Download as pdf or txt
Download as pdf or txt
You are on page 1of 27

Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV.

on September 11, 2023 - Published by Cold Spring


Harbor Laboratory Press

The Nuclear Lamina

Xianrong Wong,1 Ashley J. Melendez-Perez,2 and Karen L. Reddy2,3


1
Laboratory of Developmental and Regenerative Biology, Skin Research Institute of Singapore,
Agency for Science, Technology and Research (A∗STAR), Singapore 138648
2
Department of Biological Chemistry and Center for Epigenetics, Johns Hopkins University of Medicine,
Baltimore, Maryland 21205, USA
3
Sidney Kimmel Cancer Institute, Johns Hopkins University School of Medicine, Baltimore,
Maryland 21231, USA
Correspondence: kreddy4@jhmi.edu

Lamins interact with a host of nuclear membrane proteins, transcription factors, chromatin
regulators, signaling molecules, splicing factors, and even chromatin itself to form a nuclear
subcompartment, the nuclear lamina, that is involved in a variety of cellular processes such as
the governance of nuclear integrity, nuclear positioning, mitosis, DNA repair, DNA replica-
tion, splicing, signaling, mechanotransduction and -sensation, transcriptional regulation, and
genome organization. Lamins are the primary scaffold for this nuclear subcompartment, but
interactions with lamin-associated peptides in the inner nuclear membrane are self-reinforc-
ing and mutually required. Lamins also interact, directly and indirectly, with peripheral
heterochromatin domains called lamina-associated domains (LADs) and help to regulate
dynamic 3D genome organization and expression of developmentally regulated genes.

T he nucleus is structurally organized into func-


tional domains and, as an interpreter and reg-
ulator of cellular function, is unique between cell
Underlying the INM is the nuclear lamina, a
10- to 30-nm-thick filamentous meshwork,
with its thickness varying between different cell
types. In addition to expressing cell-type-specific types (Höger et al. 1991). The principal compo-
transcription factors and genome regulators, this nents of the lamina are the type V intermediate
diversity is also quite evident at the periphery of filament proteins—the lamins (Fig. 1; Gerace and
the nucleus. The nuclear periphery encompasses Huber 2012). In mammals, the lamins are
the nuclear envelope and the underlying nuclear grouped into two classes: A-type (LMNA,
lamina (Aebi et al. 1986). The nuclear envelope is LMNAΔ10, and LMNC) and B-type (LMNB1,
a dual membrane barrier composed of the inner LMNB2, and LMNB3) (Peter et al. 1989; Vor-
nuclear membrane (INM) and the outer nuclear burger et al. 1989). All metazoans express at least
membrane (ONM), which is contiguous with one B-type lamin, with most invertebrates ex-
the endoplasmic reticulum (ER). The nuclear pressing a single lamin gene, although Drosophila
envelope (NE) is studded with nuclear pore com- has one B-type (Dm0 or Dmel/Lam) and one
plexes (NPCs) that span the ONM and INM. A-type (DmeI/LamC). In mammals, most adult

Editors: Ana Pombo, Martin W. Hetzer, and Tom Misteli


Additional Perspectives on The Nucleus available at www.cshperspectives.org
Copyright © 2021 Cold Spring Harbor Laboratory Press; all rights reserved
Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113

1
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

Nuclear
integrity

Spindle
assembly

Transcriptional
regulation
Nucleocytoskeletal Nuclear
coupling positioning
Lamins
Splicing

Chromatin
organization

Signaling

DNA
replication DNA repair

Figure 1. Lamins—a critical nexus in the regulation of cellular processes. Lamins and their associated proteins
(lamin-associated proteins [LAPs] and nuclear envelope transmembrane proteins [NETs]) have been implicated
in a wide range of interdependent cellular processes placing lamins at the center of an integrative hub at the
nuclear lamina. Black arrows indicate lamin influences on specific pathways and processes and the gray lines
indicate cross talk between these processes. These processes include maintaining structural integrity of the
nucleus (Shin et al. 2013; Swift et al. 2013; Harada et al. 2014), regulation of transcription through direct
interactions with transcription factors as well as signaling pathways (Andrés and González 2009; Ho and
Lammerding 2012), and three-dimensional chromatin organization (scaffolding of lamina-associated domains
[LADs]) (van Steensel and Belmont 2017; Briand and Collas 2020), which in turn impacts nuclear integrity and
structure, since chromatin and lamins collaborate to confer mechanical properties to the nucleus (Chalut et al.
2012; Furusawa et al. 2015; Shimamoto et al. 2017; Stephens et al. 2017; Strickfaden et al. 2020). The nuclear
lamina and interacting proteins also regulate other DNA-based processes such as DNA replication (Dorner et al.
2006; Shumaker et al. 2008; Pope et al. 2014) and DNA repair (Redwood et al. 2011; Aymard et al. 2014; Gibbs-
Seymour et al. 2015; Lottersberger et al. 2015; Li et al. 2018; Marnef et al. 2019). Linker of nucleoskeleton and
cytoskeleton complex (LINC) proteins mediate nucleocytoskeletal coupling, which in turn influences other
processes such as signaling, chromatin organization, DNA repair, and gene regulation (through mechanosensa-
tion and mechanotransduction) as well as nuclear position through direct interaction of LINCs with cytoskeletal
motors (Razafsky et al. 2014; Wong et al. 2021a). Other identified but less commonly known roles of the lamins
include splicing and spindle pole assembly (Georgatos et al. 1997; Maison et al. 1997; Kumaran et al. 2002; Tsai
et al. 2006; Ma et al. 2009).

differentiated somatic cells contain four major pre-mRNA (Lin and Worman 1993; Machiels
lamin proteins (LMNA, LMNB1, LMNB2, and et al. 1996; DeBoy et al. 2017). A minor spliced
LMNC). A single gene, LMNA, encodes the A- variant, LMNC2, is also produced in the testis
type lamins LMNA and LMNC, along with other (Furukawa et al. 1994). Separate genes encode
minor variants including LMNAΔ10, which are LMNB1 and LMNB2 with LMNB3 being pro-
all generated by alternative splicing of a common duced as a minor spliced variant of LMNB2

2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

and, as with LMNC2, is found in the testis (Fu- residues in B-type lamins near the coiled-coil
rukawa and Hotta 1993; Lin and Worman 1993). domains induce the disassembly of the nuclear
All lamin isotypes share a similar overall struc- lamina. Intriguingly, a subset of A-type lamins
ture: a head domain, a central α-helical rod do- remains phosphorylated, soluble, and nucleo-
main comprising four coiled-coil domains that plasmic during interphase (Gerace and Blobel
enable lamin dimer formation, interspersed by 1980; Kochin et al. 2014; Torvaldson et al.
unstructured linkers, followed by a nuclear local- 2015; Ikegami et al. 2020). While S22P is neces-
ization signal (NLS), an Ig-fold domain ending in sary for lamin depolymerization, it is not suffi-
an unstructured carboxy-terminal tail (detailed cient; thus, CDK1 is likely working in concert
in Fig. 2). The basic unit of the lamins is the lamin with other kinases, such as protein kinase C
dimer, which forms protofilaments in a head-to- (PKC), cyclin-dependent kinase 5 (CDK5), and
tail manner and form higher-order assemblies to AKT to facilitate complete disassembly and to
make the final lamin filament structure (Dechat further regulate lamin functions (Torvaldson
et al. 2010). In mammals, recent high-resolution et al. 2015). It is important to note that the ma-
light and electron microscopy studies have re- jority of PTM sites of lamins have been identified
vealed that the A-, B-, and C-lamin isotypes by mass spectrometry without accompanying
form their own spatially separate but interacting mechanistic and functional studies, so there is
and overlapping filament networks of 3.5-A tet- still much to be understood about the dynamic
rameric filaments (Shimi et al. 2008; Turgay et al. regulation of lamins through these different
2017), with lamin C showing an apparent pref- modifications and modification sites.
erential association with nuclear pores (Xie et al. In addition to being regulated by PTMs (and
2016). This makes lamin organization in mam- splicing), expression of lamin isotypes is also de-
malian somatic cells more complex and less reg- velopmentally regulated, with expression of A-
ular than previously observed in the frog oocyte type lamins (LMNA and LMNC) being restrict-
lamina (Aebi et al. 1986). ed to more differentiated cells, and each cell type
displaying different relative ratios of the different
lamin isotypes. All nucleated mammalian cells
REGULATION OF LAMINS
express at least one B-type lamin, whereas A-
Lamin functions are regulated by a myriad of type lamins are absent during the early pre-
posttranslational modifications (PTMs) (see and postimplantation embryonic stages and in
Fig. 1), especially higher-order assembly into fil- embryonic stem (ES) cells, with these types ex-
aments and disassembly during nuclear enve- pressing high levels of LMNB1 and LMNB2
lope breakdown as cells enter mitosis. Most (Stewart and Burke 1987; Wong and Stewart
prominently, lamins are regulated by phosphor- 2020). A-type lamins are expressed as different
ylation, but are also subjected to farnesylation, tissues form in the postimplantation embryo,
ubiquitination, sumoylation, O-linked sugar although some tissues and cell types do not ex-
modification (O-GlcNAc), and acetylation (Ge- press A-type lamins until after birth (Solovei
race and Blobel 1980; Simon and Wilson 2013; et al. 2013). Lamins appear to be nonessential
Kochin et al. 2014; Gruenbaum and Foisner for ES cells, since ES cells and their derivative
2015; Torvaldson et al. 2015). While the roles progenitors lacking LMNA, LMNC, and
of the vast majority of phosphorylation and LMNB1 proliferate normally in culture, main-
other modifications to lamin proteins are not tain euploidy, and differentiate normally into
understood, there is a clear role for phosphory- multiple cell types (Kim et al. 2013). However,
lation in lamin disassembly in mitosis. Cyclin- it should be noted that the ES culture system
dependent kinase 1 (Cdk1 or cdc2) directly reflects early pre-organ development. The story
phosphorylates both A- and B-type lamins dur- is not as clear-cut in vivo, although A-type lam-
ing mitosis. Phosphorylation of amino-terminal ins are dispensable for early development. Mice
Serine 22 (S22P) and carboxy-terminal Serine lacking either LMNA, LMNC, or both are indis-
392 (S392P) in human lamin A/C or analogous tinguishable from normal siblings at birth, pos-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 3
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

A Ubiquitination

Phosphorylation

Acetylation
Sparse: O-GlcNAc,
SUMOylation
Coil 1A Coil 1B Coil 2A Coil 2B Ig fold
CAAX
Head L1 L12 L2 NLS

α-Helical central rod


B
Lamin C

Prelamin A
Farnesyltransferase
AAX endopeptidase
Carboxyl methyltransferase S
COCH3

Zmpste24
Mature lamin A

S
COCH3 Lamin B1 and B2

C Lamin monomer
Mitotic CDKs

Antiparallel arrangement, higher-order assembly

Lamin coiled-coil dimer—basic unit of assembly

Interphase
Phosphatases

Head-to-tail polymerization
into a protofilament
Mitotic exit

Figure 2. Assembly, processing, and regulation of lamins. (A) The general structure of a lamin protein, consisting
of a short unstructured head domain (yellow), a central α-helical rod domain comprised of four helical subre-
gions (coils 1A, 1B, 2A, and 2B in purple) interspersed by unstructured linkers (L1, L12, and L2), a tail region that
includes a nuclear localization signal (NLS) (green), an immunoglobulin-fold domain (Ig-fold) (red), and a fairly
unstructured carboxy-terminal end that in most cases terminates with a carboxy-terminal CAAX motif (Stuur-
man et al. 1998; Krimm et al. 2002; Herrmann and Aebi 2004). (Legend continues on following page.)

4 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

sibly because of some functional redundancy gion) (Jung et al. 2012). Less is known about the
with the developmentally regulated INM pro- cell and tissue variation of expression of somatic
tein lamin B receptor (LBR) that is expressed B-type lamins. Both LMNB1 and LMNB2 are
(or over- or re-expressed) in cells engineered to required for normal neuronal development
lack Lmna (Sullivan et al. 1999; Jahn et al. 2012; (Vergnes et al. 2004; Coffinier et al. 2011). In
Solovei et al. 2013). Loss of Lmna does, however, addition, both human and murine fibroblasts
result in severe postnatal growth retardation and with reduced levels of LMNB1 undergo senes-
death within 3 weeks, which is correlated to cence and display reduced proliferation and loss
some extent with the normal silencing of LBR of LMNB1 correlates with keratinocyte senes-
expression in many postnatal tissues (Sullivan cence in vivo (Shimi et al. 2011; Dreesen et al.
et al. 1999; Jahn et al. 2012; Solovei et al. 2013). 2013; Shah et al. 2013). In adults, B-type lamin
Intriguingly, the ratio of LMNA/LMNC varies expression appears to be nonessential in some
across different cell types suggesting cell-type- tissues, such as the skin epidermis and liver
specific and developmental regulation of LMNA (Yang et al. 2011). These findings reveal that
splicing or posttranscriptional regulation. One an absolute dependence on lamin expression
striking example of differential A/C expression in mammals varies among different cell types
is in neurons where LMNA (and not the LMNC and that early embryos, including pluripotent
splice variant) translation is inhibited by the mi- cells and some of their differentiated derivatives,
croRNA miR-9, which binds specifically to the may not require any of the lamins for their pro-
longer LMNA mRNA 30 UTR (untranslated re- liferation and early differentiation.

