Revisiting the Gregarious Lipid Droplet: Maintaining contacts to regulate energy homeostasis in the cell and beyond-Review

Contact Volume 3: 1–16 Seipin-Mediated Contacts as Gatekeepers ! The Author(s) 2020 Article reuse guidelines: of Lipid Flux at the Endoplasmic sagepub.com/journals-permissions DOI: 10.1177/2515256420945820 Reticulum–Lipid Droplet Nexus journals.sagepub.com/home/ctc

Veijo T. Salo1,2 , Maarit Holtt€ a-Vuori€ 1,2 and Elina Ikonen1,2

Abstract Lipid droplets (LDs) are dynamic cellular hubs of lipid metabolism. While LDs contact a plethora of organelles, they have the most intimate relationship with the (ER). Indeed, LDs are initially assembled at specialized ER subdomains, and recent work has unraveled an increasing array of proteins regulating ER-LD contacts. Among these, seipin, a highly conserved lipodystrophy protein critical for LD growth and adipogenesis, deserves special attention. Here, we review recent insights into the role of seipin in LD biogenesis and as a regulator of ER-LD contacts. These studies have also highlighted the evolving concept of ER and LDs as a functional continuum for lipid partitioning and pinpointed a role for seipin at the ER-LD nexus in controlling lipid flux between these compartments.

Keywords seipin, endoplasmic reticulum–lipid droplet contacts, lipid droplet biogenesis

Lipid droplets (LDs) are intracellular storage organelles mutations in seipin also result in hereditary spastic para- composed of a core of hydrophobic neutral lipids (NLs), plegias (Windpassinger et al., 2004) and a severe form of mainly triglycerides (TAG) and sterol esters, surrounded encephalopathy (Guillen-Navarro et al., 2013). Seipin is by a phospholipid (PL) monolayer (Henne et al., 2018). evolutionary conserved, and perturbation of seipin or its Their main function is to store excess energy in the form homologs results in aberrant LD morphology in numer- of TAGs, but LDs also play instrumental roles in many ous model systems (Szymanski et al., 2007; Fei et al., other cellular processes, including maintaining endoplas- 2008; Boutet et al., 2009; Cai et al., 2015; Salo et al., mic reticulum (ER) homeostasis (Welte, 2015; Olzmann 2016; Wang et al., 2016; Kornke and Maniak, 2017; and Carvalho, 2019). LDs are formed in the ER, and Coradetti et al., 2018; Cao et al., 2019). In accordance they retain a close physical and functional relationship with its role in lipodystrophy syndrome in humans, with it after their formation. The lipid and protein fluxes seipin is essential for adipogenesis in various organisms (Payne et al., 2008; Chen et al., 2009; Victoria et al., between these compartments are controlled at ER-LD 2010; Cui et al., 2011; Tian et al., 2011; Ebihara et al., contacts. Recent studies have begun to shed light on the 2015; Mori et al., 2016). properties of LD forming ER subdomains and ER-LD contacts. In this review, we discuss recent insights into LD biogenesis and ER-LD contacts with a special focus on the lipodystrophy protein seipin. 1Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki Seipin Structure Gives Clues to Its 2Minerva Foundation Institute for Medical Research, Helsinki, Finland Function Received April 7, 2020. Revised June 6, 2020. Accepted June 30, 2020. Seipin in an oligomeric ER transmembrane (TM) pro- Corresponding Author: tein originally identified to be mutated in Berardinelli– Elina Ikonen, Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Seip Congenital Lipodistrophy Type 2 (BSCL2) congen- Finland. ital lipodystrophy (Magre et al., 2001). Distinct Email: [email protected]

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution- NonCommercial 4.0 License (https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en- us/nam/open-access-at-sage). 2 Contact

The LD phenotype in the absence of seipin is consis- Structurally seipin contains two transmembrane tent among examined systems, with a proliferation of domains, an evolutionary conserved ER luminal loop tiny and some supersized LDs in an apparently stochas- and variable N- and C-terminal cytoplasmic regions tic manner (Cartwright and Goodman, 2012; Chen and (Lundin et al., 2006; Yang et al., 2013). The luminal Goodman, 2017). Several hypotheses have been pro- loop and its flanking two transmembrane domains are posed for seipin in relation to LDs: a role in controlling critical for seipin function in LD biogenesis, as they ER PL metabolism, sequestering NLs or regulating ER- alone are sufficient to rescue defective LD formation in LD contact sites. These hypotheses are not mutually seipin-deficient cells (Yang et al., 2013; Wang et al., exclusive, and the structure of seipin is in principle com- 2016). Seipin forms homo-oligomers, each containing patible with any of these. In addition to its role in LD 11 (human) or 12 (Drosophila) seipin monomers (Binns formation, seipin has been linked to cytoskeleton remod- et al., 2010; Sim et al., 2012; Sui et al., 2018; Yan et al., eling, calcium handling, and lipolysis (Chen et al., 2012; 2018). In yeast, seipin forms foci localizing to ER-LD Bi et al., 2014; Yang et al., 2014; Ding et al., 2018; Li contact sites (Szymanski et al., 2007; Fei et al., 2008). In et al., 2019b) with a growing number of identified inter- mammalian cells, endogenously fluorescently tagged action partners (Table 1). seipin appears as homogenous, discrete, and mobile foci in the ER, a subset of which are stably associated

Table 1. Seipin Interacting Proteins.

Proposed function Protein name Protein function Main evidence for interaction of interaction Reference

Ldb16 Regulator of LD Co-ip: endogenous proteins in Forms a complex Cartwright et al. (2015), morphology yeast with seipin to Wang et al. (2014), and regulate LDs Wolinski et al. (2015) Promethin/LDAF1/ Regulator of LD iden- Co-ip: endogenous proteins Forms a complex Castro et al. (2019), Ldo45 Ldo16 tity (yeast) (yeast and human cells), with seipin to Chung et al. (2019), comigration of endogenously regulate LDs Eisenberg-Bord et al. tagged proteins (human cells) (2018), and Teixeira et al. (2018) GPAT3-4/Gat1-2 Catalyzes G3P to LPA Co-ip: endogenous proteins Seipin negatively Pagac et al. (2016) and Sim conversion (yeast), overexpressed pro- regulates GPAT et al. (2020) teins (murine cells) enzyme activity SERCA ER calcium pump, Co-ip: endogenous proteins Seipin positively Bi et al. (2014) þ transports Ca2 (human cells), overexpressed regulates SERCA into ER lumen proteins (Drosophila cells) activity Lipin-1 Catalyzes PA to DAG Co-ip and AFM: overexpressed Seipin recruits lipin Sim et al. (2012) and conversion proteins (murine cells) to the ER Talukder et al. (2015) membrane AGPAT-2 Catalyzes LPA to PA Co-ip and AFM: overexpressed Scaffolding of lipo- Sim et al. (2012) and conversion proteins (mice cells) genic enzymes Talukder et al. (2015) Serine palmitoyl- Sphingolipid synthesis Co-ip and BiFic of overexpressed Seipin negatively Su et al. (2019) transferase protein (yeast) regulates sphin- Complex, FA- golipid elongase Tsc13 production 14-3-3-b A scaffolding protein, Co-ip: overexpressed proteins Cytoskeleton Yang et al. (2014) signal transduction (murine and human cells) remodeling Perilipin-2 LD coat protein, lipol- Co-ip: overexpressed proteins Seipin regulates Mori et al. (2016) ysis regulator (human cells) perilipin-2 locali- zation to LDs Perilipin-1 LD coat protein, lipol- Co-ip and AFM: overexpressed Seipin regulates Jiao et al. (2019) ysis regulator proteins (human cells) perilipin-1 locali- zation to LDs Reep-1 Regulates ER Co-ip: overexpressed proteins None proposed Renvoise et al. (2016) morphology (murine cells)

ER ¼ endoplasmic reticulum; AGPAT ¼ 1-acylglycerol-3-phosphate-O-acyltransferase; GPAT ¼ Glycerol-3-phosphate acyltransferase; SERCA ¼ sarco/ þ endoplasmic reticulum Ca2 -ATPase ; FA ¼ Fatty acid; G3P ¼ Glycerol 3-phosphate; LPA ¼Lysophosphatidic acid; DAG ¼ diacylglycerol; PA ¼ phosphatidic acid; LD ¼ lipid droplet; AFM ¼ Atomic force microscopy. Salo et al. 3 with ER-LD contacts (Salo et al., 2016; Wang et al., and Thiam, 2020), these seipin helices might recognize 2016). Seipin oligomerization is critical for its function, similar packing defects induced by NL lenses in the ER as evidenced by the fact that the lipodystrophy- (Sui et al., 2018). associated A212P-seipin mutant forms smaller The bulk of the seipin luminal region forms a beta- oligomers and fails to rescue the aberrant LD biogenesis sandwich-like fold, structurally similar to many lipid- of seipin-deficient cells (Binns et al., 2010; Sim et al., binding domains, including those of Niemann-Pick C2 2013; Salo et al., 2016). The same holds true for an olig- (NPC2) and PKC (Sui et al., 2018; Yan et al., 2018).This omerization deficient mutant designed based on the suggests that seipin may bind lipids in the ER lumen. In cryo-electron microscopy (EM) structure of seipin vitro work suggests that both purified full-length seipin (Yan et al., 2018). and a truncated variant containing only the putative The structures of the luminal domain of human and lipid binding domain can bind anionic PLs, such as Drosophila seipins have been solved by cryo-EM (Sui phosphatidic acid (PA; Sui et al., 2018). A mutant har- et al., 2018; Yan et al., 2018; Figure 1). They form a boring amino acid substitutions in the hydrophobic ring-like/disk-like structure of circa 15 nm in diameter, cavity of the beta sandwich was unable to complement with 11 (human) or 12 (Drosophila) seipin monomers the seipin knockout (KO) LD phenotype (Sui et al., closely intertwined. Each monomer displays two notable 2018). However, which lipids seipin interacts with in features: several hydrophobic helices (HHs) in very close intact cells remains to be determined. proximity to the bilayer membrane, and two beta sheets that exhibit an overall fold similar to certain lipid bind- Seipin Forms an LD Assembly Complex ing domains (Sui et al., 2018; Yan et al., 2018). The structure of the TM or cytoplasmic regions could not The most extensively studied binding partners of seipin be solved by cryo-EM in these studies, suggesting con- are Ldb16 and Ldo16 and its splicing isoform Ldo 45/ formational structural flexibility therein. promethin (Table 1). In yeast, seipin forms a stable com- The ER luminal HHs of seipin are able to bind to a plex with Ldb16, an ER TM protein with no known monolayer covering NLs in vitro (Sui et al., 2018) and mammalian homologues.Seipin is required for Ldb16 sta- appear to be widely conserved across species (Chapman bilization, and depletion of either Ldb16 or seipin results et al., 2019). When expressed as individual peptides in in indistinguishable LD phenotypes (Wang et al., 2014; cells, the HHs localize to LDs. This localization is Grippa et al., 2015). These LD defects can be rescued by impaired by mutating three hydrophobic residues to heterologous expression of human seipin, strongly sug- aspartic acid (Sui et al., 2018). A mutant seipin with gesting that human seipin functionally corresponds to corresponding amino acid substitutions still rescues the yeast Ldb16-seipin complex. seipin function in LD biogenesis, but further concomi- Yeast ER-LD-localized protein isoforms Ldo16 and tant deletion of the N-terminus of seipin, shown previ- Ldo45 were recently discovered to be interactors of the ously to localize to LDs when expressed alone (Wang seipin complex (Eisenberg-Bord et al., 2018; Teixeira et al., 2016), results in a dysfunctional seipin. Hence, et al., 2018). Overexpression of Ldo45 leads to prolifer- the presence of either LD-targeting luminal HHs or the ation and clustering of LDs that exhibit aberrant protein cytoplasmic N-terminus is required for seipin function. targeting. This phenotype is similar to seipin KO cells, Considering that the LD PL monolayer harbors packing suggesting that Ldo45 and seipin may play antagonistic defects (Bacle et al., 2017; Prevost et al., 2018; Chorlay roles in LD formation and maintenance (Eisenberg-Bord

