. 2019. Data from: Selective clearance of the inner nuclear membrane protein emerin by vesicular transport during ER stress. Dryad Digital Repository.

References

  1. Albert S, Schaffer M, Beck F, Mosalaganti S, Asano S, Thomas HF, Plitzko JM, Beck M, Baumeister W, Engel BD. Proteasomes tether to two distinct sites at the nuclear pore complex. PNAS. 2017;114:13726–13731. doi: 10.1073/pnas.1716305114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Barlowe CK, Miller EA. Secretory protein biogenesis and traffic in the early secretory pathway. Genetics. 2013;193:383–410. doi: 10.1534/genetics.112.142810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Boni A, Politi AZ, Strnad P, Xiang W, Hossain MJ, Ellenberg J. Live imaging and modeling of inner nuclear membrane targeting reveals its molecular requirements in mammalian cells. The Journal of Cell Biology. 2015;209:705–720. doi: 10.1083/jcb.201409133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bonne G, Quijano-Roy S. Emery-Dreifuss muscular dystrophy, Laminopathies, and other nuclear envelopathies. Pediatric Neurology. 2013:1367–1376. doi: 10.1016/B978-0-444-59565-2.00007-1. [DOI] [PubMed] [Google Scholar]
  5. Brachner A, Foisner R. Evolvement of LEM proteins as chromatin tethers at the nuclear periphery. Biochemical Society Transactions. 2011;39:1735–1741. doi: 10.1042/BST20110724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Buchwalter A, Hetzer MW. Nucleolar expansion and elevated protein translation in premature aging. Nature Communications. 2017;8:328. doi: 10.1038/s41467-017-00322-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bulbarelli A, Sprocati T, Barberi M, Pedrazzini E, Borgese N. Trafficking of tail-anchored proteins: transport from the endoplasmic reticulum to the plasma membrane and sorting between surface domains in polarised epithelial cells. Journal of Cell Science. 2002;115:1689–1702. doi: 10.1242/jcs.115.8.1689. [DOI] [PubMed] [Google Scholar]
  8. Cambridge SB, Gnad F, Nguyen C, Bermejo JL, Krüger M, Mann M. Systems-wide proteomic analysis in mammalian cells reveals conserved, functional protein turnover. Journal of Proteome Research. 2011;10:5275–5284. doi: 10.1021/pr101183k. [DOI] [PubMed] [Google Scholar]
  9. Campeau E, Ruhl VE, Rodier F, Smith CL, Rahmberg BL, Fuss JO, Campisi J, Yaswen P, Cooper PK, Kaufman PD. A versatile viral system for expression and depletion of proteins in mammalian cells. PLOS ONE. 2009;4:e6529. doi: 10.1371/journal.pone.0006529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Castellano BM, Thelen AM, Moldavski O, Feltes M, van der Welle RE, Mydock-McGrane L, Jiang X, van Eijkeren RJ, Davis OB, Louie SM, Perera RM, Covey DF, Nomura DK, Ory DS, Zoncu R. Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science. 2017;355:1306–1311. doi: 10.1126/science.aag1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Chardin P, McCormick F. Brefeldin A: the advantage of being uncompetitive. Cell. 1999;97:153–155. doi: 10.1016/s0092-8674(00)80724-2. [DOI] [PubMed] [Google Scholar]
  12. Christianson JC, Olzmann JA, Shaler TA, Sowa ME, Bennett EJ, Richter CM, Tyler RE, Greenblatt EJ, Harper JW. Defining human ERAD networks through an integrative mapping strategy. Nature Publishing Group. 2011;14:93–105. doi: 10.1038/ncb2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. D'Angelo MA, Raices M, Panowski SH, Hetzer MW. Age-dependent deterioration of nuclear pore complexes causes a loss of nuclear integrity in postmitotic cells. Cell. 2009;136:284–295. doi: 10.1016/j.cell.2008.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Deldicque L, Hespel P, Francaux M. Endoplasmic reticulum stress in skeletal muscle: origin and metabolic consequences. Exercise and Sport Sciences Reviews. 2012;40:43–49. doi: 10.1097/JES.0b013e3182355e8c. [DOI] [PubMed] [Google Scholar]
  15. Demmerle J, Koch AJ, Holaska JM. Emerin and histone deacetylase 3 (HDAC3) cooperatively regulate expression and nuclear positions of MyoD, Myf5, and Pax7 genes during myogenesis. Chromosome Research. 2013;21:765–779. doi: 10.1007/s10577-013-9381-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Deshaies RJ, Joazeiro CA. RING domain E3 ubiquitin ligases. Annual Review of Biochemistry. 2009;78:399–434. doi: 10.1146/annurev.biochem.78.101807.093809. [DOI] [PubMed] [Google Scholar]
  17. Dörrbaum AR, Kochen L, Langer JD, Schuman EM. Local and global influences on protein turnover in neurons and Glia. eLife. 2018;7:489. doi: 10.7554/eLife.34202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Fagioli C, Sitia R. Glycoprotein quality control in the endoplasmic reticulum. Journal of Biological Chemistry. 2001;276:12885–12892. doi: 10.1074/jbc.M009603200. [DOI] [PubMed] [Google Scholar]
  19. Fairley EA, Kendrick-Jones J, Ellis JA. The Emery-Dreifuss muscular dystrophy phenotype arises from aberrant targeting and binding of emerin at the inner nuclear membrane. Journal of Cell Science. 1999;112 ( Pt 15:2571–2582. doi: 10.1242/jcs.112.15.2571. [DOI] [PubMed] [Google Scholar]
  20. Farkas T, Daugaard M, Jäättelä M. Identification of small molecule inhibitors of phosphatidylinositol 3-kinase and autophagy. Journal of Biological Chemistry. 2011;286:38904–38912. doi: 10.1074/jbc.M111.269134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Fellmann C, Hoffmann T, Sridhar V, Hopfgartner B, Muhar M, Roth M, Lai DY, Barbosa IA, Kwon JS, Guan Y, Sinha N, Zuber J. An optimized microRNA backbone for effective single-copy RNAi. Cell Reports. 2013;5:1704–1713. doi: 10.1016/j.celrep.2013.11.020. [DOI] [PubMed] [Google Scholar]
  22. Foresti O, Ruggiano A, Hannibal-Bach HK, Ejsing CS, Carvalho P. Sterol homeostasis requires regulated degradation of squalene monooxygenase by the ubiquitin ligase Doa10/Teb4. eLife. 2013;2:e00953. doi: 10.7554/eLife.00953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Gao R, Schellenberg MJ, Huang SY, Abdelmalak M, Marchand C, Nitiss KC, Nitiss JL, Williams RS, Pommier Y. Proteolytic degradation of topoisomerase II (Top2) enables the processing of Top2·DNA and Top2·RNA covalent complexes by tyrosyl-DNA-phosphodiesterase 2 (TDP2) Journal of Biological Chemistry. 2014;289:17960–17969. doi: 10.1074/jbc.M114.565374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Guilluy C, Osborne LD, Van Landeghem L, Sharek L, Superfine R, Garcia-Mata R, Burridge K. Isolated nuclei adapt to force and reveal a mechanotransduction pathway in the nucleus. Nature Cell Biology. 2014;16:376–381. doi: 10.1038/ncb2927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Harding HP, Zhang Y, Ron D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 1999;397:271–274. doi: 10.1038/16729. [DOI] [PubMed] [Google Scholar]
  26. He L, Diedrich J, Chu YY, Yates JR. Extracting accurate precursor information for tandem mass spectra by RawConverter. Analytical Chemistry. 2015;87:11361–11367. doi: 10.1021/acs.analchem.5b02721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Heckman KL, Pease LR. Gene splicing and mutagenesis by PCR-driven overlap extension. Nature Protocols. 2007;2:924–932. doi: 10.1038/nprot.2007.132. [DOI] [PubMed] [Google Scholar]
  28. Hegde RS, Zavodszky E. Recognition and Degradation of Mislocalized Proteins in Health and Disease. Cold Spring Harbor Perspectives in Biology. 2019;19:a033902. doi: 10.1101/cshperspect.a033902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Hofemeister H, O'Hare P. Analysis of the localization and topology of nurim, a polytopic protein tightly associated with the inner nuclear membrane. Journal of Biological Chemistry. 2005;280:2512–2521. doi: 10.1074/jbc.M410504200. [DOI] [PubMed] [Google Scholar]
  30. Katta SS, Smoyer CJ, Jaspersen SL. Destination: inner nuclear membrane. Trends in Cell Biology. 2014;24:221–229. doi: 10.1016/j.tcb.2013.10.006. [DOI] [PubMed] [Google Scholar]