Figure 2. (Continued) While the rod domain is involved in the dimerization of lamins through coiled-coil
interactions, the head and tail domains play a significant role in regulating the higher assembly of lamins (Gieffers
and Krohne 1991; Heitlinger et al. 1992; Stuurman et al. 1996; Klapper et al. 1997; Sasse et al. 1998; Izumi et al.
2000; Moir et al. 2000a; Ben-Harush et al. 2009). Also shown are potential posttranslational modifications
(PTMs) and their documented density occurring throughout the monomer where the color intensity is propor-
tional to the probability of finding the PTMs. The most highly occurring PTMs, as shown, are phosphorylation
and ubiquitination, which exhibit an inverse distribution, with phosphorylation more often occurring near and at
the head and tail domains while ubiquitination more frequently found within the central rod domain and
extending toward the Ig fold. The distribution pattern of acetylation is similar to that of ubiquitination, albeit
less frequently occurring. In contrast, O-GlcNAcylation and SUMOylation are sparse and have been found only
on a few residues (Simon and Wilson 2013). (B) A-type lamins (LMNA and LMNC) are the result of alternative
splicing from the LMNA gene, while the major B-type isoforms (LMNB1 and LMNB2) are transcribed from two
different genes. Both LMNA and LMNB1/2 are farnesylated at the cysteine residue of the –CAAX motif by a
farnesyltransferase, followed by the removal of the last three amino acids by means of an AAX endopeptidase and
finally carboxymethylation via carboxyl methyltransferase (Dechat et al. 2008). Lamin B1 and B2 remain
farnesylated, facilitating their anchorage to the nuclear envelope. LMNA undergoes an additional step: removal
of the farnesyl group and the 15 most carboxy-terminal residues by the protease Zmpste24, rendering a fully
functional and mature, lamin A protein (Dechat et al. 2008). (C) Depicts principles of lamin assembly and their
regulation through the cell cycle. Lamin monomers associate in a parallel, head-to-head manner, leading to a
coiled-coil dimer formation—the basic unit for higher-order assembly. As cells enter mitosis, higher-order lamin
filaments localized at the nuclear lamina become phosphorylated by cyclin-dependent kinase 1 (CDK1) and
potentially other kinases triggering the disassembly of the nuclear lamina as lamins depolymerize and become
cytosolic. A-type lamins are found in the cytosol, while depolymerized B-type remain anchored to lipid mem-
branes (Gerace and Blobel 1980). As the cell approaches interphase, lamins are dephosphorylated by protein
phosphatase 1a (PP1a) and are recruited to and reassembled at the reforming NE (Thompson et al. 1997; Dechat
et al. 2010). This reincorporation at the NE is temporally regulated, with B-type lamins organizing to the lamin
first, followed by lamin A and then lamin C networks (Pugh et al. 1997; Shimi et al. 2015; Xie et al. 2016; Wong
et al. 2020). Importantly, and particularly for A-type lamins, a subpopulation of lamins can remain nucleoplas-
mic, due to the persistence of mitotic PTMs such as S22P (Simon and Wilson 2013; Gruenbaum and Medalia
2015; Ikegami et al. 2020; Wong et al. 2020).

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 5
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

LAMINS AS A SCAFFOLD FOR THE LAMINA 2007; Lee et al. 2001; Shumaker et al. 2001; Shimi
INTERFACE et al. 2004; Margalit et al. 2007; Kind and van
Steensel 2014). Mouse and human LEM domain
Lamins interact with a host of nuclear mem- proteins include LAP2 (lamina-associated poly-
brane proteins, transcription factors, chromatin peptide 2), emerin, and MAN1 along with other
regulators, signaling molecules, splicing factors, LEM and LEM-like proteins (Lin et al. 2000;
and even chromatin itself to form a nuclear sub- Schirmer et al. 2003; Lee and Wilson 2004;
compartment that is involved in a variety of cel- Brachner et al. 2005; Chen et al. 2006; Ulbert
lular processes such as the governance of nuclear et al. 2006; Simon and Wilson 2013). These
integrity, nuclear positioning, mitosis, DNA re- LEM domain proteins, like lamins, are strongly
pair, DNA replication, splicing, mechanotrans- conserved, reflecting their fundamental roles in
duction and -sensation, transcriptional regula- the nucleus. LEM proteins primarily reside in
tion, and genome organization. The INM and the INM, but LAP2 has several isoforms, at least
the nuclear lamina are interdependent struc- one of which, LAP2α, lacks a transmembrane
tures held together by mutual molecular inter- domain and is nucleoplasmic; thus, LAP2 can
actions between a myriad of INM proteins and interact with both nucleoplasmic and INM-as-
the underlying lamina meshwork (Fig. 3; Simon sociated lamins (Harris et al. 1994; Berger et al.
and Wilson 2013; Wong et al. 2014, 2021b). 1996; Dechat et al. 1998). Nucleoplasmic LAP2α
The protein composition at the nuclear peri- interacts with nucleoplasmic lamin A (likely
phery has been profiled in various cell types phosphorylated) and has been implicated in
using different approaches. Initially, lamin- gene activation (Gesson et al. 2016). LEM do-
associated proteins (LAPs) were identified by main proteins are translated into the ER mem-
cofractionation in nuclei extracted with high brane and diffuse into the ONM and then re-
concentrations of monovalent salts and ionic tained in the INM via interactions with lamin
detergents (Senior and Gerace 1988; Gerace proteins (Holmer and Worman 2001; Wilson
and Foisner 1994). More recently, nuclear enve- and Foisner 2010; Berk et al. 2013); thus, pertur-
lope transmembrane proteins (NETs) have been bations of the nuclear lamina can lead to deloc-
identified by mass spectrometry identification alization of these proteins.
proteins extracted from the NE (Korfali et al. Additional well-studied lamin-interacting
2012; de Las Heras et al. 2013, 2017; Worman proteins include INM proteins such as LBR
and Schirmer 2015). Additional LAPs have been and linker of nucleoskeleton and cytoskeleton
identified by BioID, a technique to biotinylate complex (LINC) proteins (Fig. 2). LBR is an
proximal proteins, followed by mass spectrom- eight-pass transmembrane protein localized at
etry (Kim et al. 2016; Mehus et al. 2016; Xie et the INM that contains a nucleoplasmic domain
al. 2016; Wong et al. 2021b). While the terms that codes for a sterol reductase. LBR is required
may be used interchangeably for the most part, it for nuclear shape changes, as seen in neutro-
is important to note that LAPs are presumed to phils, and is developmentally regulated, with
interact with lamins, but not necessarily the NE, higher expression in ES and progenitor cells.
since some lamins are nucleoplasmic and, con- LBR has been shown to interact with heterochro-
versely, NETs are present in the nuclear enve- matin organizers HP1 (heterochromatin protein
lope, but may not interact with lamins or be at 1) and PRR14 (proline-rich protein 14), and to
the INM. Many new tissue-specific NETs have interact with the repressive heterochromatin
not been fully characterized. modification histone H4 lysine 20 dimethylation
The LEM (Lap2-Emerin-Man1) domain (H4K20me2) (Ye et al. 1997; Polioudaki et al.
family of proteins are LAPs/NETs containing 2001; Hirano et al. 2012; Dunlevy et al. 2020).
a LEM domain that binds a conserved metazoan The LINC complex spans the NE and is formed
chromatin protein, barrier to autointegration by interactions between SUN domain proteins of
factor (BAF), allowing their indirect interaction the INM (SUN1 and SUN2 in somatic cells) and
with chromatin (Furukawa 1999; Cai et al. 2001, the KASH domain proteins of the ONM (nes-

6 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

Microtubules Cytoplasm
Intermediate
F-actin filaments

LINC Nesprin ONM


complex
NPC
SUN protein
PNS
NE INM
Ts Zone 1

NETs
Ts LEM Lamina

LAP
NE
LAP
Zone 2
LAP

HP1 Peripheral heterochromatin/LADs


Peripheral heterochromatin/LADs Zone 3
D s LAD
LAD LA tein
o proteins
proteins pr
Nucleoplasmic lamins

ct

A
iv e p Nucleoplasm
c h r o m a ti n l o o

Figure 3. Structure and complexity at the nuclear periphery. The nuclear lamina serves as an integrative hub for
various aspects of nuclear and cellular functions (Wong et al. 2021b). The lamin meshwork(s) (gray), serves to
maintain nuclear integrity as well as act as a structural scaffold, anchoring a diverse range of proteins and
heterochromatin domains (lamina-associated domains [LADs]) to the nuclear envelope (NE). Lamin-associated
proteins (LAPs) and some nuclear envelope transmembrane proteins (NETs) interact with lamins and chroma-
tin. Relatively more well studied, NETs/LAPs include the components that form the linker of cytoskeleton and
nucleoskeleton complexes (LINCs) (SUN and KASH domain-containing nesprins), LEM domain proteins, and
lamin B receptor (LBR) ( pink), shown above. Additionally, the NE also associates with interphase heterochro-
matin (red chromatin—LADs) providing a structural basis for establishing interphase chromosome topology and
at the same time providing a means for transcriptional gene regulation through repression of genes sequestered at
the periphery. The tethering of chromatin at the nuclear periphery can be achieved via LBR interactions or lamin
A/C. LBR is known to interact directly with chromatin binders (such as Hp1α and PRR14) while A-type lamins
may mediate chromatin contacts through LAPs and chromatin binders and modifiers (orange circles) interacting
with LADs. A recent comparative proteomics study querying the interaction of lamins, LAP2β and LADs, further
resolved the nuclear periphery into three functional zones with respect to chromatin regulation. Zone 1 includes
proteins at the INM/lamina that do not interact with LADs, Zone 2 is comprised of proteins that bind the
INM/lamina network and LADs—the “middlemen,” and Zone 3 contains only proteins associating uniquely
with LADs and chromatin (Wong et al. 2021b).

prins) (Méjat and Misteli 2010; Rothballer and also involved in the cytoplasmic motor-driven
Kutay 2013; Chang et al. 2015; Kim et al. 2015). movement of the nucleus within the cell (Luxton
KASH domain proteins are able to interact with et al. 2010; Wu et al. 2014) and have been impli-
cytoskeletal elements (actin, myosin, and cytosol- cated in gene regulation as well (Kim and Wirtz
ic intermediate filaments); thus, LINCs physically 2015; Tajik et al. 2016). Specific isoforms of nes-
link the nucleoplasm and lamina to the cytoskel- prins are expressed in different cell types (Duong
eton, ultimately connecting to the extracellular et al. 2014) and some KASH-less nesprin isotypes
matrix (ECM) via integrins. It is through these can be found at the INM and interact with lamins
interactions that the nuclear lamina acts as a crit- and emerin directly (Rajgor and Shanahan 2013;
ical mechanosensing and transduction node Kim et al. 2015).
(Lombardi and Lammerding 2011; Osmanagic- As mentioned above, during mitosis, lamins
Myers et al. 2015; Belaadi et al. 2018; Wang et al. are phosphorylated and disassembled. Likewise,
2018; Maurer and Lammerding 2019). LINCs are LAPs and BAFs are also phosphorylated, thus

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 7
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

leading to loss of LAP–lamin interactions analyses, during myogenesis. These types of stud-
(Haraguchi et al. 2001; Foisner 2003; Ellenberg ies will undoubtedly continue to identify addi-
2013). This leads to the vesiculation or move- tional novel and differential NET proteins and
ment into the mitotic ER of these INM proteins. profiles. How such changes to the nuclear periph-
At the onset of anaphase/telophase, these pro- eral proteome affects cellular process and behav-
teins are dephosphorylated and ER/vesicle ior and how their deregulation would allow the
membranes are recruited back onto chromatin manifestation of disease phenotypes would be of
to reform the nuclear envelope and subsequently particular interest (Wong et al. 2014).
recruit the lamins. Interestingly, the LEM do-
main proteins emerin and LAP2β appear to in-
LAMINA-ASSOCIATED DOMAINS (LADs)
teract with condensed chromatin (mediated
through BAF, which is also regulated by mitotic The genome is functionally organized, with re-
phosphorylation) and interact with different gions of late-replicating heterochromatin posi-
chromatin domains than LBR during NE assem- tioned at the nuclear lamina and at the nucleolar
bly (Haraguchi et al. 2001; Dechat et al. 2004). periphery in most metazoan cells (Cremer and
Thus, while in the interphase, nuclear lamins Cremer 2001). Conversely, more euchromatic re-
retain the LAP INM proteins and prevent their gions are positioned in the nuclear interior or
redistribution to the ONM and ER, after mitosis interact with NPC (Buchwalter et al. 2019). Not
membrane-bound LAPs scaffold on chromatin surprisingly, lamins have long been implicated in
and subsequently recruit the nuclear lamins, regulating the three-dimensional (3D) organiza-
highlighting the interdependence of interactions tion of chromatin, particularly via these hetero-
and the lamina interface. chromatin domains found at the nuclear lamina.
3D genome organization and chromatin com-
partments were initially identified and studied
CELL- AND TISSUE-SPECIFIC INM/LAMINA
by microscopy and cytological tools using DNA
PROTEOMES
stains, DNA hybridization techniques such as
Many of the proteins that are resident to the pe- fluorescent in situ hybridization (FISH), or elec-
ripheral zone of the nucleus—the lamins, LAPs, tron microscopy. These early studies led to an
and NETs—are differentially expressed (Figs. 3 understanding of nonrandom organization of
and 4; Furukawa and Hotta 1993; Furukawa chromatin, including identification of chromo-
et al. 1994; Alsheimer et al. 1999; Schütz et al. some territories (CTs) and putative regulatory
2005a,b; Chen et al. 2006; Korfali et al. 2010, subdomains within the nucleus, including
2012; Jung et al. 2012; Solovei et al. 2013). This the lamina. These studies relied heavily on iden-
has been highlighted in several proteomic studies tifying spatial relationships between a known se-
using isolated NE from different cell and tissue quence (or sequences) and a protein compart-
types that have identified many new tissue- and ment (such as the lamina) in the nucleus.
cell-specific NET proteins. These experiments More recently, deep-sequencing-based ap-
uncovered an impressive proteomic diversity of proaches have led to genome-wide and molecu-
this cellular compartment. In particular, a com- lar-level understanding of genome organization
parative study across three disparate tissues (liver, and identification of specific DNA sequences and
leukocytes, and muscle) revealed that the major- features that accompany such 3D architecture.
ity of the 598 NETs displayed distinct expression Three different molecular methods and their
profiles between the tissues examined, with only a derivatives are most routinely used to measure
modest 16% of these identified NETs being genome organization: chromatin immunoprecip-
shared across all three tissue types (Korfali et al. itation (ChIP) (Ma and Zhang 2020), the proxim-
2012). Other high throughput studies have doc- ity-labeling technique DNA adenine methyltrans-
umented differential NE proteomes even in less ferase identification (DamID) (Orian et al. 2009),
disparate cell types, such as during T-cell activa- and chromatin conformation capture methods
tion and in addition, using mRNA expression such as Hi-C (Lieberman-Aiden et al. 2009; Be-

8 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

A
Skeletal muscle: Blood
NET9, NET14a, (non-
NET25, NET32, erythrocyte):
NET37, NET39,
Emerin, Man1, Net 9, Net 25, Net 32,
Lamin B1, Lap2, Emerin, Lap2, LBR, Man1,
Lamin A, Lamin C, Lamin B1 (relative levels
LBR, Lamin B2 + not determined). Lamin
muscle-specific NETs A/C, Net 14a, Net 37, Net
39, + blood-specific NETs

Heart:
Net 39, Lamin A, Lamin C,
Liver: Emerin, Net9, Net 14a,
Net 9, Net 37, Lap2, Net 25, Net 32,
Lamin A, Lamin C, Net 37, Lamin B1,
Net14a, Net25, Net32, Lamin B2, Man1, LBR
Lamin B1, Lamin B2, (proteomic studies on
Lap2, Emerin, Man1, hearts not availble)
LBR, NET39 + liver-
specific NETs Fat tissue:
Tissue-specific
proteome speculated
(no comprehensive
proteomic study
performed)
Low High

+ Lamin mutation associated


B Wild-type
with cardiomyopathy

Cardiac-specifc changes in LAD organization

No change in apipose tissue

Wild-type lamin
Cardiomyocyte-specific NET
Cardiomyopathy lamin mutant
Dysfunctional cardiomyocyte-
(Pre) adipocyte-specific NET specific NET

Figure 4. Tissue- and cell-type proteomic diversity at the nuclear lamina. (A) The inferred tissue-specific
expression of nuclear envelope transmembrane proteins (NETs) determined by a transcriptomic analysis
(Chen et al. 2006). For this representation, the proteins were binned into three expression groups for each tissue
type examined: low (green), medium (orange), and high (red). It is clear, even given the small number of proteins
considered here, how different the NE proteome is in divergent cell types. It has been shown for a few cardio-
myopathy lamin mutations that chromatin organization is perturbed in induced pluripotent stem cells derived
cardiomyocytes in the form of gain and loss of lamina-associated domains (LADs), thereby affecting transcrip-
tional output and lineage commitment. (Legend continues on following page.)