Top view Side view

Cytosol Membrane- associated helices 160 Å

Transmembrane helices ER lumen Beta-sheets

Figure 1. Seipin Oligomer Structure. The TM regions were modeled into the cryo-EM structure of the luminal domains of human seipin (PDB: 6ds5; Yan et al., 2018) using PyMol software. ER ¼ endoplasmic reticulum. 4 Contact et al., 2018; Teixeira et al., 2018). Ldo16 and Ldo45 were surface tension and PL intrinsic curvature, while the also found to be regulators of a subpopulation of LDs directionality of LD budding to the cytosol may arise that reside in close proximity to the yeast nucleus-vacu- due to ER membrane asymmetry (Ben M’barek et al., ole junctions (NVJs).During nutritional stress, this site 2017; Choudhary et al., 2018; Chorlay et al., 2019; Zoni expands and serves as a site for LD biogenesis under et al., 2020). It therefore seems likely that the LD form- these conditions (Hariri et al., 2018).Indeed, the relative ing ER subdomain harbors a distinct PL milieu, abundance of Ldo16/Ldo45 is regulated by cellular although direct evidence for this is lacking. growth conditions, and Ldo45 is necessary for recruiting LD formation may also be controlled by the overall Pdr16, a lipid transfer protein, to NVJ-associated LDs ER architecture. LD biogenesis appears to occur at (Eisenberg-Bord et al., 2018; Teixeira et al., 2018). peripheral ER tubules in mammalian cells (Kassan Based on remote homology searches, promethin (also et al., 2013; Joshi et al., 2018; Santinho et al., 2020). In known asTMEM159 and recently renamed as Lipid accordance, many ER shaping proteins have been impli- Droplet Assembly Factor 1; LDAF1 ) was proposed as cated in LD formation (Klemm et al., 2013; Falk et al., a homolog of Ldo45 (Eisenberg-Bord et al., 2018). 2014; Papadopoulos et al., 2015). Conversely, proteins Promethin is upregulated during adipogenesis, localizes participating in LD biogenesis such as Rab18 and fat to the ER and LDs, and was found to interact with storage-inducing transmembrane 2 (FIT2) have been seipin (Castro et al., 2019). When expressed heterolo- shown to impact ER morphology (Gerondopoulos gously in yeast, human promethin was able to co- et al., 2014; Hayes et al., 2017; Becuwe et al., 2018). immunoprecipitate the yeast seipin complex, suggesting Changes in ER ultrastructure have been observed also an evolutionary conserved interaction (Castro et al., in seipin-deficient cells (Grippa et al., 2015; Salo et al., 2019). In human cells, endogenously tagged promethin 2016), and many proposed seipin collaborators, such as forms distinct foci in the ER and a high fraction of these Pex30/MCTP2, Ldo45/Promethin, Reep-1, and Aradopsis comigrate with seipin foci (Chung et al., 2019). LDs were LDIP (Table 1), have been suggested to act as ER curva- found to be formed at sites typically occupied by both ture sensors/inducers (Renvoise et al., 2016; Pyc et al., seipin and promethin. Upon subsequent growth of the 2017; Eisenberg-Bord et al., 2018; Joshi et al., 2018; LDs, promethin relocates to LD surface, due to hairpin Teixeira et al., 2018; Wang et al., 2018; Chung et al., topology allowing both ER and LD localization, while 2019). seipin remains at the ER-LD junction (Chung et al., The relationship between ER morphology and LD 2019). nucleation was recently investigated (Santinho et al., 2020). LDs were found to be preferentially generated ER Architecture Promoting LD Formation at ER tubules both in the presence and absence of LD biogenesis is a complex stepwise process in the ER seipin. Experiments in cells and model membranes sug- (Pol et al., 2014). Here, only a brief description of the gested that the presence of free TAG in highly curved currently prevailing model of LD biogenesis is presented. membranes (such as ER tubules) is energetically unfa- For detailed discussion, we kindly refer the reader to vorable compared with flat regions (such as ER sheets). recent comprehensive reviews (Chen and Goodman, This leads to either outflow of TAGs from tubules or 2017; Walther et al., 2017; Chapman et al., 2019; Gao their condensation into LDs. Accordingly, LD nucle- et al., 2019b; Jackson, 2019; Henne et al., 2020; Renne ation can be achieved in vitro by increasing membrane et al., 2020). curvature. Together, these data suggest that the During LD assembly, TAGs synthesized by ER- ER membrane curvature can catalyze LD assembly resident diacylglycerol acyltransferase (DGAT) enzymes (Santinho et al., 2020). first accumulate between the ER bilayer leaflets. A major challenge in deciphering the dynamics and However, as PL bilayers can only accommodate kinetics of LD biogenesis is the small size and possible minute concentrations of TAG, rising local concentra- instability of the formed NL clusters. Indeed, while 40- tions will lead to the formation of nm-sized TAG clus- to 60-nm sized lens-like ER-embedded structures were ters through nucleation and phase separation. These detected in yeast (Choudhary et al., 2015), LD-like struc- form lens-like structures as predicted by simulations tures in the size range of 30 nm were observed at and observed in yeast by EM (Khandelia et al., 2010; seipin-marked sites in human A431 cells during LD Choudhary et al., 2015). Subsequent growth of TAG assembly (Salo et al., 2019).Circa 40 to 50 nm vesicular aggregates will lead to deformation of the ER bilayer structures marked by LiveDrop were detected in COS-7 and budding out of a nascent LD, with the monolayer cells (Li et al., 2019a), and LDs in this size range enclosing the LD derived from the cytoplasmic leaflet of (although not necessarily made de novo) were also the ER (Kassan et al., 2013). These early steps are mod- recently observed by cryo-electron tomography ulated by the ER PL composition via effects on ER (Mahamid et al., 2019).Thus, the size limit of the Salo et al. 5 smallest forming LDs is yet unknown and may vary phenotype, arguing for more redundancy in mammalian between cell types. systems (Joshi et al., 2018). Seipin might control ER PL metabolism, especially Seipin Defines LD Formation Sites that of PA, via protein–protein interactions of major lipid synthesizing enzymes, such as AGPAT2, lipin-1, Several lines of evidence suggest that seipin is a key and GPATs (Sim et al., 2012; Talukder et al., 2015; player in early LD biogenesis and may define the ER Pagac et al., 2016; Gao et al., 2019b; Sim et al., 2020). subdomain primed for LD formation. Seipin foci are Indeed, GPAT activity was altered in a number of seipin localized at LD forming sites in the ER during the ear- depleted systems, suggesting that seipin negatively regu- liest observable steps of LD biogenesis (Wang et al., lates GPAT activity to inhibit aberrant expansion of 2016; Salo et al., 2019; Choudhary et al., 2020). LDs (Pagac et al., 2016). In line with this, defective adi- Fluorescently labeled model ER-LD peptides harboring pogenesis due to seipin-depletion was partially restored LD-targeting motifs, such as HPos and LiveDrop (Wang by GPAT inhibition (Pagac et al., 2016; Gao et al., et al., 2016), have been used to visualize nascent LDs 2020). However, in a recent study, GPAT3 deficiency prior to their detection with traditional hydrophobic failed to rescue adipogenesis in seipin-deficient differen- dyes such as BODIPY. In our hands, the bright lipophilic tiating adipocytes (Sim et al., 2020). Based on these find- dye LD540 (Spandl et al., 2009) stains nascent LDs at ings, the significance of seipin-GPAT interaction least as early as HPos or LiveDrop. However, it is not remains to be determined, but increasing evidence sug- clear whether any of these markers detect lenses or only gests that seipin can act as a scaffold for lipogenic budded-out LDs. enzymes. In A431 cells, seipin motility was found to be In the absence of seipin, PA has been suggested to decreased prior to accumulation of LD540, LiveDrop, accumulate in the ER and play an inhibitory role on or endogenously tagged acyl-CoA synthetase 3 (ACSL3) adipogenic transcriptional program (Liu et al., 2014; (Salo et al., 2019), suggesting that seipin is stabilized at Gao et al., 2019b). This notion is supported by the LD-forming sites prior to other known actors. Similar observation of increased PA levels in several seipin results were reported in SUM159 cells, where LDs depleted systems (Fei et al., 2011; Tian et al., 2011; marked by endogenously tagged Perilipin-3 emerged at Jiang et al., 2014; Liu et al., 2014; Pagac et al., 2016; seipin-defined sites (Chung et al., 2019). Seipin can also Cao et al., 2019). However, other studies have failed to spatially define LD formation sites in the ER. detect differences in cellular PA levels even in the pres- Relocalization of seipin to a subdomain of the ER, the ence of a robust LD phenotype, arguing that global PA nuclear envelope, was sufficient to relocate LD biogen- handling may not be the culprit in seipin deficiency esis to this new site (Salo et al., 2019). Analogous results (Grippa et al., 2015; Wang et al., 2016). Rather, seipin were reported when seipin was relocalized to ER-plasma might alter PA metabolism at a distinct subdomain of membrane junctions (Chung et al., 2019). These obser- the ER. This would be in line with the observation that vations not only demonstrate that seipin can define PA-sensing fluorescent probes accumulate at LD- where a LD starts to develop but also suggest consider- associated ER foci in yeast seipin KO cells, suggesting able spatial malleability in LD formation sites. localized PA accumulation (Grippa et al., 2015; Han et al., 2015; Wolinski et al., 2015). Seipin and Regulation of ER PL Metabolism In this regard, the putative PA-binding site of seipin is in the ER lumen (Yan et al., 2018) proposes a topolog- Local PL levels, especially those of PA and DAG, have ical puzzle. While at least some of the PA-generating been proposed to be vital for LD emergence (Adeyo GPAT and AGPAT enzymes may have their active et al., 2011; Cartwright et al., 2015; Wolinski et al., sites in the ER lumen or within the ER bilayer 2015; Ben M’barek et al., 2017; Choudhary et al., (Yamashita et al., 2014), the PA-utilizing lipin enzymes 2018). It is conceivable that seipin somehow directly are soluble, cytosolic proteins (Zhang and Reue, 2017), modulates local PLs at LD forming sites (Yan et al., which would necessitate PA flipping to access seipin. 2018). In line with this, LD budding is almost completely Intriguingly, there are indications that FIT2 might act abolished in a yeast mutant harboring double deletion of as a lipid phosphate phosphatase in the ER luminal side seipin and a putative ER-shaping protein Pex30, result- (Hayes et al., 2017; Becuwe et al., 2018) and that the ing in accumulation of toxic levels of TAG in the ER protein is localized at seipin-marked LD formation (Wang et al., 2018). Moreover, this phenotype was par- sites (Choudhary et al., 2020). In this scenario, seipin tially restored by modulating the ER PL composition might function to trap PA to promote its efficient catal- toward permissive for LD budding. However, codeple- ysis into DAG at the ER-LD contact. However, this tion of seipin and Pex30’s putative mammalian homo- view is challenged by the lack of robust defects in lipid logue, MCTP2 in human cells did not display the same biosynthesis upon acute seipin loss (Wang et al., 2016; 6 Contact

