Abstract
The nuclear compartment is delimited by a specialized expanded sheet of the endoplasmic reticulum (ER) known as the nuclear envelope (NE). Compared to the outer nuclear membrane and the contiguous peripheral ER, the inner nuclear membrane (INM) houses a unique set of transmembrane proteins that serve a staggering range of functions. Many of these functions reflect the exceptional position of INM proteins at the membrane–chromatin interface. Recent research revealed that numerous INM proteins perform crucial roles in chromatin organization, regulation of gene expression, genome stability, and mediation of signaling pathways into the nucleus. Other INM proteins establish mechanical links between chromatin and the cytoskeleton, help NE remodeling, or contribute to the surveillance of NE integrity and homeostasis. As INM proteins continue to gain prominence, we review these advancements and give an overview on the functional versatility of the INM proteome.
During evolution, the nuclear envelope (NE) arose as a separating membrane sheath between the genetic material and the cytoplasm. This physical barrier became one of the hallmarks of eukaryotic cells and defined nuclear compartmentalization. The emergence of the NE did not only generate a protective cover of the genome but also spatially separated transcription and translation, allowing for new forms of their regulation. Over the course of a billion years, the NE got equipped with a set of membrane-embedded proteins that endowed it with diverse tasks, yielding a functionally versatile compartment interface.
One of the most prominent features of the NE is its asymmetry. The NE is built by two double lipid bilayers, an outer nuclear membrane (ONM) and an inner nuclear membrane (INM), which are compositionally distinct with respect to their membrane protein assortment although both membranes are interconnected at numerous sites. Compared to the ONM and the connected endoplasmic reticulum (ER), the INM is enriched in a unique collection of membrane proteins that maintain close associations with chromatin and a network of intermediate filament proteins called nuclear lamins.
Clearly, the molecular identity of the INM is reflective of its functional specialization. Early work had identified only a handful of INM proteins (Senior and Gerace 1988; Worman et al. 1988; Foisner and Gerace 1993; Manilal et al. 1996; Lin et al. 2000). Later, the mass spectrometric characterization of the NE proteome from different sources, including rat liver and muscle (Schirmer et al. 2003; Wilkie et al. 2011; Korfali et al. 2012), human leukocytes (Korfali et al. 2010), and mouse neuroblastoma cells (Dreger et al. 2001), identified nearly 1000 putative nuclear envelope transmembrane proteins (NETs). However, as the NE is interconnected with ER, a fraction of the identified putative NETs may not be NE-specific and represent membrane proteins of the peripheral ER. Therefore, protein localization studies are a decisive element of NET definition. Remarkably, many of these NETs showed a high level of tissue specificity, suggesting that the NE proteome is adapted to cell functionality. Only a small fraction of NETs has so far been confirmed to be enriched at the NE, and most of those are also residents of the INM. To date, we know roughly 35 integral membrane proteins that were found to localize at the INM of mammalian cells (Table 1). These include prominent, well-studied INM proteins like the lamin B receptor (LBR), the LAP2-Emerin-Man1 (LEM)-domain family members lamina-associated polypeptide 2 (LAP2), emerin, MAN1, and LEM2, the Sad1p, Unc-84 (SUN)-domain proteins SUN1 and SUN2, as well as LAP1 and NET5 (Table 1; Fig. 1). Various others are less well characterized (Table 1) and the biological significance of their INM localization remains unexplored.
Table 1.
Well-characterized (in bold) and other putative inner nuclear membrane (INM) proteins, listing their predicted molecular weight, the number of transmembrane segments, membrane orientation, and references pertaining to their localization at the INM
Gene name/NET designation | Protein name | Predicted molecular weight (kD) | Predicted transmembrane (TM) segments | Orientation | References for localization at the INM |
---|---|---|---|---|---|
LBR | Lamin B receptor (LBR) | 58 (observed), 70.7 (predicted) | 8 | Multipass | Worman et al. 1988 |
SUN1 | SUN domain-containing protein 1 (SUN1) | 90 | 1 | Single-pass, type II | Dreger et al. 2001 |
SUN2 | SUN domain-containing protein 2 (SUN2) | 80.3 | 1 | Single-pass, type II | Hodzic et al. 2004 |
TMPO | Lamina-associated polypeptide 2 (LAP2) | 50.6 (isoform β) | 1 | Single-pass, type II | Foisner and Gerace 1993 |
EMD | Emerin (EDMD)/LEMD5 | 28.9 | 1 | Single-pass, type II | Manilal et al. 1996 |
LEMD3/MAN1 | INM protein MAN1 (MAN1)/LEMD3 | 99.9 | 2 | Multipass | Lin et al. 2000 |
LEMD2 | LEM-domain-containing protein 2 (LEMD2) | 56.9 | 2 | Multipass | Brachner et al. 2005 |
TMEM201/NET5 | TM protein 201/SAMP1 | 43.3 | 4 | Multipass | Buch et al. 2009 |
TOR1AIP1/LAP1 | Torsin-1A-interacting protein 1 (TOR1AIP1)/Lamina-associated polypeptide 1 (LAP1) | 66.2 (isoform B) | 1 | Single-pass, type II | Senior and Gerace 1988 |
NRM | Nurim | 29.3 | 6 | Multipass | Rolls et al. 1999 |
LUMA/TMEM43 | TM protein 43 (TMEM43)/LUMA | 44.8 | 4 | Multipass | Dreger et al. 2001 |
TMEM120A/NET29 | TM protein 120A (TMEM120A)/TMPIT | 42.8 | 5 | Multipass | Malik et al. 2010 |
MOSPD3/NET30 | Motile sperm domain containing 3 (MOSPD3) | 25.5 | 2 | Multipass | Malik et al. 2010 |
SCARA5/NET33a | Scavenger receptor class A, member 5 (SCARA5) | 43.2 | 1 | Single-pass, type II | Malik et al. 2010 |
SLC39A14/NET34a | Metal cation symporter ZIP14 (ZIP14)/SLC39A14 | 52.8 | 7 | Multipass | Malik et al. 2010 |
MYORG/NET37 | Myogenesis-regulating glycosidase (MYORG)/KIAA1161 | 81 | 1 | Single-pass, type II | Malik et al. 2010 |
PLPP7/NET39 | Inactive phospholipid phosphatase 7 (PLPP7)/PPAPDC3/C9orf67 | 21.9 | 4 | Multipass | Liu et al. 2009; Malik et al. 2010 |
TM7SF2/NET47b | Delta (14)-sterol reductase TM7SF2/ANG1/DHCR14A | 46.4 | 7 | Multipass | Malik et al. 2010 |
DHRS7/NET50 | Dehydrogenase/reductase (SDR family) member 7 (DHRS7) | 38.2 | 1 | N.D. | Malik et al. 2010 |
ERG28/NET51b | Ergosterol biosynthetic protein 28 (ERG28)/C14orf1 | 15.8 | 4 | Multipass | Malik et al. 2010 |
APH1B/NET55b | Anterior pharynx defective 1B (APH1B) | 28.4 | 6 | Multipass | Malik et al. 2010 |
NCLN/NET59b | Nicalin (NCLN) | 62.9 | 2 | Multipass | Malik et al. 2010 |
NCEH1b | Neutral cholesterol ester hydrolase 1 (NCEH1)/AADACL1/KIAA1363 | 45.8 | 1 | Single-pass, type II | Korfali et al. 2010 |
MAGT1a | Magnesium transporter protein 1 (MAGT1)/IAG2 | 38 | 4 | Multipass | Korfali et al. 2010 |
METTL7Ab | Methyltransferase-like protein 7A (METTL7A) | 28.3 | 1 | N.D. | Korfali et al. 2010 |
TMEM41A | TM protein 41A (TMEM41A) | 29.7 | 5 | Multipass | Korfali et al. 2010 |
STT3Ab | Dolichyl-diphosphooligosaccharide—protein glycosyltransferase subunit STT3A(STT3-A)/ITM1/TMC | 80.5 | 12 | Multipass | Korfali et al. 2010 |
TMEM38Ab | Trimeric intracellular cation channel type A (TRIC-A)/TMEM38A | 33.6 | 7 | Multipass | Wilkie et al. 2011 |
LPCAT3b | Lysophospholipid acyltransferase 5 (LPCAT3)/MBOAT5/OACT5 | 56 | 9 | Multipass | Wilkie et al. 2011 |
NEMP1 | Nuclear envelope integral membrane protein 1 (NEMP1)/TMEM194/KIAA0286 | 50.6 | 5 | Multipass | Wilkie et al. 2011 |
TMEM214c | TM protein 214 (TMEM214) | 77 | 2 | Multipass | Wilkie et al. 2011 |
LRRC59b | Leucine-rich repeat-containing protein 59 (LRRC59)/p34 | 35 | 1 | Single-pass, type II | Blenski and Kehlenbach 2019 |
All the putative INM proteins listed here fulfilled two criteria: (1) resistance to prefixation extraction with triton X-100, and (2) localization of a fraction of the total population at the INM observed through superresolution microscopy. Please note that possibly all membrane proteins that are inserted in the endoplasmic reticulum (ER) and possess extraluminal domains <60 kDa can access the INM. However, this does not ensure their enrichment and functional relevance at the INM. The other predominant cellular localizations for the putative INM proteins are annotated in the footnotes. Please note that SUN3, SUN4 (Spag4), and SUN5 (Frohnert et al. 2011; Calvi et al. 2015; Pasch et al. 2015) are specifically expressed in testis where they enrich at the NE. However, their localization to the INM remains to be formally proven.
(N.D.) No data available.
aProteins also localize at the plasma membrane as observed for SCARA5 (Huang et al. 2010), SLC39A14 (Taylor et al. 2004; Tuschl et al. 2016), and MAGT1 (Zhou and Clapham 2009).
bProteins primarily localize at the ER as observed for SLC39A14 (Malik et al. 2010), TM7SF2 (Holmer et al. 1998), ERG28 (Malik et al. 2010), APH1B (Malik et al. 2010), NCLN (Dettmer et al. 2010), NCEH1 (Igarashi et al. 2010), MAGT1 (Cherepanova et al. 2014), METTL7A (McKinnon and Mellor 2017), STT3A (Braunger et al. 2018; Zhu et al. 2019), TMEM38A (Yazawa et al. 2007; Shrestha et al. 2020), LPCAT3 (Zhao et al. 2008), and LRRC 59 (Zhen et al. 2012).
cPredominantly localizes at the ER and the Golgi complex (Li et al. 2013).
Figure 1.
Schematic representation of well-characterized inner nuclear membrane (INM) proteins indicating their membrane topology, domain organization, and known interaction partners. INM proteins bind chromatin, chromatin-associated factors, and nuclear lamins as indicated. Certain INM proteins like lamin B receptor (LBR) and LAP2-Emerin-Man1 (LEM)-domain proteins bind transcription factors and thereby modulate gene expression. Additionally, some INM proteins bind to each other while others, like LBR, self-interact, altogether creating a complex network of interactions. At the INM, Sad1p, Unc-84 (SUN) domain proteins form trimers via their coiled-coil domains and interact with three KASH peptides of outer nuclear membrane (ONM)-resident Nesprins to form the LINC (linker of nucleoskeleton and cytoskeleton) complex.
Most transmembrane proteins of the INM are cotranslationally inserted into the ER membrane and assume either a type II or a polytopic topology, with their extended amino termini facing the cytosol (Fig. 1). From the ER, these membrane proteins distribute to the ONM and INM by diffusion, passing through narrow membrane-proximal openings of nuclear pore complexes (NPCs) (Soullam and Worman 1995; Ohba et al. 2004; Theerthagiri et al. 2010; Zuleger et al. 2011; Boni et al. 2015; Ungricht et al. 2015; Pawar et al. 2017). These peripheral channels prevent the passage of membrane proteins with extraluminal domains larger than 60 kDa. The accumulation of INM-destined proteins at the nuclear face of the NE is determined by their retention on nuclear lamins and/or chromatin (Soullam and Worman 1995; Zuleger et al. 2011; Boni et al. 2015; Ungricht et al. 2015; Pawar et al. 2017). Thus, in principle, ER proteins with small extraluminal domains can also partition to the INM (Soullam and Worman 1995; Zuleger et al. 2011; Boni et al. 2015; Ungricht et al. 2015; Pawar et al. 2017), but in contrast to bona fide INM proteins, they are not enriched at the NE. Notably, in dividing cells undergoing open mitosis, both INM and ER proteins disperse in the mitotic ER network. During mitotic exit, a nearly synchronous enrichment of INM proteins on chromatin drives the process of NE reformation during late anaphase and telophase, rapidly reestablishing the INM membrane territory, representing a second, postmitotic mode of targeting proteins to the INM (Mattaj 2004).
The INM can be viewed as a functionally multifaceted membrane domain (Fig. 2). INM proteins engage in an array of genome-regulatory functions ranging from spatial genome organization (Solovei et al. 2013), epigenetic silencing (Somech et al. 2005), DNA replication (Martins et al. 2003), DNA damage repair (Lei et al. 2012), and transcriptional regulation. They also play important roles in diverse signaling pathways (Berk et al. 2013), in lipid synthesis (Tsai et al. 2016), and mediate mechanobiological tasks, including nuclear anchorage, migration, and mechanotransduction (Lammerding et al. 2005; Rothballer and Kutay 2013; Cho et al. 2017; Lee and Burke 2018). Interestingly, many of these functions converge and are essential for differentiation and development. They are realized through an intricate spectrum of biochemical interactions of INM proteins with chromatin, the nuclear lamina, and regulatory factors (Fig. 2).
Figure 2.
The diverse functions of inner nuclear membrane (INM) proteins. (A) Many INM proteins tether chromatin to the nuclear envelope (NE) and facilitate repression of genomic elements. Some INM proteins assist processes such as splicing, DNA replication, or the DNA damage response. (B) Members of the LINC (linker of nucleoskeleton and cytoskeleton) complex have mechanical and structural roles, interlinking the nucleoskeleton and cytoskeleton and thereby acting as force transmission devices across the NE. LINC complexes perform a wide range of functions, including nuclear anchorage and migration, chromosome movements, and the maintenance of NE membrane spacing. Other INM proteins (e.g., emerin and SAMP1) may functionally collaborate with LINC complexes in mechanotransduction. (C) INM proteins interact with transcription factors to regulate gene expression, thereby contributing to diverse signaling pathways. (D) Certain INM proteins possess enzymatic functions. Lamin B receptors (LBRs) exhibit C14 sterol reductase activity that is used in cholesterol synthesis. Others, like LAP1 and LEM2, directly or indirectly regulate the activities of partner proteins such as Torsins and the ESCRT-III complex, respectively.
An appreciation of the functional complexity of INM proteome is imperative to achieve an integrated view of INM biology. This is of particular importance in light of the numerous links that exist between INM protein dysfunction and human diseases. The idea that INM proteins are physiologically relevant was first realized by the discovery that emerin, mutations of which cause X-linked Emery–Dreifuss muscular dystrophy, is located at the INM (Bione et al. 1994; Manilal et al. 1996). This placed an INM protein at the causative epicenter of a disease, which paved ways for the intensification of research directed toward the cellular functions of INM proteins and their implications in human health. Presently, over 15 syndromes have been linked to the dysfunction of INM proteins (Table 2), and the list of diseases continues to expand. Every new disease link reiterates the significance of the INM proteome and stimulates novel ways to perceive how human physiology is influenced by the cellular functions of INM proteins. In this review, we use the examples of well-characterized, widely expressed INM proteins to portray their broad functional repertoire, giving priority to mammalian representatives.
Table 2.