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 9
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

laghzal et al. 2017). ChIP is used to identify chro- vanced microscopy approaches, including super-
matin domains and binding sites of chromatin resolution FISH technologies and live cell imag-
interactors, while DamID and Hi-C directly mea- ing, to complement and extend the molecular
sure 3D genome organization, with DamID and findings and to map single-cell chromatin confor-
related techniques measuring proximity to a pro- mations and locus positioning in single cells in
tein nuclear compartment and Hi-C-related tech- situ (Boettiger and Murphy 2020; Kempfer and
niques measuring chromatin–chromatin interac- Pombo 2020). While genome-wide sequencing
tions. DamID is most often used to detect approaches have the advantage of higher resolu-
peripheral heterochromatin domains, the so- tion (in base pairs) and numbers of cells pro-
called LADs (Greil et al. 2006; Vogel et al. 2007). cessed, imaging approaches are inherently single
Hi-C is used to identify both local and long-range cell, thus giving an immediate output of cell–cell
chromatin interactions (Dekker 2002; Lieberman- variability, but also provide additional contextual
Aiden et al. 2009; Dixon et al. 2012; Phillips- information, such as cellular state and changes
Cremins et al. 2013; Rao et al. 2014; Belaghzal over time. Integration of different data types is
et al. 2017), in particular, local self-interacting re- key to understanding genome organization gen-
gions called topologically associated domains erally and this also applies to LADs.
(TADs) and, in active regions of the genome, pro- As identified by DamID, LAD regions are
moter–enhancer interactions. Of importance to large (>100 kb), AT-rich, mostly heterochromatic,
LAD organization, Hi-C also identifies longer- and largely correlate with the inactive B compart-
range chromosome and genome-wide self-inter- ment as determined by Hi-C (Fig. 5; Dixon et al.
acting domains: the A (active) and B (inactive) 2012; Rao et al. 2014; Fraser et al. 2015; Robson
compartments, with activity state of the compart- et al. 2017). Although CTCF is found at the
ments being defined via intersections of other data boundaries between LADs and non- or inter-
(such as RNA-seq or chromatin state maps by LAD (iLAD) regions (Guelen et al. 2008), thus
ChIP) (Dekker 2002; Lieberman-Aiden et al. demarcating the transition between B and A com-
2009; Dixon et al. 2012; Rao et al. 2014). It is partments, it is depleted within the LADs them-
important to note that special care must be taken selves (Guelen et al. 2008; Harr et al. 2015), sug-
in ChIP assays to identify chromatin state and gesting that LADs (and the B compartment) have
lamin interactions in heterochromatic regions a fundamentally different organization than the
since these regions are resistant to traditional CTCF and cohesin-mediated looping structures
ChIP fragmentation and isolation protocols (Das found in the active iLAD regions (A compart-
et al. 2004; Gesson et al. 2016). An alternative ment). Indeed, depletion of either CTCF or cohe-
method to DamID is TSA-seq (tyramide signal sin leads to loss of observable TAD and sub-TAD
amplification–seq), another proximity-based organization, but A/B compartmentalization is
method to identify geographic chromatin do- largely maintained with only minor changes,
mains, including LADs (Chen et al. 2018b). These most strikingly the movement of some inactive
molecular methods are often combined with ad- regions previously constrained in the A compart-

Figure 4. (Continued) The same mutations, however, have no impact on LAD architecture in hepatocytes and
adipocytes, underscoring the tissue specificity of lamin mutations (Shah et al. 2021). (B) How tissue-specific
proteomes at the nuclear periphery can allow differential susceptibility to lamin mutations. In the top panel, the
expression of a lamin mutation associated with cardiomyopathy disrupts the function of a cardiomyocyte-
specific NET protein (heart shape to broken heart) that may be involved in LAD anchorage, leading to the
loss of LADs at the periphery. In adipocytes, however, a different tissue-specific NET may be involved in LAD
tethering (depicted as lipid droplets), the function of which is not affected by the same lamin mutation and hence
LAD architecture remains unperturbed. It is important to note that such structural reorganization could result
from a mutation in either lamin or the NET protein itself. Crossed-out proteins indicate a documented absence of
these NETs from the indicated tissue. (Figure adapted from Wong et al. 2014.)

10 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

Chromosome territories

Nuclear envelope
Nuclear lamina
LADs

non-LADs
H3K9me2/3
H3K27me3
CTCF

Chromatin loops

A compartment

B compartment

TADs

DNA

Figure 5. Higher-order genome organization in mammalian cells. Shown is a schematic of the levels of chromatin
folding within the higher-order 3D genome organization. The DNA interacts with histone octamers and aggre-
gates forming nucleosome arrays that are more or less compacted, depending on the histone variants present and
the posttranslational modifications (PTMs) to their amino-terminal tails. The next level of organization is the
formation of topologically associated domains (TADs), which in active chromatin domains are formed by loop
extrusion via cohesin and stabilized by CCCTC-binding factor (CTCF) (Dixon et al. 2012; Rao et al. 2014;
Sanborn et al. 2015). TADs and sub-TADs range from tens of kilobases to megabase-sized and have delimited
boundaries and high rates of interactions inside of these domains. TADs segregate based on their transcriptional
status into active A and inactive B compartments, with A compartments mostly occupying the nuclear interior
and B compartments associated with transcriptionally repressive nuclear domains enriched in histone H3 lysine
9 di- and trimethylation (H3K9me2/3) and histone H3 lysine 27 trimethylation (H3K7me3) at the NE and the
periphery of the nucleoli (Lieberman-Aiden et al. 2009; Harr et al. 2015, 2016; Guelen et al. 2008). It is important
to note that A/B compartments exist even if TADs are depleted, suggesting that segregation into A/B compart-
ments is not driven by TAD architecture (Nora et al. 2017; Schwarzer et al. 2017b). The geographic organization
of the B compartment to the nuclear envelope (NE) aids in the establishment and/or maintenance of interphase
chromosome topology and hence overall genome organization. Moreover, since the nuclear lamina is a tran-
scriptionally repressive compartment, it represents a manipulative model for transcriptional regulation via spatial
organizational changes relative to the lamina. The territorial organization of chromosomes in interphase (chro-
mosome territories [CTs]) constitutes a basic feature of nuclear architecture and these other levels of organization
occur within the context of CTs (Cremer and Cremer 2010). (Figure based on data in Maeshima et al. 2020.)

ment to the B compartment (Nora et al. 2017; 2019). It remains unclear, however, whether
Schwarzer et al. 2017a). Thus, the genome re- LADs remain geographically positioned at
mains partitioned, even if reconfigured (Nora the nuclear lamina in the absence of CTCF or
et al. 2017; Schwarzer et al. 2017b; Falk et al. cohesin.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 11
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

GEOGRAPHIC ORGANIZATION OF LADs cec-4 null worms, this second pathway was dis-
IS REGULATED BY CHROMATIN STATE covered to be a sequestration of euchromatin
away from the lamina (Cabianca et al. 2019). In
LADs are enriched in histone H3 lysine 9 di- and this case, the delocalization of CBP/p300 causes
trimethylation (H3K9me2/3) and this modifica- aberrant H3K27Ac to replace H3K27me3, sug-
tion is also required for LAD recruitment to and gesting that sequestration of active chromatin
maintenance at the nuclear lamina (Guelen et al. modifiers is a driving force for tethering of het-
2008; Towbin et al. 2012; Harr et al. 2015). In erochromatin to the lamina in differentiated cells.
mammals, proline-rich protein 14 (PRR14) has Similarly, in a recent study in mouse T-cells in
been shown to anchor these regions to the lamina which a LAD border region at the T-cell receptor
through its interactions with heterochromatin (TCR) locus was deleted, invading H3K27ac was
protein 1 (HP1), a chromodomain protein that found to drive an enhancer region away from
binds H3K9me3 (Poleshko and Katz 2014; Dun- the nuclear lamina and subsequently led to al-
levy et al. 2020), while in Caenorhabditis elegans tered enhancer–promoter interactions (Chen
this function is carried out by the INM-bound et al. 2018a). The role of histone H4 lysine 20
chromodomain protein CEC-4 (Towbin et al. dimethylation (H4K20me2), a repressive chro-
2012; Gonzalez-Sandoval et al. 2015; Gonzalez- matin mark bound by the tudor domain of LBR
Sandoval and Gasser 2016; Harr et al. 2016, 2020; (Hirano et al. 2012), in LAD regulation and
Bian et al. 2020). Intriguingly, heterochromatic organization remains unclear. H4K20me2 has
regions have been shown to accrete due to phase a wide genome distribution with enrichment
separation directed by HP1α, and the organiza- on centromeric and telomeric chromatin and
tion of LADs at the lamina likely depends upon does not appear to be particularly enriched on
such biophysical forces (Larson et al. 2017; Strom LADs (Mattout et al. 2015). However, this mod-
et al. 2017; Falk et al. 2019). In particular, bio- ification does seem to play a role in LAD regu-
physical models predict that, in the absence of an lation/organization under certain conditions,
active constraint at the nuclear lamina, LADs particularly senescence and in particular lami-
would form large clusters in the nucleoplasm nopathies (Shumaker et al. 2006; Barski et al.
(Falk et al. 2019). This is supported by studies 2007; Bártová et al. 2008; Mattout et al. 2015;
showing that in cells lacking lamin A/C and Nelson et al. 2016). Taken together, these stud-
LBR (either naturally or engineered) heterochro- ies suggest a complex interplay between chro-
matin configuration is inverted (Solovei et al. matin state and chromatin-binding proteins in
2013). Thus, the heterochromatic nature of the separation and organization of LADs to the
LADs leads to their separation from euchromatic periphery.
regions, but other forces and interactions are like-
ly at play in geographically organizing these re-
GENE REGULATION IN LADs
gions to the lamina. There is a less clear role for
the facultative heterochromatin modification his- LAD regions contain relatively few genes, but
tone H3 lysine 27 trimethylation (H3K27me3) in are, conversely, enriched in developmental and
LAD organization, since this modification is lineage-specific genes, leading to the hypothesis
found mostly outside of LADs, but is enriched that the epigenetic state of these regions is tied
at LAD borders (Guelen et al. 2008; Harr et al. to both organization and developmental con-
2015) and necessary to target an inserted test trol of gene expression (Guelen et al. 2008;
locus to the lamina in coordination with Peric-Hupkes et al. 2010; Bian et al. 2013;
H3K9me2/3 (Harr et al. 2015). In contrast, in Harr et al. 2015). Indeed, early studies of regu-
C. elegans, H3K27me3 modifications seem to lation of individual genes by the nuclear lamina
be dispensable for driving peripheral localization, focused on developmentally regulated gene loci,
but there is a second pathway for lamina locali- such as the immunoglobulin heavy chain locus
zation independent of Cec-4/H3K9me3 anchor- (Igh) in B-cell development (Kosak et al. 2002),
ing (Towbin et al. 2012). In a clever screen using the β-globin locus in erythroid development

12 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

(Bian et al. 2013), and MyoD in muscle devel- showed a two- to threefold reduction in expres-
opment (Yao et al. 2011) among others. These sion after tethering using a truncated emerin to
early studies pointed to the dynamic nature of tether a lac operator array (lacO) (Reddy et al.
loci associated with (and presumably regulated 2008). A similar study, using Lap2β to target a
by) the nuclear lamina during development, lacO array in a human cell line, found only a
but it was not until genome-wide mapping minimal reduction in the recruited reporter
through DamID that the shifting LAD organi- gene expression, while a subset of endogenous
zation, chromatin, and expression landscape flanking genes on the same chromosome did
could be fully appreciated. LADs are dynamic show reduced expression (Finlan et al. 2008). In
between cell states and in a simplistic view can Drosophila, the tethering of two lacO tagged re-
be divided into constitutive or facultative LADs, porter genes by Dme/LamC to the nuclear lamina
cLADs, and fLADs, respectively (Peric-Hupkes caused substantial repression, but the level of re-
et al. 2010; Meuleman et al. 2012; van Steensel pression was integration site and reporter gene
and Belmont 2017). cLADs are LADs that re- dependent (Dialynas et al. 2010). These early re-
main irrespective of cell type, while fLADs re- sults suggested that recruitment to the lamina
organize between cell types or developmental could cause repression, but that this repression
stages. The majority of LADs do not change was variable depending upon local chromatin
between individual cell types, although 70% of context and different promoters. A more recent
LADs are fLADs; thus, reorganization is re- study sought to investigate exactly this question
stricted by cell type. fLADs are more gene and found that upon integration into LADs some
rich and in cell types where they undergo reor- promoters are more sensitive to the more repres-
ganization away from the lamina, genes in these sive environment and are almost universally
regions are activated or poised for activation repressed, while other promoters can “escape”
(Peric-Hupkes et al. 2010). Even within a given silencing within LAD regions (Leemans et al.
cell type or population there is some cell–cell 2019). These escapers were generally less sensitive
heterogeneity in LAD organization, as detected repressive domains, particularly H3K27me3. All
by low-resolution, single-cell DamID, with promoters showed some variation in regulation
more gene-dense LADs displaying greater fluc- by LADs and less repressed promoters were
tuations in their association with the lamina, found to reside in more weakly (and perhaps
implying that these disruptions may be due to transiently) lamina-bound regions within LADs
differential gene usage between cells (Kind et al. (Leemans et al. 2019). Thus, even though LADs
2015). A level of heterogeneity in reorganizing are generally repressive, genes respond differently
to the lamina after cell division has also been depending upon promoter type and local LAD
noted, with some LADs appearing to become context.
nucleolar-proximal in the daughter cells, al-
though there is some expected overlap between
ROLE OF TRANSCRIPTION FACTORS IN LAD
nucleolar-associated domains (NADs) and
ORGANIZATION
LADs given the invaginations of nuclear lamins
at nucleoli noted in many cell types (Cremer and While both lamins and chromatin are important
Cremer 2001; Legartová et al. 2014; Padeken and for organization of LADs to the nuclear lamina,
Heun 2014). there is some evidence that transcription factors
In addition to LAD heterogeneity between and machinery may be more directly involved in
cells, it has been documented that different pro- peripheral localization (and regulation) of genes
moters respond differently to integration into a at the lamina. Several transcription factors have
LAD and to different environments within a been found to interact with lamins and/or
LAD. Early ectopic lamina tethering experiments lamin-interacting proteins, and some of these
suggested that recruitment to the nuclear lamina have been proposed to be more directly involved
reduces expression of the tethered locus. In one in organizing specific loci to the lamina. One
report, an integrated reporter gene in mouse cells study found that sequences from two different