Salo et al., 2019) and that PA appears to locally accu- formation, but promethin may aid seipin in decreasing mulate in some seipin-deficient systems. the barrier for LD formation. Therefore, in the absence Besides PA, studies have also linked seipin to sphin- of promethin, as LDs are assembled by seipin alone, the golipid metabolism and defects in acyl chain saturation flux to these LDs keeps the ER TAG concentration low (Boutet et al., 2009; Amine et al., 2017; Su et al., 2019). enough to prevent aberrant LD formation, but not high Further work is needed to define how these relate to enough to trigger the assembly of new LDs by seipin. This the LD biogenesis and adipogenesis defects of seipin would then lead to the formation of fewer but larger LDs. loss. The recent discovery that polyunsaturated fatty acids were found to facilitate seipin recruitment to Morphology of Seipin-Mediated ER-LD ER-LD contacts in C. elegans is highly interesting in this regard (Cao et al., 2019). Contacts After their formation, LDs remain functionally con- Seipin May Sequester TAGs at LD nected to the ER via ER-LD contacts, which function to facilitate cargo exchange between the two organ- Formation Sites elles. Morphologically, ER-LD contacts can be broad- Emerging studies suggest that seipin may directly control ly categorized into two overlapping classes: membrane TAG distribution in the ER and its partitioning into nascent bridges, wherein the LD monolayer appears continu- LDs. In a yeast system where NL synthesis was held con- ous with the ER bilayer; and areas of close ER-LD stant, LD biogenesis was severely delayed in seipin-deficient proximity without direct membrane continuity cells with a relative accumulation of TAG in ER mem- (Figure 2A and B; Salo and Ikonen, 2019). In addition, branes (Cartwright et al., 2015). Similarly, upon LD induc- proteinaceous tethers between the two organelles have tion in mammalian cells in the absence of seipin, numerous also been observed by EM (Wang et al., 2016; Salo tiny and growth-abortive LDs emerged from the ER, with a et al., 2019; Figure 2C). Although the functional sig- subset of these completely detaching from the ER, a phe- nificance of these different contact zones is not yet nomenon not observed in wild-type cells (Salo et al., 2016; understood, seipin appears to be primarily involved Wang et al., 2016). These tiny LDs may reflect spontaneous in the formation and maintenance of the membrane phase separation or oiling out of excess TAG, as has been bridges between ER and LDs. postulated to occur in protein-free systems (Deslandes et al., Considering the persistent localization of seipin at 2017). Thus, seipin could function to attract and foster ER-LD contact sites (Salo et al., 2016; Wang et al., TAG aggregates in the ER for controlled LD formation. 2016), the morphological features of seipin-mediated How could seipin sequester TAG at LD formation ER-LD contacts were recently characterized. Using sites? The membrane-anchored HHs may be important correlative light and EM, seipin-mediated contacts for this, as they have affinity for a monolayer enclosing were found to harbor a strikingly uniform, neck-like NLs (Sui et al., 2018). Furthermore, seipin appears to architecture (Salo et al., 2019). At these ER-LD control TAG condensation in the ER (Santinho et al., necks, the LD monolayer appeared to fuse with the 2020) and copurified seipin-promethin complexes con- ER lumen, indicating membrane continuity between tained TAG molecules (Chung et al., 2019). Therefore, the outer leaflet of the ER and the LD monolayer seipin may initially attract TAG molecules in the ER (Figure 2C and D). The diameter of the region in bilayer and foster TAG aggregates to grow into a nascent touch with the LD was circa 15 nm, a size consistent LD. This is in line with the recently reported DGAT1 with the structure of oligomeric seipin resolved with the structure placing the active TAG-generating site within cryo-EM (Sui et al., 2018; Yan et al., 2018). This sug- the ER bilayer (Sui et al., 2020; Wang et al., 2020). In gests that seipin may physically limit the diameter of the absence of seipin, efficient TAG phase separation in the ER-LD neck. High curvature of the neck implicates the ER would be delayed, leading to an increase in the ER a specific PL composition, and the lipid-binding TAG concentration, as reported (Cartwright et al., 2015; domain of seipin might be involved in stabilizing the Gao et al., 2017). This increased bilayer TAG concentra- neck. Indeed, such seipin-mediated ER-LD necks were tion eventually leads to the aberrant oiling out of LDs. disrupted by acute removal of seipin via auxin-induced The precise role of promethin in this scenario needs to degradation (Salo et al., 2019). be established, as such oiling out of LDs was not observed A morphologically similar ER-LD contact of circa in promethin KO cells. Instead, promethin KO cells gen- 15 nm in diameter, with apparent membrane continuity, erated fewer, but larger LDs, which still contained seipin was also recently observed by cryo-EM tomography of at their ER-LD contact (Chung et al., 2019). This may HeLa cells (Figure 3E of Mahamid et al., 2019) and suggest that seipin is de facto required to keep ER TAG similar neck-like structures were previously observed concentration low enough to prevent aberrant LD in, for example, Drosophila and Cos-1 cells (Kassan Salo et al. 7

A ER-LD contact topologies C Seipin NE-trapped cells

inside view 20000 nmnm side view B ER-LD contact examples D CLEM ()end-seipin-GFPx7 / LiveDrop-mCherry

inside view side view

Figure 2. Seipin-Mediated ER-LD Contacts. A: Topologies of ER-LD contacts. Left panel: membranous bridges, mediated by seipin, right panel: zones of ER-LD proximity, likely mediated by various ER-LD tethers. B: Examples of ER-LD contacts from primary human fibroblasts. Left panel is adapted from Salo et al. (2019) with permission of the publisher, and right panel is from the same data set. Scale bar: 50 nm. Orange arrowhead indicates neck-like membranous bridge. C and D: Examples of the architecture of seipin mediated ER-LD contacts, adapted from Salo et al. (2019) with permission of the publisher. Scale bars for C: 100 nm, 50 nm, 20 nm, and 20 nm. Scale bars for D: 10 mm, 50 nm, 50 nm and 20 nm. C: Examplary neck-like ER-LD contact from A431 cells with seipin trapped at the NE ER subdomain. Orange arrowhead indicates neck-like membranous bridge, blue arrowheads indicate fibrous tethers. Right panel is 3D modeling of the LD (brown), NE (blue), and NE-LD contact site (red). D: CLEM of A431 cells with seipin tagged endogenously with GFPx7 and expressing the nascent LD marker LiveDrop-mCherry. The inset of the fluorescence microscopy panel (left) depicts the same LD in the same orientation as the EM images. Orange arrowhead indicates neck-like membranous bridge. Right panel depicts 3D modeling of the LD (brown), ER (yellow) and the ER-LD contact site (red). ER ¼ endoplasmic reticulum; LD ¼ lipid droplet; NE ¼ nuclear envelope; CLEM ¼ correlative light and electron microscopy.

A Seipin antibody B Differentiated human adipocyte stem cells WT anti-seipin Projection Lipidtox (LDs)