Human diseases (in bold) related to either mutations in genes encoding for inner nuclear membrane (INM) proteins or a misregulation of INM protein levels
Disease | Pathology/clinical features | Implicated INM protein | Associated mutations/defects | References |
---|---|---|---|---|
Pelger-Huët anomaly | Hyposegmentation and abnormal chromatin organization in granulocyte nuclei | Lamin B receptor (LBR) | P119L, P569R, R377X*, W436X*, L534X*, V11EfsX24, G382DfsX39, S167TfsX176, loss of splicing of exon 3, 11, 14, altered splicing of exon 13 | Hoffmann et al. 2002; Best et al. 2003; Clayton et al. 2010; Tsai et al. 2016 |
Greenberg skeletal dysplasia | Abnormal sterol metabolism, hydrops, ectopic calcification, and moth-eaten (HEM) skeletal dysplasia | LBR | N547D, R583Q, L534X, Y468TfsX475, V11EfsX24 | Waterham et al. 2003; Clayton et al. 2010; Tsai et al. 2016; Turner and Schlieker 2016 |
Pelger-Huët anomaly with mild skeletal anomaly | Bilobed neutrophil nuclei with mild skeletal dysplasia | LBR | I218DfsX19, R586H, R76X, N547S | Borovik et al. 2013; Sobreira et al. 2015 |
Reynolds syndrome | Primary biliary cirrhosis, cutaneous systemic sclerosis | LBR | R372C | Gaudy-Marqueste et al. 2010 |
Emery–Dreifuss muscular dystrophy (EDMD) or EDMD-related myopathies | Joint contracture, muscle wasting and weakness, cardiac diseases with conduction defects, and arrhythmia | Emerin | ∼100 mutations in emerin have been associated with X-linked EDMD, including M1V, S54F, Q133H, P183H, P183T, S143X, W200X, Q44X, Q86X, S171X, Q219X, Q228X, Y41X, S66X, S120X, W226X, Δ95–99, R207GfsX30, L84fsX7, D9GfsX24, F39SfsX17, R45KfsX16, F190YfsX19, R203AfsX34, R204PfsX7, S52QfsX9, L84PfsX7, Y85LfsX8, E11SfsX2, S49LfsX11, and others | Bione et al. 1994; Mora et al. 1997; Ellis et al. 1999; Morris and Manilal 1999; Astejada et al. 2007; Ben Yaou et al. 2007; Brown et al. 2011; Dai et al. 2019; Zhou et al. 2019 |
LUMA | E85K, I91V | Liang et al. 2011 | ||
MAN1 | G88V, R230T occur along with mutations in dysferlin, plectin, ryanodine receptor 3, ankyrin2, nesprin1, integrator complex subunit 1, or titin | Meinke et al. 2020 | ||
NET39 | M92K, R252P occur along with mutations in plectin, collagen, dysferlin, or integrator complex subunit 1 | Meinke et al. 2020 | ||
SUN1 | A230V, G76A, and W377C occur along with mutations in either emerin or lamin A/C: G68D, G338S, W377C | Meinke et al. 2014 | ||
SUN2 | A56P and V378I occur along with mutations in lamin A/C: R620C, E438D | Meinke et al. 2014 | ||
TMEM201/Samp1 | G15A, G18S, and G597S occur along with mutations in lamin A/C, nesprin1, or SUN1 | Meinke et al. 2020 | ||
TMEM38A | V247M and D260N occur along with mutations in plectin, collagen, dysferlin, or integrator complex subunit 1 | Meinke et al. 2020 | ||
TMEM214 | G236S and R179H occur along with mutations in nesprin3 or collagen | Meinke et al. 2020 | ||
Buschke–Ollendorff syndrome | Connective tissue nevi combined with osteopoikilosis | MAN1 | W621X, Y441X, R625X, L611X, S619X, W855X, R655X, and L638fs mutation at exon 12/intron 12 boundary causing defective splicing | Hellemans et al. 2006; Kobayashi et al. 2007; Zhang et al. 2009; Burger et al. 2010; Korekawa et al. 2012; Brodbeck et al. 2016 |
Isolated osteopoikilosis | Small and round spots of increased bone density that are mainly located in the epiphyseal regions of the tubular bones | MAN1 | E601X, Q153X, C1323A, R537X, L478X, R678X, and R735X | Hellemans et al. 2004, 2006; Couto et al. 2007; Mumm et al. 2007; Baasanjav et al. 2010 |
Isolated melorheostosis | “Dripping wax” appearance in the cortex of long bones, accompanied by abnormalities of adjacent soft tissues | MAN1 | L638X | Hellemans et al. 2006 |
Hutterite-type cataract (with or without arrhythmic cardiomyopathy) | Clouding of the lens of the eye; some patients have suffered sudden death presumably of arrhythmogenic origin | LEM2 | L13R | Boone et al. 2016 |
Severe dystonia with cerebellar atrophy | Progressive dystonia, severe contractures of the Achilles tendons, and feet deformations | LAP1 | E482A | Dorboz et al. 2014 |
Limb–girdle muscular dystrophy type 2 | Proximal and distal weakness and atrophy, rigid spine and contractures of the proximal and distal interphalangeal hand joints, cardiomyopathy, and respiratory involvement | LAP1 | L394P, E62DfsX25, P43fsX15 | Kayman-Kurekci et al. 2014; Ghaoui et al. 2016 |
Unnamed multisystemic disease | Psychomotor retardation, cataract, heart malformation, sensorineural deafness | LAP1 | R321X | Fichtman et al. 2019 |
Arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC5) | Cardiomyocyte apoptosis, lethal ventricular tachycardia, and fibro-fatty infiltration mainly in the right ventricle | LUMA | S358L | Stroud et al. 2018 |
Dilated cardiomyopathy | Increase in left ventricular systolic and diastolic diameter and decrease in ejection fraction | LAP2α | R690C | Taylor et al. 2005 |
Hutchinson–Gilford progeria syndrome | Premature aging disease: individuals exhibit low body weight, decreased joint mobility, scleroderma, and die around the age of 13 | SUN1 | Up-regulated levels of SUN1 in patients expressing mutant lamin A/C | Chen et al. 2012 |
Hypertrophic cardiomyopathy | Cardiac arrythmia, supraventricular extrasystoles | SUN2 | M50T, V378I occur along with a mutation in myosin binding protein C | Meinke et al. 2014 |
Mandibuloacral dysplasia type A | Growth retardation, craniofacial anomalies, mottled cutaneous pigmentation, skin rigidity, partial lipodystrophy, insulin resistance | SUN2 | Mislocalization of SUN2 due to mutation in LMNA | Camozzi et al. 2012 |
Cancers | Subtype: cervical, colon, colorectal, gastric, pancreatic cancers, myeloma, lymphoma, medulloblastoma, and others | LAP2α | Up-regulated levels of LAP2α | Reviewed in Brachner and Foisner (2014) |
Subtype: digestive tract cancers | LAP2β | Up-regulated levels of LAP2β | Kim et al. 2012 | |
Subtype: ovarian cancer | Emerin | Reduced expression of emerin | Capo-chichi et al. 2009 | |
Subtype: hepatocellular carcinoma and neoplasms | NET33 | Down-regulation of NET33 | Liu et al. 2018; Ulker et al. 2018 | |
Subtype: aggressive breast, ovarian and prostate cancers | AADACL1 | Up-regulated levels/activity of AADACL1 | Chiang et al. 2006; Chang et al. 2011 |
The table does not cover diseases associated with nuclear lamins and barrier to autointegration factor (BAF).
(X) Termination codon, (fs) frameshift, (*) the amino acids at which premature termination occurs were allocated using Uniprot.
LAMIN B RECEPTOR (LBR)
When radioactive lamins were incubated with erythrocyte NE membranes, B-type lamins bound a 58 kDa protein with high specificity (Worman et al. 1988). This protein, designated as the “lamin B receptor,” was the first integral INM protein to be identified and remains one of the most well-characterized proteins of the INM to date. Structurally, LBR is a polytopic membrane protein with a hydrophilic amino-terminal domain that protrudes into the nucleoplasm and is composed of a Tudor domain, a serine/arginine-rich (RS) hinge region, and a second globular domain. This is followed by a hydrophobic region containing eight membrane-spanning helices, and a short nucleoplasmic carboxy-terminal tail (Fig. 1). Functionally, LBR is a versatile protein. Whereas its extraluminal domain contributes to chromatin organization in the nuclear periphery, the transmembrane segments exhibit sterol reductase activity.
Early biochemical studies had revealed a striking enrichment of heterochromatin marks on chromatin pulled down by LBR (Makatsori et al. 2004). Later, a combination of histone tail peptide arrays, chromatin immunoprecipitation, and direct binding experiments indicated that LBR can directly recognize the heterochromatin mark H4K20me2 via its amino-terminal Tudor domain (Hirano et al. 2012). Besides nucleosomes, LBR engages in a multitude of other interactions with nuclear partners, including lamin B1 (Ye and Worman 1994), dsDNA (Ye and Worman 1994; Duband-Goulet and Courvalin 2000), and RNA (Chen et al. 2016), which all bind to the RS domain of LBR; HA95 (Martins et al. 2000) and the methylated DNA-binding protein MeCP2 (Guarda et al. 2009), as well as the heterochromatin organizer HP1 that associates with LBR's membrane-proximal globular domain (Ye et al. 1997; Lechner et al. 2005). The nucleoplasmic domain of LBR thereby constitutes a multifaceted molecular backbone for heterochromatin tethering to the NE. In addition, homo-oligomerization of LBR may even contribute to the compaction of heterochromatin at the nuclear periphery (Hirano et al. 2012).
The functional importance of LBR's interaction with chromatin is highlighted by studies on chromatin organization during mammalian development and differentiation. The spatial segregation of transcriptionally repressed heterochromatin to the nuclear periphery of differentiated mouse cells was shown to rely on two molecular assemblies, referred to as the A-type and B-type tethers (Solovei et al. 2013). LBR constitutes the molecular pillar of the B-type tether, whereas the A-type tether consists of lamins A/C and (an) unknown protein(s) of the INM. One of the two tethers is sufficient to maintain heterochromatin at the nuclear periphery. While developing tissues primarily rely on LBR for peripheral heterochromatin tethering, differentiated tissues seem to be more dependent on lamin A/C (Solovei et al. 2013). Remarkably, loss of both LBR and lamin A/C can lead to “nuclear inversion,” an atypical collapse of heterochromatin into the nuclear interior, which is physiologically relevant for light perception in the photoreceptor rod cells of nocturnal animals (Solovei et al. 2013).
Beyond tethering and compacting chromatin at the nuclear periphery, LBR also facilitates transcriptional repression. Myoblast transcriptome analyses, for example, revealed that LBR is involved in suppression of muscle-specific gene expression during early stages of differentiation (Solovei et al. 2013). Furthermore, LBR has been suggested to promote X-chromosome inactivation (XCI) during development in mammals. The association of LBR with the long noncoding RNA Xist contributes to the recruitment of the inactivated X chromosome to the nuclear periphery and was also suggested to ensure its transcriptional silencing (McHugh et al. 2015; Chen et al. 2016). However, in a recent X-chromosome-wide analysis of Xist-mediated silencing, loss of LBR had only a minor effect on silencing (Nesterova et al. 2019). Discrepancies in these observations may be linked to the different silencing assays, different times of Xist induction, as well as allelic versus nonallelic analyses of XCI used in these studies. Thus, although the LBR–Xist interaction might be necessary for positioning of the inactive X chromosome and/or to stabilize gene expression, the exact impact of LBR on XCI still remains to be determined.
LBR–chromatin association may also be relevant for cellular senescence. Initial studies established a link between a reduction of lamin B1 and cellular senescence, suggesting that the nuclear lamina undergoes profound changes as cells enter senescence (Shimi et al. 2011; Freund et al. 2012; Dreesen et al. 2013; Shah et al. 2013; Dou et al. 2015). More recent studies have revealed that LBR is also down-regulated during transition into senescence (Ivanov et al. 2013; Lenain et al. 2015; Lukášová et al. 2017; Arai et al. 2019; En et al. 2020). The cellular transition to senescence is associated with an extensive reorganization of chromatin and changes in gene expression. Induction of senescence through γ-irradiation of cancer cells significantly decreased the levels of LBR and caused relocation of centromeric heterochromatin from the nuclear periphery to the nuclear interior (Lukášová et al. 2017). Upon induction of senescence via replicative or oncogene-induced stress, heterochromatin-rich fragments bud off from nuclei associated with a down-regulation of lamin B1 and LBR (Ivanov et al. 2013; Dou et al. 2015). The significant decrease in the levels of both LBR and lamin B1 in all these studies may imply that the regulation of their levels during senescence is interrelated. Importantly, however, down-regulation of LBR and lamin B1 levels alone is insufficient to induce senescence phenotypes (Dreesen et al. 2013; Lukášová et al. 2017), indicating that reduction of lamin B1 and LBR is a hallmark and the consequence of senescence rather than its cause.
Last, LBR–chromatin interactions have also been implicated in postmitotic NE assembly. This is supported by early observations showing that immunodepletion of LBR reduces chromatin recruitment of the NE vesicles in vitro (Pyrpasopoulou et al. 1996) and that deletion of LBR slightly delays NE assembly in cultured mammalian cells (Anderson et al. 2009).
Compared to the plethora of studies on the chromatin-associated functions of the nucleoplasmic domain of LBR, its carboxy-terminal membrane domain, which shares extensive homology to the human C14 sterol reductase TM7SF2 (Holmer et al. 1998) (also called DHCR14 or SR-1), has remained neglected for a long time. Because most enzymes involved in the cholesterol biosynthesis pathway localize to the ER, such a function seemed unexpected for an INM protein (Schuler et al. 1994). However, human LBR complemented yeast C14 reductase mutants, which provided first evidence for the enzymatic activity of LBR (Silve et al. 1998; Prakash et al. 1999). Recent findings have now unraveled that the C14-sterol reductase activity of LBR is even essential for the viability of mammalian cells, which can explain its relevance to human physiology (Tsai et al. 2016). Two congenital diseases are associated with mutations in LBR: Pelger–Huët anomaly, an autosomal dominant disorder resulting in abnormal hypolobulation of granulocyte nuclei, and Greenberg skeletal dysplasia, an autosomal-recessive condition resulting in abnormal bone development, fetal hydrops, and the ultimate nonviability of the fetus (Turner and Schlieker 2016). Most of these mutations map to the enzymatic membrane domain of LBR and strongly perturb LBR's ability to engage in cholesterol synthesis, albeit through two different mechanisms. Whereas some mutations interfere with LBR's ability to bind the enzymatic cofactor NADPH, others lead to LBR degradation. Importantly, all investigated disease-causing mutants fail to complement the cholesterol auxotrophy imposed by LBR deficiency in cultured mammalian cells, thus establishing LBR as a major sterol reductase required for cholesterol synthesis (Tsai et al. 2016). Peculiarly, the sterol reductase domain of LBR also resembles a conserved domain of isoprenylcysteine carboxyl methyltransferases, which methylate the carboxyl group of prenylated cysteine as a last step of CaaX modification. Such a function for LBR has been speculated to aid the final modification steps of substrates such as prelamin A and/or lamin B (Li et al. 2015), but still awaits supporting evidence.
SUN DOMAIN PROTEINS
Members of the conserved family of SUN (Sad1p, Unc-84)-domain proteins localize at the INM and interact with KASH (Klarsicht/ANC-1/Syne-1 homology)-domain proteins of the ONM in the perinuclear space, forming NE-spanning LINC (linker of nucleoskeleton and cytoskeleton) complexes (Crisp et al. 2006). SUN domain proteins oligomerize into homotrimers with the help of their long intraluminal coiled-coil regions that also force the carboxy-terminal SUN domains into a trimeric arrangement (Sosa et al. 2012; Wang et al. 2012). The formation of LINC complexes relies on the tight intercalation of structurally extended KASH peptides at the SUN domain interfaces, building force-resistant coupling devices within the NE. The extraluminal domains of different SUN and KASH proteins, in turn, engage in a variety of interactions with nuclear components and the cytoskeleton, respectively, conferring the LINC complex with a diverse range of functions. These include nuclear anchorage, nuclear migration, insertion of NPCs or spindle pole bodies into the NE, the coupling of centrosomes to the nucleus, NE membrane spacing, and NE remodeling at the onset of mitosis (Malone et al. 1999; Starr et al. 2001; Crisp et al. 2006; Liu et al. 2007; Hiraoka and Dernburg 2009; Friederichs et al. 2011; Talamas and Hetzer 2011; Gundersen and Worman 2013; Cain et al. 2014; Guilluy et al. 2014; Turgay et al. 2014). As these processes have been covered in elaborate reviews (Starr and Fridolfsson 2010; Gundersen and Worman 2013; Rothballer and Kutay 2013; Rothballer et al. 2013; Burke and Stewart 2014; Chang et al. 2015; Burke 2018; Lee and Burke 2018), we will limit ourselves here to some prominent intranuclear functions of LINC complexes.
From yeast to humans, SUN domain proteins establish specialized links between the INM and chromatin. The best-defined role is the anchorage of telomeres. In Saccharomyces cerevisiae, the SUN domain protein Mps3 mediates telomere tethering to the NE by chromatin silencing factors like Sir4 (Bupp et al. 2007; Horigome et al. 2011) or telomerase subunits like Est1 (Antoniacci et al. 2007; Schober et al. 2009). The association between telomeres and the NE is thought to suppress telomeric transcription and protect telomeres from harmful recombination (Gartenberg 2009; Schober et al. 2009; Mekhail and Moazed 2010).
In addition, the association between telomeres and SUN proteins is of particular importance for meiosis. During meiotic prophase I, rapid prophase movements (RPMs) led by telomeres facilitate the alignment and pairing of homologous chromosomes to ensure their faithful partitioning. Clustering of telomeres requires the concerted effort of LINC complexes, the cytoskeleton, and telomere-associated proteins. In mammalian cells, a specialized LINC complex consisting of SUN1 and KASH5 is required to couple telomeres via cytoplasmic dynein to the microtubule system (Morimoto et al. 2012; Horn et al. 2013; Lee et al. 2015). Telomere association of SUN1 relies on a complex of the membrane-associated junction protein (MAJIN1) and the telomere repeat-binding proteins 1 and 2 (TERB1 and 2) (Shibuya et al. 2014, 2015), and is regulated by cyclin-dependent kinase 2 (CDK2) (Viera et al. 2015). Mice deficient in either SUN1 or KASH5 are infertile due to meiotic arrest, highlighting the physiological relevance of RPMs and subsequent meiotic bouquet formation (Ding et al. 2007; Horn et al. 2013).
Similarly, in Schizosaccharomyces pombe, the SUN and KASH domain proteins Sad1 and Kms1 promote the formation of the meiotic chromosome bouquet. They mediate RPMs through an association of Kms1 with cytoplasmic dynein and of Sad1 with the telomere bouquet proteins Bqt1-4 and Rap1 and Taz1, components of the shelterin complex (Chikashige et al. 2006, 2009; Hiraoka and Dernburg 2009). The function of LINC complexes in bouquet formation is also conserved in the budding yeast S. cerevisiae (Trelles-Sticken et al. 2000; Conrad et al. 2008), in which the telomere-led RPMs are actin dependent (Trelles-Sticken et al. 2005; Koszul et al. 2008), and in Caenorhabditis elegans (Penkner et al. 2007, 2009; Sato et al. 2009; Woglar and Jantsch 2014), with the difference that RPMs rely on the association of specialized chromosome pairing centers with SUN1 (MacQueen et al. 2005). The evolutionary conserved meiotic chromosome movements nicely illustrate how cytoskeletal forces are transmitted via LINC complexes across the NE, here to impart changes in intranuclear organization.
SUN domain proteins have also been prominently implicated in NE-associated DNA repair. The yeast SUN domain protein Mps3, for instance, assists the tethering of persistent DNA double-strand breaks (DSBs) to the NE, potentially providing an environment supportive for alternative repair pathways (Oza et al. 2009; Horigome et al. 2014). Also in higher organisms, several lines of evidence suggest a role for LINC complexes in the DNA damage response (DDR) (Aymard et al. 2017; Marnef et al. 2019). DDR is accomplished either through error-prone nonhomologous end joining (NHEJ) or by a more precise repair pathway involving homologous recombination (HR). Initial studies revealed that mouse SUN1 and SUN2 interact with the DNA-dependent protein kinase (DNAPK), which plays a role in NHEJ repair and thereby assists the process (Lei et al. 2012). A later study then demonstrated that human cells use the DNA repair factor 53BP1, SUN1/2, and dynamic microtubules to promote the mobility of both dysfunctional telomeres and DSBs, potentially to facilitate NHEJ of DSBs (Lottersberger et al. 2015). Whether the mobility of DSBs is facilitated via direct and specific interactions between 53BP1-bound DSBs and the LINC complex or is the result of LINC complex-mediated transduction of forces onto chromatin in an untargeted manner remain interesting possibilities to be explored. On the other hand, the C. elegans SUN domain protein, UNC-84 sequesters components of the NHEJ pathway at the NE while promoting the recruitment of Fanconi anemia nuclease (FAN-1) to the sites of DNA interstrand cross-links. Thereby, UNC-84 has been proposed to guide repair pathway choice through inactivation of NHEJ and promotion of FAN-1-mediated HR at chromosomal breaks (Lawrence et al. 2016).