  31. Khmelinskii A, Blaszczak E, Pantazopoulou M, Fischer B, Omnus DJ, Le Dez G, Brossard A, Gunnarsson A, Barry JD, Meurer M, Kirrmaier D, Boone C, Huber W, Rabut G, Ljungdahl PO, Knop M. Protein quality control at the inner nuclear membrane. Nature. 2014;516:410–413. doi: 10.1038/nature14096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, Adachi H, Adams CM, Adams PD, Adeli K, Adhihetty PJ, Adler SG, Agam G, Agarwal R, Aghi MK, Agnello M, Agostinis P, Aguilar PV, Aguirre-Ghiso J, Airoldi EM, Ait-Si-Ali S, Akematsu T, Akporiaye ET, Al-Rubeai M, Albaiceta GM, Albanese C, Albani D, Albert ML, Aldudo J, Algül H, Alirezaei M, Alloza I, Almasan A, Almonte-Beceril M, Alnemri ES, Alonso C, Altan-Bonnet N, Altieri DC, Alvarez S, Alvarez-Erviti L, Alves S, Amadoro G, Amano A, Amantini C, Ambrosio S, Amelio I, Amer AO, Amessou M, Amon A, An Z, Anania FA, Andersen SU, Andley UP, Andreadi CK, Andrieu-Abadie N, Anel A, Ann DK, Anoopkumar-Dukie S, Antonioli M, Aoki H, Apostolova N, Aquila S, Aquilano K, Araki K, Arama E, Aranda A, Araya J, Arcaro A, Arias E, Arimoto H, Ariosa AR, Armstrong JL, Arnould T, Arsov I, Asanuma K, Askanas V, Asselin E, Atarashi R, Atherton SS, Atkin JD, Attardi LD, Auberger P, Auburger G, Aurelian L, Autelli R, Avagliano L, Avantaggiati ML, Avrahami L, Awale S, Azad N, Bachetti T, Backer JM, Bae D-H, Bae J-sung, Bae O-N, Bae SH, Baehrecke EH, Baek S-H, Baghdiguian S, Bagniewska-Zadworna A, Bai H, Bai J, Bai X-Y, Bailly Y, Balaji KN, Balduini W, Ballabio A, Balzan R, Banerjee R, Bánhegyi G, Bao H, Barbeau B, Barrachina MD, Barreiro E, Bartel B, Bartolomé A, Bassham DC, Bassi MT, Bast RC, Basu A, Batista MT, Batoko H, Battino M, Bauckman K, Baumgarner BL, Bayer KU, Beale R, Beaulieu J-F, Beck GR, Becker C, Beckham JD, Bédard P-A, Bednarski PJ, Begley TJ, Behl C, Behrends C, Behrens GMN, Behrns KE, Bejarano E, Belaid A, Belleudi F, Bénard G, Berchem G, Bergamaschi D, Bergami M, Berkhout B, Berliocchi L, Bernard A, Bernard M, Bernassola F, Bertolotti A, Bess AS, Besteiro S, Bettuzzi S, Bhalla S, Bhattacharyya S, Bhutia SK, Biagosch C, Bianchi MW, Biard-Piechaczyk M, Billes V, Bincoletto C, Bingol B, Bird SW, Bitoun M, Bjedov I, Blackstone C, Blanc L, Blanco GA, Blomhoff HK, Boada-Romero E, Böckler S, Boes M, Boesze-Battaglia K, Boise LH, Bolino A, Boman A, Bonaldo P, Bordi M, Bosch J, Botana LM, Botti J, Bou G, Bouché M, Bouchecareilh M, Boucher M-J, Boulton ME, Bouret SG, Boya P, Boyer-Guittaut M, Bozhkov PV, Brady N, Braga VMM, Brancolini C, Braus GH, Bravo-San Pedro JM, Brennan LA, Bresnick EH, Brest P, Bridges D, Bringer M-A, Brini M, Brito GC, Brodin B, Brookes PS, Brown EJ, Brown K, Broxmeyer HE, Bruhat A, Brum PC, Brumell JH, Brunetti-Pierri N, Bryson-Richardson RJ, Buch S, Buchan AM, Budak H, Bulavin DV, Bultman SJ, Bultynck G, Bumbasirevic V, Burelle Y, Burke RE, Burmeister M, Bütikofer P, Caberlotto L, Cadwell K, Cahova M, Cai D, Cai J, Cai Q, Calatayud S, Camougrand N, Campanella M, Campbell GR, Campbell M, Campello S, Candau R, Caniggia I, Cantoni L, Cao L, Caplan AB, Caraglia M, Cardinali C, Cardoso SM, Carew JS, Carleton LA, Carlin CR, Carloni S, Carlsson SR, Carmona-Gutierrez D, Carneiro LAM, Carnevali O, Carra S, Carrier A, Carroll B, Casas C, Casas J, Cassinelli G, Castets P, Castro-Obregon S, Cavallini G, Ceccherini I, Cecconi F, Cederbaum AI, Ceña V, Cenci S, Cerella C, Cervia D, Cetrullo S, Chaachouay H, Chae H-J, Chagin AS, Chai C-Y, Chakrabarti G, Chamilos G, Chan EYW, Chan MTV, Chandra D, Chandra P, Chang C-P, Chang RC-C, Chang TY, Chatham JC, Chatterjee S, Chauhan S, Che Y, Cheetham ME, Cheluvappa R, Chen C-J, Chen G, Chen G-C, Chen G, Chen H, Chen JW, Chen J-K, Chen M, Chen M, Chen P, Chen Q, Chen Q, Chen S-D, Chen S, Chen SS-L, Chen W, Chen W-J, Chen WQ, Chen W, Chen X, Chen Y-H, Chen Y-G, Chen Y, Chen Y, Chen Y, Chen Y-J, Chen Y-Q, Chen Y, Chen Z, Chen Z, Cheng A, Cheng CHK, Cheng H, Cheong H, Cherry S, Chesney J, Cheung CHA, Chevet E, Chi HC, Chi S-G, Chiacchiera F, Chiang H-L, Chiarelli R, Chiariello M, Chieppa M, Chin L-S, Chiong M, Chiu GNC, Cho D-H, Cho S-G, Cho WC, Cho Y-Y, Cho Y-S, Choi AMK, Choi E-J, Choi E-K, Choi J, Choi ME, Choi S-I, Chou T-F, Chouaib S, Choubey D, Choubey V, Chow K-C, Chowdhury K, Chu CT, Chuang T-H, Chun T, Chung H, Chung T, Chung Y-L, Chwae Y-J, Cianfanelli V, Ciarcia R, Ciechomska IA, Ciriolo MR, Cirone M, Claerhout S, Clague MJ, Clària J, Clarke PGH, Clarke R, Clementi E, Cleyrat C, Cnop M, Coccia EM, Cocco T, Codogno P, Coers J, Cohen EEW, Colecchia D, Coletto L, Coll NS, Colucci-Guyon E, Comincini S, Condello M, Cook KL, Coombs GH, Cooper CD, Cooper JM, Coppens I, Corasaniti MT, Corazzari M, Corbalan R, Corcelle-Termeau E, Cordero MD, Corral-Ramos C, Corti O, Cossarizza A, Costelli P, Costes S, Cotman SL, Coto-Montes A, Cottet S, Couve E, Covey LR, Cowart LA, Cox JS, Coxon FP, Coyne CB, Cragg MS, Craven RJ, Crepaldi T, Crespo JL, Criollo A, Crippa V, Cruz MT, Cuervo AM, Cuezva JM, Cui T, Cutillas PR, Czaja MJ, Czyzyk-Krzeska MF, Dagda RK, Dahmen U, Dai C, Dai W, Dai Y, Dalby KN, Dalla Valle L, Dalmasso G, D'Amelio M, Damme M, Darfeuille-Michaud A, Dargemont C, Darley-Usmar VM, Dasarathy S, Dasgupta B, Dash S, Dass CR, Davey HM, Davids LM, Dávila D, Davis RJ, Dawson TM, Dawson VL, Daza P, de Belleroche J, de Figueiredo P, de Figueiredo RCBQ, de la Fuente J, De Martino L, De Matteis A, De Meyer GRY, De Milito A, De Santi M, de Souza W, De Tata V, De Zio D, Debnath J, Dechant R, Decuypere J-P, Deegan S, Dehay B, Del Bello B, Del Re DP, Delage-Mourroux R, Delbridge LMD, Deldicque L, Delorme-Axford E, Deng Y, Dengjel J, Denizot M, Dent P, Der CJ, Deretic V, Derrien B, Deutsch E, Devarenne TP, Devenish RJ, Di Bartolomeo S, Di Daniele N, Di Domenico F, Di Nardo A, Di Paola S, Di Pietro A, Di Renzo L, DiAntonio A, Díaz-Araya G, Díaz-Laviada I, Diaz-Meco MT, Diaz-Nido J, Dickey CA, Dickson RC, Diederich M, Digard P, Dikic I, Dinesh-Kumar SP, Ding C, Ding W-X, Ding Z, Dini L, Distler JHW, Diwan A, Djavaheri-Mergny M, Dmytruk K, Dobson RCJ, Doetsch V, Dokladny K, Dokudovskaya S, Donadelli M, Dong XC, Dong X, Dong Z, Donohue TM, Doran KS, D'Orazi G, Dorn GW, Dosenko V, Dridi S, Drucker L, Du J, Du L-L, Du L, du Toit A, Dua P, Duan L, Duann P, Dubey VK, Duchen MR, Duchosal MA, Duez H, Dugail I, Dumit VI, Duncan MC, Dunlop EA, Dunn WA, Dupont N, Dupuis L, Durán RV, Durcan TM, Duvezin-Caubet S, Duvvuri U, Eapen V, Ebrahimi-Fakhari D, Echard A, Eckhart L, Edelstein CL, Edinger AL, Eichinger L, Eisenberg T, Eisenberg-Lerner A, Eissa NT, El-Deiry WS, El-Khoury V, Elazar Z, Eldar-Finkelman H, Elliott CJH, Emanuele E, Emmenegger U, Engedal N, Engelbrecht A-M, Engelender S, Enserink JM, Erdmann R, Erenpreisa J, Eri R, Eriksen JL, Erman A, Escalante R, Eskelinen E-L, Espert L, Esteban-Martínez L, Evans TJ, Fabri M, Fabrias G, Fabrizi C, Facchiano A, Færgeman NJ, Faggioni A, Fairlie WD, Fan C, Fan D, Fan J, Fang S, Fanto M, Fanzani A, Farkas T, Faure M, Favier FB, Fearnhead H, Federici M, Fei E, Felizardo TC, Feng H, Feng Y, Feng Y, Ferguson TA, Fernández Álvaro F, Fernandez-Barrena MG, Fernandez-Checa JC, Fernández-López A, Fernandez-Zapico ME, Feron O, Ferraro E, Ferreira-Halder CV, Fesus L, Feuer R, Fiesel FC, Filippi-Chiela EC, Filomeni G, Fimia GM, Fingert JH, Finkbeiner S, Finkel T, Fiorito F, Fisher PB, Flajolet M, Flamigni F, Florey O, Florio S, Floto RA, Folini M, Follo C, Fon EA, Fornai F, Fortunato F, Fraldi A, Franco R, Francois A, François A, Frankel LB, Fraser IDC, Frey N, Freyssenet DG, Frezza C, Friedman SL, Frigo DE, Fu D, Fuentes JM, Fueyo J, Fujitani Y, Fujiwara Y, Fujiya M, Fukuda M, Fulda S, Fusco C, Gabryel B, Gaestel M, Gailly P, Gajewska M, Galadari S, Galili G, Galindo I, Galindo MF, Galliciotti G, Galluzzi L, Galluzzi L, Galy V, Gammoh N, Gandy S, Ganesan AK, Ganesan S, Ganley IG, Gannagé M, Gao F-B, Gao F, Gao J-X, García Nannig L, García Véscovi E, Garcia-Macía M, Garcia-Ruiz C, Garg AD, Garg PK, Gargini R, Gassen NC, Gatica D, Gatti E, Gavard J, Gavathiotis E, Ge L, Ge P, Ge S, Gean P-W, Gelmetti V, Genazzani AA, Geng J, Genschik P, Gerner L, Gestwicki JE, Gewirtz DA, Ghavami S, Ghigo E, Ghosh D, Giammarioli AM, Giampieri F, Giampietri C, Giatromanolaki A, Gibbings DJ, Gibellini L, Gibson SB, Ginet V, Giordano A, Giorgini F, Giovannetti E, Girardin SE, Gispert S, Giuliano S, Gladson CL, Glavic A, Gleave M, Godefroy N, Gogal RM, Gokulan K, Goldman GH, Goletti D, Goligorsky MS, Gomes AV, Gomes LC, Gomez H, Gomez-Manzano C, Gómez-Sánchez R, Gonçalves DAP, Goncu E, Gong Q, Gongora C, Gonzalez CB, Gonzalez-Alegre P, Gonzalez-Cabo P, González-Polo RA, Goping IS, Gorbea C, Gorbunov NV, Goring DR, Gorman AM, Gorski SM, Goruppi S, Goto-Yamada S, Gotor C, Gottlieb RA, Gozes I, Gozuacik D, Graba Y, Graef M, Granato GE, Grant GD, Grant S, Gravina GL, Green DR, Greenhough A, Greenwood MT, Grimaldi B, Gros F, Grose C, Groulx J-F, Gruber F, Grumati P, Grune T, Guan J-L, Guan K-L, Guerra B, Guillen C, Gulshan K, Gunst J, Guo C, Guo L, Guo M, Guo W, Guo X-G, Gust AA, Gustafsson Åsa B, Gutierrez E, Gutierrez MG, Gwak H-S, Haas A, Haber JE, Hadano S, Hagedorn M, Hahn DR, Halayko AJ, Hamacher-Brady A, Hamada K, Hamai A, Hamann A, Hamasaki M, Hamer I, Hamid Q, Hammond EM, Han F, Han W, Handa JT, Hanover JA, Hansen M, Harada M, Harhaji-Trajkovic L, Harper JW, Harrath AH, Harris