Seipin KO Projection Projection Projection Single slice

Figure 3. Seipin in Human Adipocytes. A: Confocal images of human A431 WT and seipin KO cells stained with anti-seipin antibodies (Salo et al., 2019). Scale bar: 10 mm. B: Airyscan images of human adipocytes derived from visceral fat stem cells. Adipogenic differentiation was induced by a cocktail of transferrin (10 mg/ml final concentration), human insulin (Actrapid; 66 nM), cortisol (100 nM), triiodothyronine (1 nM), 3-isobutyl-1-methylxanthine (500 mM), and rosiglitazone (2 mM). After 3 days, supplements were reduced to transferrin, insulin, cortisol, and triiodothyronine with the aforementioned concentrations. After 14 days of adipogenic differentiation, cells were fixed and stained with anti-seipin antibodies followed by anti-rabbit Alexa 647 and LipidTox Green. Scale bars: 10 mm and 1 mm. LD ¼ lipid droplet; KO ¼ knockout; WT ¼wild type. et al., 2013; Wilfling et al., 2013). Membranous bridges data suggest that seipin localizes to and controls the between inner nuclear membrane and intranuclear LDs formation of such bridges. This would be a logical con- in yeast cells were also reported to be seipin-dependent sequence of early LD biogenesis occurring at seipin- (Romanauska and Kohler,€ 2018). Altogether, these marked sites: if the initial LD budding event were to 8 Contact take place through the seipin disk, this would constrain DGAT2 (LD; Xu et al., 2012); Rab18 (LD) and the the dimensions of the ER-LD neck accordingly. NRZ complex and associated SNARE proteins (ER; In nonadipogenic cell lines studied so far, LDs typi- Xu et al., 2018); and Orp2 (LD) and VAPA (ER; cally harbor a single distinct seipin focus stably associ- Weber-Boyvat et al., 2015).Snx14 is also capable of teth- ated with each LD (Salo et al., 2016; Wang et al., 2016) ering LDs to the ER, by binding to both organelles and seipin-mediated ER-LD necks appear to be long- simultaneously (Datta et al., 2019) and a similar topo- lasting structures (stable at least on timescale of hours; logical arrangement has been proposed for DGAT2 Salo et al., 2019). Moreover, seipin recruitment to pre- (McFie et al., 2018).DFCP1 has also been documented existing LDs has not been reported. These data imply to facilitate ER-LD contact formation, acting in concert that seipin does not act like a canonical membrane con- with Rab18 (Gao et al., 2019a; Li et al., 2019a). In yeast tact site tether but might rather remain at the topologi- cells, Pex30 and the yeast homolog of FIT2 localize to cally unique ER-LD contact site via virtue of LD ER-LD contacts, colocalizing with seipin (Choudhary biogenesis occurring through the seipin disk. et al., 2018; Joshi et al., 2018; Wang et al., 2018). There may, however, be species–specific differences, Newly discovered ER-LD contact site proteins with as overexpressed C. elegans seipin was shown to form putative lipid transfer activity include Orp4, VPS13A, much larger (several hundreds of nanometers in diame- and VPS13C (Kumar et al., 2018; Du et al., 2020). ter) peri-LD cages at sites where the tubular elements of At least in the case of Rab18, Snx14, DFCP1, and the ER are in close proximity to the LD (Cao et al., VPS13A, overexpression has been shown to increase 2019). Overexpression of C. elegans seipin promoted ER-LD proximity, strongly suggesting that these factors the formation of such structures also in Cos-7 cells can facilitate tethering of the two organelles (Kumar et al., and, conversely, heterologously expressed human 2018; Xu et al., 2018; Datta et al., 2019; Li et al., 2019a). seipin localized similarly in tubular peri-LD cages in the The topology of these expanded ER-LD contact zones is C. elegans intestine. Such structures have not been probably not similar to seipin-mediated ER-LD necks but rather reflects a close proximity of the two organelles reported upon overexpression of human or Drosophila without direct membrane continuity (Figure 2A). seipin in nonadipogenic cell lines (Salo et al., 2016; However, these factors are still likely to influence/modu- Wang et al., 2016), but seipin peri-LD cages do resemble late seipin-mediated ER-LD contacts. For example, the localization of overexpressed N-glycosylation defective DFCP1 or Rab18 perturbation decreased ER-LD prox- N88S-seipin mutant, which also accumulates as ring-like imities in seipin depleted Cos-7 cells (Li et al., 2019a). structures flanking LDs (Holtt€ a-Vuorietal.,2013).€ DFCP1 and Rab18 were proposed to form a complex An intriguing possibility is that the peri-LD cages of and were shown to co-immunoprecipitate seipin, suggest- C. elegans intestine, a major fat deposit in these organ- ing that seipin may participate in modulation of isms, are a consequence of LD fusion where individual DFCP1Rab18-mediated ER-LD contacts. The phenotype LDs, initially harboring one seipin-mediated neck, fuse of DFCP1 or Rab18 depletion, however, is distinct from and the resulting structure would contain multiple such that of seipin loss; overall, LD sizes were moderately contact sites in very close proximity. One could envision reduced and LD numbers were increased. However, this to be similar to the case in adipocytes, where a single Rab18 and DFCP1 seem to have cell-type specific func- unilocular LD forms via ripening-mediated coalescence tions, as their perturbation can also induce supersized of a high number of smaller ER-derived LDs (Paar et al., LDs in some cell types such as 3T3-L1 cells, while only 2012; Chu et al., 2014). These giant LDs could thus con- minor alterations in LDs are observed in others (Jayson tain a high number of seipin-mediated contacts. Indeed, et al., 2018; Xu et al., 2018; Li et al., 2019a). Clearly, our immunofluorescence analysis of endogenous seipin further investigations into the interplay between Rab18, localization in differentiated human adipocyte stem cells DFCP1, and seipin are warranted. suggests that this is the case, with numerous seipin foci DFCP1 has also been implicated in early LD juxtaposed to enlarged LDs (Figure 3). Detailed charac- biogenesis. Overexpressed DFCP1 labeled distinct terization of seipin-mediated ER-LD contacts at the EM ER-associated foci that increased in number upon fatty level in adipocytes has not been reported. acid administration (Li et al., 2019a). A subset of these developed into LDs, but DFCP1 labeling preceded that of Modulation of Seipin-Mediated ER-LD early LD marker peptides HPos or LiveDrop, suggesting Contacts by Other Proteins that DFCP1 may detect very early NL accumulations. By EM, these DFCP1 foci appeared as small vesicles or ER- Besides seipin, a number of other proteins act at ER-LD associated papillary structures. Whether seipin localizes contacts (Bohnert, 2020). These include tethering protein to these DFCP1 positive regions prior to their maturation complexes, where one partner binds to the ER while the into HPos or LiveDrop-positive organelles is an interest- other contacts the LD, such as FATP1 (ER) and ing question not yet addressed. Salo et al. 9

The interplay of Snx14 and seipin at ER-LD contacts bigger ones through a connecting phase and wherein was also recently investigated (Datta et al., 2019). LDs the directionality is governed by the higher internal pres- were found to be supersized and tiny in Snx14 KO U2OS sure of smaller droplets, imposing a flux of molecules cells, resembling the LD phenotype upon seipin loss. toward lower pressure, that is, bigger droplets (Thiam Overexpressed Snx14 dramatically increased LD-ER et al., 2013; Thiam and Foreˆt, 2016). proximity and increased TAG generation, suggesting Ostwald ripening-mediated fusion is a well-known that Snx14, similarly to its proposed yeast counterpart property of liquid-liquid phase-separated systems. A Mdm1, may regulate fatty acid homeostasis (Datta vast number of recent work has begun to elucidate the et al., 2019; Hariri et al., 2019). Conversely, loss of importance of liquid–liquid phase-separated in organiza- Snx14 decreased LD-ER proximity and Snx14 was thus tion of the cellular milieu, especially with regard to mem- proposed to function as an ER-LD tether, ensuring that braneless organelles (Brangwynne et al., 2009, 2009; the two organelles maintain a connection as ER-produced Alberti et al., 2019), but also in the context of ER- TAG is fluxed into the maturing LD. However, seipin derived organelle biogenesis such as autophagosome for- overexpression failed to rescue the LD phenotype in mation (Fujioka et al., 2020).Ripening-mediated growth Snx14 KO cells and vice versa, and concomitant depletion of LDs via direct LD-LD contacts has also been docu- of both proteins did not aggravate the LD phenotypes. mented (Gong et al., 2011; Ju¨ ngst et al., 2013). At LD- The authors therefore concluded Snx14 to function inde- LD interfaces, a larger LD grows via acquisition of lipids pendently of seipin (Datta et al., 2019). On the other from the smaller LD in a process facilitated by Fsp27/ hand, considering the different topologies of Snx14 and CIDE-C (Gong et al., 2011). Interestingly, mutations in seipin-mediated ER-LD contacts (Snx14 tethering the Fsp27/CIDE-C also lead to congenital lipodystrophy organelles in trans and seipin mediating an ER-LD (Rubio-Cabezas et al., 2009) and CIDE family members neck with membrane continuity), Snx14 might aid in are regulators of adipogenesis (Slayton et al., 2019). the stabilization of seipin-mediated ER-LD necks, espe- However, in contrast to the Fsp27-mediated process, cially during later stages of LD maturation. Indeed, ripening-mediated LD size changes upon acute seipin Snx14 localized to LDs later than seipin, hours after ini- removal take place via the ER bilayer (Salo et al., tiation of LD biogenesis (Datta et al., 2019). 2019). To decipher the role of seipin in this process, het- erologous cell fusion experiments were conducted, Seipin-Mediated Lipid Flux to LDs revealing that in a continuous ER network, LDs with seipin at their ER-LD contact site grew, while LDs with The persistent localization of seipin at ER-LD contacts seipin removed from their ER-LD contact shrunk. These and a defined architecture of seipin-mediated ER-LD data imply that seipin at the ER-LD neck facilitates the contacts suggests that seipin has a role in LDs beyond growth of the LD it is associated with (Figure 4A and B). facilitating initial LD assembly. In support of this, deple- This function is critical for preventing the shrinkage of tion of seipin from mature adipocytes in vivo results in smaller LDs (more prone to ripening) and facilitating progressive lipodystrophy and decreased adiposity (Liu their growth (Salo et al., 2019). Recent studies of C. et al., 2014; Zhou et al., 2015). However, due to the elegans seipin also implicate a droplet-autonomous role dramatic effects on initial LD biogenesis in the absence of seipin in promoting LD growth (Cao et al., 2019). of seipin, investigating its role in ER-LD contacts at How is seipin performing this LD growth promoting later stages has been challenging. To overcome this function? Considering the topological arrangement at obstacle, the plant-based auxin-inducible degron seipin-mediated ER-LD necks, an intriguing possibility system (Nishimura et al., 2009; Li et al., 2019c) was is that seipin controls lipid diffusion, possibly by attract- recently utilized to rapidly deplete seipin in human ing TAG molecules from within the ER bilayer and facil- cells. With this system seipin could be acutely removed itating their flux to the LD and/or by controlling PL from preexisting ER-LD contacts, revealing a role for density at the LD surface (Grippa et al., 2015; Salo seipin in LD maintenance (Salo et al., 2019). et al., 2019). Upon acute seipin removal, ER-LD lipid Upon acute seipin removal, LDs rapidly started to flux becomes dominated by biophysical forces favoring become more heterogeneous in size, with larger LDs the growth of larger LDs over smaller ones. In essence, growing and acquiring more lipid cargo from the ER, seipin would thus function as a lipid transfer protein. and smaller LDs shrinking, apparently donating their This is well in line with the proposed role of seipin cargo to the larger LDs. This occurred in the absence during initial LD assembly. of major alterations to overall net fatty acid handling or lipolysis (Salo et al., 2019). The observed LD size changes can be accounted for by a phenomenon called The ER-LD Network as a Joint System Ostwald ripening. It is a molecular diffusion process by The notion that LDs may ripen via the connecting lipid which smaller droplets continuously leak material to phase, the ER bilayer, implies that LDs and ER can be 10 Contact

A Seipin mediated TAG flux at the ER-LD contact Both growing Seipin LD growing Bigger growing, smaller shrinking