A surge of recent work reaffirmed the significance of LINC complexes in coupling nuclear mechanics and chromatin-associated processes. First, shear stress applied to the mammalian cell surface was found to stretch chromatin regions and led to transcriptional up-regulation of a transgene inserted in a stretched region, perhaps due to stretch-enabled chromatin opening and binding of RNA polymerase (RNAP) II (Tajik et al. 2016). Importantly, both chromatin displacement and transgene expression were affected upon depletion of SUN proteins, supporting the requirement of SUN proteins in coupling mechanical signals to transcription. Second, LINC complexes were suggested to influence a mechanically induced blockade of adipogenesis in mesenchymal stem cells (MSCs) (Uzer et al. 2018). Here, depletion of SUN1 and SUN2 diminished the nuclear localization of β-catenin, which normally counteracts adipocytic commitment by down-regulation of adipogenic transcription factors (Sen et al. 2008). Finally, extreme mechanical stimulation imposed by a high frequency tensile strain in human MSCs causes a rapid phosphorylation and turnover of SUN2. This contributes to a decoupling of the cytoskeleton and the nucleus, thereby imparting protection from strain-induced DNA damage (Gilbert et al. 2019).
Interestingly, through its association with lamin A/C, human SUN2 is also known to contribute to the maintenance of the latent HIV provirus in a transcriptionally repressed chromatin domain at the NE, thereby promoting the latency of HIV infection (Sun et al. 2018). Yet, SUN proteins may not only modulate viral latency but also affect early steps of HIV infection. The mechanistic details are, however, not well understood (Donahue et al. 2016; Lahaye et al. 2016; Schaller et al. 2017; Luo et al. 2018). Likely, even to date, many other exciting roles of LINC complexes remain to be discovered.
LEM-DOMAIN PROTEINS
LEM-domain proteins, named after the founding family members LAP2, emerin, and MAN1, are defined by the presence of a bihelical motif called the LEM domain (Lin et al. 2000). This domain confers interaction with the widespread metazoan DNA-binding protein, the barrier to autointegration factor (BAF), which has been implicated in important processes such as gene regulation, chromatin condensation, and nuclear assembly (Margalit et al. 2007; Samwer et al. 2017). Whereas BAF and LEM domains are restricted to metazoans, some INM proteins in both lower and higher eukaryotes possess a structurally related helix-extension-helix fold (Heh) that confers direct DNA interaction.
Metazoan LEM-domain proteins strongly associate with the nuclear lamina (Sullivan et al. 1999; Östlund et al. 2006; Ulbert et al. 2006) and are organizers of a complex interaction network at the NE–lamina–chromatin interface. In this network, LEM-domain proteins functionally overlap in recruiting chromatin-modifying proteins such as the histone deacetylase HDAC3 (Somech et al. 2005; Demmerle et al. 2012), and transcriptional regulators such as germ cell-less (GCL) and Btf (Holaska et al. 2003; Haraguchi et al. 2004; Mansharamani and Wilson 2005) (see also Fig. 1). Thereby, they coordinate the peripheral immobilization and repression of genomic elements, and the inhibition of transcription (Nili et al. 2001; Brachner and Foisner 2011; Ho et al. 2013; Guilluy et al. 2014; Lee et al. 2017). However, individually, they also perform some specialized functions, which will be discussed in the following sections.
LAP2
LAP2 proteins comprise at least six distinct protein isoforms (α, β, γ, δ, ε, ζ) that are generated by alternative splicing in mammals (Dechat et al. 2000). The most abundant among them are LAP2α and LAP2β, both of which interact with lamins and chromatin. LAP2α diverges significantly from LAP2β and the other LAP2 isoforms, as it is a nucleoplasmic protein, whereas the others are membrane embedded. LAP2β, the longest membrane-bound isoform, possesses an extended nucleoplasmic domain composed of an amino-terminal LEM-like domain through which it interacts with DNA, followed by the LEM domain that interacts with BAF, a low complexity region encompassing a lamina-binding domain, a single transmembrane segment, and a short luminal domain (Fig. 1). Early studies demonstrated a role for LAP2β in postmitotic targeting of membranes to chromatin and postmitotic nuclear growth (Foisner and Gerace 1993; Yang et al. 1997; Gant et al. 1999; Anderson et al. 2009).
In interphase cells, Lap2β plays an important role in fundamental cellular processes such as transcription and DNA replication. First, LAP2β contributes to the formation of a repressive chromatin environment at the INM (Cutler et al. 2019). Both on its own and in complex with the transcriptional repressor GCL, it represses the transcriptional activity of the E2F-DP3 heterodimer (Nili et al. 2001). Transcriptional repression by LAP2β is further regulated by its interaction with epigenetic modifiers like the histone deacetylase 3 (HDAC3) (Somech et al. 2005). Importantly, the LAP2β-HDAC3 complex also binds the transcriptional repressor cKrox that recognizes GAGA-type DNA elements within lamina-associated chromatin domains, for instance those spanning the developmentally regulated gene loci IgH and Cyp3a in human cells, leading to their peripheral localization and transcriptional silencing (Zullo et al. 2012). Second, LAP2β has also been suggested to control DNA replication by direct association with the chromatin factor HA95 that in turn interacts with Cdc6, an essential component of the prereplication complex (preRC). Abolishing the HA95-LAP2β interaction induces proteasome-mediated degradation of Cdc6 and thereby inhibits the initiation of DNA replication (Martins et al. 2003).
In LAP2α, the transmembrane domain is replaced by a unique coiled-coil domain that confers A-type lamin-binding activity (Dechat et al. 1998; Vlcek et al. 1999). Notably, both LAP2α and the nucleoplasmic pool of lamin A/C associate with euchromatin (Gesson et al. 2016). Furthermore, LAP2α is known to stabilize the tumor suppressor retinoblastoma protein pRb by anchoring it to nucleoplasmic lamin A/C (Markiewicz et al. 2002; Dorner et al. 2006). Thereby LAP2α regulates several functions of pRb such as cell cycle control and terminal differentiation of adipose and muscle tissues (Hansen et al. 2004; Huh et al. 2004). Overexpression of LAP2α in pre-adipocytes promotes cell cycle exit and initiation of differentiation to adipocytes in vitro (Dorner et al. 2006). On the other hand, LAP2α loss impairs pRb function causing inefficient cell cycle arrest in mouse fibroblast cultures and hyperproliferation of epidermal and erythroid progenitor cells in vivo (Naetar et al. 2008).
But do the soluble and membrane-bound LAP2 isoforms have completely independent functions? A recent study has demonstrated that LAP2α and LAP2β use their common LEM-like domain to modulate the activity of GLI1 (Mirza et al. 2019), a zinc finger transcription factor that controls the hedgehog pathway during tumorigenesis (Oro et al. 1997). Acetylated GLI1 is anchored at the INM by LAP2β, creating an inactive but dynamic nuclear reserve of GLI1 at the NE. Nucleoplasmic LAP2α does not only compete for binding of GLI1 but also drives GLI1 activation together with its binding partner HDAC1, which converts GLI1 into the active, deacetylated form on chromatin. Thus, LAP2 proteins form an isoform-based nuclear chaperoning system that controls the balance between inactive GLI1 in the nuclear periphery and active GLI1 in the nucleoplasm, promoted by the release of GLI1 from LAP2β by aPKC to ensure maximal GLI activation. As LAP2 proteins were found to interact quite broadly with zinc-finger proteins via their LEM-like domain, this may suggest a novel and general nuclear scaffolding function toward zinc-finger transcription factors (Mirza et al. 2019).
EMERIN
Emerin is one of the best-studied INM proteins, as mutations in the EMD gene causes X-linked Emery–Dreifuss muscular dystrophy (EDMD) (Bione et al. 1994). It is a tail-anchored membrane protein possessing an amino-terminal LEM domain followed by a so-called regulator-binding domain (RBD), a lamin-binding region, and a second RBD overlapping with an adenomatous polyposis coli-like (APC-L) domain (Berk et al. 2013; Koch and Holaska 2014).
Emerin perfectly epitomizes the degree of biochemical complexity at the NE as it can interact with a plethora of partners and is involved in diverse cellular processes. First, it associates with factors involved in genome organization and regulation. These include epigenetic regulators, proteins involved in signaling, transcription, mRNA splicing, and of course BAF (Holaska and Wilson 2007; Wilson and Foisner 2010; Koch and Holaska 2014). Following the paradigm of formation of repressive chromatin at the NE, emerin epigenetically modulates gene expression by interacting with and regulating the enzymatic activity of HDAC3, which forms the catalytic subunit of nuclear corepressor complex (NCoR). Emerin-null fibroblasts exhibit epigenetic changes that strikingly resemble those of HDAC3 knockout (KO) cells. These include a global increase in H4K5 acetylation and decreased H3K27 and H3K9 trimethylation (Demmerle et al. 2012). Besides epigenetic modulation, emerin influences gene expression by scaffolding a variety of gene-regulatory partners at the INM through its RBDs and APC-L domain. Prominent among them are GCL (Holaska et al. 2003), Btf (Haraguchi et al. 2004), Lim domain only (Lmo7) (Holaska et al. 2006), β-catenin (Markiewicz et al. 2006), and the splicing factor YT521-B (Wilkinson et al. 2003). Through these interactions, emerin regulates fundamental processes such as cell cycle progression, apoptosis, myogenic differentiation, and mRNA splicing. Not surprisingly, due to its involvement in several gene regulatory networks, emerin also has an established role in signaling. A large number of genes regulated by the transforming growth factor β (TGF-β), Notch, JNK, MAPK, NF-κB, integrin, and IGF pathways, are misexpressed upon loss of emerin (Muchir et al. 2007; Koch and Holaska 2012; Berk et al. 2013). Interestingly, most of these signaling pathways regulate myogenic differentiation—an intriguing aspect given the involvement of emerin in EDMD (Massague et al. 1986; Bione et al. 1994; Polesskaya et al. 2003).
Second, emerin associates with a variety of structural components such as lamin A/C (Clements et al. 2000), the LINC complex (Mislow et al. 2002; Haque et al. 2010), actin (Holaska et al. 2004), myosin (Holaska and Wilson 2007), and spectrin (Holaska and Wilson 2007). Owing to these interactions, emerin is perfectly positioned to integrate mechanical impetuses at the INM. Emerin-null mouse embryonic fibroblasts display alterations in nuclear morphology, NE plasticity, and response to mechanical stimulation as well as an impaired viability under mechanical strain (Lammerding et al. 2005; Rowat et al. 2006). This suggests that emerin may help cells to adapt to mechanical load. Indeed, upon mechanical stimulation, emerin gets phosphorylated at tyrosine residues 74 and 95. This strengthens the interaction between the LINC complex and lamin A/C, and initiates actin bundle formation at the nuclear periphery, which may provide structural rigidity to the nucleus (Guilluy et al. 2014). Additionally, in response to the mechanically induced increase in actin polymerization, emerin regulates nuclear accumulation and activity of mechanosensitive transcription factors such as MLK1-SRF, and thus stimulates mechanosensitive gene expression (Olson and Nordheim 2010; Ho et al. 2013; Willer and Carroll 2017). Interestingly, the function of emerin as a mechanosensor also impinges on the positioning of chromosome territories. Growing cells on softer matrices leads to phosphorylation of emerin at tyrosine residue 99, a spatial repositioning of chromatin domains to the nuclear interior and transcriptional deregulation of associated genes, albeit to different extents (Pradhan et al. 2018).
Further investigations into how perturbation of signaling pathways and mechanosensing influence muscle regeneration and cardiac conduction may prove beneficial for the development of EDMD treatments.
MAN1
MAN1/LEMD3 is the longest of all LEM family members. It is anchored at the INM by two transmembrane segments encompassing a 130-residues-long protein segment that is located in the perinuclear space. The LEM domain of MAN1 resides at the amino terminus of its first, long nucleoplasmic domain, whereas the shorter carboxy-terminal nucleoplasmic domain comprises a DNA-binding winged-helix domain, also known as the Man1-Src1p carboxy-terminal (MSC) domain, and a carboxy-terminal RNA recognition motif (RRM) (Lin et al. 2000; Caputo et al. 2006).
MAN1 has been shown to interact with lamin A/C, BAF, R-SMADs, and PPM1A (Smad2/3 phosphatase). Much of the cellular function of MAN1 is attributed to the interaction of its RRM with R-SMADs (Osada et al. 2003; Raju et al. 2003; Hellemans et al. 2004; Lin et al. 2005; Pan et al. 2005; Cohen et al. 2007). R-SMAD proteins are key regulators of multiple signaling pathway. Two types of R-SMADs exist in mammals: TGF-β-responsive (SMAD2 and SMAD3) and bone morphogenic protein (BMP)-responsive (SMAD1, SMAD5, and SMAD8) Smads. Interaction of the carboxyl terminus of MAN1 with SMAD2/3 sequesters them at the NE and competes for their binding to transcriptional activator complexes, thereby regulating expression of TGF-β target genes. Indeed, MAN1 has been shown to antagonize TGF-β signaling in mammalian cells (Lin et al. 2005; Pan et al. 2005; Chambers et al. 2018) and mice embryos (Ishimura et al. 2006; Cohen et al. 2007). Inactivation of MAN1 in mouse embryos is associated with increased transcriptional activity of SMAD2/3 and an increased expression of TGF-β, causing perturbations in vascular remodeling and angiogenesis, which leads to embryonic lethality (Ishimura et al. 2006; Cohen et al. 2007). Similarly, MAN1 also negatively regulates the BMP signaling pathway via interactions with SMADs 1, 5, and 8. In Drosophila, MAN1-mediated inhibition of the BMP pathway has been shown to ensure proper synaptic growth and the integrity of neuromuscular junctions, which is required for proper locomotor activity (Wagner et al. 2010; Laugks et al. 2017).
Notably, MAN1 has also been found to activate the promoter of BMAL1, which is one of the small numbers of “clock genes” that are responsible for generating the internal circadian rhythm. Consistently, depletion of MAN1 resulted in a prolonged circadian period, whereas its overexpression led to a reduced period length in mammalian cells and Drosophila (Lin et al. 2014). This study brings to light two interesting findings: first, it presents the possibility that INM proteins can also affect gene transcription in an activating manner. Second, it reveals an unexpected function of an INM protein in determining the circadian rhythm.
Beyond transcriptional control, MAN1 also influences the dynamic organization of the NE during the cell cycle in various organisms. In the fission yeast Schizosaccharomyces japonicus, the NE partially ruptures during late anaphase and reseals following mitotic exit. During anaphase, NPCs redistribute toward the spindle poles where they cosegregate with chromatin prior to NE breakage. Here, Man1 has been implicated in connecting segregating chromatin to NPCs to ensure equal partitioning of the nucleus and NPCs between daughters. Cells lacking Man1 exhibit a failure in anaphase NPC distribution, evident by an irregular clustering of NPCs between segregated chromosomes in daughter nuclei (Yam et al. 2013). In S. pombe, Man1 collaborates with Lem2 to perform some key functions of the missing nuclear lamina, including the maintenance of NE structure and stability as well as the anchorage of telomeres at the nuclear periphery (Gonzalez et al. 2012). Finally, MAN1 functionally cooperates with other LEM-domain proteins also in C. elegans, where Ce-MAN1 and Ce-emerin ensure proper chromosome segregation and cell division (Liu et al. 2003).
LEM2
Similar to MAN1, LEM2 is anchored in the INM by two transmembrane segments and possesses two nucleoplasmic domains (Fig. 1), which comprise an amino-terminal LEM domain and a winged-helix MSC domain, respectively. Both domains have been proposed to mediate interactions with DNA and/or chromatin. The LEM2 homolog of fission yeast is part of a network of silencing factors at the nuclear periphery that collectively ensure perinuclear heterochromatin repression (Banday et al. 2016; Barrales et al. 2016). S. pombe Lem2 mediates centromere tethering via its LEM domain, whereas heterochromatin silencing and the anchorage of telomeres requires its MSC domain. For transcriptional silencing, Lem2 cooperates with other factors, such as the RNAi machinery and the telomere-associated protein Taz1 (Barrales et al. 2016). Additionally, Lem2, via its MSC domain, promotes the binding of the Snf2-like/HDAC repressor complex (SHREC) to chromatin, which in turn suppresses the recruitment of the anti-silencing factor Epe1 (Banday et al. 2016; Barrales et al. 2016). This function seems key to the role of Lem2 in silencing of telomeres, as deletion of epe1+ completely suppresses the respective defect of a lem2Δ mutant. Lem2 has also been implicated in the maintenance of genome stability in S. cerevisiae, where the Lem2 homolog Heh1 along with its interacting partner Nur1 forms the “chromosome linkage INM protein” (CLIP) complex that physically links rDNA repeats to the nuclear periphery (Mekhail et al. 2008). Deletion of either heh1 or nur1 causes release of rDNA repeats from the NE and leads to chromosome instability by promoting aberrant recombination events in the rDNA repeats.
A suite of recent studies in both yeast and mammalian cells have placed LEM2 into a new spotlight for its role in the recruitment of the ESCRT-III complex to the NE. Here, ESCRT-III supports the constriction of tubular membrane structures culminating in membrane fission at different occasions, in line with the general role of ESCRT-III in membrane scission events (Henne et al. 2013; McCullough et al. 2018). NE-associated functions of ESCRT-III include the surveillance of NPC assembly (Webster et al. 2014), postmitotic NE closure (Olmos et al. 2015; Vietri et al. 2015), repair of NE ruptures (Denais et al. 2016; Raab et al. 2016), and the remodeling of heterochromatin-INM contacts (Pieper et al. 2020). Recruitment of the ESCRT-III complex to its site of action depends on specific adaptor proteins, and LEM2 has emerged as one player that promotes the recruitment of ESCRT-III to a specific site at the NE.
The role of LEM2/Lem2 in directing the ESCRT-III complex to the NE initially emerged from studies on NPC assembly in yeast (Webster et al. 2014). The inhibition of NPC assembly in S. cerevisiae leads to formation of an NE subdomain named “storage of improperly assembled NPCs” (SINC) compartment, which is enriched in NPCs that may represent improper assembly intermediates. Heh1, the yeast homolog of LEM2, recruits Chm7 (the CHMP7 homolog) to these sites and promotes SINC formation (Webster et al. 2016). Recent work has now even revealed that the Heh1-Chm7 axis does not only support the surveillance of NPC formation but also the sealing of NE ruptures. Heh1 and Chm7 are usually physically separated on opposite sides of the nuclear border. However, damage of the NE leads to the local encounter of Heh1 and Chm7 at the site of perturbation. Heh1 then activates the membrane shaping function of Chm7, leading to rapid repair of the nuclear border (Thaller et al. 2019). However, there seems to be an additional backup mechanism for the repair of NE ruptures in cultured mammalian cells where repair of laser-induced NE ruptures can occur independently of Chmp7 but requires recruitment of LEM-domain proteins including LEM2 to chromatin through BAF (Halfmann et al. 2019).