AL, Harris J, Hasler U, Hasselblatt P, Hasui K, Hawley RG, Hawley TS, He C, He CY, He F, He G, He R-R, He X-H, He Y-W, He Y-Y, Heath JK, Hébert M-J, Heinzen RA, Helgason GV, Hensel M, Henske EP, Her C, Herman PK, Hernández A, Hernandez C, Hernández-Tiedra S, Hetz C, Hiesinger PR, Higaki K, Hilfiker S, Hill BG, Hill JA, Hill WD, Hino K, Hofius D, Hofman P, Höglinger GU, Höhfeld J, Holz MK, Hong Y, Hood DA, Hoozemans JJM, Hoppe T, Hsu C, Hsu C-Y, Hsu L-C, Hu D, Hu G, Hu H-M, Hu H, Hu MC, Hu Y-C, Hu Z-W, Hua F, Hua Y, Huang C, Huang H-L, Huang K-H, Huang K-Y, Huang S, Huang S, Huang W-P, Huang Y-R, Huang Y, Huang Y, Huber TB, Huebbe P, Huh W-K, Hulmi JJ, Hur GM, Hurley JH, Husak Z, Hussain SNA, Hussain S, Hwang JJ, Hwang S, Hwang TIS, Ichihara A, Imai Y, Imbriano C, Inomata M, Into T, Iovane V, Iovanna JL, Iozzo RV, Ip NY, Irazoqui JE, Iribarren P, Isaka Y, Isakovic AJ, Ischiropoulos H, Isenberg JS, Ishaq M, Ishida H, Ishii I, Ishmael JE, Isidoro C, Isobe K-ichi, Isono E, Issazadeh-Navikas S, Itahana K, Itakura E, Ivanov AI, Iyer AKV, Izquierdo JM, Izumi Y, Izzo V, Jäättelä M, Jaber N, Jackson DJ, Jackson WT, Jacob TG, Jacques TS, Jagannath C, Jain A, Jana NR, Jang BK, Jani A, Janji B, Jannig PR, Jansson PJ, Jean S, Jendrach M, Jeon J-H, Jessen N, Jeung E-B, Jia K, Jia L, Jiang H, Jiang H, Jiang L, Jiang T, Jiang X, Jiang X, Jiang X, Jiang Y, Jiang Y, Jiménez A, Jin C, Jin H, Jin L, Jin M, Jin S, Jinwal UK, Jo E-K, Johansen T, Johnson DE, Johnson GVW, Johnson JD, Jonasch E, Jones C, Joosten LAB, Jordan J, Joseph A-M, Joseph B, Joubert AM, Ju D, Ju J, Juan H-F, Juenemann K, Juhász G, Jung HS, Jung JU, Jung Y-K, Jungbluth H, Justice MJ, Jutten B, Kaakoush NO, Kaarniranta K, Kaasik A, Kabuta T, Kaeffer B, Kågedal K, Kahana A, Kajimura S, Kakhlon O, Kalia M, Kalvakolanu DV, Kamada Y, Kambas K, Kaminskyy VO, Kampinga HH, Kandouz M, Kang C, Kang R, Kang T-C, Kanki T, Kanneganti T-D, Kanno H, Kanthasamy AG, Kantorow M, Kaparakis-Liaskos M, Kapuy O, Karantza V, Karim MR, Karmakar P, Kaser A, Kaushik S, Kawula T, Kaynar AM, Ke P-Y, Ke Z-J, Kehrl JH, Keller KE, Kemper JK, Kenworthy AK, Kepp O, Kern A, Kesari S, Kessel D, Ketteler R, Kettelhut IdoC, Khambu B, Khan MM, Khandelwal VKM, Khare S, Kiang JG, Kiger AA, Kihara A, Kim AL, Kim CH, Kim DR, Kim D-H, Kim EK, Kim HY, Kim H-R, Kim J-S, Kim JH, Kim JC, Kim JH, Kim KW, Kim MD, Kim M-M, Kim PK, Kim SW, Kim S-Y, Kim Y-S, Kim Y, Kimchi A, Kimmelman AC, Kimura T, King JS, Kirkegaard K, Kirkin V, Kirshenbaum LA, Kishi S, Kitajima Y, Kitamoto K, Kitaoka Y, Kitazato K, Kley RA, Klimecki WT, Klinkenberg M, Klucken J, Knævelsrud H, Knecht E, Knuppertz L, Ko J-L, Kobayashi S, Koch JC, Koechlin-Ramonatxo C, Koenig U, Koh YH, Köhler K, Kohlwein SD, Koike M, Komatsu M, Kominami E, Kong D, Kong HJ, Konstantakou EG, Kopp BT, Korcsmaros T, Korhonen L, Korolchuk VI, Koshkina NV, Kou Y, Koukourakis MI, Koumenis C, Kovács AL, Kovács T, Kovacs WJ, Koya D, Kraft C, Krainc D, Kramer H, Kravic-Stevovic T, Krek W, Kretz-Remy C, Krick R, Krishnamurthy M, Kriston-Vizi J, Kroemer G, Kruer MC, Kruger R, Ktistakis NT, Kuchitsu K, Kuhn C, Kumar AP, Kumar A, Kumar A, Kumar D, Kumar D, Kumar R, Kumar S, Kundu M, Kung H-J, Kuno A, Kuo S-H, Kuret J, Kurz T, Kwok T, Kwon TK, Kwon YT, Kyrmizi I, La Spada AR, Lafont F, Lahm T, Lakkaraju A, Lam T, Lamark T, Lancel S, Landowski TH, Lane DJR, Lane JD, Lanzi C, Lapaquette P, Lapierre LR, Laporte J, Laukkarinen J, Laurie GW, Lavandero S, Lavie L, LaVoie MJ, Law BYK, Law HK-wai, Law KB, Layfield R, Lazo PA, Le Cam L, Le Roch KG, Le Stunff H, Leardkamolkarn V, Lecuit M, Lee B-H, Lee C-H, Lee EF, Lee GM, Lee H-J, Lee H, Lee JK, Lee J, Lee J-hyun, Lee JH, Lee M, Lee M-S, Lee PJ, Lee SW, Lee S-J, Lee S-J, Lee SY, Lee SH, Lee SS, Lee S-J, Lee S, Lee Y-R, Lee YJ, Lee YH, Leeuwenburgh C, Lefort S, Legouis R, Lei J, Lei Q-Y, Leib DA, Leibowitz G, Lekli I, Lemaire SD, Lemasters JJ, Lemberg MK, Lemoine A, Leng S, Lenz G, Lenzi P, Lerman LO, Lettieri Barbato D, Leu JI-J, Leung HY, Levine B, Lewis PA, Lezoualc'h F, Li C, Li F, Li F-J, Li J, Li K, Li L, Li M, Li M, Li Q, Li R, Li S, Li W, Li W, Li X, Li Y, Lian J, Liang C, Liang Q, Liao Y, Liberal J, Liberski PP, Lie P, Lieberman AP, Lim HJ, Lim K-L, Lim K, Lima RT, Lin C-S, Lin C-F, Lin F, Lin F, Lin F-C, Lin K, Lin K-H, Lin P-H, Lin T, Lin W-W, Lin Y-S, Lin Y, Linden R, Lindholm D, Lindqvist LM, Lingor P, Linkermann A, Liotta LA, Lipinski MM, Lira VA, Lisanti MP, Liton PB, Liu B, Liu C, Liu C-F, Liu F, Liu H-J, Liu J, Liu J-J, Liu J-L, Liu K, Liu L, Liu L, Liu Q, Liu R-Y, Liu S, Liu S, Liu W, Liu X-D, Liu X, Liu X-H, Liu X, Liu X, Liu X, Liu Y, Liu Y, Liu Z, Liu Z, Liuzzi JP, Lizard G, Ljujic M, Lodhi IJ, Logue SE, Lokeshwar BL, Long YC, Lonial S, Loos B, López-Otín C, López-Vicario C, Lorente M, Lorenzi PL, Lõrincz P, Los M, Lotze MT, Lovat PE, Lu B, Lu B, Lu J, Lu Q, Lu S-M, Lu S, Lu Y, Luciano F, Luckhart S, Lucocq JM, Ludovico P, Lugea A, Lukacs NW, Lum JJ, Lund AH, Luo H, Luo J, Luo S, Luparello C, Lyons T, Ma J, Ma Y, Ma Y, Ma Z, Machado J, Machado-Santelli GM, Macian F, MacIntosh GC, MacKeigan JP, Macleod KF, MacMicking JD, MacMillan-Crow LA, Madeo F, Madesh M, Madrigal-Matute J, Maeda A, Maeda T, Maegawa G, Maellaro E, Maes H, Magariños M, Maiese K, Maiti TK, Maiuri L, Maiuri MC, Maki CG, Malli R, Malorni W, Maloyan A, Mami-Chouaib F, Man N, Mancias JD, Mandelkow E-M, Mandell MA, Manfredi AA, Manié SN, Manzoni C, Mao K, Mao Z, Mao Z-W, Marambaud P, Marconi AM, Marelja Z, Marfe G, Margeta M, Margittai E, Mari M, Mariani FV, Marin C, Marinelli S, Mariño G, Markovic I, Marquez R, Martelli AM, Martens S, Martin KR, Martin SJ, Martin S, Martin-Acebes MA, Martín-Sanz P, Martinand-Mari C, Martinet W, Martinez J, Martinez-Lopez N, Martinez-Outschoorn U, Martínez-Velázquez M, Martinez-Vicente M, Martins WK, Mashima H, Mastrianni JA, Matarese G, Matarrese P, Mateo R, Matoba S, Matsumoto N, Matsushita T, Matsuura A, Matsuzawa T, Mattson MP, Matus S, Maugeri N, Mauvezin C, Mayer A, Maysinger D, Mazzolini GD, McBrayer MK, McCall K, McCormick C, McInerney GM, McIver SC, McKenna S, McMahon JJ, McNeish IA, Mechta-Grigoriou F, Medema JP, Medina DL, Megyeri K, Mehrpour M, Mehta JL, Mei Y, Meier U-C, Meijer AJ, Meléndez A, Melino G, Melino S, de Melo EJT, Mena MA, Meneghini MD, Menendez JA, Menezes R, Meng L, Meng L-hua, Meng S, Menghini R, Menko AS, Menna-Barreto RFS, Menon MB, Meraz-Ríos MA, Merla G, Merlini L, Merlot AM, Meryk A, Meschini S, Meyer JN, Mi M-tian, Miao C-Y, Micale L, Michaeli S, Michiels C, Migliaccio AR, Mihailidou AS, Mijaljica D, Mikoshiba K, Milan E, Miller-Fleming L, Mills GB, Mills IG, Minakaki G, Minassian BA, Ming X-F, Minibayeva F, Minina EA, Mintern JD, Minucci S, Miranda-Vizuete A, Mitchell CH, Miyamoto S, Miyazawa K, Mizushima N, Mnich K, Mograbi B, Mohseni S, Moita LF, Molinari M, Molinari M, Møller AB, Mollereau B, Mollinedo F, Mongillo M, Monick MM, Montagnaro S, Montell C, Moore DJ, Moore MN, Mora-Rodriguez R, Moreira PI, Morel E, Morelli MB, Moreno S, Morgan MJ, Moris A, Moriyasu Y, Morrison JL, Morrison LA, Morselli E, Moscat J, Moseley PL, Mostowy S, Motori E, Mottet D, Mottram JC, Moussa CE-H, Mpakou VE, Mukhtar H, Mulcahy Levy JM, Muller S, Muñoz-Moreno R, Muñoz-Pinedo C, Münz C, Murphy ME, Murray JT, Murthy A, Mysorekar IU, Nabi IR, Nabissi M, Nader GA, Nagahara Y, Nagai Y, Nagata K, Nagelkerke A, Nagy P, Naidu SR, Nair S, Nakano H, Nakatogawa H, Nanjundan M, Napolitano G, Naqvi NI, Nardacci R, Narendra DP, Narita M, Nascimbeni AC, Natarajan R, Navegantes LC, Nawrocki ST, Nazarko TY, Nazarko VY, Neill T, Neri LM, Netea MG, Netea-Maier RT, Neves BM, Ney PA, Nezis IP, Nguyen HTT, Nguyen HP, Nicot A-S, Nilsen H, Nilsson P, Nishimura M, Nishino I, Niso-Santano M, Niu H, Nixon RA, Njar VCO, Noda T, Noegel AA, Nolte EM, Norberg E, Norga KK, Noureini SK, Notomi S, Notterpek L, Nowikovsky K, Nukina N, Nürnberger T, O'Donnell VB, O'Donovan T, O'Dwyer PJ, Oehme I, Oeste CL, Ogawa M, Ogretmen B, Ogura Y, Oh YJ, Ohmuraya M, Ohshima T, Ojha R, Okamoto K, Okazaki T, Oliver FJ, Ollinger K, Olsson S, Orban DP, Ordonez P, Orhon I, Orosz L, O'Rourke EJ, Orozco H, Ortega AL, Ortona E, Osellame LD, Oshima J, Oshima S, Osiewacz HD, Otomo T, Otsu K, Ou J-hsiungJ, Outeiro TF, Ouyang D-yun, Ouyang H, Overholtzer M, Ozbun MA, Ozdinler PH, Ozpolat B, Pacelli C, Paganetti P, Page G, Pages G, Pagnini U, Pajak B, Pak SC, Pakos-Zebrucka K, Pakpour N, Palková Z, Palladino F, Pallauf K, Pallet N, Palmieri M, Paludan SR, Palumbo C, Palumbo S, Pampliega O, Pan H, Pan W, Panaretakis T, Pandey A, Pantazopoulou A, Papackova Z, Papademetrio