B Seipin mediated TAG flux and ripening in the ER network

Seipin facilitating Seipin removed: Additional tiny LDs TAG flux to LDs ripening mediated emerge as ER TAG heterogeneous concentration rises growth

C The effect of pre-existing LDs on newly emerging LDs

TAG flux

ER TAG concentration

New LD

Figure 4. Seipin-Mediated TAG Partitioning. A: Schematic of seipin (green ellipse) function in promoting LD growth, adapted from Salo et al.(2019). B: Seipin removal evokes ripening-mediated LD size changes via the ER. C: The effect of preexisting LDs on nascent LD formation sites: seipin-mediated TAG flux to LDs decreases the nearby TAG concentration in the ER, and thus new LDs tend to be formed at sites distant to preexisting LDs. ER ¼ endoplasmic reticulum; LD ¼ lipid droplet; TAG ¼ triglycerides. Salo et al. 11 considered as a joint system. This continuity is evident in providing human visceral fat biopsies and Dr. Xavier the case of many proteins, typically harboring hairpin Prasanna for providing the structures for Figure 1. We motifs, which exhibit dual localization on ER and LDs acknowledge the use of HiLIFE and Biocenter Finland imag- (Kory et al., 2016). However, also NL fluxes through the ing core facilities (Biomedicum Imaging Unit and Electron connecting ER bilayer appear to contribute to LD-LD Microscopy Unit). Figures 1A, 2A and 3C were generated communication. For example, during early LD biogen- with the help of BioRender.com. esis, NLs are likely synthesized at highly dispersed sites along the ER (Poppelreuther et al., 2018). Upon Declaration of Conflicting Interests increased local concentrations, a NL lens may nucleate The author(s) declared no potential conflicts of interest at a site marked by seipin and start to grow. The flux of with respect to the research, authorship, and/or publication NL to the growing LD, enhanced by seipin, may thus of this article. reduce nearby TAG concentration in the ER, disfavoring the formation of other LDs nearby. Thus, a preexisting Funding LD would inhibit nearby LD formation (Figure 4C). The author(s) disclosed receipt of the following financial sup- As a consequence, LDs would be expected to form at port for the research, authorship, and/or publication of this sites more distant to one another than expected by article: This work was supported by Sigrid Juseliuksen chance, which is the case at least in human A431 cells Sa€ati€ o,€ Jane ja Aatos Erkon Sa€ati€ o,€ Helsingin Yliopisto, and (Salo et al., 2019). Indeed, nascent LDs are formed at Academy of Finland (grant no. 307415 and 312491). highly dispersed sites in multiple cell types (Kassan et al., 2013; Chung et al., 2019). ORCID iDs While direct observation of NL fluxes within the ER Veijo T. Salo https://orcid.org/0000-0002-6991-2142 is technically challenging, rapid advances in imaging and Maarit Holtt€ a-Vuori€ https://orcid.org/0000-0002-0488-4486 probe technologies (Valm et al., 2017; Adhikari et al., 2019) are likely to facilitate this in the near future. Findings from a recent study, employing elegant imaging References techniques (holo-tomographic microscopy), support the Adeyo O, Horn PJ, Lee S, Binns DD, Chandrahas A, notion of a fluid connection of LDs through the ER Chapman KD, Goodman JM (2011). The yeast lipin ortho- (Sandoz et al., 2019). It was found that LD growth in logue Pah1p is important for biogenesis of lipid droplets. J a given cell during fatty acid administration was highly Cell Biol 192, 1043–1055. doi: 10.1083/jcb.201010111 Adhikari S, Moscatelli J, Smith EM, Banerjee, C, Puchner EM synchronous, with both shrinking and growing of LDs (2019). Single-molecule localization microscopy and track- happening in concert, as if sharing a common pool of ing with red-shifted states of conventional BODIPY conju- lipid precursors.Furthermore, upon initial LD assembly, gates in living cells. Nat Commun 10, 1–12. doi: https://doi. preexisting LDs actually initially slightly shrunk in size org/10.1038/s41467-019-11384-6 at the same time as numerous new LDs emerged. This Alberti S, Gladfelter A, Mittag T (2019). Considerations and suggests that preexisting LDs unload material into a challenges in studying Liquid-Liquid phase separation and common lipid pool, the ER (Sandoz et al., 2019). biomolecular condensates. Cell 176, 419–434. doi: 10.1016/j. cell.2018.12.035 Amine M, Simon L, Sabri L, Rial A, Bergeron KF, Ralston Concluding Remarks JC, Mutch DM, Mounier C (2017). Hepatic BSCL2 (seipin) Overall, these are exciting times for seipin and LD biol- deficiency disrupts lipid droplet homeostasis and increases ogy. Through concentrated efforts of cell biological, lipid metabolism via SCD1 activity. Lipids 52, 129–150. doi: structural, and biophysical endeavors, we are beginning 10.1007/s11745-016-4210-5 to get clues of the molecular mechanisms underlying LD Bacle A, Gautier R, Jackson CL, Fuchs PF, Vanni S (2017). biogenesis in the ER-LD network. Bridging these emerg- Interdigitation between triglycerides and lipids modulates sur- ing concepts to in vivo models of fat storage will likely face properties of lipid droplets. Biophys J 112, 1417–1430. doi: 10.1016/j.bpj.2017.02.032 yield powerful insight and new ways to tackle lipid Becuwe M, Bond LM, Mejhert N, Boland S, Shane D, imbalance beyond that related to the loss of seipin Cicconet M, Liu XN, Graham MM, Walther TC Jr, function. Farese RV (2018). FIT2 is a lipid phosphate phosphatase crucial for endoplasmic reticulum homeostasis. Biorxiv. doi: Acknowledgements https://doi.org/10.1101/291765 We thank Prof. Hannele Yki-Jarvinen,€ Helsinki University Ben M’barek K, Ajjaji D, Chorlay A, Vanni S, Foreˆt, L, Thiam Hospital and University of Helsinki, for collaboration in AR (2017). ER membrane phospholipids and surface 12 Contact

tension control cellular lipid droplet formation. Dev Cell 41, Chorlay A, Monticelli L, Verıssimo Ferreira J, Ben M’barek K, 591.e7–604.e7. doi: 10.1016/j.devcel.2017.05.012 Ajjaji D, Wang S, Johnson E, Beck R, Omrane M, Beller Bi J, Wang W, Liu Z, Huang X, Jiang Q, Liu G, Wang, Y, M, et al. (2019). Membrane asymmetry imposes direction- Huang, X (2014). Seipin promotes adipose tissue fat storage ality on lipid droplet emergence from the ER. Dev Cell 50, 2þ through the ER Ca -ATPase SERCA. Cell Metab 19, 25.e7–42.e7. doi: 10.1016/j.devcel.2019.05.003 861–871. doi: 10.1016/j.cmet.2014.03.028 Chorlay A, Thiam AR (2020). Neutral lipids regulate amphi- Binns D, Lee S, Hilton CL, Jiang Q-X, Goodman JM (2010). pathic helix affinity for model lipid droplets. J Cell Biol. doi: Seipin is a discrete homooligomer. 49, 10.1083/jcb.201907099 10747–10755. doi: 10.1021/bi1013003 Choudhary V, El Atab O, Mizzon G, Prinz WA, Schneiter R Bohnert M (2020). Tethering fat: tethers in lipid droplet con- (2020). Seipin and Nem1 establish discrete ER subdomains tact sites. Contact. doi: https://doi.org/10.1177/ to initiate yeast lipid droplet biogenesis. J Cell Biol 219. doi: 2515256420908142 10.1083/jcb.201910177 Boutet E, El Mourabit H, Prot M, Nemani M, Khallouf E, Choudhary V, Golani G, Joshi AS, Cottier S, Schneiter R, Colard O, Maurice M, Durand-Schneider A-M, Chretien Y, Prinz WA, Kozlov MM (2018). Architecture of lipid drop- Gres S, et al. (2009). Seipin deficiency alters fatty acid lets in endoplasmic reticulum is determined by phospholipid Delta9 desaturation and lipid droplet formation in intrinsic curvature. Curr Biol 28, 915.e9–926.e9. doi: Berardinelli-Seip congenital lipodystrophy. Biochimie 91, 10.1016/j.cub.2018.02.020 796–803. doi: 10.1016/j.biochi.2009.01.011 Choudhary V, Ojha N, Golden A, Prinz WA (2015). A con- Brangwynne CP, Eckmann CR, Courson DS, Rybarska A, served family of proteins facilitates nascent lipid droplet Hoege C, Gharakhani J, Julicher F, Hyman AA (2009). budding from the ER. J Cell Biol 211, 261–271. doi: Germline P granules are liquid droplets that localize 10.1083/jcb.201505067 by controlled dissolution/condensation. Science 324, Chu M, Sampath H, Cahana DY, Kahl CA, Somwar R, 1729–1732. doi: 10.1126/science.1172046 Cornea A, Roberts CT, Varlamov O (2014). Cai Y, Goodman JM, Pyc M, Mullen RT, Dyer JM, Chapman Spatiotemporal dynamics of triglyceride storage in uniloc- KD (2015). Arabidopsis SEIPIN proteins modulate triacyl- ular adipocytes. Mol Biol Cell 25, 4096–4105. doi: 10.1091/ glycerol accumulation and influence lipid droplet prolifera- mbc.E14-06-1085 tion. Plant Cell 27, 2616–2636. doi: 10.1105/tpc.15.00588 Chung J, Wu X, Lambert TJ, Lai ZW, Walther TC, Farese RV Cao Z, Hao Y, Lee YY, Wang P, Li X, Xie K, Lam WJ, Qiu Y, (2019). LDAF1 and seipin form a lipid droplet assembly Shui G, Liu P, et al. (2019). Dietary fatty acids promote complex. Dev Cell 51, 1–13. doi: 10.1016/j. lipid droplet diversity through seipin enrichment in an ER devcel.2019.10.006 subdomain. Nat Commun 10, 2902. doi: 10.1038/s41467- Coradetti ST, Pinel D, Geiselman GM, Ito M, Mondo SJ, 019-10835-4 Reilly MC, Cheng YF, Bauer S, Grigoriev IV, Gladden Cartwright BR, Binns DD, Hilton CL, Han S, Gao Q, JM, et al. (2018). Functional genomics of lipid metabolism Goodman JM (2015). Seipin performs dissectible functions 7 in promoting lipid droplet biogenesis and regulating droplet in the oleaginous yeast Rhodosporidium toruloides. Elife , morphology. Mol Biol Cell 26, 726–739. doi: 10.1091/mbc. 1–55. doi: 10.7554/elife.32110 E14-08-1303 Cui X, Wang Y, Tang Y, Liu Y, Zhao L, Deng J, Xu G, Peng Cartwright BR, Goodman JM (2012). Seipin: from human dis- X, Ju S, Liu G, et al. (2011). Seipin ablation in mice results ease to molecular mechanism. J Lipid Res 53, 1042–1055. in severe generalized lipodystrophy. Hum Mol Genet 20, doi: 10.1194/jlr.R023754 3022–3030. doi: 10.1093/hmg/ddr205 Castro IG, Eisenberg-Bord M, Persiani E, Rochford JJ, Datta S, Liu Y, Hariri H, Bowerman, J, Henne WM (2019). Schuldiner M, Bohnert M (2019). Promethin is a conserved Cerebellar ataxia disease-associated Snx14 promotes lipid seipin partner protein. Cells 8, 268. doi: 10.3390/ droplet growth at ER-droplet contacts. J Cell Biol 218, cells8030268 1335–1351. doi: 10.1083/jcb.201808133 Chapman KD, Aziz M, Dyer JM, Mullen RT (2019). Deslandes F, Thiam AR, Foreˆt L (2017). Lipid droplets can Mechanisms of lipid droplet biogenesis. Biochem J 476, spontaneously bud off from a symmetric bilayer. Biophys J 1929–1942. doi: 10.1042/BCJ20180021 113, 15–18. doi: 10.1016/j.bpj.2017.05.045 Chen, X, Goodman JM (2017). The collaborative work of Ding L, Yang X, Tian H, Liang J, Zhang F, Wang G, Wang Y, droplet assembly. Biochim Biophys Acta Mol Cell Biol Ding M, Shui G, Huang X (2018). Seipin regulates lipid Lipids 1862, 1205–1211. doi: 10.1016/j.bbalip.2017.07.003 homeostasis by ensuring calcium-dependent mitochondrial Chen W, Chang B, Saha P, Hartig SM, Li L, Reddy VT, Yang metabolism. EMBO J 37, 1–17. doi: 10.15252/ Y, Yechoor V, Mancini MA, Chan L (2012). Berardinelli- embj.201797572 seip congenital lipodystrophy 2/seipin is a cell-autonomous Du X, Zhou L, Aw YC, Mak HY, Xu Y, Rae J, Wang W, regulator of lipolysis essential for adipocyte differentiation. Zadoorian A, Hancock SE, Osborne B, et al. (2020). ORP5 Mol Cell Biol 32, 1099–1111. doi: 10.1128/MCB.06465-11 localizes to ER-lipid droplet contacts and regulates the level Chen W, Yechoor VK, Chang BH-J, Li MV, March KL, Chan of PI(4)P on lipid droplets. J Cell Biol 219, 1–16. doi: L (2009). The human lipodystrophy gene product 10.1083/jcb.201905162 Berardinelli-Seip congenital lipodystrophy 2/seipin plays a Ebihara C, Ebihara K, Aizawa-Abe M, Mashimo T, Tomita T, key role in adipocyte differentiation. Endocrinology 150, Zhao M, Gumbilai V, Kusakabe T, Yamamoto Y, Aotani 4552–4561. doi: 10.1210/en.2009-0236 D, et al. (2015). Seipin is necessary for normal brain Salo et al. 13