LEM2 does not only cooperate with ESCRT-III in interphase cells, but also during mitosis. In mammalian cells, postmitotic NE sealing depends on ESCRT-III, which is recruited to sites where spindle microtubules penetrate the reforming NE during anaphase (Olmos et al. 2015; Vietri et al. 2015). ESCRT-III recruitment is mediated by its component CHMP7, which interacts with the MSC domain of LEM2 (Gu et al. 2017). Initially, LEM2 tethers membranes to postmitotic chromatin disks by binding of its LEM domain to BAF. At the same time, a low-complexity domain within LEM2 undergoes liquid–liquid phase separation to coat bundles of spindle microtubules that need to be cleared. Finally, the MSC domain of LEM2 activates CHMP7. These two proteins copolymerize around microtubule bundles to form a molecular O-ring that initiates recruitment of spastin and ESCRT-III to severe the microtubules and promote nuclear compartmentalization, respectively (von Appen et al. 2020).
The contribution of LEM2 in establishing nuclear compartmentalization is also evident in S. japonicus that undergoes semi-open mitosis during which the NE is broken and then resealed at a single site. For NE sealing, the intersecting spindle becomes tightly enwrapped by the nuclear membrane, and both Lem2 and its interacting partner Nur1 are enriched at these sites called NE “tails.” However, the accumulation of LEM2 at these “tails” demands the release of a pool of LEM2 from their interaction with pericentromeric and telomeric heterochromatin. This is mediated by the ESCRT-III-associated AAA+-ATPase Vps4, which ensures that the association between LEM2 and peripheral heterochromatin is transient and subject to continuous remodeling. In the absence of Vps4, the interactions between Lem2 and heterochromatin are locked, causing a defect in bulk release of chromosomes from the NE at mitotic entry and a failure in the reestablishment of nuclear compartmentalization during mitotic exit (Yam et al. 2011; Pieper et al. 2020). Taken together, LEM2/Lem2 safeguards the integrity of the cell nucleus in response to numerous insults that are associated with a lack of proper nuclear compartmentalization.
LEM2 might also protect cells from DNA damage by interacting with several components of the nucleotide excision repair (NER) machinery. Indeed, cells depleted of LEM2 show increased sensitivity to UV-induced DNA damage and increased phosphorylated γ-H2AX protein levels, indicating an impaired or delayed DNA damage response (Moser et al. 2020). Furthermore, LEM2 plays a role in regulating MAP kinase and AKT signaling pathway during embryonic development in mice (Tapia et al. 2015) and acts together with emerin to regulate the ERK signaling during myoblast differentiation (Huber et al. 2009). And, last but not least, S. pombe Lem2 was suggested to restrict changes in nuclear size imposed by alterations in membrane synthesis and nucleocytoplasmic transport, thereby contributing to the maintenance of a constant nucleus to cell volume ratio, perhaps by acting as a barrier to membrane flow into and out of the NE (Kume et al. 2019).
NET5 ALIAS SAMP1
SAMP1 is a polytopic membrane protein with four central transmembrane segments that is well conserved from yeast to human. The amino-terminal nucleoplasmic domain of SAMP1 contains two conserved zinc-finger domains that are required for NE localization (Gudise et al. 2011). From a functional perspective, the zinc-fingers might be used for chromatin interaction. In line with this, Ima1, the SAMP1 homolog in S. pombe, attaches centromeric heterochromatin to the nuclear periphery (King et al. 2008). Likewise, human SAMP1 has been suggested to contribute to the organization of peripheral heterochromatin in U2OS cells (Bergqvist et al. 2019) and its overexpression was shown to enhance the peripheral localization of chromosome 5 (Zuleger et al. 2013).
Originally, mammalian SAMP1 had been linked to mitotic functions (Buch et al. 2009; Larsson et al. 2018). During mitosis, it localizes to membranes in the vicinity of the spindle poles. The depletion of SAMP1 was associated with some spindle defects, prolonged mitosis, and chromosome mis-segregation. Because SAMP1 interacts with γ-tubulin and HAUS6, a subunit of the augmin complex involved in microtubule nucleation, it has been suggested that SAMP1 may promote the recruitment of the augmin complex and γ-tubulin to the mitotic spindle (Larsson et al. 2018).
Other interaction partners of SAMP1 include the LINC complex, emerin, and lamin A/C (Gudise et al. 2011). The association of SAMP1 with the LINC complex plays an important role in nuclear movement during fibroblast polarization and centrosome positioning (Gudise et al. 2011; Borrego-Pinto et al. 2012). Similar to the defects observed upon depletion of lamin A and emerin, knockdown of SAMP1 perturbs the differentiation of myoblasts (Jafferali et al. 2017; Le Thanh et al. 2017). Supporting the role of SAMP1 in early differentiation, recent studies have revealed that SAMP1 levels increase in differentiating induced pluripotent stem cells (iPSCs) concomitantly with those of lamin A/C and ectopic expression of SAMP1 induces a rapid differentiation of iPSCs even under pluripotent culturing conditions. Whereas the idea that an INM protein can drive differentiation of iPSCs is interesting, mechanistic insights into how Samp1 may regulate this process are lacking and require further investigation (Bergqvist et al. 2017).
LAP1
LAP1/TOR1AIP1 is a lamina-binding type II membrane protein that is restricted to metazoans. It exists in three isoforms (A, B, C) in rodents (Foisner and Gerace 1993), of which only two, LAP1B and LAP1C, are expressed in human cells (Santos et al. 2014). Whereas LAP1 is a bona fide component of the INM, its paralog LULL1 is distributed in the peripheral ER. Both possess a well-conserved luminal domain that adopts a RecA-like fold and is used for the activation of Torsin AAA+ ATPase family members, first and foremost Torsin-1A and Torsin-1B (Goodchild and Dauer 2005; Jungwirth et al. 2010; Kim et al. 2010; Zhao et al. 2013; Brown et al. 2014; Sosa et al. 2014). Torsins reside in the perinuclear space and the ER lumen; however, their precise molecular function remains elusive to date. Torsin activation by LAP1 and LULL1 relies on an active site complementation mechanism in which a critical arginine finger complements the active site of Torsins (Brown et al. 2014; Sosa et al. 2014; Rose et al. 2015). Interestingly, Torsin-1A, the predominant Torsin isoform in the brain, is prominently associated with early onset torsion dystonia (DYT1), a severe movement disorder (Ozelius et al. 1997). Similarly, mutations in the TOR1AIP1 gene have been linked to primary dystonia (Table 2; Dorboz et al. 2014; Rebelo et al. 2015), and the ablation of Lap1 phenocopies both the nuclear blebbing and perinatal lethality observed in Tor1A KO mice (Kim et al. 2010).
In comparison, the nucleoplasmic domain of LAP1, which interacts with lamins A/C and B1 (Foisner and Gerace 1993; Senior and Gerace 1988) and emerin (Shin et al. 2013), has remained less well studied. Functional investigations have suggested that LAP1 collaborates with emerin in the maintenance of skeletal muscle cells, supported by the observation that combined loss of LAP1 and emerin causes a significantly more pronounced myopathy than that observed upon loss of the individual proteins (Shin et al. 2013). Some recent work uncovered that the nucleoplasmic domain of LAP1 also directly interacts with chromatin, similar to other INM proteins. Surprisingly, however, it was observed that LAP1 cannot be released from chromatin during mitosis if Torsin functionality is compromised or LAP1 is overexpressed, resulting in chromosome segregation defects and binucleation (Luithle et al. 2020), revealing an unexpected function of Torsins in the ER lumen in modulating chromatin association of LAP1 in the nucleoplasm.
Future work aimed at characterizing the molecular function of LAP1, LULL1, and the enigmatic Torsin family of proteins will hopefully allow rationalizing the cellular and organismal phenotypes associated with their dysfunction.
CONCLUDING REMARKS
In the past years, a wealth of research has pushed our perception of the NE from a mere shield for the genome to a dynamic biochemical factory that supports a multitude of essential cellular processes. Several converging themes emerge from the functional characterization of INM proteins. Principally, most of the INM proteins can be appreciated for their association with chromatin and for their role in the establishment of a repressive environment for gene expression. Yet, we still miss the full picture of the mechanisms governing the formation, organization, and functionality of the peripheral heterochromatin vis-à-vis INM proteins. That being said, there have been many fresh insights into important roles of INM proteins like lipid synthesis, mechanosensing, and functional collaborations with the ESCRT-III and Torsin machineries. Understanding the molecular principles governing these functions remains an exciting challenge for the future. Finally, a large number of INM proteins, especially those expressed in a tissue-specific manner, remain uncharacterized. This demands attention in light of nuclear envelopathies, many of which are characterized by tissue-specific defects. We emphasize that convergence of fundamental research driven toward unraveling the mechanistic basis of INM protein functions with translational research directed toward understanding disease etiology will provide a great promise toward developing future therapeutic strategies.
ACKNOWLEDGMENTS
We thank Dr. Madhav Jagannathan, Jelmi uit de Bos, and Renard Lewis for comments on the manuscript, and the Swiss National Science Foundation (SNSF, Grant 310030_184801) for financial support. We apologize for not being able to cite all original publications.
Footnotes
Editors: Ana Pombo, Martin W. Hetzer, and Tom Misteli
Additional Perspectives on The Nucleus available at www.cshperspectives.org
REFERENCES
- Anderson DJ, Vargas JD, Hsiao JP, Hetzer MW. 2009. Recruitment of functionally distinct membrane proteins to chromatin mediates nuclear envelope formation in vivo. J Cell Biol 186: 183–191. 10.1083/jcb.200901106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Antoniacci LM, Kenna MA, Skibbens RV. 2007. The nuclear envelope and spindle pole body-associated Mps3 protein bind telomere regulators and function in telomere clustering. Cell Cycle 6: 75–79. 10.4161/cc.6.1.3647 [DOI] [PubMed] [Google Scholar]
- Arai R, En A, Takauji Y, Maki K, Miki K, Fujii M, Ayusawa D. 2019. Lamin B receptor (LBR) is involved in the induction of cellular senescence in human cells. Mech Ageing Dev 178: 25–32. 10.1016/j.mad.2019.01.001 [DOI] [PubMed] [Google Scholar]
- Astejada MN, Goto K, Nagano A, Ura S, Noguchi S, Nonaka I, Nishino I, Hayashi YK. 2007. Emerinopathy and laminopathy clinical, pathological and molecular features of muscular dystrophy with nuclear envelopathy in Japan. Acta Myol 26: 159–164. [PMC free article] [PubMed] [Google Scholar]
- Aymard F, Aguirrebengoa M, Guillou E, Javierre BM, Bugler B, Arnould C, Rocher V, Iacovoni JS, Biernacka A, Skrzypczak M, et al. 2017. Genome-wide mapping of long-range contacts unveils clustering of DNA double-strand breaks at damaged active genes. Nat Struct Mol Biol 24: 353–361. 10.1038/nsmb.3387 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baasanjav S, Jamsheer A, Kolanczyk M, Horn D, Latos T, Hoffmann K, Latos-Bielenska A, Mundlos S. 2010. Osteopoikilosis and multiple exostoses caused by novel mutations in LEMD3 and EXT1 genes respectively—coincidence within one family. BMC Med Genet 11: 110. 10.1186/1471-2350-11-110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banday S, Farooq Z, Rashid R, Abdullah E, Altaf M. 2016. Role of inner nuclear membrane protein complex Lem2-Nur1 in heterochromatic gene silencing. J Biol Chem 291: 20021–20029. 10.1074/jbc.M116.743211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrales RR, Forn M, Georgescu PR, Sarkadi Z, Braun S. 2016. Control of heterochromatin localization and silencing by the nuclear membrane protein Lem2. Genes Dev 30: 133–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ben Yaou R, Toutain A, Arimura T, Demay L, Massart C, Peccate C, Muchir A, Llense S, Deburgrave N, Leturcq F, et al. 2007. Multitissular involvement in a family with LMNA and EMD mutations: role of digenic mechanism? Neurology 68: 1883–1894. 10.1212/01.wnl.0000263138.57257.6a [DOI] [PubMed] [Google Scholar]
- Bergqvist C, Jafferali MH, Gudise S, Markus R, Hallberg E. 2017. An inner nuclear membrane protein induces rapid differentiation of human induced pluripotent stem cells. Stem Cell Res 23: 33–38. 10.1016/j.scr.2017.06.008 [DOI] [PubMed] [Google Scholar]
- Bergqvist C, Niss F, Figueroa RA, Beckman M, Maksel D, Jafferali MH, Kulyté A, Ström AL, Hallberg E. 2019. Monitoring of chromatin organization in live cells by FRIC. Effects of the inner nuclear membrane protein Samp1. Nucleic Acids Res 47: e49. 10.1093/nar/gkz123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berk JM, Tifft KE, Wilson KL. 2013. The nuclear envelope LEM-domain protein emerin. Nucleus 4: 298–314. 10.4161/nucl.25751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Best S, Salvati F, Kallo J, Garner C, Height S, Thein SL, Rees DC. 2003. Lamin B-receptor mutations in Pelger-Huët anomaly. Br J Haematol 123: 542–544. 10.1046/j.1365-2141.2003.04621.x [DOI] [PubMed] [Google Scholar]
- Bione S, Maestrini E, Rivella S, Mancini M, Regis S, Romeo G, Toniolo D. 1994. Identification of a novel X-linked gene responsible for Emery–Dreifuss muscular dystrophy. Nat Genet 8: 323–327. 10.1038/ng1294-323 [DOI] [PubMed] [Google Scholar]
- Blenski M, Kehlenbach RH. 2019. Targeting of LRRC59 to the endoplasmic reticulum and the inner nuclear membrane. Int J Mol Sci 20: 334. 10.3390/ijms20020334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boni A, Politi AZ, Strnad P, Xiang W, Hossain MJ, Ellenberg J. 2015. Live imaging and modeling of inner nuclear membrane targeting reveals its molecular requirements in mammalian cells. J Cell Biol 209: 705–720. 10.1083/jcb.201409133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boone PM, Yuan B, Gu S, Ma Z, Gambin T, Gonzaga-Jauregui C, Jain M, Murdock TJ, White JJ, Jhangiani SN, et al. 2016. Hutterite-type cataract maps to chromosome 6p21.32-p21.31, cosegregates with a homozygous mutation in LEMD2, and is associated with sudden cardiac death. Mol Genet Genomic Med 4: 77–94. 10.1002/mgg3.181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borovik L, Modaff P, Waterham HR, Krentz AD, Pauli RM. 2013. Pelger–Huet anomaly and a mild skeletal phenotype secondary to mutations in LBR. Am J Med Genet A 161A: 2066–2073. 10.1002/ajmg.a.36019 [DOI] [PubMed] [Google Scholar]
- Borrego-Pinto J, Jegou T, Osorio DS, Aurade F, Gorjanacz M, Koch B, Mattaj IW, Gomes ER. 2012. Samp1 is a component of TAN lines and is required for nuclear movement. J Cell Sci 125: 1099–1105. 10.1242/jcs.087049 [DOI] [PubMed] [Google Scholar]
- Brachner A, Foisner R. 2011. Evolvement of LEM proteins as chromatin tethers at the nuclear periphery. Biochem Soc Trans 39: 1735–1741. 10.1042/BST20110724 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brachner A, Foisner R. 2014. Lamina-associated polypeptide (LAP)2α and other LEM proteins in cancer biology. Adv Exp Med Biol 773: 143–163. 10.1007/978-1-4899-8032-8_7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brachner A, Reipert S, Foisner R, Gotzmann J. 2005. LEM2 is a novel MAN1-related inner nuclear membrane protein associated with A-type lamins. J Cell Sci 118: 5797–5810. 10.1242/jcs.02701 [DOI] [PubMed] [Google Scholar]
- Braunger K, Pfeffer S, Shrimal S, Gilmore R, Berninghausen O, Mandon EC, Becker T, Förster F, Beckmann R. 2018. Structural basis for coupling protein transport and N-glycosylation at the mammalian endoplasmic reticulum. Science 360: 215–219. 10.1126/science.aar7899 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brodbeck M, Yousif Q, Diener PA, Zweier M, Gruenert J. 2016. The Buschke–Ollendorff syndrome: a case report of simultaneous osteo-cutaneous malformations in the hand. BMC Res Notes 9: 294. 10.1186/s13104-016-2095-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown CA, Scharner J, Felice K, Meriggioli MN, Tarnopolsky M, Bower M, Zammit PS, Mendell JR, Ellis JA. 2011. Novel and recurrent EMD mutations in patients with Emery–Dreifuss muscular dystrophy, identify exon 2 as a mutation hot spot. J Hum Genet 56: 589–594. 10.1038/jhg.2011.65 [DOI] [PubMed] [Google Scholar]
- Brown RS, Zhao C, Chase AR, Wang J, Schlieker C. 2014. The mechanism of Torsin ATPase activation. Proc Natl Acad Sci 111: E4822–E4831. 10.1073/pnas.1415271111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buch C, Lindberg R, Figueroa R, Gudise S, Onischenko E, Hallberg E. 2009. An integral protein of the inner nuclear membrane localizes to the mitotic spindle in mammalian cells. J Cell Sci 122: 2100–2107. 10.1242/jcs.047373 [DOI] [PubMed] [Google Scholar]