DL, Papassideri I, Papini A, Parajuli N, Pardo J, Parekh VV, Parenti G, Park J-I, Park J, Park OK, Parker R, Parlato R, Parys JB, Parzych KR, Pasquet J-M, Pasquier B, Pasumarthi KBS, Patschan D, Patterson C, Pattingre S, Pattison S, Pause A, Pavenstädt H, Pavone F, Pedrozo Z, Peña FJ, Peñalva MA, Pende M, Peng J, Penna F, Penninger JM, Pensalfini A, Pepe S, Pereira GJS, Pereira PC, Pérez-de la Cruz V, Pérez-Pérez ME, Pérez-Rodríguez D, Pérez-Sala D, Perier C, Perl A, Perlmutter DH, Perrotta I, Pervaiz S, Pesonen M, Pessin JE, Peters GJ, Petersen M, Petrache I, Petrof BJ, Petrovski G, Phang JM, Piacentini M, Pierdominici M, Pierre P, Pierrefite-Carle V, Pietrocola F, Pimentel-Muiños FX, Pinar M, Pineda B, Pinkas-Kramarski R, Pinti M, Pinton P, Piperdi B, Piret JM, Platanias LC, Platta HW, Plowey ED, Pöggeler S, Poirot M, Polčic P, Poletti A, Poon AH, Popelka H, Popova B, Poprawa I, Poulose SM, Poulton J, Powers SK, Powers T, Pozuelo-Rubio M, Prak K, Prange R, Prescott M, Priault M, Prince S, Proia RL, Proikas-Cezanne T, Prokisch H, Promponas VJ, Przyklenk K, Puertollano R, Pugazhenthi S, Puglielli L, Pujol A, Puyal J, Pyeon D, Qi X, Qian W-bin, Qin Z-H, Qiu Y, Qu Z, Quadrilatero J, Quinn F, Raben N, Rabinowich H, Radogna F, Ragusa MJ, Rahmani M, Raina K, Ramanadham S, Ramesh R, Rami A, Randall-Demllo S, Randow F, Rao H, Rao VA, Rasmussen BB, Rasse TM, Ratovitski EA, Rautou P-E, Ray SK, Razani B, Reed BH, Reggiori F, Rehm M, Reichert AS, Rein T, Reiner DJ, Reits E, Ren J, Ren X, Renna M, Reusch JEB, Revuelta JL, Reyes L, Rezaie AR, Richards RI, Richardson DR, Richetta C, Riehle MA, Rihn BH, Rikihisa Y, Riley BE, Rimbach G, Rippo MR, Ritis K, Rizzi F, Rizzo E, Roach PJ, Robbins J, Roberge M, Roca G, Roccheri MC, Rocha S, Rodrigues CMP, Rodríguez CI, de Cordoba SR, Rodriguez-Muela N, Roelofs J, Rogov VV, Rohn TT, Rohrer B, Romanelli D, Romani L, Romano PS, Roncero MIG, Rosa JL, Rosello A, Rosen KV, Rosenstiel P, Rost-Roszkowska M, Roth KA, Roué G, Rouis M, Rouschop KM, Ruan DT, Ruano D, Rubinsztein DC, Rucker EB, Rudich A, Rudolf E, Rudolf R, Ruegg MA, Ruiz-Roldan C, Ruparelia AA, Rusmini P, Russ DW, Russo GL, Russo G, Russo R, Rusten TE, Ryabovol V, Ryan KM, Ryter SW, Sabatini DM, Sacher M, Sachse C, Sack MN, Sadoshima J, Saftig P, Sagi-Eisenberg R, Sahni S, Saikumar P, Saito T, Saitoh T, Sakakura K, Sakoh-Nakatogawa M, Sakuraba Y, Salazar-Roa M, Salomoni P, Saluja AK, Salvaterra PM, Salvioli R, Samali A, Sanchez AMJ, Sánchez-Alcázar JA, Sanchez-Prieto R, Sandri M, Sanjuan MA, Santaguida S, Santambrogio L, Santoni G, dos Santos CN, Saran S, Sardiello M, Sargent G, Sarkar P, Sarkar S, Sarrias MR, Sarwal MM, Sasakawa C, Sasaki M, Sass M, Sato K, Sato M, Satriano J, Savaraj N, Saveljeva S, Schaefer L, Schaible UE, Scharl M, Schatzl HM, Schekman R, Scheper W, Schiavi A, Schipper HM, Schmeisser H, Schmidt J, Schmitz I, Schneider BE, Schneider EM, Schneider JL, Schon EA, Schönenberger MJ, Schönthal AH, Schorderet DF, Schröder B, Schuck S, Schulze RJ, Schwarten M, Schwarz TL, Sciarretta S, Scotto K, Scovassi AI, Screaton RA, Screen M, Seca H, Sedej S, Segatori L, Segev N, Seglen PO, Seguí-Simarro JM, Segura-Aguilar J, Seki E, Seiliez I, Sell C, Semenkovich CF, Semenza GL, Sen U, Serra AL, Serrano-Puebla A, Sesaki H, Setoguchi T, Settembre C, Shacka JJ, Shajahan-Haq AN, Shapiro IM, Sharma S, She H, Shen C-KJ, Shen C-C, Shen H-M, Shen S, Shen W, Sheng R, Sheng X, Sheng Z-H, Shepherd TG, Shi J, Shi Q, Shi Q, Shi Y, Shibutani S, Shibuya K, Shidoji Y, Shieh J-J, Shih C-M, Shimada Y, Shimizu S, Shin DW, Shinohara ML, Shintani M, Shintani T, Shioi T, Shirabe K, Shiri-Sverdlov R, Shirihai O, Shore GC, Shu C-W, Shukla D, Sibirny AA, Sica V, Sigurdson CJ, Sigurdsson EM, Sijwali PS, Sikorska B, Silveira WA, Silvente-Poirot S, Silverman GA, Simak J, Simmet T, Simon AK, Simon H-U, Simone C, Simons M, Simonsen A, Singh R, Singh SV, Singh SK, Sinha D, Sinha S, Sinicrope FA, Sirko A, Sirohi K, Sishi BJN, Sittler A, Siu PM, Sivridis E, Skwarska A, Slack R, Slaninová I, Slavov N, Smaili SS, Smalley KSM, Smith DR, Soenen SJ, Soleimanpour SA, Solhaug A, Somasundaram K, Son JH, Sonawane A, Song C, Song F, Song HK, Song J-X, Song W, Soo KY, Sood AK, Soong TW, Soontornniyomkij V, Sorice M, Sotgia F, Soto-Pantoja DR, Sotthibundhu A, Sousa MJ, Spaink HP, Span PN, Spang A, Sparks JD, Speck PG, Spector SA, Spies CD, Springer W, Clair DS, Stacchiotti A, Staels B, Stang MT, Starczynowski DT, Starokadomskyy P, Steegborn C, Steele JW, Stefanis L, Steffan J, Stellrecht CM, Stenmark H, Stepkowski TM, Stern ST, Stevens C, Stockwell BR, Stoka V, Storchova Z, Stork B, Stratoulias V, Stravopodis DJ, Strnad P, Strohecker AM, Ström A-L, Stromhaug P, Stulik J, Su Y-X, Su Z, Subauste CS, Subramaniam S, Sue CM, Suh SW, Sui X, Sukseree S, Sulzer D, Sun F-L, Sun J, Sun J, Sun S-Y, Sun Y, Sun Y, Sun Y, Sundaramoorthy V, Sung J, Suzuki H, Suzuki K, Suzuki N, Suzuki T, Suzuki YJ, Swanson MS, Swanton C, Swärd K, Swarup G, Sweeney ST, Sylvester PW, Szatmari Z, Szegezdi E, Szlosarek PW, Taegtmeyer H, Tafani M, Taillebourg E, Tait SWG, Takacs-Vellai K, Takahashi Y, Takáts S, Takemura G, Takigawa N, Talbot NJ, Tamagno E, Tamburini J, Tan C-P, Tan L, Tan ML, Tan M, Tan Y-J, Tanaka K, Tanaka M, Tang D, Tang D, Tang G, Tanida I, Tanji K, Tannous BA, Tapia JA, Tasset-Cuevas I, Tatar M, Tavassoly I, Tavernarakis N, Taylor A, Taylor GS, Taylor GA, Taylor JP, Taylor MJ, Tchetina EV, Tee AR, Teixeira-Clerc F, Telang S, Tencomnao T, Teng B-B, Teng R-J, Terro F, Tettamanti G, Theiss AL, Theron AE, Thomas KJ, Thomé MP, Thomes PG, Thorburn A, Thorner J, Thum T, Thumm M, Thurston TLM, Tian L, Till A, Ting JP-yun, Titorenko VI, Toker L, Toldo S, Tooze SA, Topisirovic I, Torgersen ML, Torosantucci L, Torriglia A, Torrisi MR, Tournier C, Towns R, Trajkovic V, Travassos LH, Triola G, Tripathi DN, Trisciuoglio D, Troncoso R, Trougakos IP, Truttmann AC, Tsai K-J, Tschan MP, Tseng Y-H, Tsukuba T, Tsung A, Tsvetkov AS, Tu S, Tuan H-Y, Tucci M, Tumbarello DA, Turk B, Turk V, Turner RFB, Tveita AA, Tyagi SC, Ubukata M, Uchiyama Y, Udelnow A, Ueno T, Umekawa M, Umemiya-Shirafuji R, Underwood BR, Ungermann C, Ureshino RP, Ushioda R, Uversky VN, Uzcátegui NL, Vaccari T, Vaccaro MI, Váchová L, Vakifahmetoglu-Norberg H, Valdor R, Valente EM, Vallette F, Valverde AM, Van den Berghe G, Van Den Bosch L, van den Brink GR, van der Goot FG, van der Klei IJ, van der Laan LJW, van Doorn WG, van Egmond M, van Golen KL, Van Kaer L, van Lookeren Campagne M, Vandenabeele P, Vandenberghe W, Vanhorebeek I, Varela-Nieto I, Vasconcelos MH, Vasko R, Vavvas DG, Vega-Naredo I, Velasco G, Velentzas AD, Velentzas PD, Vellai T, Vellenga E, Vendelbo MH, Venkatachalam K, Ventura N, Ventura S, Veras PST, Verdier M, Vertessy BG, Viale A, Vidal M, Vieira HLA, Vierstra RD, Vigneswaran N, Vij N, Vila M, Villar M, Villar VH, Villarroya J, Vindis C, Viola G, Viscomi MT, Vitale G, Vogl DT, Voitsekhovskaja OV, von Haefen C, von Schwarzenberg K, Voth DE, Vouret-Craviari V, Vuori K, Vyas JM, Waeber C, Walker CL, Walker MJ, Walter J, Wan L, Wan X, Wang B, Wang C, Wang C-Y, Wang C, Wang C, Wang C, Wang D, Wang F, Wang F, Wang G, Wang H-jie, Wang H, Wang H-G, Wang H, Wang H-D, Wang J, Wang J, Wang M, Wang M-Q, Wang P-Y, Wang P, Wang RC, Wang S, Wang T-F, Wang X, Wang X-jia, Wang X-W, Wang X, Wang X, Wang Y, Wang Y, Wang Y, Wang Y-J, Wang Y, Wang Y, Wang YT, Wang Y, Wang Z-N, Wappner P, Ward C, Ward DM, Warnes G, Watada H, Watanabe Y, Watase K, Weaver TE, Weekes CD, Wei J, Weide T, Weihl CC, Weindl G, Weis SN, Wen L, Wen X, Wen Y, Westermann B, Weyand CM, White AR, White E, Whitton JL, Whitworth AJ, Wiels J, Wild F, Wildenberg ME, Wileman T, Wilkinson DS, Wilkinson S, Willbold D, Williams C, Williams K, Williamson PR, Winklhofer KF, Witkin SS, Wohlgemuth SE, Wollert T, Wolvetang EJ, Wong E, Wong GW, Wong RW, Wong VKW, Woodcock EA, Wright KL, Wu C, Wu D, Wu GS, Wu J, Wu J, Wu M, Wu M, Wu S, Wu WKK, Wu Y, Wu Z, Xavier CPR, Xavier RJ, Xia G-X, Xia T, Xia W, Xia Y, Xiao H, Xiao J, Xiao S, Xiao W, Xie C-M, Xie Z, Xie Z, Xilouri M, Xiong Y, Xu C, Xu C, Xu F, Xu H, Xu H, Xu J, Xu J, Xu J, Xu L, Xu X, Xu Y, Xu Y, Xu Z-X, Xu Z, Xue Y, Yamada T, Yamamoto A, Yamanaka K, Yamashina S, Yamashiro S, Yan B, Yan B, Yan X, Yan Z, Yanagi Y, Yang D-S, Yang J-M, Yang L, Yang M, Yang P-M, Yang P, Yang Q, Yang W, Yang WY, Yang X, Yang Y, Yang Y, Yang Z, Yang Z, Yao M-C, Yao PJ, Yao X, Yao Z, Yao Z, Yasui LS, Ye M, Yedvobnick B, Yeganeh B, Yeh ES, Yeyati PL, Yi F, Yi L, Yin X-M, Yip CK, Yoo Y-M, Yoo YH, Yoon S-Y, Yoshida K-I, Yoshimori T, Young KH, Yu H, Yu JJ, Yu J-T, Yu J, Yu L, Yu WH, Yu X-F, Yu Z, Yuan J, Yuan Z-M, Yue BYJT, Yue J, Yue Z, Zacks DN, Zacksenhaus E, Zaffaroni N, Zaglia T, Zakeri Z, Zecchini V, Zeng J, Zeng M, Zeng Q, Zervos AS, Zhang DD, Zhang F, Zhang G, Zhang G-C, Zhang H, Zhang H, Zhang H, Zhang H, Zhang J, Zhang J, Zhang J, Zhang J, Zhang J-pu, Zhang L, Zhang L, Zhang L, Zhang L, Zhang M-Y, Zhang X, Zhang XD, Zhang Y, Zhang Y, Zhang Y, Zhang Y, Zhang Y, Zhao M, Zhao W-L, Zhao X, Zhao YG, Zhao Y, Zhao Y, Zhao Y-xia, Zhao Z, Zhao ZJ, Zheng D, Zheng X-L, Zheng X, Zhivotovsky B, Zhong Q, Zhou G-Z, Zhou G, Zhou H, Zhou S-F, Zhou X-jie, Zhu H, Zhu H, Zhu W-G, Zhu W, Zhu X-F, Zhu Y, Zhuang S-M, Zhuang X, Ziparo E, Zois CE, Zoladek T, Zong W-X, Zorzano A, Zughaier SM. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition) Autophagy. 2016;12:1–222. doi: 10.1080/15548627.2015.1100356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Knott SRV, Maceli A, Erard N, Chang K, Marran K, Zhou X, Gordon A, Demerdash OE, Wagenblast E, Kim S, Fellmann C, Hannon GJ. A computational algorithm to predict shRNA potency. Molecular Cell. 2014;56:796–807. doi: 10.1016/j.molcel.2014.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Krogh A, Larsson B, von Heijne G, Sonnhammer EL. Predicting transmembrane protein topology with a hidden markov model: application to complete genomes. Journal of Molecular Biology. 2001;305:567–580. doi: 10.1006/jmbi.2000.4315. [DOI] [PubMed] [Google Scholar]
  35. Lee KK, Haraguchi T, Lee RS, Koujin T, Hiraoka Y, Wilson KL. Distinct functional domains in Emerin Bind Lamin A and DNA-bridging protein BAF. Journal of Cell Science. 2001;114:4567–4573. doi: 10.1242/jcs.114.24.4567. [DOI] [PubMed] [Google Scholar]
  36. Leto DE, Morgens DW, Zhang L, Walczak CP, Elias JE, Bassik MC, Kopito RR. Genome-wide CRISPR analysis identifies Substrate-Specific conjugation modules in ER-Associated degradation. Molecular Cell. 2019;73:377–389. doi: 10.1016/j.molcel.2018.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Ong SE, Mann M. A practical recipe for stable isotope labeling by amino acids in cell culture (SILAC) Nature Protocols. 2006;1:2650–2660. doi: 10.1038/nprot.2006.427. [DOI] [PubMed] [Google Scholar]
  38. Park SK, Aslanian A, McClatchy DB, Han X, Shah H, Singh M, Rauniyar N, Moresco JJ, Pinto AF, Diedrich JK, Delahunty C, Yates JR. Census 2: isobaric labeling data analysis. Bioinformatics. 2014;30:2208–2209. doi: 10.1093/bioinformatics/btu151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Pryszcz LP, Huerta-Cepas J, Gabaldón T. MetaPhOrs: orthology and paralogy predictions from multiple phylogenetic evidence using a consistency-based confidence score. Nucleic Acids Research. 2011;39:e32. doi: 10.1093/nar/gkq953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Rolls MM, Stein PA, Taylor SS, Ha E, McKeon F, Rapoport TA. A visual screen of a Gfp-Fusion library identifies a new type of nuclear envelope membrane protein. The Journal of Cell Biology. 1999;146:29–44. doi: 10.1083/jcb.146.1.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Ruggiano A, Foresti O, Carvalho P. Quality control: er-associated degradation: protein quality control and beyond. The Journal of Cell Biology. 2014;204:869–879. doi: 10.1083/jcb.201312042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Saftig P, Klumperman J. Lysosome biogenesis and lysosomal membrane proteins: trafficking meets function. Nature Reviews Molecular Cell Biology. 2009;10:623–635. doi: 10.1038/nrm2745. [DOI] [PubMed] [Google Scholar]
  43. Satpute-Krishnan P, Ajinkya M, Bhat S, Itakura E, Hegde RS, Lippincott-Schwartz J. ER stress-induced clearance of misfolded GPI-anchored proteins via the secretory pathway. Cell. 2014;158:522–533. doi: 10.1016/j.cell.2014.06.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Schirmer EC, Florens L, Guan T, Yates JR, Gerace L. Nuclear membrane proteins with potential disease links found by subtractive proteomics. Science. 2003;301:1380–1382. doi: 10.1126/science.1088176. [DOI] [PubMed] [Google Scholar]
  45. Schreiber KH, Kennedy BK. When lamins go bad: nuclear structure and disease. Cell. 2013;152:1365–1375. doi: 10.1016/j.cell.2013.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Shimi T, Koujin T, Segura-Totten M, Wilson KL, Haraguchi T, Hiraoka Y. Dynamic interaction between BAF and emerin revealed by FRAP, FLIP, and FRET analyses in living HeLa cells. Journal of Structural Biology. 2004;147:31–41. doi: 10.1016/j.jsb.2003.11.013. [DOI] [PubMed] [Google Scholar]
  47. Smith MD, Harley ME, Kemp AJ, Wills J, Lee M, Arends M, von Kriegsheim A, Behrends C, Wilkinson S. CCPG1 is a Non-canonical autophagy cargo receptor essential for ER-Phagy and pancreatic ER proteostasis. Developmental Cell. 2018;44:217–232. doi: 10.1016/j.devcel.2017.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Tabb DL, McDonald WH, Yates JR. DTASelect and contrast: tools for assembling and comparing protein identifications from shotgun proteomics. Journal of Proteome Research. 2002;1:21–26. doi: 10.1021/pr015504q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Toyama BH, Savas JN, Park SK, Harris MS, Ingolia NT, Yates JR, Hetzer MW. Identification of long-lived proteins reveals exceptional stability of essential cellular structures. Cell. 2013;154:971–982. doi: 10.1016/j.cell.2013.07.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Toyama BH, Arrojo E Drigo R, Lev-Ram V, Ramachandra R, Deerinck TJ, Lechene C, Ellisman MH, Hetzer MW. Visualization of long-lived proteins reveals age mosaicism within nuclei of postmitotic cells. The Journal of Cell Biology. 2019;218:433–444. doi: 10.1083/jcb.201809123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Tsai P-L, Zhao C, Turner E, Schlieker C. The Lamin B receptor is essential for cholesterol synthesis and perturbed by disease-causing mutations. eLife. 2016;5:4181. doi: 10.7554/eLife.16011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjostedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA-K, Odeberg J, Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist P-H, Berling H, Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, Zwahlen M, von Heijne G, Nielsen J, Ponten F. Tissue-based map of the human proteome. Science. 2015;347:1260419. doi: 10.1126/science.1260419. [DOI] [PubMed] [Google Scholar]
  53. Ungricht R, Klann M, Horvath P, Kutay U. Diffusion and retention are major determinants of protein targeting to the inner nuclear membrane. The Journal of Cell Biology. 2015;209:687–704. doi: 10.1083/jcb.201409127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Vaughan A, Alvarez-Reyes M, Bridger JM, Broers JL, Ramaekers FC, Wehnert M, Morris GE, Whitfield WGF, Hutchison CJ. Both emerin and lamin C depend on lamin A for localization at the nuclear envelope. Journal of Cell Science. 2001;114:2577–2590. doi: 10.1242/jcs.114.14.2577. [DOI] [PubMed] [Google Scholar]
  55. Verzijlbergen KF, Menendez-Benito V, van Welsem T, van Deventer SJ, Lindstrom DL, Ovaa H, Neefjes J, Gottschling DE, van Leeuwen F. Recombination-induced tag exchange to track old and new proteins. PNAS. 2010;107:64–68. doi: 10.1073/pnas.0911164107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Wang Q, Li L, Ye Y. Inhibition of p97-dependent protein degradation by eeyarestatin I. Journal of Biological Chemistry. 2008;283:7445–7454. doi: 10.1074/jbc.M708347200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Xu T, Park SK, Venable JD, Wohlschlegel JA, Diedrich JK, Cociorva D, Lu B, Liao L, Hewel J, Han X, Wong CCL, Fonslow B, Delahunty C, Gao Y, Shah H, Yates JR. ProLuCID: an improved SEQUEST-like algorithm with enhanced sensitivity and specificity. Journal of Proteomics. 2015;129:16–24. doi: 10.1016/j.jprot.2015.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Zattas D, Berk JM, Kreft SG, Hochstrasser M. A conserved C-terminal element in the yeast Doa10 and human MARCH6 ubiquitin ligases required for selective substrate degradation. Journal of Biological Chemistry. 2016;291:12105–12118. doi: 10.1074/jbc.M116.726877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Zavodszky E, Hegde RS. Misfolded GPI-anchored proteins are escorted through the secretory pathway by ER-derived factors. eLife. 2019;8:32. doi: 10.7554/eLife.46740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Zhang K, Sha J, Harter ML. Activation of Cdc6 by MyoD is associated with the expansion of quiescent myogenic satellite cells. The Journal of Cell Biology. 2010;188:39–48. doi: 10.1083/jcb.200904144. [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision letter