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 13
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

LAD-regulated loci enriched in GA dinucleo- LAD ORGANIZATION HELPS ENFORCE


tides were able to drive an ectopic region to CHROMATIN AND GENE EXPRESSION
the lamina (Zullo et al. 2012). This is interesting PROGRAMS
given that GA dinucleotides are not enriched in
the relatively AT-rich LADs (Guelen et al. 2008). If chromatin state drives lamin association, is as-
These motifs were found to bind a cell-type-spe- sociation with the nuclear lamina simply a by-
cific transcription factor (ZBTB7B, also known product of this chromatin state and biophysical
as THPOK) in complex with a histone deacety- properties? It is difficult to tease apart the role
lase (HDAC3), and an inner nuclear membrane that chromatin state itself versus organization to
protein (LAP2β) to mediate de novo interac- the lamina plays. As discussed previously, re-
tions with the nuclear lamina. Both ZBTB7B cruiting inserted reporter genes to the lamina is
and HDAC3 were also found to be important able to repress some genes, depending on pro-
for myogenesis and in regulating myogenic moters and integration site (Finlan et al. 2008;
genes and their organization to the lamina Reddy et al. 2008). In addition, biophysical mod-
(Poleshko et al. 2017), suggesting that specific eling shows that in the absence of nuclear periph-
transcription factor complexes can regulate lam- ery “tethers,” heterochromatin will not organize
ina interactions, potentially directly. ZBTB7B to the lamina, but is instead predicted to collapse
features a BTB–POZ domain that recruits Poly- into the center of the nucleus and form an “in-
comb repressive complex 2 (PRC2) to chroma- verted” type of chromatin organization (Falk
tin, thereby initiating trimethylation of histone et al. 2019). This implies that there are active
H3 on lysine 27 (H3K27me3) (Boulay et al. mechanisms to organize LADs to the nuclear
2012). Interestingly, there are numerous BTB- lamina. In support of this, several studies have
POZ domain transcription factors that are ex- implicated A-type lamins as especially important
pressed in a cell-type-specific manner, and these for the geographical organization of LADs to the
have been found to both activate and repress lamina in differentiated cells (Solovei et al. 2013;
gene expression and to interact with HDAC3. Harr et al. 2015; Zheng et al. 2018). In particular,
HDAC3 itself, which interacts with numerous the predicted inverted chromatin organization is
transcription factor complexes, has been shown noted in the absence of both A-type lamins and
in independent studies to interact with both LBRs (Solovei et al. 2013); thus, chromatin orga-
LAP2β and emerin (Somech et al. 2005; Dem- nization to the lamina is not the default. But is
merle et al. 2012). Whether this complex of there evidence that loss of lamin association af-
ZBTB7B, HDAC3, and LAP2β initiates locus fects gene regulation—that such localization is
interaction at the lamina or simply maintains a itself important? In C. elegans, a global reduction
heterochromatin state is unclear. Other studies of lamin–chromatin interactions through deple-
suggest that altered chromatin state, including tion of the chromobox protein CEC-4, which is
both H3K9me2/3 and H3K27me3, drive lamina required for lamina association, resulted in
association (Bian et al. 2013, 2020; Kind et al. up-regulation of only one single gene, while de-
2013; Harr et al. 2015). Taken together, these pletion of H3K9 methylation caused widespread
findings suggest that the epigenetic state may disruption (Gonzalez-Sandoval et al. 2015).
be the ultimate factor in determining organiza- From this experiment, one could conclude that
tion to the lamina. Lamina-associating sequenc- organization of chromatin into LADs at the lam-
es may recruit repressive transcriptional com- ina is not important for gene repression (al-
plexes to promote a local chromatin signature though there is a second non-CEC-4-dependent
favorable for lamina association. While at the anchoring pathway that confounds these results
lamina, these complexes would then maintain (Cabianca et al. 2019). However, loss of CEC-4
or reinforce this repressive state, in agreement did impair induced differentiation into muscle
with studies showing that integration of pro- cells, reminiscent of mouse studies, in which
moter sequences into LADs leads to their re- the effect of loss of lamins is seen later in muscle
pression. differentiation (Sullivan et al. 1999). In addition,

14 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

several developmentally regulated tissue-restrict- in the B compartment, such findings highlight a


ed NETs have been shown to cause reorganiza- dynamic process of organization of LADs post-
tion of lineage-specific genes and altered expres- mitosis. In a clever study using an antibody var-
sion when ectopically expressed, suggesting that iant of DamID technology called pA-DamID on
these proteins directly influence organization and synchronized cells, dynamics of LAD organiza-
regulation (de Las Heras et al. 2017). In cells in- tion after mitotic exit were revealed (van Schaik
duced to undergo senescence, alterations in LAD et al. 2020). LADs organize to the nuclear lamina
organization were noted, including altered in early G1, with more telomere-proximal LADs
chromatin state outside and inside of LADs, par- interacting first, followed by more centromere-
ticularly an encroachment across boundaries, proximal LADs. Because these studies were
suggesting a “blurring” of chromatin domains done in human cells, this stepwise association
(Shah et al. 2013). This disorganization and dys- of different chromosomal positions suggests
regulation could be mimicked by acute depletion that the orientation of the chromosomes postmi-
of lamin B1, suggesting that, unlike ES cells, more tosis influences LAD organization to the lamina.
differentiated cells may rely on the geographical These associations generally increase as cells pro-
organization of heterochromatin to the lamina to gress further into interphase, in agreement with
maintain a cell-type-specific chromatin land- the increased A/B compartmentalization noted
scape. This is in agreement with the idea of in the earlier Hi-C studies. The relationship be-
LAD organization functioning as a mechanism tween LAD organization and lamin reorganiza-
to maintain a cell-type-specific epigenetic state; tion after mitosis remains to be determined.
some cell types and states will be more sensitive to
disruption of LAD organization.
LAMINOPATHIES
Disruption of the unique protein environment
DYNAMIC ORGANIZATION OF LADs
of the nuclear lamina often leads to disease
THROUGH THE CELL CYCLE
(Table 1). The diseases that result from mutations
Interphase genome organization, including LAD in LMNA, or other NET or LAP proteins that
and lamin organization, is ablated during mitosis heavily interact with lamins, are collectively
and re-established after mitosis (Burke and Ellen- termed “laminopathies” or “envelopathies.” Be-
berg 2002; Salina et al. 2003; Kind et al. 2013; cause of differential expression of lamins during
Naumova et al. 2013; Kind and van Steensel development and in different cell types along with
2014; Gibcus et al. 2018; Luperchio et al. 2018). the complex and cell-type-specific interactomes
Several studies have suggested that A-type lamins (NETs and LAPs), laminopathies display a wide
organize to the reforming nuclear envelope with breadth of phenotypes. Most laminopathies are
different kinetics, although there is some discrep- autosomal dominant and generally cause late-
ancy on how lamin A and C might differ in their onset degeneration of mesenchymal-derived cells
timing of association (Pugh et al. 1997; Moir et al. (Wong et al. 2021a). Nearly 500 disease-causing
2000b; Vaughan et al. 2001; González-Cruz et al. mutations have mapped to the LMNA gene, each
2018). Recent work has not only highlighted how with its own specific phenotype, and manyof these
chromosomes condense into their mitotic con- mutations are dominant. A variety of models have
figuration, but also how these regions reorganize been suggested to explain the variety of cell- and
into their interphase configuration postmitosis tissue-specific phenotypes. Indeed, it appears that
(Gibcus et al. 2018; Zhang et al. 2019). These disruption of lamins or lamin-binding proteins
studies focused on chromatin configuration as affect numerous cellular functions, depending
measured by Hi-C and it is notable that chroma- upon the particular mutation and cell type, includ-
tin A/B compartment organization precedes ing perturbations in mechanosensation and resil-
TAD organization, and that compartmentaliza- ience, DNA repair, signaling pathways, interac-
tion intensifies as the cells proceed further into tions with specific transcription factor, altered
interphase. Given that LADs are highly enriched interactions with NETs/LAPs, altered chromatin

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 15
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

Table 1. Diseases of the nuclear periphery


Gene Disease/anomaly Other notes
Primary laminopathies
LMNA Autosomal-dominant form of Emery– Affects striated muscle tissues
Dreifuss muscular dystrophy (AD-
EDMD)
Dilated cardiomyopathy (DCM)
Limb-girdle muscular dystrophy 1B
(LMG1B).
Peripheral neuropathy (R298C), a
recessive form of Charcot-Marie-
Tooth disease (AR-CMT2A)
Dunnigan-type familial partial Affects fat distribution and skeletal
lipodystrophy (FPLD2) development
Mandibuloacral dysplasia (MAD)
Hutchinson–Gilford progeria Premature aging
syndrome (HGPS)
Atypical Werner’s syndrome
LMNB1 Adult-onset autosomal-dominant Neural
leukodystrophy (ADLD)
Ataxia telangiectasia
Gene Function Disease/anomaly
Secondary envelopathies
Emerin (EMD) NE associated, may regulate β-catenin X-linked Emery–Dreifuss muscular
nuclear entry and MKL1 nuclear dystrophy
localization
Man1 (LEMD3) Regulates TGF-β signaling by Buschke–Ollendorff syndrome; excessive
modulating Smad phosphorylation bone nodule formation, required for
development of the vascular system
Lap1/Traf3/TOR1AIP1 Interacting protein with Torsin, Myopathy exacerbated by EMD loss
LULL1, and emerin
LEMD2/Net25 Chromatin organization MAP/AKT Homozygote nulls embryonic lethal
signaling progeroid; symptoms result from
missense mutations
Lap2 Chromatin organization, telomere Dilated cardiomyopathy, reduced
maintenance epidermal proliferation, ameliorates
LMNA-induced MD/DCM
Torsin AAA+ATPAse interacts closely with DYT dystonia in CNS, steatohepatitis
Lap1
Lamin B receptor (LBR) Multifunctional-reduced sterol Pelger–Huët anomaly, Greenberg
reductase activity, heterochromatin dysplasia
organization
Nesprin1 (SYNE1) LINC complex tethers nucleus to Limb girdle muscular dystrophy; ARCA1
cytoskeleton cerebellar ataxia required for nuclear
migration during CNS development
Nesprin2 (SYNE2) LINC complex tethers nucleus to EDMD variants required for nuclear
cytoskeleton migration during CNS development
Nesprin/Kash4 Interacts with MT motor proteins Required for nuclear positioning in
cochlear hair cells, mutations result in
deafness
Continued

16 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

Table 1. Continued
Gene Disease/anomaly Other notes
SUN1 Anchors LINC complex to the INM, Ameliorates LMNA-induced DCM/MD,
regulates miRNA synthesis during missense mutations associated with
muscle regeneration MD
BANF1 Postmitotic nuclear reassembly, Nestor–Guillermo progeria
chromatin organization
Data adapted from Wong and Stewart (2020).

state, and genome organization, none of which are REFERENCES


mutually exclusive (Mewborn et al. 2010; Osma-
Aebi U, Cohn J, Buhle L, Gerace L. 1986. The nuclear lamina
nagic-Myers et al. 2015; Vadrot et al. 2015; Wor- is a meshwork of intermediate-type filaments. Nature
man and Schirmer 2015; Perovanovic et al. 2016; 323: 560–564. doi:10.1038/323560a0
Le Dour et al. 2017; Briand and Collas 2018; Bian- Alsheimer M, von Glasenapp E, Hock R, Benavente R. 1999.
chi et al. 2018; and reviewed in Wong et al. 2021a). Architecture of the nuclear periphery of rat pachytene
spermatocytes: distribution of nuclear envelope proteins
in relation to synaptonemal complex attachment sites.
CONCLUDING REMARKS Mol Biol Cell 10: 1235–1245. doi:10.1091/mbc.10.4.1235
Andrés V, González JM. 2009. Role of A-type lamins in
The multiple functions of lamins and the nuclear signaling, transcription, and chromatin organization. J
lamina make it very difficult to assess the impact Cell Biol 187: 945–957. doi:10.1083/jcb.200904124
of lamina abnormalities on diseases initiated by Aymard F, Bugler B, Schmidt CK, Guillou E, Caron P, Briois
LMNA or NET mutations. In laminopathies, S, Iacovoni JS, Daburon V, Miller KM, Jackson SP, et al.
2014. Transcriptionally active chromatin recruits homol-
both gene dysregulation and the loss of nuclear ogous recombination at DNA double-strand breaks. Nat
integrity seem to play a role in observed pheno- Struct Mol Biol 21: 366–374. doi:10.1038/nsmb.2796
types. Since this interface controls both the me- Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z,
chanical “fitness” of the nucleus (through Wei G, Chepelev I, Zhao K. 2007. High-resolution profil-
ing of histone methylations in the human genome. Cell
LINCs, lamins, and heterochromatin scaffold- 129: 823–837. doi:10.1016/j.cell.2007.05.009
ing) as well as genome regulation, it will be in- Bártová E, Krejcí J, Harnicarová A, Galiová G, Kozubek S.
teresting to determine how mechanical force re- 2008. Histone modifications and nuclear architecture: a
lates to genome organization. In addition, while review. J Histochem Cytochem 56: 711–721. doi:10.1369/
jhc.2008.951251
it is clear that lamina association frequently re-
Belaadi N, Millon-Frémillon A, Aureille J, Guilluy C. 2018.
presses genes, how LADs are mechanistically Analyzing mechanotransduction through the LINC com-
maintained at and/or reorganized to the nuclear plex in isolated nuclei. The LINC Complex 1840: 73–80.
periphery during interphase and particularly doi:10.1007/978-1-4939-8691-0_7
during mitotic exit are only recently getting un- Belaghzal H, Dekker J, Gibcus JH. 2017. Hi-C 2.0: an opti-
raveled. Systematic study of how candidate mized Hi-C procedure for high-resolution genome-wide
mapping of chromosome conformation. Methods 123:
factors (including lamin isotypes, chromatin 56–65. doi:10.1016/j.ymeth.2017.04.004
interactors, NETs, and mechanical inputs) influ- Ben-Harush K, Wiesel N, Frenkiel-Krispin D, Moeller D,
ence LAD and lamina organization during mi- Soreq E, Aebi U, Herrmann H, Gruenbaum Y, Medalia
tosis to G1 may help provide more definitive O. 2009. The supramolecular organization of the C.
elegans nuclear lamin filament. J Mol Biol 386: 1392–
answers on the role of these regulators in lamina 1402. doi:10.1016/j.jmb.2008.12.024
and LAD organization and regulation, and the Berger R, Theodor L, Shoham J, Gokkel E, Brok-Simoni F,
consequences of their disruption in disease. Avraham KB, Copeland NG, Jenkins NA, Rechavi G, Si-
mon AJ. 1996. The characterization and localization of
the mouse thymopoietin/lamina-associated polypeptide
ACKNOWLEDGMENTS 2 gene and its alternatively spliced products. Genome Res
6: 361–370. doi:10.1101/gr.6.5.361
Funding sources were NIHR01GM132427 to
Berk JM, Tifft KE, Wilson KL. 2013. The nuclear envelope
K.L.R. and A.J.M-P. and NIH R25GM109441 LEM-domain protein emerin. Nucleus 4: 298–314. doi:10
to A.J.M.-P. .4161/nucl.25751