- Bupp JM, Martin AE, Stensrud ES, Jaspersen SL. 2007. Telomere anchoring at the nuclear periphery requires the budding yeast Sad1-UNC-84 domain protein Mps3. J Cell Biol 179: 845–854. 10.1083/jcb.200706040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burger B, Hershkovitz D, Indelman M, Kovac M, Galambos J, Haeusermann P, Sprecher E, Itin PH. 2010. Buschke–Ollendorff syndrome in a three-generation family: influence of a novel LEMD3 mutation to tropoelastin expression. Eur J Dermatol 20: 693–697. [DOI] [PubMed] [Google Scholar]
- Burke B. 2018. LINC complexes as regulators of meiosis. Curr Opin Cell Biol 52: 22–29. 10.1016/j.ceb.2018.01.005 [DOI] [PubMed] [Google Scholar]
- Burke B, Stewart CL. 2014. Functional architecture of the cell's nucleus in development, aging, and disease. Curr Top Dev Biol 109: 1–52. 10.1016/B978-0-12-397920-9.00006-8 [DOI] [PubMed] [Google Scholar]
- Cain NE, Tapley EC, McDonald KL, Cain BM, Starr DA. 2014. The SUN protein UNC-84 is required only in force-bearing cells to maintain nuclear envelope architecture. J Cell Biol 206: 163–172. 10.1083/jcb.201405081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calvi A, Wong AS, Wright G, Wong ES, Loo TH, Stewart CL, Burke B. 2015. SUN4 is essential for nuclear remodeling during mammalian spermiogenesis. Dev Biol 407: 321–330. 10.1016/j.ydbio.2015.09.010 [DOI] [PubMed] [Google Scholar]
- Camozzi D, D'Apice MR, Schena E, Cenni V, Columbaro M, Capanni C, Maraldi NM, Squarzoni S, Ortolani M, Novelli G, et al. 2012. Altered chromatin organization and SUN2 localization in mandibuloacral dysplasia are rescued by drug treatment. Histochem Cell Biol 138: 643–651. 10.1007/s00418-012-0977-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Capo-chichi CD, Cai KQ, Testa JR, Godwin AK, Xu XX. 2009. Loss of GATA6 leads to nuclear deformation and aneuploidy in ovarian cancer. Mol Cell Biol 29: 4766–4777. 10.1128/MCB.00087-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Caputo S, Couprie J, Duband-Goulet I, Kondé E, Lin F, Braud S, Gondry M, Gilquin B, Worman HJ, Zinn-Justin S. 2006. The carboxyl-terminal nucleoplasmic region of MAN1 exhibits a DNA binding winged helix domain. J Biol Chem 281: 18208–18215. 10.1074/jbc.M601980200 [DOI] [PubMed] [Google Scholar]
- Chambers DM, Moretti L, Zhang JJ, Cooper SW, Chambers DM, Santangelo PJ, Barker TH. 2018. LEM domain-containing protein 3 antagonizes TGFβ-SMAD2/3 signaling in a stiffness-dependent manner in both the nucleus and cytosol. J Biol Chem 293: 15867–15886. 10.1074/jbc.RA118.003658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang JW, Nomura DK, Cravatt BF. 2011. A potent and selective inhibitor of KIAA1363/AADACL1 that impairs prostate cancer pathogenesis. Chem Biol 18: 476–484. 10.1016/j.chembiol.2011.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang W, Worman HJ, Gundersen GG. 2015. Accessorizing and anchoring the LINC complex for multifunctionality. J Cell Biol 208: 11–22. 10.1083/jcb.201409047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen CY, Chi YH, Mutalif RA, Starost MF, Myers TG, Anderson SA, Stewart CL, Jeang KT. 2012. Accumulation of the inner nuclear envelope protein Sun1 is pathogenic in progeric and dystrophic laminopathies. Cell 149: 565–577. 10.1016/j.cell.2012.01.059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen CK, Blanco M, Jackson C, Aznauryan E, Ollikainen N, Surka C, Chow A, Cerase A, McDonel P, Guttman M. 2016. Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing. Science 354: 468–472. 10.1126/science.aae0047 [DOI] [PubMed] [Google Scholar]
- Cherepanova NA, Shrimal S, Gilmore R. 2014. Oxidoreductase activity is necessary for N-glycosylation of cysteine-proximal acceptor sites in glycoproteins. J Cell Biol 206: 525–539. 10.1083/jcb.201404083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiang KP, Niessen S, Saghatelian A, Cravatt BF. 2006. An enzyme that regulates ether lipid signaling pathways in cancer annotated by multidimensional profiling. Chem Biol 13: 1041–1050. 10.1016/j.chembiol.2006.08.008 [DOI] [PubMed] [Google Scholar]
- Chikashige Y, Tsutsumi C, Yamane M, Okamasa K, Haraguchi T, Hiraoka Y. 2006. Meiotic proteins bqt1 and bqt2 tether telomeres to form the bouquet arrangement of chromosomes. Cell 125: 59–69. 10.1016/j.cell.2006.01.048 [DOI] [PubMed] [Google Scholar]
- Chikashige Y, Yamane M, Okamasa K, Tsutsumi C, Kojidani T, Sato M, Haraguchi T, Hiraoka Y. 2009. Membrane proteins Bqt3 and -4 anchor telomeres to the nuclear envelope to ensure chromosomal bouquet formation. J Cell Biol 187: 413–427. 10.1083/jcb.200902122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cho S, Irianto J, Discher DE. 2017. Mechanosensing by the nucleus: from pathways to scaling relationships. J Cell Biol 216: 305–315. 10.1083/jcb.201610042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clayton P, Fischer B, Mann A, Mansour S, Rossier E, Veen M, Lang C, Baasanjav S, Kieslich M, Brossuleit K, et al. 2010. Mutations causing Greenberg dysplasia but not Pelger anomaly uncouple enzymatic from structural functions of a nuclear membrane protein. Nucleus 1: 354–366. 10.4161/nucl.1.4.12435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clements L, Manilal S, Love DR, Morris GE. 2000. Direct interaction between emerin and lamin A. Biochem Biophys Res Commun 267: 709–714. 10.1006/bbrc.1999.2023 [DOI] [PubMed] [Google Scholar]
- Cohen TV, Kosti O, Stewart CL. 2007. The nuclear envelope protein MAN1 regulates TGFβ signaling and vasculogenesis in the embryonic yolk sac. Development 134: 1385–1395. 10.1242/dev.02816 [DOI] [PubMed] [Google Scholar]
- Conrad MN, Lee CY, Chao G, Shinohara M, Kosaka H, Shinohara A, Conchello JA, Dresser ME. 2008. Rapid telomere movement in meiotic prophase is promoted by NDJ1, MPS3, and CSM4 and is modulated by recombination. Cell 133: 1175–1187. 10.1016/j.cell.2008.04.047 [DOI] [PubMed] [Google Scholar]
- Couto AR, Bruges-Armas J, Peach CA, Chapman K, Brown MA, Wordsworth BP, Zhang Y. 2007. A novel LEMD3 mutation common to patients with osteopoikilosis with and without melorheostosis. Calcif Tissue Int 81: 81–84. 10.1007/s00223-007-9043-z [DOI] [PubMed] [Google Scholar]
- Crisp M, Liu Q, Roux K, Rattner JB, Shanahan C, Burke B, Stahl PD, Hodzic D. 2006. Coupling of the nucleus and cytoplasm: role of the LINC complex. J Cell Biol 172: 41–53. 10.1083/jcb.200509124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cutler JA, Wong X, Hoskins VE, Gordon M, Madugundu AK, Pandey A, Reddy KL. 2019. Mapping the micro-proteome of the nuclear lamina and lamin associated domains. bioRxiv 10.1101/828210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai X, Zheng C, Chen X, Tang Y, Zhang H, Yan C, Ma H, Li X. 2019. Targeted next-generation sequencing identified a known EMD mutation in a Chinese patient with Emery–Dreifuss muscular dystrophy. Hum Genome Var 6: 42. 10.1038/s41439-019-0072-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dechat T, Gotzmann J, Stockinger A, Harris CA, Talle MA, Siekierka JJ, Foisner R. 1998. Detergent-salt resistance of LAP2α in interphase nuclei and phosphorylation-dependent association with chromosomes early in nuclear assembly implies functions in nuclear structure dynamics. EMBO J 17: 4887–4902. 10.1093/emboj/17.16.4887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dechat T, Vlcek S, Foisner R. 2000. Review: lamina-associated polypeptide 2 isoforms and related proteins in cell cycle-dependent nuclear structure dynamics. J Struct Biol 129: 335–345. 10.1006/jsbi.2000.4212 [DOI] [PubMed] [Google Scholar]
- Demmerle J, Koch AJ, Holaska JM. 2012. The nuclear envelope protein emerin binds directly to histone deacetylase 3 (HDAC3) and activates HDAC3 activity. J Biol Chem 287: 22080–22088. 10.1074/jbc.M111.325308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Denais CM, Gilbert RM, Isermann P, McGregor AL, te Lindert M, Weigelin B, Davidson PM, Friedl P, Wolf K, Lammerding J. 2016. Nuclear envelope rupture and repair during cancer cell migration. Science 352: 353–358. 10.1126/science.aad7297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dettmer U, Kuhn PH, Abou-Ajram C, Lichtenthaler SF, Krüger M, Kremmer E, Haass C, Haffner C. 2010. Transmembrane protein 147 (TMEM147) is a novel component of the Nicalin-NOMO protein complex. J Biol Chem 285: 26174–26181. 10.1074/jbc.M110.132548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ding X, Xu R, Yu J, Xu T, Zhuang Y, Han M. 2007. SUN1 is required for telomere attachment to nuclear envelope and gametogenesis in mice. Dev Cell 12: 863–872. 10.1016/j.devcel.2007.03.018 [DOI] [PubMed] [Google Scholar]
- Donahue DA, Amraoui S, di Nunzio F, Kieffer C, Porrot F, Opp S, Diaz-Griffero F, Casartelli N, Schwartz O. 2016. SUN2 overexpression deforms nuclear shape and inhibits HIV. J Virol 90: 4199–4214. 10.1128/JVI.03202-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorboz I, Coutelier M, Bertrand AT, Caberg JH, Elmaleh-Bergès M, Lainé J, Stevanin G, Bonne G, Boespflug-Tanguy O, Servais L. 2014. Severe dystonia, cerebellar atrophy, and cardiomyopathy likely caused by a missense mutation in TOR1AIP1. Orphanet J Rare Dis 9: 174. 10.1186/s13023-014-0174-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorner D, Vlcek S, Foeger N, Gajewski A, Makolm C, Gotzmann J, Hutchison CJ, Foisner R. 2006. Lamina-associated polypeptide 2α regulates cell cycle progression and differentiation via the retinoblastoma-E2F pathway. J Cell Biol 173: 83–93. 10.1083/jcb.200511149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dou Z, Xu C, Donahue G, Shimi T, Pan JA, Zhu J, Ivanov A, Capell BC, Drake AM, Shah PP, et al. 2015. Autophagy mediates degradation of nuclear lamina. Nature 527: 105–109. 10.1038/nature15548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dreesen O, Chojnowski A, Ong PF, Zhao TY, Common JE, Lunny D, Lane EB, Lee SJ, Vardy LA, Stewart CL, et al. 2013. Lamin B1 fluctuations have differential effects on cellular proliferation and senescence. J Cell Biol 200: 605–617. 10.1083/jcb.201206121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dreger M, Bengtsson L, Schoneberg T, Otto H, Hucho F. 2001. Nuclear envelope proteomics: novel integral membrane proteins of the inner nuclear membrane. Proc Natl Acad Sci 98: 11943–11948. 10.1073/pnas.211201898 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duband-Goulet I, Courvalin JC. 2000. Inner nuclear membrane protein LBR preferentially interacts with DNA secondary structures and nucleosomal linker. Biochemistry 39: 6483–6488. 10.1021/bi992908b [DOI] [PubMed] [Google Scholar]
- Ellis JA, Yates JR, Kendrick-Jones J, Brown CA. 1999. Changes at P183 of emerin weaken its protein–protein interactions resulting in X-linked Emery–Dreifuss muscular dystrophy. Hum Genet 104: 262–268. 10.1007/s004390050946 [DOI] [PubMed] [Google Scholar]
- En A, Takauji Y, Ayusawa D, Fujii M. 2020. The role of lamin B receptor in the regulation of senescence-associated secretory phenotype (SASP). Exp Cell Res 390: 111927. 10.1016/j.yexcr.2020.111927 [DOI] [PubMed] [Google Scholar]
- Fichtman B, Zagairy F, Biran N, Barsheshet Y, Chervinsky E, Ben Neriah Z, Shaag A, Assa M, Elpeleg O, Harel A, et al. 2019. Combined loss of LAP1B and LAP1C results in an early onset multisystemic nuclear envelopathy. Nat Commun 10: 605. 10.1038/s41467-019-08493-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foisner R, Gerace L. 1993. Integral membrane proteins of the nuclear envelope interact with lamins and chromosomes, and binding is modulated by mitotic phosphorylation. Cell 73: 1267–1279. 10.1016/0092-8674(93)90355-T [DOI] [PubMed] [Google Scholar]
- Freund A, Laberge RM, Demaria M, Campisi J. 2012. Lamin B1 loss is a senescence-associated biomarker. Mol Biol Cell 23: 2066–2075. 10.1091/mbc.e11-10-0884 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friederichs JM, Ghosh S, Smoyer CJ, McCroskey S, Miller BD, Weaver KJ, Delventhal KM, Unruh J, Slaughter BD, Jaspersen SL. 2011. The SUN protein Mps3 is required for spindle pole body insertion into the nuclear membrane and nuclear envelope homeostasis. PLoS Genet 7: e1002365. 10.1371/journal.pgen.1002365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frohnert C, Schweizer S, Hoyer-Fender S. 2011. SPAG4L/SPAG4L-2 are testis-specific SUN domain proteins restricted to the apical nuclear envelope of round spermatids facing the acrosome. Mol Hum Reprod 17: 207–218. 10.1093/molehr/gaq099 [DOI] [PubMed] [Google Scholar]
- Gant TM, Harris CA, Wilson KL. 1999. Roles of LAP2 proteins in nuclear assembly and DNA replication: truncated LAP2β proteins alter lamina assembly, envelope formation, nuclear size, and DNA replication efficiency in Xenopus laevis extracts. J Cell Biol 144: 1083–1096. 10.1083/jcb.144.6.1083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gartenberg MR. 2009. Life on the edge: telomeres and persistent DNA breaks converge at the nuclear periphery. Genes Dev 23: 1027–1031. 10.1101/gad.1805309 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gaudy-Marqueste C, Roll P, Esteves-Vieira V, Weiller PJ, Grob JJ, Cau P, Levy N, De Sandre-Giovannoli A. 2010. LBR mutation and nuclear envelope defects in a patient affected with Reynolds syndrome. J Med Genet 47: 361–370. 10.1136/jmg.2009.071696 [DOI] [PubMed] [Google Scholar]
- Gesson K, Rescheneder P, Skoruppa MP, von Haeseler A, Dechat T, Foisner R. 2016. A-type lamins bind both hetero- and euchromatin, the latter being regulated by lamina-associated polypeptide 2α. Genome Res 26: 462–473. 10.1101/gr.196220.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghaoui R, Benavides T, Lek M, Waddell LB, Kaur S, North KN, MacArthur DG, Clarke NF, Cooper ST. 2016. TOR1AIP1 as a cause of cardiac failure and recessive limb-girdle muscular dystrophy. Neuromuscul Disord 26: 500–503. 10.1016/j.nmd.2016.05.013 [DOI] [PubMed] [Google Scholar]
- Gilbert HTJ, Mallikarjun V, Dobre O, Jackson MR, Pedley R, Gilmore AP, Richardson SM, Swift J. 2019. Nuclear decoupling is part of a rapid protein-level cellular response to high-intensity mechanical loading. Nat Commun 10: 4149. 10.1038/s41467-019-11923-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez Y, Saito A, Sazer S. 2012. Fission yeast Lem2 and Man1 perform fundamental functions of the animal cell nuclear lamina. Nucleus 3: 60–76. 10.4161/nucl.18824 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodchild RE, Dauer WT. 2005. The AAA+ protein torsinA interacts with a conserved domain present in LAP1 and a novel ER protein. J Cell Biol 168: 855–862. 10.1083/jcb.200411026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu M, LaJoie D, Chen OS, von Appen A, Ladinsky MS, Redd MJ, Nikolova L, Bjorkman PJ, Sundquist WI, Ullman KS, et al. 2017. LEM2 recruits CHMP7 for ESCRT-mediated nuclear envelope closure in fission yeast and human cells. Proc Natl Acad Sci 114: E2166–E2175. 10.1073/pnas.1613916114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guarda A, Bolognese F, Bonapace IM, Badaracco G. 2009. Interaction between the inner nuclear membrane lamin B receptor and the heterochromatic methyl binding protein, MeCP2. Exp Cell Res 315: 1895–1903. 10.1016/j.yexcr.2009.01.019 [DOI] [PubMed] [Google Scholar]
- Gudise S, Figueroa RA, Lindberg R, Larsson V, Hallberg E. 2011. Samp1 is functionally associated with the LINC complex and A-type lamina networks. J Cell Sci 124: 2077–2085. 10.1242/jcs.078923 [DOI] [PubMed] [Google Scholar]
- Guilluy C, Osborne LD, Van Landeghem L, Sharek L, Superfine R, Garcia-Mata R, Burridge K. 2014. Isolated nuclei adapt to force and reveal a mechanotransduction pathway in the nucleus. Nat Cell Biol 16: 376–381. 10.1038/ncb2927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gundersen GG, Worman HJ. 2013. Nuclear positioning. Cell 152: 1376–1389. 10.1016/j.cell.2013.02.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halfmann CT, Sears RM, Katiyar A, Busselman BW, Aman LK, Zhang Q, O'Bryan CS, Angelini TE, Lele TP, Roux KJ. 2019. Repair of nuclear ruptures requires barrier-to-autointegration factor. J Cell Biol 218: 2136–2149. 10.1083/jcb.201901116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen JB, Jorgensen C, Petersen RK, Hallenborg P, De Matteis R, Boye HA, Petrovic N, Enerback S, Nedergaard J, Cinti S, et al. 2004. Retinoblastoma protein functions as a molecular switch determining white versus brown adipocyte differentiation. Proc Natl Acad Sci 101: 4112–4117. 10.1073/pnas.0301964101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haque F, Mazzeo D, Patel JT, Smallwood DT, Ellis JA, Shanahan CM, Shackleton S. 2010. Mammalian SUN protein interaction networks at the inner nuclear membrane and their role in laminopathy disease processes. J Biol Chem 285: 3487–3498. 10.1074/jbc.M109.071910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haraguchi T, Holaska JM, Yamane M, Koujin T, Hashiguchi N, Mori C, Wilson KL, Hiraoka Y. 2004. Emerin binding to Btf, a death-promoting transcriptional repressor, is disrupted by a missense mutation that causes Emery–Dreifuss muscular dystrophy. Eur J Biochem 271: 1035–1045. 10.1111/j.1432-1033.2004.04007.x [DOI] [PubMed] [Google Scholar]