Editor: Elizabeth A Miller1
Reviewed by: Elizabeth A Miller2, Maurizio Molinari3

In the interests of transparency, eLife includes the editorial decision letter and accompanying author responses. A lightly edited version of the letter sent to the authors after peer review is shown, indicating the most substantive concerns; minor comments are not usually included.

Thank you for submitting your article "Selective clearance of the inner nuclear membrane protein emerin by vesicular transport during ER stress" for consideration by eLife. Your article has been reviewed by three peer reviewers, including Elizabeth A Miller as the Reviewing Editor and Reviewer #1, and the evaluation has been overseen by David Ron as the Senior Editor. The following individuals involved in review of your submission have agreed to reveal their identity: Maurizio Molinari (Reviewer #2).

The reviewers have discussed the reviews with one another, and the Reviewing Editor has drafted this decision to help you prepare a revised submission.

This manuscript from the Hetzer lab uses SILAC pulse-chase mass spectrometry to measure the lifetime of nuclear envelope proteins, finding a range of turnover rates. For a short-lived protein, the authors go on to characterize the potential degradation pathways, finding no clear ubiquitination machinery that can be assigned to proteasome-mediated degradation, suggesting redundancy in this pathway. The authors then go on to characterize a second mode of degradation, mediated by vesicle trafficking machineries, and triggered by ER stress. This has hallmarks of the RESET pathway described by others and is exciting in presenting a new substrate for this pathway. The conclusions are largely supported by the data, and the description of a new RESET client is an exciting advance. It would have been nice to also define the proteasome-mediated pathway, but clearly this will require more dissection to overcome problems with redundancy.

The primary shared concern is over the use of chemical compounds (which have potential pleiotropic effects) over long time periods, with additional concern over the potential for gene expression effects to explain observed changes (e.g. Figure 3E). To this end, pulse-chase experiments are preferable for being quantitative and taking into account gene expression effects. Coupled with the glycosylation site addition (as suggested by reviewer 3), this should give a more detailed view of the fate of EMD under ER stress. We do not suggest that all experiments need to be repeated using this type of analysis, but a representative set of experiments covering the key conditions is required to strengthen the authors' model.

Reviewers also propose to more directly visualize the fate of EMD using the tag switching method to more acutely observe the transition of the "old" protein from the INM, to the ER, Golgi and then to the lysosome upon different treatments. A time course of imaging and co-localization would strengthen this argument. Indeed, some co-localization experiments are required even for the steady state observations, most notably for a lysosomal marker in the Baf A1 experiment. Similarly, a control IF experiment of unstressed cells treated with Baf A1 is required to demonstrate the requirement for ER stress in this remobilization.

Reviewer #2:

In this paper, the authors examine the intracellular fate of ectopically expressed emerin (EMD) and of a disease-causing variant thereof. EMD is used as model to investigate proteasomal and lysosomal pathways that regulate turnover of inner nuclear membrane (INM) proteins. They report that both wild type and mutant EMD are ERAD substrates. During acute ER stress, both wild type and mutant EMD are exported from the INM and the ER, via the Golgi and the plasma membrane, to the lysosomes for clearance.

There are few major issues to consider:

1) The authors monitor variations in intracellular level and localization of ectopically expressed EMD proteins in response to cell exposure to various compounds that inhibit ERAD, jeopardize ER to Golgi transport, induce ER stress, block protein synthesis. Note that all these compounds have pleiotropic effects and are to some extent toxic to cells at the concentrations and times (up to 24 hours) used in the experiments. Nevertheless, the authors solely ascribe these drug-induced variations in EMD's intracellular level and localization, to changes in EMD's turnover.