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 17
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

Bian Q, Khanna N, Alvikas J, Belmont AS. 2013. β-Globin Chang W, Worman HJ, Gundersen GG. 2015. Accessorizing
cis-elements determine differential nuclear targeting and anchoring the LINC complex for multifunctionality.
through epigenetic modifications. J Cell Biol 203: 767– J Cell Biol 208: 11–22. doi:10.1083/jcb.201409047
783. doi:10.1083/jcb.201305027 Chen IHB, Huber M, Guan T, Bubeck A, Gerace L. 2006.
Bian Q, Anderson EC, Yang Q, Meyer BJ. 2020. Histone Nuclear envelope transmembrane proteins (NETs) that
H3K9 methylation promotes formation of genome com- are up-regulated during myogenesis. BMC Cell Biol 7: 38.
partments in Caenorhabditis elegans via chromosome doi:10.1186/1471-2121-7-38
compaction and perinuclear anchoring. Proc Natl Acad Chen S, Luperchio TR, Wong X, Doan EB, Byrd AT, Roy
Sci 117: 11459–11470. doi:10.1073/pnas.2002068117 Choudhury K, Reddy KL, Krangel MS. 2018a. A lamina-
Bianchi A, Manti PG, Lucini F, Lanzuolo C. 2018. Mecha- associated domain border governs nuclear lamina inter-
notransduction, nuclear architecture and epigenetics in actions, transcription, and recombination of the Tcrb lo-
emery dreifuss muscular dystrophy: tous pour un, un cus. Cell Rep 25: 1729–1740.e6. doi:10.1016/j.celrep.2018
pour tous. Nucleus 9: 321–335. doi:10.1080/19491034 .10.052
.2018.1460044 Chen Y, Zhang Y, Wang Y, Zhang L, Brinkman EK, Adam
Boettiger A, Murphy S. 2020. Advances in chromatin imag- SA, Goldman R, van Steensel B, Ma J, Belmont AS. 2018b.
ing at kilobase-scale resolution. Trends Genet 36: 273– Mapping 3D genome organization relative to nuclear
287. doi:10.1016/j.tig.2019.12.010 compartments using TSA-Seq as a cytological ruler. J
Cell Biol 217: 4025–4048. doi: 10.1083/jcb.201807108.
Boulay G, Dubuissez M, Van Rechem C, Forget A, Helin K,
Ayrault O, Leprince D. 2012. Hypermethylated in cancer Coffinier C, Jung HJ, Nobumori C, Chang S, Tu Y, Barnes
1 (HIC1) recruits polycomb repressive complex 2 (PRC2) RH II, Yoshinaga Y, de Jong PJ, Vergnes L, Reue K, et al.
to a subset of its target genes through interaction with 2011. Deficiencies in lamin B1 and lamin B2 cause neuro-
human polycomb-like (hPCL) proteins. J Biol Chem developmental defects and distinct nuclear shape abnor-
malities in neurons. Mol Biol Cell 22: 4683–4693. doi:10
287: 10509–10524. doi:10.1074/jbc.M111.320234
.1091/mbc.e11-06-0504
Brachner A, Reipert S, Foisner R, Gotzmann J. 2005. LEM2 is
Cremer T, Cremer C. 2001. Chromosome territories, nuclear
a novel MAN1-related inner nuclear membrane protein
architecture and gene regulation in mammalian cells. Nat
associated with A-type lamins. J Cell Sci 118: 5797–5810.
Rev Genet 2: 292–301. doi:10.1038/35066075
doi:10.1242/jcs.02701
Cremer T, Cremer M. 2010. Chromosome territories. Cold
Briand N, Collas P. 2018. Laminopathy-causing lamin A
Spring Harb Perspect Biol 2: a003889. doi:10.1101/cshper
mutations reconfigure lamina-associated domains and spect.a003889
local spatial chromatin conformation. Nucleus 9: 216–
226. doi:10.1080/19491034.2018.1449498 Das PM, Ramachandran K, vanWert J, Singal R. 2004. Chro-
matin immunoprecipitation assay. BioTechniques 37:
Briand N, Collas P. 2020. Lamina-associated domains: pe- 961–969. doi:10.2144/04376RV01
ripheral matters and internal affairs. Genome Biol 21: 85.
DeBoy E, Puttaraju M, Jailwala P, Kasoji M, Cam M, Misteli
doi:10.1186/s13059-020-02003-5
T. 2017. Identification of novel RNA isoforms of LMNA.
Buchwalter A, Kaneshiro JM, Hetzer MW. 2019. Coaching Nucleus 8: 573–582. doi:10.1080/19491034.2017.1348449
from the sidelines: the nuclear periphery in genome reg-
Dechat T, Gotzmann J, Stockinger A, Harris CA, Talle MA,
ulation. Nat Rev Genet 20: 39–50. doi:10.1038/s41576-
Siekierka JJ, Foisner R. 1998. Detergent-salt resistance of
018-0063-5
LAP2α in interphase nuclei and phosphorylation-depen-
Burke B, Ellenberg J. 2002. Remodelling the walls of the dent association with chromosomes early in nuclear as-
nucleus. Nat Rev Mol Cell Biol 3: 487–497. doi:10.1038/ sembly implies functions in nuclear structure dynamics.
nrm860 EMBO J 17: 4887–4902. doi:10.1093/emboj/17.16.4887
Cabianca DS, Muñoz-Jiménez C, Kalck V, Gaidatzis D, Pa- Dechat T, Gajewski A, Korbei B, Gerlich D, Daigle N, Ha-
deken J, Seeber A, Askjaer P, Gasser SM. 2019. Active raguchi T, Furukawa K, Ellenberg J, Foisner R. 2004.
chromatin marks drive spatial sequestration of hetero- LAP2α and BAF transiently localize to telomeres and
chromatin in C. elegans nuclei. Nature 569: 734–739. specific regions on chromatin during nuclear assembly.
doi:10.1038/s41586-019-1243-y J Cell Sci 117: 6117–6128. doi:10.1242/jcs.01529
Cai M, Huang Y, Ghirlando R, Wilson KL, Craigie R, Clore Dechat T, Pfleghaar K, Sengupta K, Shimi T, Shumaker DK,
GM. 2001. Solution structure of the constant region of Solimando L, Goldman RD. 2008. Nuclear lamins: major
nuclear envelope protein LAP2 reveals two LEM-domain factors in the structural organization and function of the
structures: one binds BAF and the other binds DNA. nucleus and chromatin. Genes Dev 22: 832–853. doi:10
EMBO J 20: 4399–4407. doi:10.1093/emboj/20.16.4399 .1101/gad.1652708
Cai M, Huang Y, Suh JY, Louis JM, Ghirlando R, Craigie R, Dechat T, Adam SA, Taimen P, Shimi T, Goldman RD. 2010.
Clore GM. 2007. Solution NMR structure of the barrier- Nuclear lamins. Cold Spring Harb Perspect Biol 2:
to-autointegration factor-Emerin complex. J Biol Chem a000547. doi:10.1101/cshperspect.a000547
282: 14525–14535. doi:10.1074/jbc.M700576200 Dekker J. 2002. Capturing chromosome conformation. Sci-
Chalut KJ, Höpfler M, Lautenschläger F, Boyde L, Chan CJ, ence 295: 1306–1311. doi:10.1126/science.1067799
Ekpenyong A, Martinez-Arias A, Guck J. 2012. Chroma- de Las Heras JI, Meinke P, Batrakou DG, Srsen V, Zuleger N,
tin decondensation and nuclear softening accompany Kerr AR, Schirmer EC. 2013. Tissue specificity in the
Nanog downregulation in embryonic stem cells. Biophys nuclear envelope supports its functional complexity. Nu-
J 103: 2060–2070. doi:10.1016/j.bpj.2012.10.015 cleus 4: 460–477. doi:10.4161/nucl.26872

18 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

de Las Heras JI, Zuleger N, Batrakou DG, Czapiewski R, Kerr Furukawa K, Hotta Y. 1993. cDNA cloning of a germ cell
ARW, Schirmer EC. 2017. Tissue-specific NETs alter ge- specific lamin B3 from mouse spermatocytes and analysis
nome organization and regulation even in a heterologous of its function by ectopic expression in somatic cells.
system. Nucleus 8: 81–97. doi:10.1080/19491034.2016 EMBO J 12: 97–106. doi:10.1002/j.1460-2075.1993.tb0
.1261230 5635.x
Demmerle J, Koch AJ, Holaska JM. 2012. The nuclear enve- Furukawa K, Inagaki H, Hotta Y. 1994. Identification and
lope protein emerin binds directly to histone deacetylase 3 cloning of an mRNA coding for a germ cell-specific A-
(HDAC3) and activates HDAC3 activity. J Biol Chem 287: type lamin in mice. Exp Cell Res 212: 426–430. doi:10
22080. doi:10.1074/jbc.M111.325308 .1006/excr.1994.1164
Dialynas G, Speese S, Budnik V, Geyer PK, Wallrath LL. Furusawa T, Rochman M, Taher L, Dimitriadis EK, Naga-
2010. The role of Drosophila lamin C in muscle function shima K, Anderson S, Bustin M. 2015. Chromatin decom-
and gene expression. Development 137: 3067–3077. paction by the nucleosomal binding protein HMGN5
doi:10.1242/dev.048231 impairs nuclear sturdiness. Nat Commun 6: 6138.
doi:10.1038/ncomms7138
Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, Hu M, Liu
JS, Ren B. 2012. Topological domains in mammalian ge- Georgatos SD, Pyrpasopoulou A, Theodoropoulos PA. 1997.
nomes identified by analysis of chromatin interactions. Nuclear envelope breakdown in mammalian cells in-
Nature 485: 376–380. doi:10.1038/nature11082 volves stepwise lamina disassembly and microtubule-
drive deformation of the nuclear membrane. J Cell Sci
Dorner D, Vlcek S, Foeger N, Gajewski A, Makolm C, Gotz- 110: 2129–2140. doi:10.1242/jcs.110.17.2129
mann J, Hutchison CJ, Foisner R. 2006. Lamina-associ-
ated polypeptide 2α regulates cell cycle progression and Gerace L, Blobel G. 1980. The nuclear envelope lamina is
differentiation via the retinoblastoma-E2F pathway. J Cell reversibly depolymerized during mitosis. Cell 19: 277–
Biol 173: 83–93. doi:10.1083/jcb.200511149 287. doi:10.1016/0092-8674(80)90409-2
Gerace L, Foisner R. 1994. Integral membrane proteins and
Dreesen O, Chojnowski A, Ong PF, Zhao TY, Common JE,
dynamic organization of the nuclear envelope. Trends Cell
Lunny D, Lane EB, Lee SJ, Vardy LA, Stewart CL, et al.
Biol 4: 127–131. doi:10.1016/0962-8924(94)90067-1
2013. Lamin B1 fluctuations have differential effects on
cellular proliferation and senescence. J Cell Biol 200: 605– Gerace L, Huber MD. 2012. Nuclear lamina at the crossroads
617. doi:10.1083/jcb.201206121 of the cytoplasm and nucleus. J Struct Biol 177: 24–31.
doi:10.1016/j.jsb.2011.11.007
Dunlevy KL, Medvedeva V, Wilson JE, Hoque M, Pellegrin
T, Maynard A, Kremp MM, Wasserman JS, Poleshko A, Gesson K, Rescheneder P, Skoruppa MP, von Haeseler A,
Katz RA. 2020. The PRR14 heterochromatin tether en- Dechat T, Foisner R. 2016. A-type lamins bind both het-
codes modular domains that mediate and regulate nuclear ero- and euchromatin, the latter being regulated by lam-
lamina targeting. J Cell Sci 133: jcs240416. doi:10.1242/jcs ina-associated polypeptide 2α. Genome Res 26: 462–473.
.240416 doi:10.1101/gr.196220.115
Gibbs-Seymour I, Markiewicz E, Bekker-Jensen S, Mailand
Duong NT, Morris GE, Lam LT, Zhang Q, Sewry CA, Sha-
N, Hutchison CJ. 2015. Lamin A/C-dependent interac-
nahan CM, Holt I. 2014. Nesprins: tissue-specific expres-
tion with 53BP1 promotes cellular responses to DNA
sion of epsilon and other short isoforms. PLoS ONE 9:
damage. Aging Cell 14: 162–169. doi:10.1111/acel.12258
e94380. doi:10.1371/journal.pone.0094380
Gibcus JH, Samejima K, Goloborodko A, Samejima I, Nau-
Ellenberg J. 2013. Dynamics of nuclear envelope proteins
mova N, Nuebler J, Kanemaki MT, Xie L, Paulson JR,
during the cell cycle in mammalian cells. Landes Bio-
Earnshaw WC, et al. 2018. A pathway for mitotic chro-
science, Austin, TX. mosome formation. Science 359: eaao6135. doi:10.1126/
Falk M, Feodorova Y, Naumova N, Imakaev M, Lajoie BR, science.aao6135
Leonhardt H, Joffe B, Dekker J, Fudenberg G, Solovei I, et Gieffers C, Krohne G. 1991. In vitro reconstitution of recom-
al. 2019. Heterochromatin drives compartmentalization binant lamin A and a lamin A mutant lacking the car-
of inverted and conventional nuclei. Nature 570: 395– boxy-terminal tail. Eur J Cell Biol 55: 191–199.
399. doi:10.1038/s41586-019-1275-3
González-Cruz RD, Dahl KN, Darling EM. 2018. The
Finlan LE, Sproul D, Thomson I, Boyle S, Kerr E, Perry P, emerging role of lamin C as an important LMNA isoform
Ylstra B, Chubb JR, Bickmore WA. 2008. Recruitment to in mechanophenotype. Front Cell Dev Biol 6: 151. doi:10
the nuclear periphery can alter expression of genes in .3389/fcell.2018.00151
human cells. PLoS Genet 4: e1000039. doi:10.1371/jour
Gonzalez-Sandoval A, Gasser SM. 2016. Mechanism of
nal.pgen.1000039
chromatin segregation to the nuclear periphery in
Foisner R. 2003. Cell cycle dynamics of the nuclear envelope. C. elegans embryos. Worm 5: e1190900. doi:10.1080/
ScientificWorldJournal 3: 1–20. doi:10.1100/tsw.2003.06 21624054.2016.1190900
Fraser J, Ferrai C, Chiariello AM, Schueler M, Rito T, Lau- Gonzalez-Sandoval A, Towbin BD, Kalck V, Cabianca DS,
danno G, Barbieri M, Moore BL, Kraemer DCA, Aitken S, Gaidatzis D, Hauer MH, Geng L, Wang L, Yang T, Wang
et al. 2015. Hierarchical folding and reorganization of X, et al. 2015. Perinuclear anchoring of H3K9-methylated
chromosomes are linked to transcriptional changes in chromatin stabilizes induced cell fate in C. elegans embry-
cellular differentiation. Mol Syst Biol 11: 852. doi:10 os. Cell 163: 1333–1347. doi:10.1016/j.cell.2015.10.066
.15252/msb.20156492 Greil F, Moorman C, van Steensel B. 2006. DamID: mapping
Furukawa K. 1999. LAP2 binding protein 1 (L2BP1/BAF) is of in vivo protein-genome interactions using tethered
a candidate mediator of LAP2-chromatin interaction. J DNA adenine methyltransferase. Methods Enzymol 410:
Cell Sci 112: 2485–2492. doi:10.1242/jcs.112.15.2485 342–359. doi:10.1016/S0076-6879(06)10016-6