- Hellemans J, Preobrazhenska O, Willaert A, Debeer P, Verdonk PC, Costa T, Janssens K, Menten B, Van Roy N, Vermeulen SJ, et al. 2004. Loss-of-function mutations in LEMD3 result in osteopoikilosis, Buschke–Ollendorff syndrome and melorheostosis. Nat Genet 36: 1213–1218. 10.1038/ng1453 [DOI] [PubMed] [Google Scholar]
- Hellemans J, Debeer P, Wright M, Janecke A, Kjaer KW, Verdonk PC, Savarirayan R, Basel L, Moss C, Roth J, et al. 2006. Germline LEMD3 mutations are rare in sporadic patients with isolated melorheostosis. Hum Mutat 27: 290. 10.1002/humu.9403 [DOI] [PubMed] [Google Scholar]
- Henne WM, Stenmark H, Emr SD. 2013. Molecular mechanisms of the membrane sculpting ESCRT pathway. Cold Spring Harb Perspect Biol 5: a016766. 10.1101/cshperspect.a016766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hirano Y, Hizume K, Kimura H, Takeyasu K, Haraguchi T, Hiraoka Y. 2012. Lamin B receptor recognizes specific modifications of histone H4 in heterochromatin formation. J Biol Chem 287: 42654–42663. 10.1074/jbc.M112.397950 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hiraoka Y, Dernburg AF. 2009. The SUN rises on meiotic chromosome dynamics. Dev Cell 17: 598–605. 10.1016/j.devcel.2009.10.014 [DOI] [PubMed] [Google Scholar]
- Ho CY, Jaalouk DE, Vartiainen MK, Lammerding J. 2013. Lamin A/C and emerin regulate MKL1-SRF activity by modulating actin dynamics. Nature 497: 507–511. 10.1038/nature12105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hodzic DM, Yeater DB, Bengtsson L, Otto H, Stahl PD. 2004. Sun2 is a novel mammalian inner nuclear membrane protein. J Biol Chem 279: 25805–25812. 10.1074/jbc.M313157200 [DOI] [PubMed] [Google Scholar]
- Hoffmann K, Dreger CK, Olins AL, Olins DE, Shultz LD, Lucke B, Karl H, Kaps R, Müller D, Vayá A, et al. 2002. Mutations in the gene encoding the lamin B receptor produce an altered nuclear morphology in granulocytes (Pelger–Huët anomaly). Nat Genet 31: 410–414. 10.1038/ng925 [DOI] [PubMed] [Google Scholar]
- Holaska JM, Wilson KL. 2007. An emerin “proteome”: purification of distinct emerin-containing complexes from HeLa cells suggests molecular basis for diverse roles including gene regulation, mRNA splicing, signaling, mechanosensing, and nuclear architecture. Biochemistry 46: 8897–8908. 10.1021/bi602636m [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holaska JM, Lee KK, Kowalski AK, Wilson KL. 2003. Transcriptional repressor germ cell-less (GCL) and barrier to autointegration factor (BAF) compete for binding to emerin in vitro. J Biol Chem 278: 6969–6975. 10.1074/jbc.M208811200 [DOI] [PubMed] [Google Scholar]
- Holaska JM, Kowalski AK, Wilson KL. 2004. Emerin caps the pointed end of actin filaments: evidence for an actin cortical network at the nuclear inner membrane. PLoS Biol 2: E231. 10.1371/journal.pbio.0020231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holaska JM, Rais-Bahrami S, Wilson KL. 2006. Lmo7 is an emerin-binding protein that regulates the transcription of emerin and many other muscle-relevant genes. Hum Mol Genet 15: 3459–3472. 10.1093/hmg/ddl423 [DOI] [PubMed] [Google Scholar]
- Holmer L, Pezhman A, Worman HJ. 1998. The human lamin B receptor/sterol reductase multigene family. Genomics 54: 469–476. 10.1006/geno.1998.5615 [DOI] [PubMed] [Google Scholar]
- Horigome C, Okada T, Shimazu K, Gasser SM, Mizuta K. 2011. Ribosome biogenesis factors bind a nuclear envelope SUN domain protein to cluster yeast telomeres. EMBO J 30: 3799–3811. 10.1038/emboj.2011.267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horigome C, Oma Y, Konishi T, Schmid R, Marcomini I, Hauer MH, Dion V, Harata M, Gasser SM. 2014. SWR1 and INO80 chromatin remodelers contribute to DNA double-strand break perinuclear anchorage site choice. Mol Cell 55: 626–639. 10.1016/j.molcel.2014.06.027 [DOI] [PubMed] [Google Scholar]
- Horn HF, Kim DI, Wright GD, Wong ES, Stewart CL, Burke B, Roux KJ. 2013. A mammalian KASH domain protein coupling meiotic chromosomes to the cytoskeleton. J Cell Biol 202: 1023–1039. 10.1083/jcb.201304004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang J, Zheng DL, Qin FS, Cheng N, Chen H, Wan BB, Wang YP, Xiao HS, Han ZG. 2010. Genetic and epigenetic silencing of SCARA5 may contribute to human hepatocellular carcinoma by activating FAK signaling. J Clin Invest 120: 223–241. 10.1172/JCI38012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huber MD, Guan T, Gerace L. 2009. Overlapping functions of nuclear envelope proteins NET25 (Lem2) and emerin in regulation of extracellular signal-regulated kinase signaling in myoblast differentiation. Mol Cell Biol 29: 5718–5728. 10.1128/MCB.00270-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huh MS, Parker MH, Scimè A, Parks R, Rudnicki MA. 2004. Rb is required for progression through myogenic differentiation but not maintenance of terminal differentiation. J Cell Biol 166: 865–876. 10.1083/jcb.200403004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Igarashi M, Osuga J, Isshiki M, Sekiya M, Okazaki H, Takase S, Takanashi M, Ohta K, Kumagai M, Nishi M, et al. 2010. Targeting of neutral cholesterol ester hydrolase to the endoplasmic reticulum via its N-terminal sequence. J Lipid Res 51: 274–285. 10.1194/jlr.M900201-JLR200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishimura A, Ng JK, Taira M, Young SG, Osada S. 2006. Man1, an inner nuclear membrane protein, regulates vascular remodeling by modulating transforming growth factor β signaling. Development 133: 3919–3928. 10.1242/dev.02538 [DOI] [PubMed] [Google Scholar]
- Ivanov A, Pawlikowski J, Manoharan I, van Tuyn J, Nelson DM, Rai TS, Shah PP, Hewitt G, Korolchuk VI, Passos JF, et al. 2013. Lysosome-mediated processing of chromatin in senescence. J Cell Biol 202: 129–143. 10.1083/jcb.201212110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jafferali MH, Figueroa RA, Hasan M, Hallberg E. 2017. Spindle associated membrane protein 1 (Samp1) is required for the differentiation of muscle cells. Sci Rep 7: 16655. 10.1038/s41598-017-16746-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jungwirth M, Dear ML, Brown P, Holbrook K, Goodchild R. 2010. Relative tissue expression of homologous torsinB correlates with the neuronal specific importance of DYT1 dystonia-associated torsinA. Hum Mol Genet 19: 888–900. 10.1093/hmg/ddp557 [DOI] [PubMed] [Google Scholar]
- Kayman-Kurekci G, Talim B, Korkusuz P, Sayar N, Sarioglu T, Oncel I, Sharafi P, Gundesli H, Balci-Hayta B, Purali N, et al. 2014. Mutation in TOR1AIP1 encoding LAP1B in a form of muscular dystrophy: a novel gene related to nuclear envelopathies. Neuromuscul Disord 24: 624–633. 10.1016/j.nmd.2014.04.007 [DOI] [PubMed] [Google Scholar]
- Kim CE, Perez A, Perkins G, Ellisman MH, Dauer WT. 2010. A molecular mechanism underlying the neural-specific defect in torsinA mutant mice. Proc Natl Acad Sci 107: 9861–9866. 10.1073/pnas.0912877107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim HJ, Hwang SH, Han ME, Baek S, Sim HE, Yoon S, Baek SY, Kim BS, Kim JH, Kim SY, et al. 2012. LAP2 is widely overexpressed in diverse digestive tract cancers and regulates motility of cancer cells. PLoS ONE 7: e39482. 10.1371/journal.pone.0039482 [DOI] [PMC free article] [PubMed] [Google Scholar]
- King MC, Drivas TG, Blobel G. 2008. A network of nuclear envelope membrane proteins linking centromeres to microtubules. Cell 134: 427–438. 10.1016/j.cell.2008.06.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kobayashi H, Kasahara M, Hino M, Takahara S, Ikeda K, Son C, Iwakura T, Matsuoka N, Yoshimoto A, Ohgo N, et al. 2007. A novel heterozygous splice-site mutation of LEM domain-containing 3 in a Japanese kindred with Buschke–Ollendorff syndrome. J Endocrinol Invest 30: 263–265. 10.1007/BF03347437 [DOI] [PubMed] [Google Scholar]
- Koch AJ, Holaska JM. 2012. Loss of emerin alters myogenic signaling and miRNA expression in mouse myogenic progenitors. PLoS ONE 7: e37262. 10.1371/journal.pone.0037262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koch AJ, Holaska JM. 2014. Emerin in health and disease. Semin Cell Dev Biol 29: 95–106. 10.1016/j.semcdb.2013.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korekawa A, Nakano H, Toyomaki Y, Takiyoshi N, Rokunohe D, Akasaka E, Nakajima K, Sawamura D. 2012. Buschke–Ollendorff syndrome associated with hypertrophic scar formation: a possible role for LEMD3 mutation. Br J Dermatol 166: 900–903. 10.1111/j.1365-2133.2011.10691.x [DOI] [PubMed] [Google Scholar]
- Korfali N, Wilkie GS, Swanson SK, Srsen V, Batrakou DG, Fairley EA, Malik P, Zuleger N, Goncharevich A, de Las Heras J, et al. 2010. The leukocyte nuclear envelope proteome varies with cell activation and contains novel transmembrane proteins that affect genome architecture. Mol Cell Proteomics 9: 2571–2585. 10.1074/mcp.M110.002915 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korfali N, Wilkie GS, Swanson SK, Srsen V, de Las Heras J, Batrakou DG, Malik P, Zuleger N, Kerr AR, Florens L, et al. 2012. The nuclear envelope proteome differs notably between tissues. Nucleus 3: 552–564. 10.4161/nucl.22257 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koszul R, Kim KP, Prentiss M, Kleckner N, Kameoka S. 2008. Meiotic chromosomes move by linkage to dynamic actin cables with transduction of force through the nuclear envelope. Cell 133: 1188–1201. 10.1016/j.cell.2008.04.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kume K, Cantwell H, Burrell A, Nurse P. 2019. Nuclear membrane protein Lem2 regulates nuclear size through membrane flow. Nat Commun 10: 1871. 10.1038/s41467-019-09623-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lahaye X, Satoh T, Gentili M, Cerboni S, Silvin A, Conrad C, Ahmed-Belkacem A, Rodriguez EC, Guichou JF, Bosquet N, et al. 2016. Nuclear envelope protein SUN2 promotes cyclophilin-A-dependent steps of HIV replication. Cell Rep 15: 879–892. 10.1016/j.celrep.2016.03.074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lammerding J, Hsiao J, Schulze PC, Kozlov S, Stewart CL, Lee RT. 2005. Abnormal nuclear shape and impaired mechanotransduction in emerin-deficient cells. J Cell Biol 170: 781–791. 10.1083/jcb.200502148 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larsson VJ, Jafferali MH, Vijayaraghavan B, Figueroa RA, Hallberg E. 2018. Mitotic spindle assembly and γ-tubulin localisation depend on the integral nuclear membrane protein Samp1. J Cell Sci 131: jcs211664. 10.1242/jcs.211664 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laugks U, Hieke M, Wagner N. 2017. MAN1 restricts BMP signaling during synaptic growth in Drosophila. Cell Mol Neurobiol 37: 1077–1093. 10.1007/s10571-016-0442-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lawrence KS, Tapley EC, Cruz VE, Li Q, Aung K, Hart KC, Schwartz TU, Starr DA, Engebrecht J. 2016. LINC complexes promote homologous recombination in part through inhibition of nonhomologous end joining. J Cell Biol 215: 801–821. 10.1083/jcb.201604112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lechner MS, Schultz DC, Negorev D, Maul GG, Rauscher FJ III. 2005. The mammalian heterochromatin protein 1 binds diverse nuclear proteins through a common motif that targets the chromoshadow domain. Biochem Biophys Res Commun 331: 929–937. 10.1016/j.bbrc.2005.04.016 [DOI] [PubMed] [Google Scholar]
- Lee YL, Burke B. 2018. LINC complexes and nuclear positioning. Semin Cell Dev Biol 82: 67–76. 10.1016/j.semcdb.2017.11.008 [DOI] [PubMed] [Google Scholar]
- Lee CY, Horn HF, Stewart CL, Burke B, Bolcun-Filas E, Schimenti JC, Dresser ME, Pezza RJ. 2015. Mechanism and regulation of rapid telomere prophase movements in mouse meiotic chromosomes. Cell Rep 11: 551–563. 10.1016/j.celrep.2015.03.045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee B, Lee TH, Shim J. 2017. Emerin suppresses Notch signaling by restricting the Notch intracellular domain to the nuclear membrane. Biochim Biophys Acta Mol Cell Res 1864: 303–313. 10.1016/j.bbamcr.2016.11.013 [DOI] [PubMed] [Google Scholar]
- Lei K, Zhu X, Xu R, Shao C, Xu T, Zhuang Y, Han M. 2012. Inner nuclear envelope proteins SUN1 and SUN2 play a prominent role in the DNA damage response. Curr Biol 22: 1609–1615. 10.1016/j.cub.2012.06.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lenain C, Gusyatiner O, Douma S, van den Broek B, Peeper DS. 2015. Autophagy-mediated degradation of nuclear envelope proteins during oncogene-induced senescence. Carcinogenesis 36: 1263–1274. 10.1093/carcin/bgv124 [DOI] [PubMed] [Google Scholar]
- Le Thanh P, Meinke P, Korfali N, Srsen V, Robson MI, Wehnert M, Schoser B, Sewry CA, Schirmer EC. 2017. Immunohistochemistry on a panel of Emery–Dreifuss muscular dystrophy samples reveals nuclear envelope proteins as inconsistent markers for pathology. Neuromuscul Disord 27: 338–351. 10.1016/j.nmd.2016.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li C, Wei J, Li Y, He X, Zhou Q, Yan J, Zhang J, Liu Y, Liu Y, Shu HB. 2013. Transmembrane protein 214 (TMEM214) mediates endoplasmic reticulum stress-induced caspase 4 enzyme activation and apoptosis. J Biol Chem 288: 17908–17917. 10.1074/jbc.M113.458836 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Roberti R, Blobel G. 2015. Structure of an integral membrane sterol reductase from Methylomicrobium alcaliphilum. Nature 517: 104–107. 10.1038/nature13797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang WC, Mitsuhashi H, Keduka E, Nonaka I, Noguchi S, Nishino I, Hayashi YK. 2011. TMEM43 mutations in Emery–Dreifuss muscular dystrophy-related myopathy. Ann Neurol 69: 1005–1013. 10.1002/ana.22338 [DOI] [PubMed] [Google Scholar]
- Lin F, Blake DL, Callebaut I, Skerjanc IS, Holmer L, McBurney MW, Paulin-Levasseur M, Worman HJ. 2000. MAN1, an inner nuclear membrane protein that shares the LEM domain with lamina-associated polypeptide 2 and emerin. J Biol Chem 275: 4840–4847. 10.1074/jbc.275.7.4840 [DOI] [PubMed] [Google Scholar]
- Lin F, Morrison JM, Wu W, Worman HJ. 2005. MAN1, an integral protein of the inner nuclear membrane, binds Smad2 and Smad3 and antagonizes transforming growth factor-β signaling. Hum Mol Genet 14: 437–445. 10.1093/hmg/ddi040 [DOI] [PubMed] [Google Scholar]
- Lin ST, Zhang L, Lin X, Zhang LC, Garcia VE, Tsai CW, Ptáček L, Fu YH. 2014. Nuclear envelope protein MAN1 regulates clock through BMAL1. eLife 3: e02981. 10.7554/eLife.02981 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J, Lee KK, Segura-Totten M, Neufeld E, Wilson KL, Gruenbaum Y. 2003. MAN1 and emerin have overlapping function(s) essential for chromosome segregation and cell division in Caenorhabditis elegans. Proc Natl Acad Sci 100: 4598–4603. 10.1073/pnas.0730821100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Q, Pante N, Misteli T, Elsagga M, Crisp M, Hodzic D, Burke B, Roux KJ. 2007. Functional association of Sun1 with nuclear pore complexes. J Cell Biol 178: 785–798. 10.1083/jcb.200704108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu GH, Guan T, Datta K, Coppinger J, Yates J III, Gerace L. 2009. Regulation of myoblast differentiation by the nuclear envelope protein NET39. Mol Cell Biol 29: 5800–5812. 10.1128/MCB.00684-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu H, Hu J, Wei R, Zhou L, Pan H, Zhu H, Huang M, Luo J, Xu W. 2018. SPAG5 promotes hepatocellular carcinoma progression by downregulating SCARA5 through modifying β-catenin degradation. J Exp Clin Cancer Res 37: 229. 10.1186/s13046-018-0891-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lottersberger F, Karssemeijer RA, Dimitrova N, de Lange T. 2015. 53BP1 and the LINC complex promote microtubule-dependent DSB mobility and DNA repair. Cell 163: 880–893. 10.1016/j.cell.2015.09.057 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luithle N, de Bos JU, Hovius R, Maslennikova D, Lewis RT, Ungricht R, Fierz B, Kutay U. 2020. Torsin ATPases influence chromatin interaction of the torsin regulator LAP1. eLife 9: e63614. 10.7554/eLife.63614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lukášová E, Kovařík A, Bačíková A, Falk M, Kozubek S. 2017. Loss of lamin B receptor is necessary to induce cellular senescence. Biochem J 474: 281–300. 10.1042/BCJ20160459 [DOI] [PubMed] [Google Scholar]
- Luo X, Yang W, Gao G. 2018. SUN1 regulates HIV-1 nuclear import in a manner dependent on the interaction between the viral capsid and cellular cyclophilin A. J Virol 92: JVI.00229-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MacQueen AJ, Phillips CM, Bhalla N, Weiser P, Villeneuve AM, Dernburg AF. 2005. Chromosome sites play dual roles to establish homologous synapsis during meiosis in C. elegans. Cell 123: 1037–1050. 10.1016/j.cell.2005.09.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Makatsori D, Kourmouli N, Polioudaki H, Shultz LD, McLean K, Theodoropoulos PA, Singh PB, Georgatos SD. 2004. The inner nuclear membrane protein lamin B receptor forms distinct microdomains and links epigenetically marked chromatin to the nuclear envelope. J Biol Chem 279: 25567–25573. 10.1074/jbc.M313606200 [DOI] [PubMed] [Google Scholar]