development and spermatogenesis in addition to adipogen- Guillen-Navarro E, Sa´ nchez-Iglesias S, Domingo-Jimenez R, esis. Hum Mol Genet 24, 4238–4249. doi: 10.1093/hmg/ Victoria B, Ruiz-Riquelme A, Ra´ bano A, Loidi L, Beiras A, ddv156 Gonza´ lez-Mendez B, Ramos A, , et al. (2013). A new seipin- Eisenberg-Bord M, Mari M, Weill U, Rosenfeld-Gur E, associated neurodegenerative syndrome. J Med Moldavski O, Castro IG, Soni KG, Harpaz N, Levine Genet 1–9. doi: 10.1136/jmedgenet-2013-101525 TP, Futerman AH, et al. (2018). Identification of seipin- Han S, Binns DD, Chang Y-F, Goodman JM (2015). linked factors that act as determinants of a lipid droplet Dissecting seipin function: the localized accumulation of subpopulation. J Cell Biol 217, 269–282. doi: 10.1083/ phosphatidic acid at ER/LD junctions in the absence of jcb.201704122 seipin is suppressed by Sei1pDNterm only in combination Falk J, Rohde M, Bekhite MM, Neugebauer S, Hemmerich P, with Ldb16p. BMC Cell Biol 16, 29. doi: 10.1186/s12860- Kiehntopf M, Deufel T, Hu¨ bner C a, Beetz, C (2014). 015-0075-3 Functional mutation analysis provides evidence for a role Hariri H, Rogers S, Ugrankar R, Liu YL, Feathers JR, Henne of REEP1 in lipid droplet . Hum Mutat 35, 497–504. WM (2018). Lipid droplet biogenesis is spatially coordinat- doi: 10.1002/humu.22521 ed at ER–vacuole contacts under nutritional stress. EMBO Fei W, Shui G, Gaeta B, Du X, Kuerschner L, Li P, Brown AJ, Rep 19, 57–72. doi: 10.15252/embr.201744815 Wenk MR, Parton RG, Yang H (2008). Fld1p, a functional Hariri H, Speer N, Bowerman J, Rogers S, Fu G, Reetz E, homologue of human seipin, regulates the size of lipid drop- Datta S, Feathers JR, Ugrankar R, Nicastro D, et al. lets in yeast. J Cell Biol 180, 473–482. doi: 10.1083/ (2019). Mdm1 maintains endoplasmic reticulum homeosta- jcb.200711136 sis by spatially regulating lipid droplet biogenesis. J Cell Fei W, Shui G, Zhang Y, Krahmer N, Ferguson C, Kapterian Biol 218, 1319–1334. doi: 10.1083/jcb.201808119 TS, Lin RC, Dawes IW, Brown AJ, Li P, et al. (2011). A Hayes MJ, Choudhary V, Ojha N, Shin JJ, Han G-S., Carman role for phosphatidic acid in the formation of “supersized” GM, Loewen CJ, Prinz WA, Levine TP (2017). Fat storage- lipid droplets. PLoS Genet 7, e1002201. doi: 10.1371/jour- inducing transmembrane (FIT or FITM) proteins are relat- nal.pgen.1002201 ed to lipid phosphatase/phosphotransferase enzymes. Fujioka Y, Alam JM, Noshiro D, Mouri K, Ando T, Okada Y, Microb Cell 5, 88–103. doi: 10.15698/mic2018.02.614 May AI, Knorr RL, Suzuki K, Ohsumi Y, et al. (2020). Henne M, Goodman JM, Hariri H (2020). Spatial compart- Phase separation organizes the site of autophagosome for- mentalization of lipid droplet biogenesis. Biochim Biophys mation. Nature 578, 301–305. doi: 10.1038/s41586-020- Acta Mol Cell Biol Lipids 1865, 158499. doi: 10.1016/j. 1977-6 bbalip.2019.07.008 Gao G, Sheng Y, Yang H, Chua BT, Xu L (2019a). DFCP1 Henne WM, Reese ML, Goodman JM (2018). The assembly of associates with lipid droplets. Cell Biol Int 43, 1492–1504. lipid droplets and their roles in challenged cells. EMBO J doi: 10.1002/cbin.11199 37, e98947. doi: 10.15252/embj.201898947 Gao M, Huang X, Song B-L, Yang H (2019b). The biogenesis Holtt€ a-Vuori€ M, Salo VT, Ohsaki Y, Suster ML, Ikonen E of lipid droplets: lipids take center stage. Prog Lipid Res 75, (2013). Alleviation of seipinopathy-related ER stress by 100989. doi: 10.1016/j.plipres.2019.100989 triglyceride storage. Hum Mol Genet 22, 1157–1166. doi: Gao M, Liu L, Wang X, Mak HY, Liu G, Yang H (2020). 10.1093/hmg/dds523 GPAT3 deficiency alleviates insulin resistance and hepatic Jackson CL (2019). Lipid droplet biogenesis. Curr Opin Cell steatosis in a mouse model of severe congenital generalized Biol 59, 88–96. doi: 10.1016/j.ceb.2019.03.018 lipodystrophy. Hum Mol Genet 29, 432–443. doi: 10.1093/ Jayson CBK, Arlt H, Fischer AW, Lai ZW, Farese RV, hmg/ddz300 Walther TC (2018). Rab18 is not necessary for lipid droplet Gao Q, Binns DD, Kinch LN, Grishin NV, Ortiz N, Chen X, biogenesis or turnover in human mammary carcinoma cells. Goodman JM (2017). Pet10p is a yeast perilipin that stabil- Mol Biol Cell 29, 2037–2136. doi: 10.1091/mbc.E18-05-0282 izes lipid droplets and promotes their assembly. J Cell Biol Jiang M, Gao M, Wu C, He H, Guo X, Zhou Z, Yang H, Xiao 216, 3199–3217. doi: 10.1083/jcb.201610013 X, Liu G, Sha J (2014). Lack of testicular seipin causes Gerondopoulos A, Bastos RN, Yoshimura SI, Anderson R, teratozoospermia syndrome in men. Proc Natl Acad Sci Carpanini S, Aligianis I, Handley MT, Barr FA (2014). USA 111, 7054–7059. doi: 10.1073/pnas.1324025111 Rab18 and a Rab18 GEF complex are required for Jiao Y, Ahmed U, Sim MFM, Bejar A, Zhang X, Talukder normal ER structure. J Cell Biol 205, 707–720. doi: MMU, Rice R, Flannick J, Podgornaia AI, Reilly DF, et al. 10.1083/jcb.201403026 (2019). Discovering metabolic disease gene interactions by Gong J, Sun Z, Wu L, Xu W, Schieber N, Xu D, Shui G, Yang correlated effects on cellular morphology. Mol Metab 24, H, Parton RG, Li P (2011). Fsp27 promotes lipid droplet 108–119. doi: 10.1016/j.molmet.2019.03.001 growth by lipid exchange and transfer at lipid droplet con- Joshi AS, Nebenfuehr B, Choudhary V, Satpute-Krishnan P, tact sites. J Cell Biol 195, 953–963. doi: 10.1083/ Levine TP, Golden A, Prinz WA (2018). Lipid droplet and jcb.201104142 peroxisome biogenesis occur at the same ER subdomains. Grippa A, Buxo L, Mora G, Funaya C, Idrissi F-Z, Nat Commun 9, 2940. doi: 10.1038/s41467-018-05277-3 Mancuso F, Gomez R, Muntanya` J, SabidoE, Ju¨ ngst C, Klein M, Zumbusch A (2013). Long-term live cell Carvalho P (2015). The seipin complex Fld1/Ldb16 microscopy studies of lipid droplet fusion dynamics in adi- stabilizes ER-lipid droplet contact sites. J Cell Biol pocytes. J Lipid Res 54, 3419–3429. doi: 10.1194/jlr. 211, 829–844. doi: 10.1083/jcb.201502070 M042515 14 Contact