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 19
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

Gruenbaum Y, Foisner R. 2015. Lamins: nuclear intermedi- Holmer L, Worman HJ. 2001. Inner nuclear membrane pro-
ate filament proteins with fundamental functions in nu- teins: functions and targeting. Cell Mol Life Sci 58: 1741–
clear mechanics and genome regulation. Annu Rev Bio- 1747. doi:10.1007/PL00000813
chem 84: 131–164. doi:10.1146/annurev-biochem-060 Ikegami K, Secchia S, Almakki O, Lieb JD, Moskowitz IP.
614-034115 2020. Phosphorylated lamin A/C in the nuclear interior
Gruenbaum Y, Medalia O. 2015. Lamins: the structure and binds active enhancers associated with abnormal tran-
protein complexes. Curr Opin Cell Biol 32: 7–12. doi:10 scription in progeria. Dev Cell 52: 699–713.e11. doi:10
.1016/j.ceb.2014.09.009 .1016/j.devcel.2020.02.011
Guelen L, Pagie L, Brasset E, Meuleman W, Faza MB, Tal- Izumi M, Vaughan OA, Hutchison CJ, Gilbert DM. 2000.
hout W, Eussen BH, de Klein A, Wessels L, de Laat W, et Head and/or CaaX domain deletions of lamin proteins
al. 2008. Domain organization of human chromosomes disrupt preformed lamin A and C but not lamin B struc-
revealed by mapping of nuclear lamina interactions. Na- ture in mammalian cells. Mol Biol Cell 11: 4323–4337.
ture 453: 948–951. doi:10.1038/nature06947 doi:10.1091/mbc.11.12.4323
Harada T, Swift J, Irianto J, Shin J-W, Spinler KR, Athirasala Jahn D, Schramm S, Schnölzer M, Heilmann CJ, de Koster
A, Diegmiller R, Dingal PCDP, Ivanovska IL, Discher DE. CG, Schütz W, Benavente R, Alsheimer M. 2012. A trun-
2014. Nuclear lamin stiffness is a barrier to 3D migration,
cated lamin A in the Lmna–/– mouse line: implications for
but softness can limit survival. J Cell Biol 204: 669–682.
the understanding of laminopathies. Nucleus 3: 463–474.
doi:10.1083/jcb.201308029
doi:10.4161/nucl.21676
Haraguchi T, Koujin T, Segura-Totten M, Lee KK, Matsuoka
Jung HJ, Coffinier C, Choe Y, Beigneux AP, Davies BS, Yang
Y, Yoneda Y, Wilson KL, Hiraoka Y. 2001. BAF is required
for emerin assembly into the reforming nuclear envelope. SH, Barnes RH II, Hong J, Sun T, Pleasure SJ, et al. 2012.
J Cell Sci 114: 4575–4585. doi:10.1242/jcs.114.24.4575 Regulation of prelamin A but not lamin C by miR-9, a
brain-specific microRNA. Proc Natl Acad Sci 109: E423–
Harr JC, Luperchio TR, Wong X, Cohen E, Wheelan SJ,
E431. doi:10.1073/pnas.1111780109
Reddy KL. 2015. Directed targeting of chromatin to the
nuclear lamina is mediated by chromatin state and A-type Kempfer R, Pombo A. 2020. Methods for mapping 3D chro-
lamins. J Cell Biol 208: 33–52. doi:10.1083/jcb.201405110 mosome architecture. Nat Rev Genet 21: 207–226. doi:10
.1038/s41576-019-0195-2
Harr JC, Gonzalez-Sandoval A, Gasser SM. 2016. Histones
and histone modifications in perinuclear chromatin an- Kim DH, Wirtz D. 2015. Cytoskeletal tension induces the
choring: from yeast to man. EMBO Rep 17: 139–155. polarized architecture of the nucleus. Biomaterials 48:
doi:10.15252/embr.201541809 161–172. doi:10.1016/j.biomaterials.2015.01.023
Harr JC, Schmid CD, Muñoz-Jiménez C, Romero-Bueno R, Kim Y, Zheng X, Zheng Y. 2013. Proliferation and differen-
Kalck V, Gonzalez-Sandoval A, Hauer MH, Padeken J, tiation of mouse embryonic stem cells lacking all lamins.
Askjaer P, Mattout A, et al. 2020. Loss of an H3K9me Cell Res 23: 1420–1423. doi:10.1038/cr.2013.118
anchor rescues laminopathy-linked changes in nuclear Kim DI, Birendra KC, Roux KJ. 2015. Making the LINC:
organization and muscle function in an Emery–Dreifuss SUN and KASH protein interactions. Biol Chem 396:
muscular dystrophy model. Genes Dev 34: 560–579. 295–310. doi:10.1515/hsz-2014-0267
doi:10.1101/gad.332213.119
Kim DI, Jensen SC, Roux KJ. 2016. Identifying protein-pro-
Harris CA, Andryuk PJ, Cline S, Chan HK, Natarajan A, tein associations at the nuclear envelope with BioID.
Siekierka JJ, Goldstein G. 1994. Three distinct human Methods Mol Biol 1411: 133–146. doi:10.1007/978-1-
thymopoietins are derived from alternatively spliced 4939-3530-7_8
mRNAs. Proc Natl Acad Sci 91: 6283–6287. doi:10
.1073/pnas.91.14.6283 Kind J, van Steensel B. 2014. Stochastic genome-nuclear
lamina interactions: modulating roles of lamin A and
Heitlinger E, Peter M, Lustig A, Villiger W, Nigg EA, Aebi U.
BAF. Nucleus 5: 124–130. doi:10.4161/nucl.28825
1992. The role of the head and tail domain in lamin
structure and assembly: analysis of bacterially expressed Kind J, Pagie L, Ortabozkoyun H, Boyle S, de Vries SS,
chicken lamin A and truncated B2 lamins. J Struct Biol Janssen H, Amendola M, Nolen LD, Bickmore WA, van
108: 74–91. doi:10.1016/1047-8477(92)90009-Y Steensel B. 2013. Single-cell dynamics of genome-nuclear
Herrmann H, Aebi U. 2004. Intermediate filaments: molec- lamina interactions. Cell 153: 178–192. doi:10.1016/j.cell
ular structure, assembly mechanism, and integration into .2013.02.028
functionally distinct intracellular scaffolds. Annu Rev Bi- Kind J, Pagie L, de Vries SS, Nahidiazar L, Dey SS, Bienko M,
ochem 73: 749–789. doi:10.1146/annurev.biochem.73 Zhan Y, Lajoie B, de Graaf CA, Amendola M, et al. 2015.
.011303.073823 Genome-wide maps of nuclear lamina interactions in
Hirano Y, Hizume K, Kimura H, Takeyasu K, Haraguchi T, single human cells. Cell 163: 134–147. doi:10.1016/j.cell
Hiraoka Y. 2012. Lamin B receptor recognizes specific .2015.08.040
modifications of histone H4 in heterochromatin forma- Klapper M, Exner K, Kempf A, Gehrig C, Stuurman N,
tion. J Biol Chem 287: 42654–42663. doi:10.1074/jbc Fisher PA, Krohne G. 1997. Assembly of A- and B-type
.M112.397950 lamins studied in vivo with the baculovirus system. J Cell
Ho CY, Lammerding J. 2012. Lamins at a glance. J Cell Sci Sci 110: 2519–2532. doi:10.1242/jcs.110.20.2519
125: 2087–2093. doi:10.1242/jcs.087288 Kochin V, Shimi T, Torvaldson E, Adam SA, Goldman A,
Höger TH, Grund C, Franke WW, Krohne G. 1991. Immu- Pack CG, Melo-Cardenas J, Imanishi SY, Goldman RD,
nolocalization of lamins in the thick nuclear lamina of Eriksson JE. 2014. Interphase phosphorylation of lamin
human synovial cells. Eur J Cell Biol 54: 150–156. A. J Cell Sci 127: 2683–2696. doi:10.1242/jcs.141820

20 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

Korfali N, Wilkie GS, Swanson SK, Srsen V, Batrakou DG, human genome. Science 326: 289–293. doi:10.1126/sci
Fairley EAL, Malik P, Zuleger N, Goncharevich A, de las ence.1181369
Heras J, et al. 2010. The leukocyte nuclear envelope pro- Lin F, Worman HJ. 1993. Structural organization of the hu-
teome varies with cell activation and contains novel trans- man gene encoding nuclear lamin A and nuclear lamin C.
membrane proteins that affect genome architecture. Mol J Biol Chem 268: 16321–16326. doi:10.1016/S0021-9258
Cell Proteomics 9: 2571–2585. doi:10.1074/mcp.M110 (19)85424-8
.002915 Lin F, Blake DL, Callebaut I, Skerjanc IS, Holmer L, Mc-
Korfali N, Wilkie GS, Swanson SK, Srsen V, de Las Heras J, Burney MW, Paulin-Levasseur M, Worman HJ. 2000.
Batrakou DG, Malik P, Zuleger N, Kerr AR, Florens L, et MAN1, an inner nuclear membrane protein that shares
al. 2012. The nuclear envelope proteome differs notably the LEM domain with lamina-associated polypeptide 2
between tissues. Nucleus 3: 552–564. and emerin. J Biol Chem 275: 4840–4847. doi:10.1074/jbc
Kosak ST, Skok JA, Medina KL, Riblet R, Le Beau MM, .275.7.4840
Fisher AG, Singh H. 2002. Subnuclear compartmentali- Lombardi ML, Lammerding J. 2011. Keeping the LINC: the
zation of immunoglobulin loci during lymphocyte devel- importance of nucleocytoskeletal coupling in intracellular
opment. Science 296: 158–162. doi:10.1126/science.106 force transmission and cellular function. Biochem Soc
8768 Trans 39: 1729–1734. doi:10.1042/BST20110686
Krimm I, Östlund C, Gilquin B, Couprie J, Hossenlopp P, Lottersberger F, Karssemeijer RA, Dimitrova N, de Lange T.
Mornon J-P, Bonne G, Courvalin J-C, Worman HJ, Zinn- 2015. 53BP1 and the LINC complex promote microtu-
Justin S. 2002. The Ig-like structure of the C-terminal bule-dependent DSB mobility and DNA repair. Cell 163:
domain of lamin A/C, mutated in muscular dystrophies, 880–893. doi:10.1016/j.cell.2015.09.057
cardiomyopathy, and partial lipodystrophy. Structure 10: Luperchio TR, Sauria MEG, Hoskins VE, Wong X, DeBoy E,
811–823. doi:10.1016/S0969-2126(02)00777-3 Gaillard M-C, Tsang P, Pekrun K, Ach RA, Yamada NA,
Kumaran RI, Muralikrishna B, Parnaik VK. 2002. Lamin A/ et al. 2018. The repressive genome compartment is estab-
C speckles mediate spatial organization of splicing factor lished early in the cell cycle before forming the lamina
compartments and RNA polymerase II transcription. J associated domains. bioRxiv doi:10.1101/481598v1
Cell Biol 159: 783–793. doi:10.1083/jcb.200204149 Luxton GWG, Gomes ER, Folker ES, Vintinner E, Gun-
Larson AG, Elnatan D, Keenen MM, Trnka MJ, Johnston JB, dersen GG. 2010. Linear arrays of nuclear envelope pro-
Burlingame AL, Agard DA, Redding S, Narlikar GJ. 2017. teins harness retrograde actin flow for nuclear movement.
Liquid droplet formation by HP1α suggests a role for Science 329: 956–959. doi:10.1126/science.1189072
phase separation in heterochromatin. Nature 547: 236– Ma S, Zhang Y. 2020. Profiling chromatin regulatory land-
240. doi:10.1038/nature22822 scape: insights into the development of ChIP-seq and
Le Dour C, Wu W, Béréziat V, Capeau J, Vigouroux C, Wor- ATAC-seq. Molecular Biomedicine 1: 9. doi:10.1186/
man HJ. 2017. Extracellular matrix remodeling and trans- s43556-020-00009-w
forming growth factor-β signaling abnormalities induced Ma L, Tsai MY, Wang S, Lu B, Chen R, Yates JR III, Zhu X,
by lamin A/C variants that cause lipodystrophy. J Lipid Zheng Y. 2009. Requirement for nudel and dynein for
Res 58: 151–163. doi:10.1194/jlr.M071381 assembly of the lamin B spindle matrix. Nat Cell Biol
Lee KK, Wilson KL. 2004. All in the family: evidence for four 11: 247–256. doi:10.1038/ncb1832
new LEM-domain proteins Lem2 (NET-25), Lem3, Lem4 Machiels BM, Zorenc AH, Endert JM, Kuijpers HJ, van Eys
and Lem5 in the human genome. Symp Soc Exp Biol 56: GJ, Ramaekers FC, Broers JL. 1996. An alternative splic-
329–339. ing product of the lamin A/C gene lacks exon 10. J Biol
Lee KK, Haraguchi T, Lee RS, Koujin T, Hiraoka Y, Wilson Chem 271: 9249–9253. doi:10.1074/jbc.271.16.9249
KL. 2001. Distinct functional domains in emerin bind Maeshima K, Tamura S, Hansen JC, Itoh Y. 2020. Fluid-like
lamin A and DNA-bridging protein BAF. J Cell Sci 114: chromatin: toward understanding the real chromatin or-
4567–4573. doi:10.1242/jcs.114.24.4567 ganization present in the cell. Curr Opin Cell Biol 64: 77–
Leemans C, van der Zwalm MCH, Brueckner L, Comoglio F, 89. doi:10.1016/j.ceb.2020.02.016
van Schaik T, Pagie L, van Arensbergen J, van Steensel B. Maison C, Pyrpasopoulou A, Theodoropoulos PA, Georga-
2019. Promoter-intrinsic and local chromatin features tos SD. 1997. The inner nuclear membrane protein LAP1
determine gene repression in LADs. Cell 177: 852–864. forms a native complex with B-type lamins and partitions
e14. doi:10.1016/j.cell.2019.03.009 with spindle-associated mitotic vesicles. EMBO J 16:
Legartová S, Stixová L, Laur O, Kozubek S, Sehnalová P, 4839–4850. doi:10.1093/emboj/16.16.4839
Bártová E. 2014. Nuclear structures surrounding internal Margalit A, Brachner A, Gotzmann J, Foisner R, Gruenbaum
lamin invaginations. J Cell Biochem 115: 476–487. doi:10 Y. 2007. Barrier-to-autointegration factor—a BAFfling
.1002/jcb.24681 little protein. Trends Cell Biol 17: 202–208. doi:10.1016/j
Li BX, Chen J, Chao B, Zheng Y, Xiao X. 2018. A lamin- .tcb.2007.02.004
binding ligand inhibits homologous recombination re- Marnef A, Finoux AL, Arnould C, Guillou E, Daburon V,
pair of DNA double-strand breaks. ACS Cent Sci 4: Rocher V, Mangeat T, Mangeot PE, Ricci EP, Legube G.
1201–1210. 2019. A cohesin/HUSH- and LINC-dependent pathway
Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev controls ribosomal DNA double-strand break repair.
M, Ragoczy T, Telling A, Amit I, Lajoie BR, Sabo PJ, Genes Dev 33: 1175–1190. doi:10.1101/gad.324012.119
Dorschner MO, et al. 2009. Comprehensive mapping of Mattout A, Cabianca DS, Gasser SM. 2015. Chromatin states
long-range interactions reveals folding principles of the and nuclear organization in development—a view from