2) At this stage, it cannot be excluded that the various drugs (Figure 2E-G, Figure 3B-D, Figure 4…), or the induction of gene silencing (Figure 3E) modify the expression (rather than the clearance) of the EMDs. For example, it has been reported that MG132 enhances CMV promoter-regulated expression of ectopic genes (and EMDs gene expression is placed here under control of a CMV promoter). I suggest using Bortezomib, a more specific proteasome inhibitor. Also, in Figure 3E I notice that high levels of GFP are expressed only in cells were gene silencing has been activated. GFP expression could reduce expression of the second transgene (EMD), thereby offering an alternative explanation to the one proposed by the authors for the reduction of the EMD level upon E2 ligases knockdown.

All in all, the authors should systematically check synthesis of EMDs in their experiments and how EMDs synthesis changes under the experimental set-up. Moreover, they should measure EMDs stability directly, via quantitative methods such as pulse-chase analyses.

3) A major point of the paper (and the most interesting one) is that changes in cellular (ER) homeostasis trigger lysosomal clearance of proteins from the INM. I am not sure that this is (convincingly enough) supported by the results shown here. To demonstrate that EMD is cleared from the INM, the authors should monitor (by exploiting the epitope-exchange technology) the fate of "old" EMD and show that it re-localizes from the nuclear membrane, to the ER, Golgi and then to the lysosomes (i.e., old EMD should accumulate in the endo-lysosomes (=LAMP1-positive organelle) during ER stress, in the presence of Baf A1). Since EMDs are retained in the INM by association with Lamin A, is EMD:Lamin A complex regulated by ER stress?

4) I miss some experiment with endogenous proteins (e.g., EMD, Sun2, Lamin A). Is their turnover affected by ER stress? Are they delivered to endolysosomes upon ER stress induction?

Reviewer #3:

Buchwalter et al. investigate protein turnover in a mammalian cell system with a focus on proteins of the nuclear envelope and inner nuclear membrane (INM), an area of significant contemporary interest. In brief, the novelty of the manuscript lies in: (i) in the determination of half-lives of INM proteins in a tissue culture model of resting cells, hence minimizing the contribution of "canonical" ERAD via mixing of ER and INM through open mitosis; (ii) the application of RITE analysis in this context to also monitor protein localization, not only half-lives; (iii) chiefly the proposal of a novel route for degradation via trafficking through the Golgi-PM-lysosomal route and a definition a novel role for the LEM domain in this context.

In the opinion of this reviewer, the manuscript should be of considerable interest for the broad readership of eLife. While the identification of the responsible E3 ligase(s) should not be a key requirement for publication, a concern is that many of the key experiments rely solely on pharmacological inhibitors that often have pleiotropic/toxic effects, especially when used in combination. Some relatively straightforward experiments are suggested below that could help to strengthen the authors' proposal of a novel degradation route.

1) It is suggested to append a N-Glycosylation sequence (Asn-X-Ser/Thr) to the C terminus of emerin, a readout that is commonly used by laboratories studying tail-anchored protein biogenesis. This readout would be extremely useful for several reasons:

(i) inserted and preinserted "immature" variants and their degradative fate can be distinguished with ease on immunoblots;

(ii) trafficking from the INM or ER to and through the Golgi can be monitored with ease by monitoring the acquisition of Endo H resistance (vs. PNGase sensitivity), and;

(iii) most importantly, a glycosylation-competent variant is useful to reinforce the interpretation of trafficking to Golgi/lysosome: this EMD variant should accumulate as Endo H-resistant species upon lysosomal deacidification. Moreover, this observation would help to rule out ER-Phagy. In the opinion of this reviewer, it would not be necessary to repeat each and every experiment with this construct, but its application to a few key experiments would considerably strengthen the authors' proposal of a novel degradation route.

2) Can the authors rule out that only a selective sub-pool of "new" EMD variants enter the lysosomal pathway while another "old" subset is locally degraded? Perhaps the authors could consider demonstrating (via RITE) that "old", INM-resident EMD-variants are subject to lysosomal degradation? Alternatively, all formal possibilities could be stated/deconvoluted more clearly in the Discussion section.

eLife. 2019 Oct 10;8:e49796. doi: 10.7554/eLife.49796.031

Author response


This manuscript from the Hetzer lab uses SILAC pulse-chase mass spectrometry to measure the lifetime of nuclear envelope proteins, finding a range of turnover rates. For a short-lived protein, the authors go on to characterize the potential degradation pathways, finding no clear ubiquitination machinery that can be assigned to proteasome-mediated degradation, suggesting redundancy in this pathway. The authors then go on to characterize a second mode of degradation, mediated by vesicle trafficking machineries, and triggered by ER stress. This has hallmarks of the RESET pathway described by others and is exciting in presenting a new substrate for this pathway. The conclusions are largely supported by the data, and the description of a new RESET client is an exciting advance. It would have been nice to also define the proteasome-mediated pathway, but clearly this will require more dissection to overcome problems with redundancy.

We would like to thank the reviewers for their constructive comments. We have extensively reorganized the manuscript and included new data in response to the reviewers’ critiques. For clarity, we have reorganized the manuscript to focus mostly on the more rapidly degraded EMDΔ95-99 mutant. This makes it possible for us to show more data and controls for this mutant, while we show key comparisons with wild type EMD and the EMDΔLEM mutant. We feel that this more streamlined organization of the manuscript and the new data included with this revision strengthen the manuscript considerably.

The primary shared concern is over the use of chemical compounds (which have potential pleiotropic effects) over long time periods, with additional concern over the potential for gene expression effects to explain observed changes (e.g. Figure 3E).

We now include shorter timepoints of drug treatments (2-8 hours in many cases) which exhibit effects consistent with our observations at longer timepoints.

The reviewers expressed concern that over the time period of RNAi induction, expression of the free GFP encoded by the inducible miR-E RNAi vector might interfere with the synthesis of EMDΔ95-99-GFP. We have included supplemental data (Figure 3—figure supplement 2) showing that two methods of knockdown, siRNA transfection and miR-E RNAi induction, exhibit consistent effects. Neither siRNA transfection (Figure 3—figure supplement 2A-B) nor doxycycline-inducible miRNA expression (Figure 3C) stabilize EMDΔ95-99-GFP. In the siRNA transfection experiments, we observed some decrease of EMDΔ95-99-GFP levels in cells transfected with a scramble RNAi control, but no difference between this condition and specific knockdown conditions.

In the miR-E induction experiments, a GFP marker is co-expressed when the miRNA is induced. In some miR-E knockdown conditions (UBE2G1 or UBE2G2 miR-E, Figure 3C; MARCH6, Figure 3—figure supplement 2D), we observe loss of EMDΔ95-99-GFP. This could indicate that the protein is degraded by another pathway, or alternatively that protein synthesis is suppressed because of promoter competition between the EMDΔ95-99-GFP and the free GFP. We think the latter interpretation is unlikely because an additional knockdown (UBE2J1, Figure 3—figure supplement 2) also had no effect on emerin-GFP levels, even though free GFP is also expressed in this condition. These siRNA and miR-E experiments are overall unified in their outcome: knockdown of individual ERAD-implicated E2 or E3 ubiquitin ligases does not cause emerin-GFP to accumulate within cells.

We agree that it is likely that multiple E2 and/or E3 ligases are redundant in ERAD-mediated degradation of EMD. However, these results were an initial hint that an alternative pathway for EMD degradation exists. We acknowledge this possible redundancy in subsection “Proteasome-dependent and proteasome-independent modes of emerin clearance”:

We depleted MARCH6, Rnf26, and CGRRF1 with siRNA, but observed no effect on EMDΔ95-99 protein levels, suggesting that these ligases do not catalyze EMD turnover, or alternatively that multiple E3 ligases are redundant in this process.”

To this end, pulse-chase experiments are preferable for being quantitative and taking into account gene expression effects. Coupled with the glycosylation site addition (as suggested by reviewer 3), this should give a more detailed view of the fate of EMD under ER stress. We do not suggest that all experiments need to be repeated using this type of analysis, but a representative set of experiments covering the key conditions is required to strengthen the authors' model.

We now include RITE time courses for both wild type EMD and EMDΔ95-99, which show that EMDΔ95-99 disappears more rapidly than wild type EMD, but that both variants are stabilized by proteasome inhibition at the NE (Figure 2E-H).

We also include a cycloheximide time-course showing the relative stabilities of wild type EMD-GFP, EMDΔ95-99-GFP (Figure 3—figure supplement 1C-D), and EMDΔLEM-GFP (Figure 8—figure supplement 1). Importantly, deletion of the LEM domain makes EMD more stable, consistent with our model that the LEM domain mediates targeting of emerin variants to the lysosome.

We now also include a cycloheximide + thapsigargin time-course of EMDΔ95-99-GFP which indicates that under ER stress, EMDΔ95-99-GFP disappears more quickly and is detectable in a higher molecular weight modified form (Figure 3D).

Reviewers also propose to more directly visualize the fate of EMD using the tag switching method to more acutely observe the transition of the "old" protein from the INM, to the ER, Golgi and then to the lysosome upon different treatments.

Unfortunately, the RITE tag switching method lacks the temporal control needed to dissect induced turnover of “old” protein in response to ER stress on timescales of minutes to hours. This is because the tag switching event involves first editing of the plasmid by Cre recombinase within cells, then production of new RNA and finally protein. In the meantime, any remaining RNA that encodes the “old” protein will remain in the cell until it degrades. These factors introduce an inevitable lag time in turnover and make it difficult to pinpoint a clearly defined “old” population of protein for tracking on short timescales.

Using stable cell lines that express GFP-tagged EMD variants at roughly endogenous levels, we observe that NE-localized EMD rapidly decreases, while the protein enriches in the Golgi (Figure 4), is detectable at the plasma membrane (Figure 6) and is found in the lysosome (Figure 5) over time. We infer from this that a significant proportion of EMD leaves the INM and moves through the secretory pathway upon ER stress, but we cannot rule out at this point that some of the “old” protein, or maturely folded protein, is degraded in situ at the INM.

A time course of imaging and co-localization would strengthen this argument. Indeed, some co-localization experiments are required even for the steady state observations, most notably for a lysosomal marker in the Baf A1 experiment.

Figure 4 shows time- and stress-dependent colocalization of EMDΔ95-99-GFP with the Golgi resident protein giantin. We now include costaining with the lysosomal protein LAMP1, which clearly shows that emerin accumulates in lysosomes when lysosome acidification is blocked by bafilomycin A1 treatment (Figure 5D-F). LAMP1, which marks the limiting membrane of the lysosome, can clearly be seen encircling the EMDΔ95-99-GFP signal within the interior of the lysosome.