- Malik P, Korfali N, Srsen V, Lazou V, Batrakou DG, Zuleger N, Kavanagh DM, Wilkie GS, Goldberg MW, Schirmer EC. 2010. Cell-specific and lamin-dependent targeting of novel transmembrane proteins in the nuclear envelope. Cell Mol Life Sci 67: 1353–1369. 10.1007/s00018-010-0257-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malone CJ, Fixsen WD, Horvitz HR, Han M. 1999. UNC-84 localizes to the nuclear envelope and is required for nuclear migration and anchoring during C. elegans development. Development 126: 3171–3181. [DOI] [PubMed] [Google Scholar]
- Manilal S, Nguyen TM, Sewry CA, Morris GE. 1996. The Emery–Dreifuss muscular dystrophy protein, emerin, is a nuclear membrane protein. Hum Mol Genet 5: 801–808. 10.1093/hmg/5.6.801 [DOI] [PubMed] [Google Scholar]
- Mansharamani M, Wilson KL. 2005. Direct binding of nuclear membrane protein MAN1 to emerin in vitro and two modes of binding to barrier-to-autointegration factor. J Biol Chem 280: 13863–13870. 10.1074/jbc.M413020200 [DOI] [PubMed] [Google Scholar]
- Margalit A, Brachner A, Gotzmann J, Foisner R, Gruenbaum Y. 2007. Barrier-to-autointegration factor—a BAFfling little protein. Trends Cell Biol 17: 202–208. 10.1016/j.tcb.2007.02.004 [DOI] [PubMed] [Google Scholar]
- Markiewicz E, Dechat T, Foisner R, Quinlan RA, Hutchison CJ. 2002. Lamin A/C binding protein LAP2α is required for nuclear anchorage of retinoblastoma protein. Mol Biol Cell 13: 4401–4413. 10.1091/mbc.e02-07-0450 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Markiewicz E, Tilgner K, Barker N, van de Wetering M, Clevers H, Dorobek M, Hausmanowa-Petrusewicz I, Ramaekers FC, Broers JL, Blankesteijn WM, et al. 2006. The inner nuclear membrane protein emerin regulates β-catenin activity by restricting its accumulation in the nucleus. EMBO J 25: 3275–3285. 10.1038/sj.emboj.7601230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marnef A, Finoux AL, Arnould C, Guillou E, Daburon V, Rocher V, Mangeat T, Mangeot PE, Ricci EP, Legube G. 2019. A cohesin/HUSH- and LINC-dependent pathway controls ribosomal DNA double-strand break repair. Genes Dev 33: 1175–1190. 10.1101/gad.324012.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martins SB, Eide T, Steen RL, Jahnsen T, Skalhegg BS, Collas P. 2000. HA95 is a protein of the chromatin and nuclear matrix regulating nuclear envelope dynamics. J Cell Sci 113 (Pt 21): 3703–3713. [DOI] [PubMed] [Google Scholar]
- Martins S, Eikvar S, Furukawa K, Collas P. 2003. HA95 and LAP2β mediate a novel chromatin-nuclear envelope interaction implicated in initiation of DNA replication. J Cell Biol 160: 177–188. 10.1083/jcb.200210026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Massague J, Cheifetz S, Endo T, Nadal-Ginard B. 1986. Type β transforming growth factor is an inhibitor of myogenic differentiation. Proc Natl Acad Sci 83: 8206–8210. 10.1073/pnas.83.21.8206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mattaj IW. 2004. Sorting out the nuclear envelope from the endoplasmic reticulum. Nat Rev Mol Cell Biol 5: 65–69. 10.1038/nrm1263 [DOI] [PubMed] [Google Scholar]
- McCullough J, Frost A, Sundquist WI. 2018. Structures, functions, and dynamics of ESCRT-III/Vps4 membrane remodeling and fission complexes. Annu Rev Cell Dev Biol 34: 85–109. 10.1146/annurev-cellbio-100616-060600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McHugh CA, Chen CK, Chow A, Surka CF, Tran C, McDonel P, Pandya-Jones A, Blanco M, Burghard C, Moradian A, et al. 2015. The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature 521: 232–236. 10.1038/nature14443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKinnon CM, Mellor H. 2017. The tumor suppressor RhoBTB1 controls Golgi integrity and breast cancer cell invasion through METTL7B. BMC Cancer 17: 145. 10.1186/s12885-017-3138-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meinke P, Mattioli E, Haque F, Antoku S, Columbaro M, Straatman KR, Worman HJ, Gundersen GG, Lattanzi G, Wehnert M, et al. 2014. Muscular dystrophy-associated SUN1 and SUN2 variants disrupt nuclear-cytoskeletal connections and myonuclear organization. PLoS Genet 10: e1004605. 10.1371/journal.pgen.1004605 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meinke P, Kerr ARW, Czapiewski R, de Las Heras JI, Dixon CR, Harris E, Kölbel H, Muntoni F, Schara U, Straub V, et al. 2020. A multistage sequencing strategy pinpoints novel candidate alleles for Emery–Dreifuss muscular dystrophy and supports gene misregulation as its pathomechanism. EBioMedicine 51: 102587. 10.1016/j.ebiom.2019.11.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mekhail K, Moazed D. 2010. The nuclear envelope in genome organization, expression and stability. Nat Rev Mol Cell Biol 11: 317–328. 10.1038/nrm2894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mekhail K, Seebacher J, Gygi SP, Moazed D. 2008. Role for perinuclear chromosome tethering in maintenance of genome stability. Nature 456: 667–670. 10.1038/nature07460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mirza AN, McKellar SA, Urman NM, Brown AS, Hollmig T, Aasi SZ, Oro AE. 2019. LAP2 proteins chaperone GLI1 movement between the lamina and chromatin to regulate transcription. Cell 176: 198–212.e15. 10.1016/j.cell.2018.10.054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mislow JM, Holaska JM, Kim MS, Lee KK, Segura-Totten M, Wilson KL, McNally EM. 2002. Nesprin-1α self-associates and binds directly to emerin and lamin A in vitro. FEBS Lett 525: 135–140. 10.1016/S0014-5793(02)03105-8 [DOI] [PubMed] [Google Scholar]
- Mora M, Cartegni L, Di Blasi C, Barresi R, Bione S, Raffaele di Barletta M, Morandi L, Merlini L, Nigro V, Politano L, et al. 1997. X-linked Emery–Dreifuss muscular dystrophy can be diagnosed from skin biopsy or blood sample. Ann Neurol 42: 249–253. 10.1002/ana.410420218 [DOI] [PubMed] [Google Scholar]
- Morimoto A, Shibuya H, Zhu X, Kim J, Ishiguro K, Han M, Watanabe Y. 2012. A conserved KASH domain protein associates with telomeres, SUN1, and dynactin during mammalian meiosis. J Cell Biol 198: 165–172. 10.1083/jcb.201204085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris GE, Manilal S. 1999. Heart to heart: from nuclear proteins to Emery–Dreifuss muscular dystrophy. Hum Mol Genet 8: 1847–1851. 10.1093/hmg/8.10.1847 [DOI] [PubMed] [Google Scholar]
- Moser B, Basílio J, Gotzmann J, Brachner A, Foisner R. 2020. Comparative interactome analysis of emerin, MAN1 and LEM2 reveals a unique role for LEM2 in nucleotide excision repair. Cells 9: 463. 10.3390/cells9020463 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muchir A, Pavlidis P, Bonne G, Hayashi YK, Worman HJ. 2007. Activation of MAPK in hearts of EMD null mice: similarities between mouse models of X-linked and autosomal dominant Emery Dreifuss muscular dystrophy. Hum Mol Genet 16: 1884–1895. 10.1093/hmg/ddm137 [DOI] [PubMed] [Google Scholar]
- Mumm S, Wenkert D, Zhang X, McAlister WH, Mier RJ, Whyte MP. 2007. Deactivating germline mutations in LEMD3 cause osteopoikilosis and Buschke–Ollendorff syndrome, but not sporadic melorheostosis. J Bone Miner Res 22: 243–250. 10.1359/jbmr.061102 [DOI] [PubMed] [Google Scholar]
- Naetar N, Korbei B, Kozlov S, Kerenyi MA, Dorner D, Kral R, Gotic I, Fuchs P, Cohen TV, Bittner R, et al. 2008. Loss of nucleoplasmic LAP2α-lamin A complexes causes erythroid and epidermal progenitor hyperproliferation. Nat Cell Biol 10: 1341–1348. 10.1038/ncb1793 [DOI] [PubMed] [Google Scholar]
- Nesterova TB, Wei G, Coker H, Pintacuda G, Bowness JS, Zhang T, Almeida M, Bloechl B, Moindrot B, Carter EJ, et al. 2019. Systematic allelic analysis defines the interplay of key pathways in X chromosome inactivation. Nat Commun 10: 3129. 10.1038/s41467-019-11171-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nili E, Cojocaru GS, Kalma Y, Ginsberg D, Copeland NG, Gilbert DJ, Jenkins NA, Berger R, Shaklai S, Amariglio N, et al. 2001. Nuclear membrane protein LAP2β mediates transcriptional repression alone and together with its binding partner GCL (germ-cell-less). J Cell Sci 114: 3297–3307. [DOI] [PubMed] [Google Scholar]
- Ohba T, Schirmer EC, Nishimoto T, Gerace L. 2004. Energy- and temperature-dependent transport of integral proteins to the inner nuclear membrane via the nuclear pore. J Cell Biol 167: 1051–1062. 10.1083/jcb.200409149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olmos Y, Hodgson L, Mantell J, Verkade P, Carlton JG. 2015. ESCRT-III controls nuclear envelope reformation. Nature 522: 236–239. 10.1038/nature14503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olson EN, Nordheim A. 2010. Linking actin dynamics and gene transcription to drive cellular motile functions. Nat Rev Mol Cell Biol 11: 353–365. 10.1038/nrm2890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oro AE, Higgins KM, Hu Z, Bonifas JM, Epstein EH Jr, Scott MP. 1997. Basal cell carcinomas in mice overexpressing sonic hedgehog. Science 276: 817–821. 10.1126/science.276.5313.817 [DOI] [PubMed] [Google Scholar]
- Osada S, Ohmori SY, Taira M. 2003. XMAN1, an inner nuclear membrane protein, antagonizes BMP signaling by interacting with Smad1 in Xenopus embryos. Development 130: 1783–1794. 10.1242/dev.00401 [DOI] [PubMed] [Google Scholar]
- Östlund C, Sullivan T, Stewart CL, Worman HJ. 2006. Dependence of diffusional mobility of integral inner nuclear membrane proteins on A-type lamins. Biochemistry 45: 1374–1382. 10.1021/bi052156n [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oza P, Jaspersen SL, Miele A, Dekker J, Peterson CL. 2009. Mechanisms that regulate localization of a DNA double-strand break to the nuclear periphery. Genes Dev 23: 912–927. 10.1101/gad.1782209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ozelius LJ, Hewett JW, Page CE, Bressman SB, Kramer PL, Shalish C, de Leon D, Brin MF, Raymond D, Corey DP, et al. 1997. The early-onset torsion dystonia gene (DYT1) encodes an ATP-binding protein. Nat Genet 17: 40–48. 10.1038/ng0997-40 [DOI] [PubMed] [Google Scholar]
- Pan D, Estévez-Salmerón LD, Stroschein SL, Zhu X, He J, Zhou S, Luo K. 2005. The integral inner nuclear membrane protein MAN1 physically interacts with the R-Smad proteins to repress signaling by the transforming growth factor-β superfamily of cytokines. J Biol Chem 280: 15992–16001. 10.1074/jbc.M411234200 [DOI] [PubMed] [Google Scholar]
- Pasch E, Link J, Beck C, Scheuerle S, Alsheimer M. 2015. The LINC complex component Sun4 plays a crucial role in sperm head formation and fertility. Biol Open 4: 1792–1802. 10.1242/bio.015768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawar S, Ungricht R, Tiefenboeck P, Leroux JC, Kutay U. 2017. Efficient protein targeting to the inner nuclear membrane requires Atlastin-dependent maintenance of ER topology. eLife 6: e28202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Penkner A, Tang L, Novatchkova M, Ladurner M, Fridkin A, Gruenbaum Y, Schweizer D, Loidl J, Jantsch V. 2007. The nuclear envelope protein Matefin/SUN-1 is required for homologous pairing in C. elegans meiosis. Dev Cell 12: 873–885. 10.1016/j.devcel.2007.05.004 [DOI] [PubMed] [Google Scholar]
- Penkner AM, Fridkin A, Gloggnitzer J, Baudrimont A, Machacek T, Woglar A, Csaszar E, Pasierbek P, Ammerer G, Gruenbaum Y, et al. 2009. Meiotic chromosome homology search involves modifications of the nuclear envelope protein Matefin/SUN-1. Cell 139: 920–933. 10.1016/j.cell.2009.10.045 [DOI] [PubMed] [Google Scholar]
- Pieper GH, Sprenger S, Teis D, Oliferenko S. 2020. ESCRT-III/Vps4 controls heterochromatin-nuclear envelope attachments. Dev Cell 53: 27–41.e6. 10.1016/j.devcel.2020.01.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polesskaya A, Seale P, Rudnicki MA. 2003. Wnt signaling induces the myogenic specification of resident CD45+ adult stem cells during muscle regeneration. Cell 113: 841–852. 10.1016/S0092-8674(03)00437-9 [DOI] [PubMed] [Google Scholar]
- Pradhan R, Ranade D, Sengupta K. 2018. Emerin modulates spatial organization of chromosome territories in cells on softer matrices. Nucleic Acids Res 46: 5561–5586. 10.1093/nar/gky288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prakash A, Sengupta S, Aparna K, Kasbekar DP. 1999. The erg-3 (sterol Δ14,15-reductase) gene of Neurospora crassa: generation of null mutants by repeat-induced point mutation and complementation by proteins chimeric for human lamin B receptor sequences. Microbiology 145: 1443–1451. 10.1099/13500872-145-6-1443 [DOI] [PubMed] [Google Scholar]
- Pyrpasopoulou A, Meier J, Maison C, Simos G, Georgatos SD. 1996. The lamin B receptor (LBR) provides essential chromatin docking sites at the nuclear envelope. EMBO J 15: 7108–7119. 10.1002/j.1460-2075.1996.tb01102.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raab M, Gentili M, de Belly H, Thiam HR, Vargas P, Jimenez AJ, Lautenschlaeger F, Voituriez R, Lennon-Dumenil AM, Manel N, et al. 2016. ESCRT III repairs nuclear envelope ruptures during cell migration to limit DNA damage and cell death. Science 352: 359–362. 10.1126/science.aad7611 [DOI] [PubMed] [Google Scholar]
- Raju GP, Dimova N, Klein PS, Huang HC. 2003. SANE, a novel LEM domain protein, regulates bone morphogenetic protein signaling through interaction with Smad1. J Biol Chem 278: 428–437. 10.1074/jbc.M210505200 [DOI] [PubMed] [Google Scholar]
- Rebelo S, da Cruz ESEF, da Cruz ESOA. 2015. Genetic mutations strengthen functional association of LAP1 with DYT1 dystonia and muscular dystrophy. Mutat Res Rev Mutat Res 766: 42–47. 10.1016/j.mrrev.2015.07.004 [DOI] [PubMed] [Google Scholar]
- Rolls MM, Stein PA, Taylor SS, Ha E, McKeon F, Rapoport TA. 1999. A visual screen of a GFP-fusion library identifies a new type of nuclear envelope membrane protein. J Cell Biol 146: 29–44. 10.1083/jcb.146.1.29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rose AE, Brown RS, Schlieker C. 2015. Torsins: not your typical AAA+ ATPases. Crit Rev Biochem Mol Biol 50: 532–549. 10.3109/10409238.2015.1091804 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rothballer A, Kutay U. 2013. The diverse functional LINCs of the nuclear envelope to the cytoskeleton and chromatin. Chromosoma 122: 415–429. 10.1007/s00412-013-0417-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rothballer A, Schwartz TU, Kutay U. 2013. LINCing complex functions at the nuclear envelope: what the molecular architecture of the LINC complex can reveal about its function. Nucleus 4: 29–36. 10.4161/nucl.23387 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rowat AC, Lammerding J, Ipsen JH. 2006. Mechanical properties of the cell nucleus and the effect of emerin deficiency. Biophys J 91: 4649–4664. 10.1529/biophysj.106.086454 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samwer M, Schneider MWG, Hoefler R, Schmalhorst PS, Jude JG, Zuber J, Gerlich DW. 2017. DNA cross-bridging shapes a single nucleus from a set of mitotic chromosomes. Cell 170: 956–972.e23. 10.1016/j.cell.2017.07.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santos M, Domingues SC, Costa P, Muller T, Galozzi S, Marcus K, da Cruz e Silva EF, da Cruz e Silva OA, Rebelo S. 2014. Identification of a novel human LAP1 isoform that is regulated by protein phosphorylation. PLoS ONE 9: e113732. 10.1371/journal.pone.0113732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato A, Isaac B, Phillips CM, Rillo R, Carlton PM, Wynne DJ, Kasad RA, Dernburg AF. 2009. Cytoskeletal forces span the nuclear envelope to coordinate meiotic chromosome pairing and synapsis. Cell 139: 907–919. 10.1016/j.cell.2009.10.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaller T, Bulli L, Pollpeter D, Betancor G, Kutzner J, Apolonia L, Herold N, Burk R, Malim MH. 2017. Effects of inner nuclear membrane proteins SUN1/UNC-84A and SUN2/UNC-84B on the early steps of HIV-1 infection. J Virol 91: e00463-17. 10.1128/JVI.00463-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schirmer EC, Florens L, Guan T, Yates JR III, Gerace L. 2003. Nuclear membrane proteins with potential disease links found by subtractive proteomics. Science 301: 1380–1382. 10.1126/science.1088176 [DOI] [PubMed] [Google Scholar]
- Schober H, Ferreira H, Kalck V, Gehlen LR, Gasser SM. 2009. Yeast telomerase and the SUN domain protein Mps3 anchor telomeres and repress subtelomeric recombination. Genes Dev 23: 928–938. 10.1101/gad.1787509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schuler E, Lin F, Worman HJ. 1994. Characterization of the human gene encoding LBR, an integral protein of the nuclear envelope inner membrane. J Biol Chem 269: 11312–11317. 10.1016/S0021-9258(19)78127-7 [DOI] [PubMed] [Google Scholar]
- Sen B, Xie Z, Case N, Ma M, Rubin C, Rubin J. 2008. Mechanical strain inhibits adipogenesis in mesenchymal stem cells by stimulating a durable β-catenin signal. Endocrinology 149: 6065–6075. 10.1210/en.2008-0687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Senior A, Gerace L. 1988. Integral membrane proteins specific to the inner nuclear membrane and associated with the nuclear lamina. J Cell Biol 107: 2029–2036. 10.1083/jcb.107.6.2029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shah PP, Donahue G, Otte GL, Capell BC, Nelson DM, Cao K, Aggarwala V, Cruickshanks HA, Rai TS, McBryan T, et al. 2013. Lamin B1 depletion in senescent cells triggers large-scale changes in gene expression and the chromatin landscape. Genes Dev 27: 1787–1799. 10.1101/gad.223834.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shibuya H, Ishiguro K, Watanabe Y. 2014. The TRF1-binding protein TERB1 promotes chromosome movement and telomere rigidity in meiosis. Nat Cell Biol 16: 145–156. 10.1038/ncb2896 [DOI] [PubMed] [Google Scholar]