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 21
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

the nuclear lamina. Genome Biol 16: 174. doi:10.1186/ differentiation. Mol Cell 38: 603–613. doi:10.1016/j
s13059-015-0747-5 .molcel.2010.03.016
Maurer M, Lammerding J. 2019. The driving force: nuclear Perovanovic J, Dell’Orso S, Gnochi VF, Jaiswal JK, Sartorelli
mechanotransduction in cellular function, fate, and dis- V, Vigouroux C, Mamchaoui K, Mouly V, Bonne G, Hoff-
ease. Annu Rev Biomed Eng 21: 443–468. doi:10.1146/ man EP. 2016. Laminopathies disrupt epigenomic devel-
annurev-bioeng-060418-052139 opmental programs and cell fate. Sci Transl Med 8:
Mehus AA, Anderson RH, Roux KJ. 2016. BioID identifica- 335ra58. doi:10.1126/scitranslmed.aad4991
tion of lamin-associated proteins. Methods Enzymol 569: Peter M, Kitten GT, Lehner CF, Vorburger K, Bailer SM,
3–22. doi:10.1016/bs.mie.2015.08.008 Maridor G, Nigg EA. 1989. Cloning and sequencing of
Méjat A, Misteli T. 2010. LINC complexes in health and cDNA clones encoding chicken lamins A and B1 and
disease. Nucleus 1: 40–52. doi:10.4161/nucl.1.1.10530 comparison of the primary structures of vertebrate A-
Meuleman W, Peric-Hupkes D, Kind J, Beaudry JBB, Pagie and B-type lamins. J Mol Biol 208: 393–404. doi:10
L, Kellis M, Reinders M, Wessels L, van Steensel B. 2012. .1016/0022-2836(89)90504-4
Constitutive nuclear lamina–genome interactions are Phillips-Cremins JE, Sauria MEG, Sanyal A, Gerasimova TI,
highly conserved and associated with A/T-rich sequence. Lajoie BR, Bell JSK, Ong CT, Hookway TA, Guo C, Sun Y,
Genome Res 23: 270–280. doi:10.1101/gr.141028.112 et al. 2013. Architectural protein subclasses shape 3D
Mewborn SK, Puckelwartz MJ, Abuisneineh F, Fahrenbach organization of genomes during lineage commitment.
JP, Zhang Y, MacLeod H, Dellefave L, Pytel P, Selig S, Cell 153: 1281–1295. doi:10.1016/j.cell.2013.04.053
Labno CM, et al. 2010. Altered chromosomal positioning, Poleshko A, Katz RA. 2014. Specifying peripheral hetero-
compaction, and gene expression with a lamin A/C gene chromatin during nuclear lamina reassembly. Nucleus
mutation. PLoS ONE 5: e14342. doi:10.1371/journal.pone 5: 32–39. doi:10.4161/nucl.28167
.0014342 Poleshko A, Shah PP, Gupta M, Babu A, Morley MP, Man-
Moir RD, Spann TP, Herrmann H, Goldman RD. 2000a. derfield LJ, Ifkovits JL, Calderon D, Aghajanian H, Sierra-
Disruption of nuclear lamin organization blocks the elon- Pagán JE, et al. 2017. Genome-nuclear lamina interac-
gation phase of DNA replication. J Cell Biol 149: 1179– tions regulate cardiac stem cell lineage restriction. Cell
1192. doi:10.1083/jcb.149.6.1179 171: 573–587.e14. doi:10.1016/j.cell.2017.09.018
Moir RD, Yoon M, Khuon S, Goldman RD. 2000b. Nuclear Polioudaki H, Kourmouli N, Drosou V, Bakou A, Theo-
lamins A and B1: different pathways of assembly during doropoulos PA, Singh PB, Giannakouros T, Georgatos
nuclear envelope formation in living cells. J Cell Biol 151: SD. 2001. Histones H3/H4 form a tight complex with
1155–1168. doi:10.1083/jcb.151.6.1155 the inner nuclear membrane protein LBR and hetero-
Naumova N, Imakaev M, Fudenberg G, Zhan Y, Lajoie BR, chromatin protein 1. EMBO Rep 2: 920–925. doi:10
Mirny LA, Dekker J. 2013. Organization of the mitotic .1093/embo-reports/kve199
chromosome. Science 342: 948–953. doi:10.1126/science Pope BD, Ryba T, Dileep V, Yue F, Wu W, Denas O, Vera DL,
.1236083 Wang Y, Hansen RS, Canfield TK, et al. 2014. Topologi-
Nelson DM, Jaber-Hijazi F, Cole JJ, Robertson NA, Pawli- cally associating domains are stable units of replication-
kowski JS, Norris KT, Criscione SW, Pchelintsev NA, timing regulation. Nature 515: 402–405. doi:10.1038/na
Piscitello D, Stong N, et al. 2016. Mapping H4K20me3 ture13986
onto the chromatin landscape of senescent cells indicates Pugh GE, Coates PJ, Lane EB, Raymond Y, Quinlan RA.
a function in control of cell senescence and tumor sup- 1997. Distinct nuclear assembly pathways for lamins A
pression through preservation of genetic and epigenetic and C lead to their increase during quiescence in Swiss
stability. Genome Biol 17: 158. doi:10.1186/s13059- 3T3 cells. J Cell Sci 110: 2483–2493. doi:10.1242/jcs.110
016-1017-x .19.2483
Nora EP, Goloborodko A, Valton AL, Gibcus JH, Uebersohn Rajgor D, Shanahan CM. 2013. Nesprins: from the nuclear
A, Abdennur N, Dekker J, Mirny LA, Bruneau BG. envelope and beyond. Expert Rev Mol Med 15: e5. doi:10
2017. Targeted degradation of CTCF decouples local in- .1017/erm.2013.6
sulation of chromosome domains from genomic com- Rao SSP, Huntley MH, Durand NC, Stamenova EK, Bochkov
partmentalization. Cell 169: 930–944.e22. doi:10.1016/j ID, Robinson JT, Sanborn AL, Machol I, Omer AD, Lan-
.cell.2017.05.004 der ES, et al. 2014. A 3D map of the human genome at
Orian A, Abed M, Kenyagin-Karsenti D, Boico O. 2009. kilobase resolution reveals principles of chromatin loop-
DamID: a methylation-based chromatin profiling ap- ing. Cell 159: 1665–1680. doi:10.1016/j.cell.2014.11.021
proach. Methods Mol Biol 567: 155–169. doi:10.1007/ Razafsky D, Wirtz D, Hodzic D. 2014. Nuclear envelope in
978-1-60327-414-2_11 nuclear positioning and cell migration. Adv Exp Med Biol
Osmanagic-Myers S, Dechat T, Foisner R. 2015. Lamins at 773: 471–490. doi:10.1007/978-1-4899-8032-8_21
the crossroads of mechanosignaling. Genes Dev 29: 225– Reddy KL, Zullo JM, Bertolino E, Singh H. 2008. Transcrip-
237. doi:10.1101/gad.255968.114 tional repression mediated by repositioning of genes to
Padeken J, Heun P. 2014. Nucleolus and nuclear periphery: the nuclear lamina. Nature 452: 243–247. doi:10.1038/
velcro for heterochromatin. Curr Opin Cell Biol 28: 54– nature06727
60. doi:10.1016/j.ceb.2014.03.001 Redwood AB, Perkins SM, Vanderwaal RP, Feng Z, Biehl KJ,
Peric-Hupkes D, Meuleman W, Pagie L, Bruggeman SWM, Gonzalez-Suarez I, Morgado-Palacin L, Shi W, Sage J,
Solovei I, Brugman W, Gräf S, Flicek P, Kerkhoven RM, Roti-Roti JL, et al. 2011. A dual role for A-type lamins
van Lohuizen M, et al. 2010. Molecular maps of the reor- in DNA double-strand break repair. Cell Cycle 10: 2549–
ganization of genome-nuclear lamina interactions during 2560. doi:10.4161/cc.10.15.16531