Similarly, a control IF experiment of unstressed cells treated with Baf A1 is required to demonstrate the requirement for ER stress in this remobilization.

We now include this data in Figure 5E; EMDΔ95-99-GFP does not enrich in LAMP1-marked lysosomes without ER stress induction.

Reviewer #2

[…]

2) At this stage, it cannot be excluded that the various drugs (Figure 2E-G, Figure 3B-D, Figure 4…), or the induction of gene silencing (Figure 3E) modify the expression (rather than the clearance) of the EMDs. For example, it has been reported that MG132 enhances CMV promoter-regulated expression of ectopic genes (and EMDs gene expression is placed here under control of a CMV promoter). I suggest using Bortezomib, a more specific proteasome inhibitor. Also, in Figure 3E I notice that high levels of GFP are expressed only in cells were gene silencing has been activated. GFP expression could reduce expression of the second transgene (EMD), thereby offering an alternative explanation to the one proposed by the authors for the reduction of the EMD level upon E2 ligases knockdown.

All in all, the authors should systematically check synthesis of EMDs in their experiments and how EMDs synthesis changes under the experimental set-up. Moreover, they should measure EMDs stability directly, via quantitative methods such as pulse-chase analyses.

We now include two types of pulse-chase experiments which indicate that EMD is stabilized by proteasome inhibition. In Figure 2, we use the RITE system to directly track the localization and levels of “old” EMD variants in the absence or presence of MG132 and observe that MG132 treatment stabilizes “old” EMD. Secondly, we include representative images of cells stably expressing EMDΔ95-99-GFP and treated with CHX, MG132, or CHX + MG132 (Figure 2—figure supplement 1A). This indicates that MG132 inhibits degradation of mature protein (CHX + MG132 condition). We also observe a widespread increase in EMDΔ95-99-GFP signal throughout cell compartments in the + MG132 alone condition. In this condition, it is possible that MG132 is enhancing expression of EMDΔ95-99-GFP. We think it is more likely that significant amounts of EMDΔ95-99-GFP are cotranslationally degraded, as is known to occur for misfolded tail-anchored proteins (Hessa and Hegde, Nature, 2011). We also observe that MG132 treatment causes the accumulation of higher molecular weight, likely poly-ubiquitinated variants of EMDΔ95-99-GFP (Figure 3C), which we interpret as stalled degradation rather than increased synthesis of the protein.

3) A major point of the paper (and the most interesting one) is that changes in cellular (ER) homeostasis trigger lysosomal clearance of proteins from the INM. I am not sure that this is (convincingly enough) supported by the results shown here. To demonstrate that EMD is cleared from the INM, the authors should monitor (by exploiting the epitope-exchange technology) the fate of "old" EMD and show that it re-localizes from the nuclear membrane, to the ER, Golgi and then to the lysosomes (i.e., old EMD should accumulate in the endo-lysosomes (=LAMP1-positive organelle) during ER stress, in the presence of Baf A1). Since EMDs are retained in the INM by association with Lamin A, is EMD:Lamin A complex regulated by ER stress?

This manuscript shows that a protein that localizes to the INM can also be targeted to the lysosome, which is a surprising and novel finding. We observe that INM-localized EMDΔ95-99-GFP is undetectable at the NE after ~8 hours of ER stress (Figure 3E-F). This clearly indicates that EMDΔ95-99 is quantitatively degraded during ER stress. However, we also see evidence that EMD can be degraded by a proteasome-dependent pathway under some conditions, and we think it is likely that this represents ERAD. We cannot definitively rule out the possibility that some proportion of EMD is degraded by each of these pathways during ER stress. Defining the spectrum of EMD interactions in normal vs. stressed cells will likely help to illuminate this, and future work will address this important question. We do know that disruption of the EMD:lamin A complex is not sufficient to cause EMD to be degraded by the lysosome, as we see in lmna -/- MEFs that EMD-GFP is still stably expressed but loses its affinity for the INM and instead localizes to the peripheral ER (Figure 8—figure supplement 2). Rather, our data suggest that the LEM domain:BAF interface is likely to be the binding interaction that is regulated by ER stress.

4) I miss some experiment with endogenous proteins (e.g., EMD, Sun2, Lamin A). Is their turnover affected by ER stress? Are they delivered to endolysosomes upon ER stress induction?

Our explorations so far suggest that endogenous emerin is not quantitatively degraded during ER stress, although its levels do modestly increase at the NE after brefeldin A treatment. This could be consistent with our LEM domain competition model (see Figure 9 and Discussion); if emerin is expressed more highly than its nucleoplasmic BAF binding partner, more of emerin’s LEM domains may be unbound to BAF and may be able to associate with other factors, including factors that might enable ER export. However, we think it is possible that endogenous wild type or disease-mutant emerin may flux through this pathway at some lower level under certain conditions, and future experiments will explore this possibility.

Reviewer #3

1) It is suggested to append a N-Glycosylation sequence (Asn-X-Ser/Thr) to the C terminus of emerin, a readout that is commonly used by laboratories studying tail-anchored protein biogenesis. This readout would be extremely useful for several reasons:

(i) inserted and preinserted "immature" variants and their degradative fate can be distinguished with ease on immunoblots;

(ii) trafficking from the INM or ER to and through the Golgi can be monitored with ease by monitoring the acquisition of Endo H resistance (vs. PNGase sensitivity);

(iii) most importantly, a glycosylation-competent variant is useful to reinforce the interpretation of trafficking to Golgi/lysosome: this EMD variant should accumulate as Endo H-resistant species upon lysosomal deacidification. Moreover, this observation would help to rule our ER-Phagy. In the opinion of this reviewer, it would not be necessary to repeat each and every experiment with this construct, but its application to a few key experiments would considerably strengthen the authors' proposal of a novel degradation route.

We would like to thank this reviewer for this suggestion, which has provided important supporting data for this revised manuscript. We have generated glycosylation reporter cell lines for EMDΔ95-99-GFP* (Figure 3H, Figure 5G), EMD-WT-GFP* (Figure 7E-F), and EMDΔLEM-GFP* (Figure 8) and performed (i) THG and CHX co-treatments and (ii) THG and Baf A1 cotreatments with each of these lines. We tracked the response of a pool of mature protein during ER stress by co-treating cells with cycloheximide to block new protein synthesis, and thapsigargin to induce ER stress. These experiments clearly indicate that within 2 hours of ER stress induction EMDΔ95-99-GFP* (Figure 3G-H) and EMD-WT-GFP* (Figure 7E) shift from predominantly Endo H-sensitive species to predominantly Endo H-resistant species. This indicates that these proteins progressively leave the ER and enter post-ER compartments.

In a short 4 hour THG + Baf A1 treatment time-course, the Endo H-resistant pool of these proteins also increases over time (EMDΔ95-99-GFP*, Figure 5G; EMD-WT-GFP*, Figure 7F). The conversion from Endo H-sensitive to Endo H-resistant is not as quantitative here, likely because we did not include inhibition of protein synthesis along with these two other treatments. Nevertheless, Endo H-resistant species become more abundant when lysosome acidification is inhibited along with ER stress induction.

Both EMD-WT and EMDΔ95-99 are partially Endo H-resistant under homeostatic conditions. This likely indicates that these proteins are exiting the NE/ER with some frequency and potentially being degraded by the same lysosome-dependent pathway. Importantly, however, EMDΔLEM-GFP* shows a distinct phenotype. EMDΔLEM-GFP* exists in only one modified species that is Endo H-sensitive and does not become Endo H-resistant during ER stress (Figure 8I-J). This outcome indicates, in line with our other data, that the LEM domain is required for EMD variants to exit the NE/ER network and target to lysosomes for degradation.

2) Can the authors rule out that only a selective sub-pool of "new" EMD variants enter the lysosomal pathway while another "old" subset is locally degraded? Perhaps the authors could consider demonstrating (via RITE) that "old", INM-resident EMD-variants are subject to lysosomal degradation? Alternatively, all formal possibilities could be stated/deconvoluted more clearly in the Discussion section.

We infer from our data that a significant proportion of EMD leaves the INM and moves through the secretory pathway upon ER stress, but we cannot rule out at this point that some of the “old” protein, or maturely folded protein, is degraded in situ at the INM. We now address this in the Discussion section:

“While our data indicate that a significant proportion of EMD leaves the NE/ER during ER stress, we cannot rule out the possibility that ERAD-mediated degradation of some proportion of EMD takes place within the NE/ER network in parallel to the lysosome-mediated pathway that we have identified.”

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Data Citations

    1. Buchwalter A, Schulte R, Tsai H, Capitanio J, Hetzer MW. 2019. Data from: Selective clearance of the inner nuclear membrane protein emerin by vesicular transport during ER stress. Dryad Digital Repository. [DOI] [PMC free article] [PubMed]

    Supplementary Materials

    Source data 1. Filtered peptide data for half life calculations.

    Peptide turnover data for all peptides passing quality control filters. See R script and Materials and methods for details.

    elife-49796-data1.xlsx (2.2MB, xlsx)
    DOI: 10.7554/eLife.49796.021
    Source data 2. Filtered protein data for half life calculations.

    Filtered and averaged protein turnover data. See R script and Materials and methods for details.

    elife-49796-data2.xlsx (459.4KB, xlsx)
    DOI: 10.7554/eLife.49796.022
    Supplementary file 1. Results of half life fits passing quality filters.
    elife-49796-supp1.xlsx (131KB, xlsx)
    DOI: 10.7554/eLife.49796.023
    Supplementary file 2. Complete list of half life fits.
    elife-49796-supp2.xlsx (386.6KB, xlsx)
    DOI: 10.7554/eLife.49796.024
    Supplementary file 3. Half lives and protein topology data.

    Selected data related to Figure 1G-H.

    elife-49796-supp3.xlsx (24.7KB, xlsx)
    DOI: 10.7554/eLife.49796.025
    Transparent reporting form
    DOI: 10.7554/eLife.49796.026

    Data Availability Statement

    Raw and analyzed mass spectrometric data and associated scripts and tables have been deposited in Dryad. Analyzed data are also included with the manuscript as supplementary tables.

    The following dataset was generated:

    Buchwalter A, Schulte R, Tsai H, Capitanio J, Hetzer MW. 2019. Data from: Selective clearance of the inner nuclear membrane protein emerin by vesicular transport during ER stress. Dryad Digital Repository.


    Articles from eLife are provided here courtesy of eLife Sciences Publications, Ltd

    RESOURCES