Kassan A, Herms A, Ferna´ ndez-Vidal A, Bosch M, Schieber McFie PJ, Banman SL, Stone SJ (2018). Diacylglycerol NL, Reddy BJN, Fajardo A, Gelabert-Baldrich M, Tebar acyltransferase-2 contains a c-terminal sequence that inter- F, Enrich C, et al. (2013). Acyl-CoA synthetase 3 promotes acts with lipid droplets. Biochim Biophys Acta Mol Cell lipid droplet biogenesis in ER microdomains. J Cell Biol Biol Lipids 1863, 1068–1081. doi: 10.1016/j. 203, 985–1001. doi: 10.1083/jcb.201305142 bbalip.2018.06.008 Khandelia H, Duelund L, Pakkanen KI, Ipsen JH (2010). Mori E, Fujikura J, Noguchi M, Nakao K, Matsubara M, Triglyceride blisters in lipid bilayers: implications for lipid Sone M, Taura D, Kusakabe T, Ebihara K, Tanaka T, droplet biogenesis and the mobile lipid signal in cancer cell et al. (2016). Impaired adipogenic capacity in induced plu- membranes. PLoS One 5, e12811. doi: 10.1371/journal. ripotent stem cells from lipodystrophic patients with BSCL2 pone.0012811 mutations. Metab Clin Exp 65, 543–556. doi: 10.1016/j. Klemm RW, Norton JP, Cole RA, Li CS, Park SH, Crane metabol.2015.12.015 MM, Li L, Jin D, Boye-Doe A, Liu TY, et al. (2013). A Nishimura K, Fukagawa T, Takisawa H, Kakimoto T, conserved role for atlastin GTPases in regulating lipid drop- Kanemaki M (2009). An auxin-based degron system for let size. Cell Rep 3, 1465–1475. doi: 10.1016/j. the rapid depletion of proteins in nonplant cells. Nat celrep.2013.04.015 Methods 6, 917–922. doi: 10.1038/nmeth.1401 Kornke JM, Maniak M (2017). Fat-containing cells are elim- Olzmann JA, Carvalho P (2019). Dynamics and functions of inated during dictyostelium development. Biol Open, lipid droplets. Nat Rev Mol Cell Biol 20, 137–155. doi: 1–11. doi: 10.1242/bio.025478 10.1038/s41580-018-0085-z Kory N, Farese RV, Walther TC (2016). Targeting fat: mech- Paar M, Ju¨ ngst C, Steiner NA, Magnes C, Sinner F, Kolb D, anisms of protein localization to lipid droplets. Trends Cell Lass A, Zimmermann R, Zumbusch A, Kohlwein SD, et al. Biol 26, 535–546. doi: 10.1016/j.tcb.2016.02.007 (2012). Remodeling of lipid droplets during lipolysis and Kumar N, Leonzino M, Hancock-Cerutti W, Horenkamp FA, growth in adipocytes. J Biol Chem 287, 11164–11173. doi: Li P, Lees JA, Wheeler H, Reinisch KM, De Camilli P 10.1074/jbc.M111.316794 (2018). VPS13A and VPS13C are lipid transport proteins Pagac M, Cooper DE, Qi Y, Lukmantara IE, Mak HY, Wu Z, differentially localized at ER contact sites. J Cell Biol 217, Tian Y, Liu Z, Lei M, Du X, et al. (2016). SEIPIN regulates 3625–3639. doi: 10.1083/jcb.201807019 lipid droplet expansion and adipocyte development by mod- Li D, Zhao YG, Li D, Zhao H, Huang J, Miao G, Feng D, Liu ulating the activity of glycerol-3-phosphate acyltransferase. P, Li D, Zhang H (2019a). The ER-Localized protein Cell Rep 17, 1546–1559. doi: 10.1016/j.celrep.2016.10.037 DFCP1 modulates ER-lipid droplet contact formation. Papadopoulos C, Orso G, Mancuso G, Herholz M, Gumeni S, Cell Rep 27, 343.e5–358.e5. doi: 10.1016/j. Tadepalle N, Ju¨ ngst C, Tzschichholz A, Schauss A, Honing€ celrep.2019.03.025 S, et al. (2015). Spastin binds to lipid droplets and affects Li Q, Li Y, Zhang Z, Kang H, Zhang L, Zhang Y, Zhou L lipid metabolism. PLoS Genet 11, e1005149. doi: 10.1371/ (2019b). SEIPIN overexpression in the liver may alleviate journal.pgen.1005149 hepatic steatosis by influencing the intracellular calcium Payne VA, Grimsey N, Tuthill A, Virtue S, Gray SL, Nora level. Mol Cell Endocrinol 488, 70–78. doi: 10.1016/j. ED, Semple RK, Rahilly SO, Rochford JJ (2008). The mce.2019.03.005 human lipodystrophy gene BSCL2/seipin may be essential Li S, Prasanna X, Salo VT, Vattulainen I, Ikonen E (2019c). for normal adipocyte differentiation. Diabetes 57, An efficient auxin-inducible degron system with low basal 2055–2060. doi: 10.2337/db08-0184 degradation in human cells. Nat Methods 16, 866–869. doi: Pol A, Gross SP, Parton RG (2014). Review: biogenesis of the 10.1038/s41592-019-0512-x multifunctional lipid droplet: lipids, proteins, and sites. Liu L, Jiang Q, Wang X, Zhang Y, Lin RCY, Lam SM, Shui J Cell Biol 204, 635–646. doi: 10.1083/jcb.201311051 G, Zhou L, Li P, Wang Y, et al. (2014). Adipose-specific Poppelreuther M, Sander S, Minden F, Dietz MS, Exner T, Du knockout of seipin/Bscl2 results in progressive lipodystro- C, Zhang I, Ehehalt F, Knu¨ ppel L, Domschke S, et al. phy. Diabetes 63, 2320–2331. doi: 10.2337/db13-0729 (2018). The metabolic capacity of lipid droplet localized Lundin C, Nordstrom€ R, Wagner K, Windpassinger C, acyl-CoA synthetase 3 is not sufficient to support local tri- Andersson H, von Heijne G, Nilsson I (2006). Membrane glyceride synthesis independent of the endoplasmic reticu- topology of the human seipin protein. FEBS Lett 580, lum in A431 cells. Biochim Biophys Acta Mol Cell Biol 2281–2284. doi: 10.1016/j.febslet.2006.03.040 Lipids 1863, 614–624. doi: 10.1016/j.bbalip.2018.03.003 Magre J, Delepine M, Khallouf E, Gedde-Dahl T, Van Prevost C, Sharp ME, Kory N, Lin Q, Voth GA, Farese RV, Maldergem L, Sobel E, Papp J, Meier M, MegarbaneA, Walther TC (2018). Mechanism and determinants of amphi- Bachy A, et al. (2001). Identification of the gene altered in pathic helix-containing to lipid droplets. Berardinelli-Seip congenital lipodystrophy on chromosome Dev Cell 44, 73.e4–86.e4. doi: 10.1016/j.devcel.2017.12.011 11q13. Nat Genet 28, 365–370. doi: 10.1038/ng585 Pyc M, Cai Y, Gidda SK, Yurchenko O, Park S, Kretzschmar Mahamid J, Tegunov D, Maiser A, Arnold J, Leonhardt H, FK, Ischebeck T, Valerius O, Braus GH, Chapman KD, Plitzko JM, Baumeister W (2019). Liquid-crystalline phase et al. (2017). Arabidopsis lipid droplet-associated protein transitions in lipid droplets are related to cellular states and (LDAP) – interacting protein (LDIP) influences lipid drop- specific organelle association. Proc Natl Acad Sci 116, let size and neutral lipid homeostasis in both leaves and 201903642. doi: 10.1073/pnas.1903642116 seeds. Plant J 92, 1182–1201. doi: 10.1111/tpj.13754 Salo et al. 15