- Shibuya H, Hernández-Hernández A, Morimoto A, Negishi L, Höög C, Watanabe Y. 2015. MAJIN links telomeric DNA to the nuclear membrane by exchanging telomere cap. Cell 163: 1252–1266. 10.1016/j.cell.2015.10.030 [DOI] [PubMed] [Google Scholar]
- Shimi T, Butin-Israeli V, Adam SA, Hamanaka RB, Goldman AE, Lucas CA, Shumaker DK, Kosak ST, Chandel NS, Goldman RD. 2011. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev 25: 2579–2593. 10.1101/gad.179515.111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shin JY, Méndez-López I, Wang Y, Hays AP, Tanji K, Lefkowitch JH, Schulze PC, Worman HJ, Dauer WT. 2013. Lamina-associated polypeptide-1 interacts with the muscular dystrophy protein emerin and is essential for skeletal muscle maintenance. Dev Cell 26: 591–603. 10.1016/j.devcel.2013.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shrestha N, Bacsa B, Ong HL, Scheruebel S, Bischof H, Malli R, Ambudkar IS, Groschner K. 2020. TRIC-A shapes oscillatory Ca2+ signals by interaction with STIM1/Orai1 complexes. PLoS Biol 18: e3000700. 10.1371/journal.pbio.3000700 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silve S, Dupuy PH, Ferrara P, Loison G. 1998. Human lamin B receptor exhibits sterol C14-reductase activity in Saccharomyces cerevisiae. Biochim Biophys Acta 1392: 233–244. 10.1016/S0005-2760(98)00041-1 [DOI] [PubMed] [Google Scholar]
- Sobreira N, Modaff P, Steel G, You J, Nanda S, Hoover-Fong J, Valle D, Pauli RM. 2015. An anadysplasia-like, spontaneously remitting spondylometaphyseal dysplasia secondary to lamin B receptor (LBR) gene mutations: further definition of the phenotypic heterogeneity of LBR-bone dysplasias. Am J Med Genet A 167: 159–163. 10.1002/ajmg.a.36808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Solovei I, Wang AS, Thanisch K, Schmidt CS, Krebs S, Zwerger M, Cohen TV, Devys D, Foisner R, Peichl L, et al. 2013. LBR and lamin A/C sequentially tether peripheral heterochromatin and inversely regulate differentiation. Cell 152: 584–598. 10.1016/j.cell.2013.01.009 [DOI] [PubMed] [Google Scholar]
- Somech R, Shaklai S, Geller O, Amariglio N, Simon AJ, Rechavi G, Gal-Yam EN. 2005. The nuclear-envelope protein and transcriptional repressor LAP2β interacts with HDAC3 at the nuclear periphery, and induces histone H4 deacetylation. J Cell Sci 118: 4017–4025. 10.1242/jcs.02521 [DOI] [PubMed] [Google Scholar]
- Sosa BA, Rothballer A, Kutay U, Schwartz TU. 2012. LINC complexes form by binding of three KASH peptides to domain interfaces of trimeric SUN proteins. Cell 149: 1035–1047. 10.1016/j.cell.2012.03.046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sosa BA, Demircioglu FE, Chen JZ, Ingram J, Ploegh HL, Schwartz TU. 2014. How lamina-associated polypeptide 1 (LAP1) activates Torsin. eLife 3: e03239. 10.7554/eLife.03239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soullam B, Worman HJ. 1995. Signals and structural features involved in integral membrane protein targeting to the inner nuclear membrane. J Cell Biol 130: 15–27. 10.1083/jcb.130.1.15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Starr DA, Fridolfsson HN. 2010. Interactions between nuclei and the cytoskeleton are mediated by SUN-KASH nuclear-envelope bridges. Annu Rev Cell Dev Biol 26: 421–444. 10.1146/annurev-cellbio-100109-104037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Starr DA, Hermann GJ, Malone CJ, Fixsen W, Priess JR, Horvitz HR, Han M. 2001. unc-83 encodes a novel component of the nuclear envelope and is essential for proper nuclear migration. Development 128: 5039–5050. [DOI] [PubMed] [Google Scholar]
- Stroud MJ, Fang X, Zhang J, Guimarães-Camboa N, Veevers J, Dalton ND, Gu Y, Bradford WH, Peterson KL, Evans SM, et al. 2018. Luma is not essential for murine cardiac development and function. Cardiovasc Res 114: 378–388. 10.1093/cvr/cvx205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sullivan T, Escalante-Alcalde D, Bhatt H, Anver M, Bhat N, Nagashima K, Stewart CL, Burke B. 1999. Loss of A-type lamin expression compromises nuclear envelope integrity leading to muscular dystrophy. J Cell Biol 147: 913–920. 10.1083/jcb.147.5.913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun WW, Jiao S, Sun L, Zhou Z, Jin X, Wang JH. 2018. SUN2 modulates HIV-1 infection and latency through association with lamin A/C to maintain the repressive chromatin. MBio 9: e02408-17. 10.1128/mBio.02408-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tajik A, Zhang Y, Wei F, Sun J, Jia Q, Zhou W, Singh R, Khanna N, Belmont AS, Wang N. 2016. Transcription upregulation via force-induced direct stretching of chromatin. Nat Mater 15: 1287–1296. 10.1038/nmat4729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Talamas JA, Hetzer MW. 2011. POM121 and Sun1 play a role in early steps of interphase NPC assembly. J Cell Biol 194: 27–37. 10.1083/jcb.201012154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tapia O, Fong LG, Huber MD, Young SG, Gerace L. 2015. Nuclear envelope protein Lem2 is required for mouse development and regulates MAP and AKT kinases. PLoS ONE 10: e0116196. 10.1371/journal.pone.0116196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor KM, Morgan HE, Johnson A, Nicholson RI. 2004. Structure-function analysis of HKE4, a member of the new LIV-1 subfamily of zinc transporters. Biochem J 377: 131–139. 10.1042/bj20031183 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor MR, Slavov D, Gajewski A, Vlcek S, Ku L, Fain PR, Carniel E, Di Lenarda A, Sinagra G, Boucek MM, et al. 2005. Thymopoietin (lamina-associated polypeptide 2) gene mutation associated with dilated cardiomyopathy. Hum Mutat 26: 566–574. 10.1002/humu.20250 [DOI] [PubMed] [Google Scholar]
- Thaller DJ, Allegretti M, Borah S, Ronchi P, Beck M, Lusk CP. 2019. An ESCRT-LEM protein surveillance system is poised to directly monitor the nuclear envelope and nuclear transport system. eLife 8: e45284. 10.7554/eLife.45284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Theerthagiri G, Eisenhardt N, Schwarz H, Antonin W. 2010. The nucleoporin Nup188 controls passage of membrane proteins across the nuclear pore complex. J Cell Biol 189: 1129–1142. 10.1083/jcb.200912045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trelles-Sticken E, Dresser ME, Scherthan H. 2000. Meiotic telomere protein Ndj1p is required for meiosis-specific telomere distribution, bouquet formation and efficient homologue pairing. J Cell Biol 151: 95–106. 10.1083/jcb.151.1.95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trelles-Sticken E, Adelfalk C, Loidl J, Scherthan H. 2005. Meiotic telomere clustering requires actin for its formation and cohesin for its resolution. J Cell Biol 170: 213–223. 10.1083/jcb.200501042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai PL, Zhao C, Turner E, Schlieker C. 2016. The lamin B receptor is essential for cholesterol synthesis and perturbed by disease-causing mutations. eLife 5: e16011. 10.7554/eLife.16011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turgay Y, Champion L, Balazs C, Held M, Toso A, Gerlich DW, Meraldi P, Kutay U. 2014. SUN proteins facilitate the removal of membranes from chromatin during nuclear envelope breakdown. J Cell Biol 204: 1099–1109. 10.1083/jcb.201310116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turner EM, Schlieker C. 2016. Pelger-Huët anomaly and Greenberg skeletal dysplasia: LBR-associated diseases of cholesterol metabolism. Rare Dis 4: e1241363. 10.1080/21675511.2016.1241363 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tuschl K, Meyer E, Valdivia LE, Zhao N, Dadswell C, Abdul-Sada A, Hung CY, Simpson MA, Chong WK, Jacques TS, et al. 2016. Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun 7: 11601. 10.1038/ncomms11601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ulbert S, Antonin W, Platani M, Mattaj IW. 2006. The inner nuclear membrane protein Lem2 is critical for normal nuclear envelope morphology. FEBS Lett 580: 6435–6441. 10.1016/j.febslet.2006.10.060 [DOI] [PubMed] [Google Scholar]
- Ulker D, Ersoy YE, Gucin Z, Muslumanoglu M, Buyru N. 2018. Downregulation of SCARA5 may contribute to breast cancer via promoter hypermethylation. Gene 673: 102–106. 10.1016/j.gene.2018.06.036 [DOI] [PubMed] [Google Scholar]
- Ungricht R, Klann M, Horvath P, Kutay U. 2015. Diffusion and retention are major determinants of protein targeting to the inner nuclear membrane. J Cell Biol 209: 687–704. 10.1083/jcb.201409127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uzer G, Bas G, Sen B, Xie Z, Birks S, Olcum M, McGrath C, Styner M, Rubin J. 2018. Sun-mediated mechanical LINC between nucleus and cytoskeleton regulates β catenin nuclear access. J Biomech 74: 32–40. 10.1016/j.jbiomech.2018.04.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viera A, Alsheimer M, Gomez R, Berenguer I, Ortega S, Symonds CE, Santamaria D, Benavente R, Suja JA. 2015. CDK2 regulates nuclear envelope protein dynamics and telomere attachment in mouse meiotic prophase. J Cell Sci 128: 88–99. 10.1242/jcs.154922 [DOI] [PubMed] [Google Scholar]
- Vietri M, Schink KO, Campsteijn C, Wegner CS, Schultz SW, Christ L, Thoresen SB, Brech A, Raiborg C, Stenmark H. 2015. Spastin and ESCRT-III coordinate mitotic spindle disassembly and nuclear envelope sealing. Nature 522: 231–235. 10.1038/nature14408 [DOI] [PubMed] [Google Scholar]
- Vlcek S, Just H, Dechat T, Foisner R. 1999. Functional diversity of LAP2α and LAP2β in postmitotic chromosome association is caused by an α-specific nuclear targeting domain. EMBO J 18: 6370–6384. 10.1093/emboj/18.22.6370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- von Appen A, LaJoie D, Johnson IE, Trnka MJ, Pick SM, Burlingame AL, Ullman KS, Frost A. 2020. LEM2 phase separation promotes ESCRT-mediated nuclear envelope reformation. Nature 582: 115–118. 10.1038/s41586-020-2232-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wagner N, Weyhersmüller A, Blauth A, Schuhmann T, Heckmann M, Krohne G, Samakovlis C. 2010. The Drosophila LEM-domain protein MAN1 antagonizes BMP signaling at the neuromuscular junction and the wing crossveins. Dev Biol 339: 1–13. 10.1016/j.ydbio.2009.11.036 [DOI] [PubMed] [Google Scholar]
- Wang W, Shi Z, Jiao S, Chen C, Wang H, Liu G, Wang Q, Zhao Y, Greene MI, Zhou Z. 2012. Structural insights into SUN-KASH complexes across the nuclear envelope. Cell Res 22: 1440–1452. 10.1038/cr.2012.126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waterham HR, Koster J, Mooyer P, Noort Gv G, Kelley RI, Wilcox WR, Wanders RJ, Hennekam RC, Oosterwijk JC. 2003. Autosomal recessive HEM/Greenberg skeletal dysplasia is caused by 3β-hydroxysterol Δ14-reductase deficiency due to mutations in the lamin B receptor gene. Am J Hum Genet 72: 1013–1017. 10.1086/373938 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Webster BM, Colombi P, Jäger J, Lusk CP. 2014. Surveillance of nuclear pore complex assembly by ESCRT-III/Vps4. Cell 159: 388–401. 10.1016/j.cell.2014.09.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Webster BM, Thaller DJ, Jäger J, Ochmann SE, Borah S, Lusk CP. 2016. Chm7 and Heh1 collaborate to link nuclear pore complex quality control with nuclear envelope sealing. EMBO J 35: 2447–2467. 10.15252/embj.201694574 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilkie GS, Korfali N, Swanson SK, Malik P, Srsen V, Batrakou DG, de las Heras J, Zuleger N, Kerr AR, Florens L, et al. 2011. Several novel nuclear envelope transmembrane proteins identified in skeletal muscle have cytoskeletal associations. Mol Cell Proteomics 10: M110.003129. 10.1074/mcp.M110.003129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilkinson FL, Holaska JM, Zhang Z, Sharma A, Manilal S, Holt I, Stamm S, Wilson KL, Morris GE. 2003. Emerin interacts in vitro with the splicing-associated factor, YT521-B. Eur J Biochem 270: 2459–2466. 10.1046/j.1432-1033.2003.03617.x [DOI] [PubMed] [Google Scholar]
- Willer MK, Carroll CW. 2017. Substrate stiffness-dependent regulation of the SRF-Mkl1 co-activator complex requires the inner nuclear membrane protein Emerin. J Cell Sci 130: 2111–2118. 10.1242/jcs.197517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson KL, Foisner R. 2010. Lamin-binding proteins. Cold Spring Harb Perspect Biol 2: a000554. 10.1101/cshperspect.a000554 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woglar A, Jantsch V. 2014. Chromosome movement in meiosis I prophase of Caenorhabditis elegans. Chromosoma 123: 15–24. 10.1007/s00412-013-0436-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Worman HJ, Yuan J, Blobel G, Georgatos SD. 1988. A lamin B receptor in the nuclear envelope. Proc Natl Acad Sci 85: 8531–8534. 10.1073/pnas.85.22.8531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yam C, He Y, Zhang D, Chiam KH, Oliferenko S. 2011. Divergent strategies for controlling the nuclear membrane satisfy geometric constraints during nuclear division. Curr Biol 21: 1314–1319. 10.1016/j.cub.2011.06.052 [DOI] [PubMed] [Google Scholar]
- Yam C, Gu Y, Oliferenko S. 2013. Partitioning and remodeling of the Schizosaccharomyces japonicus mitotic nucleus require chromosome tethers. Curr Biol 23: 2303–2310. 10.1016/j.cub.2013.09.057 [DOI] [PubMed] [Google Scholar]
- Yang L, Guan T, Gerace L. 1997. Lamin-binding fragment of LAP2 inhibits increase in nuclear volume during the cell cycle and progression into S phase. J Cell Biol 139: 1077–1087. 10.1083/jcb.139.5.1077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yazawa M, Ferrante C, Feng J, Mio K, Ogura T, Zhang M, Lin PH, Pan Z, Komazaki S, Kato K, et al. 2007. TRIC channels are essential for Ca2+ handling in intracellular stores. Nature 448: 78–82. 10.1038/nature05928 [DOI] [PubMed] [Google Scholar]
- Ye Q, Worman HJ. 1994. Primary structure analysis and lamin B and DNA binding of human LBR, an integral protein of the nuclear envelope inner membrane. J Biol Chem 269: 11306–11311. 10.1016/S0021-9258(19)78126-5 [DOI] [PubMed] [Google Scholar]
- Ye Q, Callebaut I, Pezhman A, Courvalin JC, Worman HJ. 1997. Domain-specific interactions of human HP1-type chromodomain proteins and inner nuclear membrane protein LBR. J Biol Chem 272: 14983–14989. 10.1074/jbc.272.23.14983 [DOI] [PubMed] [Google Scholar]
- Zhang Y, Castori M, Ferranti G, Paradisi M, Wordsworth BP. 2009. Novel and recurrent germline LEMD3 mutations causing Buschke–Ollendorff syndrome and osteopoikilosis but not isolated melorheostosis. Clin Genet 75: 556–561. 10.1111/j.1399-0004.2009.01177.x [DOI] [PubMed] [Google Scholar]
- Zhao Y, Chen YQ, Bonacci TM, Bredt DS, Li S, Bensch WR, Moller DE, Kowala M, Konrad RJ, Cao G. 2008. Identification and characterization of a major liver lysophosphatidylcholine acyltransferase. J Biol Chem 283: 8258–8265. 10.1074/jbc.M710422200 [DOI] [PubMed] [Google Scholar]
- Zhao C, Brown RS, Chase AR, Eisele MR, Schlieker C. 2013. Regulation of Torsin ATPases by LAP1 and LULL1. Proc Natl Acad Sci 110: E1545–E1554. 10.1073/pnas.1300676110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhen Y, Sørensen V, Skjerpen CS, Haugsten EM, Jin Y, Wälchli S, Olsnes S, Wiedlocha A. 2012. Nuclear import of exogenous FGF1 requires the ER-protein LRRC59 and the importins Kpnα1 and Kpnβ1. Traffic 13: 650–664. 10.1111/j.1600-0854.2012.01341.x [DOI] [PubMed] [Google Scholar]
- Zhou H, Clapham DE. 2009. Mammalian MagT1 and TUSC3 are required for cellular magnesium uptake and vertebrate embryonic development. Proc Natl Acad Sci 106: 15750–15755. 10.1073/pnas.0908332106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou J, Li H, Li X, Li Y, Yang M, Shi G, Xu D, Shi X. 2019. A novel EMD mutation in a Chinese family with initial diagnosis of conduction cardiomyopathy. Brain Behav 9: e01167. 10.1002/brb3.1167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu S, Wan W, Zhang Y, Shang W, Pan X, Zhang LK, Xiao G. 2019. Comprehensive interactome analysis reveals that STT3B is required for N-glycosylation of Lassa virus glycoprotein. J Virol 93: e01443-19. 10.1128/JVI.01443-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuleger N, Kelly DA, Richardson AC, Kerr AR, Goldberg MW, Goryachev AB, Schirmer EC. 2011. System analysis shows distinct mechanisms and common principles of nuclear envelope protein dynamics. J Cell Biol 193: 109–123. 10.1083/jcb.201009068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuleger N, Boyle S, Kelly DA, de las Heras JI, Lazou V, Korfali N, Batrakou DG, Randles KN, Morris GE, Harrison DJ, et al. 2013. Specific nuclear envelope transmembrane proteins can promote the location of chromosomes to and from the nuclear periphery. Genome Biol 14: R14. 10.1186/gb-2013-14-2-r14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zullo JM, Demarco IA, Piqué-Regi R, Gaffney DJ, Epstein CB, Spooner CJ, Luperchio TR, Bernstein BE, Pritchard JK, Reddy KL, et al. 2012. DNA sequence-dependent compartmentalization and silencing of chromatin at the nuclear lamina. Cell 149: 1474–1487. 10.1016/j.cell.2012.04.035 [DOI] [PubMed] [Google Scholar]