Renne MF, Klug YA, Carvalho P (2020). Lipid droplet bio- Slayton M, Gupta A, Balakrishnan B, Puri V (2019). CIDE genesis : a mystery “unmixing”? Semin Cell Dev Biol. doi: proteins in human health and disease. Cells 8, 238. doi: 10.1016/j.semcdb.2020.03.001 10.3390/cells8030238 Renvoise B, Malone B, Falgairolle M, Munasinghe J, Stadler J, Spandl J, White DJ, Peychl J, Thiele C (2009). Live cell multi- Sibilla C, Park SH, Blackstone C (2016). Reep1 null mice color imaging of lipid droplets with a new dye, LD540. reveal a converging role for hereditary spastic paraplegia Traffic 10, 1579–1584. doi: 10.1111/j.1600- proteins in lipid droplet regulation. Hum Mol Genet 25, 0854.2009.00980.x 5111–5125. doi: 10.1093/hmg/ddw315 Su W-C, Lin Y-H, Pagac M, Wang C-W (2019). Seipin nega- Romanauska A, Kohler€ A (2018). The inner nuclear mem- tively regulates sphingolipid production at the ER–LD con- brane is a metabolically active territory that generates tact site. J Cell Biol. jcb.201902072. doi: 10.1083/ nuclear lipid droplets. Cell 174, 700–715. doi: 10.1016/j. jcb.201902072 cell.2018.05.047 Sui X, Arlt H, Brock KP, Lai ZW, DiMaio F, Marks DS, Liao Rubio-Cabezas O, Puri V, Murano I, Saudek V, Semple RK, M, Farese RV, Walther TC (2018). Cryo-electron micros- Dash S, Hyden CSS, Bottomley W, Vigouroux C, MagreJ, copy structure of the lipid droplet-formation protein seipin. et al. (2009). Partial lipodystrophy and insulin resistant dia- J Cell Biol 217, 4080–4091. doi: 10.1083/jcb.201809067 betes in a patient with a homozygous nonsense mutation in Sui X, Wang K, Gluchowski NL, Elliott SD, Liao M, Walther CIDEC. EMBO Mol Med 1, 280–287. doi: 10.1002/ TC, Farese RV (2020). Structure and catalytic mechanism emmm.200900037 of a human triacylglycerol-synthesis enzyme. Nature 581, Salo VT, Belevich I, Li S, Karhinen L, Vihinen H, Vigouroux 323–328. doi: 10.1038/s41586-020-2289-6 C, Magre J, Thiele C, Holtt€ a-Vuori€ M, Jokitalo E, et al. Szymanski KM, Binns D, Bartz R, Grishin NV, Li W, Agarwal (2016). Seipin regulates ER–lipid droplet contacts and cargo AK, Garg A, Anderson RG, Goodman JM (2007). The delivery. EMBO J 35, 2699–2716. doi: 10.15252/ lipodystrophy protein seipin is found at endoplasmic retic- embj.201695170 ulum lipid droplet junctions and is important for droplet Salo VT, Ikonen E (2019). Moving out but keeping in touch: morphology. Proc Natl Acad Sci USA 104, 20890–20895. contacts between endoplasmic reticulum and lipid droplets. doi: 10.1073/pnas.0704154104 Talukder MMU, Sim MFM, O’Rahilly S, Edwardson JM, Curr Opin Cell Biol 57, 64–70. doi: 10.1016/j. Rochford JJ (2015). Seipin oligomers can interact directly ceb.2018.11.002 with AGPAT2 and lipin 1, physically scaffolding critical Salo VT, Li S, Vihinen H, Holtt€ a-Vuori€ M, Szkalisity A, regulators of adipogenesis. Mol Metab 4, 199–209. doi: Horvath P, Belevich I, Peranen€ J, Thiele C, Somerharju P, 10.1016/j.molmet.2014.12.013 et al (2019). Seipin facilitates triglyceride flow to lipid drop- Teixeira V, Johnsen L, Martınez-Montan~es F, Grippa A, Buxo let and counteracts droplet ripening via ER contact. Dev L, Idrissi FZ, Ejsing CS, Carvalho P (2018). Regulation of Cell 50, 1–16. doi: 10.1016/j.devcel.2019.05.016 lipid droplets by metabolically controlled ldo isoforms. Sandoz PA, Tremblay C, Gisou van der Goot F, Frechin M J Cell Biol 217, 127–138. doi: 10.1083/jcb.201704115 (2019). Image-based analysis of living mammalian cells Thiam AR, Farese RV, Walther TC (2013). The biophysics and using label-free 3D refractive index maps reveals new organ- of lipid droplets. Nat Rev Mol Cell Biol 14, elle dynamics and dry mass flux. PLoS Biol 17, e3000553. 775–786. doi: 10.1038/nrm3699 doi: 10.1371/journal.pbio.3000553 Thiam AR, Foreˆt L (2016). The physics of lipid droplet nucle- Santinho A, Salo VT, Chorlay A, Li S, Zhou X, Omrane M, ation, growth and budding. Biochim Biophys Acta 1861, Ikonen E, Thiam AR (2020). Membrane curvature catalyzes 715–722. doi: 10.1016/j.bbalip.2016.04.018 lipid droplet assembly. Curr Biol, 1–14. doi: 10.1016/j. Tian Y, Bi J, Shui G, Liu Z, Xiang Y, Liu Y, Wenk MR, Yang cub.2020.04.066 H, Huang X (2011). Tissue-autonomous function of dro- Sim MFM, Dennis RJ, Aubry EM, Ramanathan N, Sembongi sophila seipin in preventing ectopic lipid droplet formation. H, Saudek V, Ito D, O’Rahilly S, Siniossoglou S, Rochford PLoS Genet 7, e1001364. doi: 10.1371/journal. JJ (2012). The human lipodystrophy protein seipin is an ER pgen.1001364 membrane adaptor for the adipogenic PA phosphatase lipin Valm AM, Cohen S, Legant WR, Melunis J, Hershberg U, 1. Mol Metab 2, 38–46. doi: 10.1016/j.molmet.2012.11.002 Wait E, Cohen AR, Davidson MW, Betzig E, Lippincott- Sim MFM, Persiani E, Talukder MMU, Mcilroy GD, Schwartz J (2017). Applying systems-level spectral imaging Roumane A, Edwardson JM, Rochford JJ (2020). and analysis to reveal the organelle interactome. Nature Oligomers of the lipodystrophy protein seipin may co- 546, 162–167. doi: 10.1038/nature22369 ordinate GPAT3 and AGPAT2 enzymes to facilitate adipo- Victoria B, Cabezas-Agrıcola JM, Gonza´ lez-Mendez B, Lattanzi cyte differentiation. Sci Rep 10, 1–14. doi: 10.1038/s41598- G, Del Coco R, Loidi L, Barreiro F, Calvo C, Lado-Abeal J, 020-59982-5 Araujo-Vilar D (2010). Reduced adipogenic gene expression Sim MFM, Talukder MMU, Dennis RJ, O’Rahilly S, in fibroblasts from a patient with type 2 congenital general- Edwardson JM, Rochford JJ (2013). Analysis of naturally ized lipodystrophy. Diabet Med 27, 1178–1187. doi: 10.1111/ occurring mutations in the human lipodystrophy protein j.1464-5491.2010.03052.x seipin reveals multiple potential pathogenic mechanisms. Walther T, Chung J, Farese RV (2017). Lipid droplet biogen- Diabetologia 56, 2498–2506. doi: 10.1007/s00125-013- esis. Annu Rev Cell Dev Biol 33, 491–510. doi: 10.1146/ 3029-3 annurev-cellbio-100616-060608 16 Contact

Wang C-W, Miao Y-H, Chang Y-S (2014). Control of lipid nuclear envelope in yeast. Biochim Biophys Acta 1851, droplet size in budding yeast requires the collaboration 1450–1464. doi: 10.1016/j.bbalip.2015.08.003 between Fld1 and Ldb16. J Cell Sci 127, 1214–1228. doi: Xu D, Li Y, Wu L, Li Y, Zhao D, Yu J, Huang T, Ferguson C, 10.1242/jcs.137737 Parton RG, Yang H, Li P (2018). Rab18 promotes lipid Wang H, Becuwe M, Housden BE, Chitraju C, Porras AJ, droplet (LD) growth by tethering the ER to LDs through Graham MM, Liu XN, Thiam AR, Savage DB, Agarwal SNARE and NRZ interactions. J Cell Biol 217, 975–995. AK, et al. (2016). Seipin is required for converting nascent doi: 10.1083/jcb.201704184 to mature lipid droplets. Elife, 5, 133–150. doi: 10.7554/ Xu N, Zhang SO, Cole RA, McKinney SA, Guo F, Haas JT, eLife.16582 Bobba S, Farese Jr RV, Mak HY (2012). The FATP1- Wang L, Qian H, Nian Y, Han Y, Ren Z, Zhang H, Hu L, DGAT2 complex facilitates lipid droplet expansion at the Prasad BVV, Laganowsky A, Yan N, Zhou M (2020). ER-lipid droplet interface. J Cell Biol 198, 895–911. doi: Structure and catalytic mechanism of human diacylglycerol 10.1083/jcb.201201139 O-acyltransferase 1. Nature 581, 329–332. doi: 10.1038/ Yamashita A, Hayashi Y, Matsumoto N, Nemoto-Sasaki Y, s41586-020-2280-2 Oka S, Tanikawa T, Sugiura T (2014). Glycerophosphate/ Wang S, Idrissi FZ, Hermansson M, Grippa A, Ejsing CS, acylglycerophosphate acyltransferases. Biology (Basel) 3, Carvalho P (2018). Seipin and the membrane-shaping pro- 801–830. doi: 10.3390/biology3040801 Yan R, Qian H, Lukmantara I, Gao M, Du X, Yan NYH tein Pex30 cooperate in organelle budding from the endo- (2018). Human SEIPIN binds anionic phospholipids. Dev plasmic reticulum. Nat Commun 9, 1–12. doi: 10.1038/ Cell 47, 1–9. doi: 10.1016/j.devcel.2018.09.010 s41467-018-05278-2 Yang W, Thein S, Guo X, Xu F, Venkatesh B, Sugii S, Radda Weber-Boyvat M, Kentala H, Peranen€ J, Olkkonen VM GK, Han W (2013). Seipin differentially regulates lipogen- (2015). Ligand-dependent localization and function of esis and adipogenesis through a conserved core sequence ORP-VAP complexes at membrane contact sites. Cell Mol and an evolutionarily acquired C-terminus. Biochem J Life Sci 72, 1967–1987. doi: 10.1007/s00018-014-1786-x 452, 37–44. doi: 10.1042/bj20121870 Welte MA (2015). Expanding roles for lipid droplets. Curr Biol Yang W, Thein S, Wang X, Bi X, Ericksen RE, Xu F, Han W 25, R470–R481. doi: 10.1016/j.cub.2015.04.004 (2014). BSCL2/seipin regulates adipogenesis through actin Wilfling F, Wang H, Haas JT, Krahmer N, Gould TJ, Uchida cytoskeleton remodelling. Hum Mol Genet 23, € A, Cheng J-X, Graham M, Christiano R, Frohlich F, et al. 502–513. doi: 10.1093/hmg/ddt444 (2013). Triacylglycerol synthesis enzymes mediate lipid Zhang P, Reue K (2017). Lipin proteins and glycerolipid droplet growth by relocalizing from the ER to lipid drop- metabolism: roles at the ER membrane and beyond. lets. Dev Cell 24, 384–399. doi: 10.1016/j.devcel.2013.01.013 Biochim Biophys Acta Biomembr 1859, 1583–1595. doi: Windpassinger C, Auer-Grumbach M, Irobi J, Patel H, Petek 10.1016/j.bbamem.2017.04.007 € E, Horl G, Malli R, Reed JA, Dierick I, Verpoorten N, Zhou H, Lei X, Benson T, Mintz J, Xu X, Harris RB, et al. (2004). Heterozygous missense mutations in BSCL2 Weintraub NL, Wang X, Chen W (2015). Berardinelli- are associated with distal hereditary motor neuropathy and Seip congenital lipodystrophy 2 regulates adipocyte lipoly- silver syndrome. Nat Genet 36, 271–276. doi: 10.1038/ sis, browning, and energy balance in adult animals. J Lipid ng1313 Res 56, 1912–1925. doi: 10.1194/jlr.M060244 Wolinski H, Hofbauer HF, Hellauer K, Cristobal-Sarramian Zoni V, Khaddaj R, Campomanes P, Thiam R, Schneiter R, A, Kolb D, Radulovic M, Knittelfelder OL, Rechberger Vanni S (2020). Lipid droplet biogenesis is driven by liquid- GN, Kohlwein SD (2015). Seipin is involved in the regula- liquid phase separation. BioRxiv 1–30. doi: https://doi.org/ tion of phosphatidic acid metabolism at a subdomain of the 10.1101/777466