22 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

Robson MI, de Las Heras JI, Czapiewski R, Sivakumar A, Shimamoto Y, Tamura S, Masumoto H, Maeshima K. 2017.
Kerr ARW, Schirmer EC. 2017. Constrained release of Nucleosome-nucleosome interactions via histone tails
lamina-associated enhancers and genes from the nuclear and linker DNA regulate nuclear rigidity. Mol Biol Cell
envelope during T-cell activation facilitates their associa- 28: 1580–1589. doi:10.1091/mbc.e16-11-0783
tion in chromosome compartments. Genome Res 27: Shimi T, Koujin T, Segura-Totten M, Wilson KL, Haraguchi
1126–1138. doi:10.1101/gr.212308.116 T, Hiraoka Y. 2004. Dynamic interaction between BAF
Rothballer A, Kutay U. 2013. The diverse functional LINCs and emerin revealed by FRAP, FLIP, and FRET analyses
of the nuclear envelope to the cytoskeleton and chroma- in living HeLa cells. J Struct Biol 147: 31–41. doi:10.1016/j
tin. Chromosoma 122: 415–429. doi:10.1007/s00412-013- .jsb.2003.11.013
0417-x Shimi T, Pfleghaar K, Kojima S, Pack CG, Solovei I, Goldman
Salina D, Enarson P, Rattner JB, Burke B. 2003. Nup358 AE, Adam SA, Shumaker DK, Kinjo M, Cremer T, et al.
integrates nuclear envelope breakdown with kinetochore 2008. The A- and B-type nuclear lamin networks: micro-
assembly. J Cell Biol 162: 991–1001. doi:10.1083/jcb domains involved in chromatin organization and tran-
.200304080 scription. Genes Dev 22: 3409–3421. doi:10.1101/gad
Sanborn AL, Rao SSP, Huang S-C, Durand NC, Huntley .1735208
MH, Jewett AI, Bochkov ID, Chinnappan D, Cutkosky Shimi T, Butin-Israeli V, Adam SA, Hamanaka RB, Gold-
A, Li J, et al. 2015. Chromatin extrusion explains key man AE, Lucas CA, Shumaker DK, Kosak ST, Chandel
features of loop and domain formation in wild-type and NS, Goldman RD. 2011. The role of nuclear lamin B1 in
engineered genomes. Proc Natl Acad Sci 112: E6456– cell proliferation and senescence. Genes Dev 25: 2579–
E6465. doi:10.1073/pnas.1518552112 2593. doi:10.1101/gad.179515.111
Sasse B, Aebi U, Stuurman N. 1998. A tailless Drosophila Shimi T, Kittisopikul M, Tran J, Goldman AE, Adam SA,
lamin Dm0 fragment reveals lateral associations of di- Zheng Y, Jaqaman K, Goldman RD. 2015. Structural or-
mers. J Struct Biol 123: 56–66. doi:10.1006/jsbi.1998.4006 ganization of nuclear lamins A, C, B1, and B2 revealed by
Schirmer EC, Florens L, Guan T, Yates JR 3rd, Gerace L. superresolution microscopy. Mol Biol Cell 26: 4075–4086.
2003. Nuclear membrane proteins with potential disease doi:10.1091/mbc.E15-07-0461
links found by subtractive proteomics. Science 301: 1380– Shin J-W, Spinler KR, Swift J, Chasis JA, Mohandas N, Dis-
1382. doi:10.1126/science.1088176 cher DE. 2013. Lamins regulate cell trafficking and lineage
Schütz W, Alsheimer M, Öllinger R, Benavente R. 2005a. maturation of adult human hematopoietic cells. Proc Natl
Nuclear envelope remodeling during mouse spermiogen- Acad Sci 110: 18892–18897. doi:10.1073/pnas.1304
esis: postmeiotic expression and redistribution of germ- 996110
line lamin B3. Exp Cell Res 307: 285–291. doi:10.1016/j
Shumaker DK, Lee KK, Tanhehco YC, Craigie R, Wilson KL.
.yexcr.2005.03.023
2001. LAP2 binds to BAFDNA complexes: requirement
Schütz W, Benavente R, Alsheimer M. 2005b. Dynamic for the LEM domain and modulation by variable regions.
properties of germ line-specific lamin B3: the role of the EMBO J 20: 1754–1764. doi:10.1093/emboj/20.7.1754
shortened rod domain. Eur J Cell Biol 84: 649–662. doi:10
.1016/j.ejcb.2005.03.001 Shumaker DK, Dechat T, Kohlmaier A, Adam SA, Bozovsky
MR, Erdos MR, Eriksson M, Goldman AE, Khuon S,
Schwarzer W, Abdennur N, Goloborodko A, Pekowska A, Collins FS, et al. 2006. Mutant nuclear lamin A leads to
Fudenberg G, Loe-Mie Y, Fonseca NA, Huber W, Haering progressive alterations of epigenetic control in premature
CH, Mirny L, et al. 2017a. Two independent modes of aging. Proc Natl Acad Sci 103: 8703–8708. doi:10.1073/
chromatin organization revealed by cohesin removal. Na- pnas.0602569103
ture 551: 51–56. doi:10.1038/nature24281
Shumaker DK, Solimando L, Sengupta K, Shimi T, Adam
Schwarzer W, Abdennur N, Goloborodko A, Pekowska A,
SA, Grunwald A, Strelkov SV, Aebi U, Cardoso MC,
Fudenberg G, Loe-Mie Y, Fonseca NA, Huber W, Haering
Goldman RD. 2008. The highly conserved nuclear lamin
CH, Mirny L, et al. 2017b. Two independent modes of
Ig-fold binds to PCNA: its role in DNA replication. J Cell
chromatin organization revealed by cohesin removal. Na-
Biol 181: 269–280. doi:10.1083/jcb.200708155
ture 551: 51–56. doi:10.1038/nature24281
Simon DN, Wilson KL. 2013. Partners and post-translation-
Senior A, Gerace L. 1988. Integral membrane proteins spe-
al modifications of nuclear lamins. Chromosoma 122: 13–
cific to the inner nuclear membrane and associated with
the nuclear lamina. J Cell Biol 107: 2029–2036. doi:10 31. doi:10.1007/s00412-013-0399-8
.1083/jcb.107.6.2029 Solovei I, Wang AS, Thanisch K, Schmidt CS, Krebs S,
Shah PP, Donahue G, Otte GL, Capell BC, Nelson DM, Cao Zwerger M, Cohen TV, Devys D, Foisner R, Peichl L, et
K, Aggarwala V, Cruickshanks HA, Rai TS, McBryan T, et al. 2013. LBR and lamin A/C sequentially tether periph-
al. 2013. Lamin B1 depletion in senescent cells triggers eral heterochromatin and inversely regulate differentia-
large-scale changes in gene expression and the chromatin tion. Cell 152: 584–598. doi:10.1016/j.cell.2013.01.009
landscape. Genes Dev 27: 1787–1799. doi:10.1101/gad Somech R, Shaklai S, Geller O, Amariglio N, Simon AJ,
.223834.113 Rechavi G, Gal-Yam EN. 2005. The nuclear-envelope
Shah PP, Lv W, Rhoades JH, Poleshko A, Abbey D, Capor- protein and transcriptional repressor LAP2β interacts
izzo MA, Linares-Saldana R, Heffler JG, Sayed N, Thomas with HDAC3 at the nuclear periphery, and induces his-
D, et al. 2021. Pathogenic LMNA variants disrupt cardiac tone H4 deacetylation. J Cell Sci 118: 4017–4025. doi:10
lamina-chromatin interactions and de-repress alternative .1242/jcs.02521
fate genes. Cell Stem Cell 28: 938–954.e9. doi:10.1016/j Stephens AD, Banigan EJ, Adam SA, Goldman RD, Marko
.stem.2020.12.016 JF. 2017. Chromatin and lamin A determine two different

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 23
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

X. Wong et al.

mechanical response regimes of the cell nucleus. Mol Biol Vadrot N, Duband-Goulet I, Cabet E, Attanda W, Barateau
Cell 28: 1984–1996. doi:10.1091/mbc.e16-09-0653 A, Vicart P, Gerbal F, Briand N, Vigouroux C, Oldenburg
Stewart C, Burke B. 1987. Teratocarcinoma stem cells and AR, et al. 2015. The p.R482W substitution in A-type lam-
early mouse embryos contain only a single major lamin ins deregulates SREBP1 activity in Dunnigan-type fami-
polypeptide closely resembling lamin B. Cell 51: 383–392. lial partial lipodystrophy. Hum Mol Genet 24: 2096–2109.
doi:10.1016/0092-8674(87)90634-9 doi:10.1093/hmg/ddu728
Strickfaden H, Tolsma TO, Sharma A, Underhill DA, Han- van Schaik T, Vos M, Peric-Hupkes D, Celie PHN, van
sen JC, Hendzel MJ. 2020. Condensed chromatin behaves Steensel B. 2020. Cell cycle dynamics of lamina-associat-
like a solid on the mesoscale in vitro and in living cells. ed DNA. EMBO Rep 21: e50636. doi:10.15252/embr
Cell 183: 1772–1784.e13. doi:10.1016/j.cell.2020.11.027 .202050636
Strom AR, Emelyanov AV, Mir M, Fyodorov DV, Darzacq X, van Steensel B, Belmont AS. 2017. Lamina-associated do-
Karpen GH. 2017. Phase separation drives heterochroma- mains: links with chromosome architecture, heterochro-
tin domain formation. Nature 547: 241–245. doi:10.1038/ matin, and gene repression. Cell 169: 780–791. doi:10
nature22989 .1016/j.cell.2017.04.022
Stuurman N, Sasse B, Fisher PA. 1996. Intermediate filament Vaughan A, Alvarez-Reyes M, Bridger JM, Broers JL, Ra-
protein polymerization: molecular analysis of Drosophila maekers FC, Wehnert M, Morris GE, Whitfield WGF,
nuclear lamin head-to-tail binding. J Struct Biol 117: 1– Hutchison CJ. 2001. Both emerin and lamin C depend
15. doi:10.1006/jsbi.1996.0064 on lamin A for localization at the nuclear envelope. J Cell
Sci 114: 2577–2590. doi:10.1242/jcs.114.14.2577
Stuurman N, Heins S, Aebi U. 1998. Nuclear lamins: their
structure, assembly, and interactions. J Struct Biol 122: Vergnes L, Peterfy M, Bergo MO, Young SG, Reue K. 2004.
42–66. doi:10.1006/jsbi.1998.3987 Lamin B1 is required for mouse development and nuclear
integrity. Proc Natl Acad Sci 101: 10428–10433. doi:10
Sullivan T, Escalante-Alcalde D, Bhatt H, Anver M, Bhat N,
Nagashima K, Stewart CL, Burke B. 1999. Loss of A-type .1073/pnas.0401424101
lamin expression compromises nuclear envelope integrity Vogel MJ, Peric-Hupkes D, van Steensel B. 2007. Detection
leading to muscular dystrophy. J Cell Biol 147: 913–920. of in vivo protein–DNA interactions using DamID in
doi:10.1083/jcb.147.5.913 mammalian cells. Nat Protoc 2: 1467–1478. doi:10
Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PC, .1038/nprot.2007.148
Pinter J, Pajerowski JD, Spinler KR, Shin JW, Tewari M, Vorburger K, Lehner CF, Kitten GT, Eppenberger HM, Nigg
et al. 2013. Nuclear lamin-A scales with tissue stiffness EA. 1989. A second higher vertebrate B-type lamin.
and enhances matrix-directed differentiation. Science cDNA sequence determination and in vitro processing
341: 1240104. doi:10.1126/science.1240104 of chicken lamin B2. J Mol Biol 208: 405–415. doi:10
Tajik A, Zhang Y, Wei F, Sun J, Jia Q, Zhou W, Singh R, .1016/0022-2836(89)90505-6
Khanna N, Belmont AS, Wang N. 2016. Transcription Wang S, Stoops E, Cp U, Markus B, Reuveny A, Ordan E,
upregulation via force-induced direct stretching of chro- Volk T. 2018. Mechanotransduction via the LINC com-
matin. Nat Mater 15: 1287–1296. doi:10.1038/nmat4729 plex regulates DNA replication in myonuclei. J Cell Biol
217: 2005–2018. doi:10.1083/jcb.201708137
Thompson LJ, Bollen M, Fields AP. 1997. Identification
of protein phosphatase 1 as a mitotic lamin phosphatase. Wilson KL, Foisner R. 2010. Lamin-binding proteins. Cold
J Biol Chem 272: 29693–29697. doi:10.1074/jbc.272.47 Spring Harb Perspect Biol 2: a000554. doi:10.1101/cshper
.29693 spect.a000554
Torvaldson E, Kochin V, Eriksson JE. 2015. Phosphorylation Wong X, Stewart CL. 2020. The laminopathies and the in-
of lamins determine their structural properties and sig- sights they provide into the structural and functional or-
naling functions. Nucleus 6: 166–171. doi:10.1080/ ganization of the nucleus. Annu Rev Genomics Hum Ge-
19491034.2015.1017167 net 21: 263–288. doi:10.1146/annurev-genom-121219-
Towbin BD, González-Aguilera C, Sack R, Gaidatzis D, 083616
Kalck V, Meister P, Askjaer P, Gasser SM. 2012. Step- Wong X, Luperchio TR, Reddy KL. 2014. NET gains and
wise methylation of histone H3K9 positions heterochro- losses: the role of changing nuclear envelope proteomes
matin at the nuclear periphery. Cell 150: 934–947. doi:10 in genome regulation. Curr Opin Cell Biol 28: 105–120.
.1016/j.cell.2012.06.051 doi:10.1016/j.ceb.2014.04.005
Tsai M-Y, Wang S, Heidinger JM, Shumaker DK, Adam SA, Wong X, Hoskins VE, Harr JC, Gordon M, Reddy KL. 2020.
Goldman RD, Zheng Y. 2006. A mitotic lamin B matrix Lamin C regulates genome organization after mitosis.
induced by RanGTP required for spindle assembly. Sci- bioRxiv doi:10.1101/2020.07.28.213884.
ence 311: 1887–1893. doi:10.1126/science.1122771 Wong X, Loo TH, Stewart CL. 2021a. LINC complex regu-
Turgay Y, Eibauer M, Goldman AE, Shimi T, Khayat M, Ben- lation of genome organization and function. Curr Opin
Harush K, Dubrovsky-Gaupp A, Sapra KT, Goldman RD, Genet Dev 67: 130–141. doi:10.1016/j.gde.2020.12.007
Medalia O. 2017. The molecular architecture of lamins Wong X, Cutler JA, Hoskins VE, Gordon M, Madugundu
in somatic cells. Nature 543: 261–264. doi:10.1038/na AK, Pandey A, Reddy KL. 2021b. Mapping the micro-
ture21382 proteome of the nuclear lamina and lamina-associated
Ulbert S, Antonin W, Platani M, Mattaj IW. 2006. The inner domains. Life Sci Alliance 4: e202000774. doi:10.26508/
nuclear membrane protein Lem2 is critical for normal lsa.202000774
nuclear envelope morphology. FEBS Lett 580: 6435– Worman HJ, Schirmer EC. 2015. Nuclear membrane diver-
6441. doi:10.1016/j.febslet.2006.10.060 sity: underlying tissue-specific pathologies in disease?

24 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

Nuclear Lamina

Curr Opin Cell Biol 34: 101–112. doi:10.1016/j.ceb.2015 Ye Q, Callebaut I, Pezhman A, Courvalin JC, Worman HJ.
.06.003 1997. Domain-specific interactions of human HP1-type
Wu J, Kent IA, Shekhar N, Chancellor TJ, Mendonca A, chromodomain proteins and inner nuclear membrane
Dickinson RB, Lele TP. 2014. Actomyosin pulls to ad- protein LBR. J Biol Chem 272: 14983–14989. doi:10
vance the nucleus in a migrating tissue cell. Biophys J .1074/jbc.272.23.14983
106: 7–15. doi:10.1016/j.bpj.2013.11.4489 Zhang H, Emerson DJ, Gilgenast TG, Titus KR, Lan Y,
Xie W, Chojnowski A, Boudier T, Lim JS, Ahmed S, Ser Z, Huang P, Zhang D, Wang H, Keller CA, Giardine B, et
Stewart C, Burke B. 2016. A-type lamins form distinct al. 2019. Chromatin structure dynamics during the mito-
filamentous networks with differential nuclear pore com- sis-to-G1 phase transition. Nature 576: 158–162. doi:10
plex associations. Curr Biol 26: 2651–2658. doi:10.1016/j .1038/s41586-019-1778-y
.cub.2016.07.049
Zheng X, Hu J, Yue S, Kristiani L, Kim M, Sauria M, Taylor J,
Yang SH, Chang SY, Yin L, Tu Y, Hu Y, Yoshinaga Y, de Kim Y, Zheng Y. 2018. Lamins organize the global three-
Jong PJ, Fong LG, Young SG. 2011. An absence of
dimensional genome from the nuclear periphery. Mol Cell
both lamin B1 and lamin B2 in keratinocytes has no
71: 802–815.e7. doi:10.1016/j.molcel.2018.05.017
effect on cell proliferation or the development of skin
and hair. Hum Mol Genet 20: 3537–3544. doi:10.1093/ Zullo JM, Demarco IA, Piqué-Regi R, Gaffney DJ, Epstein
hmg/ddr266 CB, Spooner CJ, Luperchio TR, Bernstein BE, Pritchard
Yao J, Fetter RD, Hu P, Betzig E, Tjian R. 2011. Subnuclear JK, Reddy KL, et al. 2012. DNA sequence-dependent
segregation of genes and core promoter factors in myo- compartmentalization and silencing of chromatin at the
genesis. Genes Dev 25: 569–580. doi:10.1101/gad.202 nuclear lamina. Cell 149: 1474–1487. doi:10.1016/j.cell
1411 .2012.04.035

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a040113 25
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

The Nuclear Lamina


Xianrong Wong, Ashley J. Melendez-Perez and Karen L. Reddy

Cold Spring Harb Perspect Biol published online August 16, 2021

Subject Collection The Nucleus

Nuclear Compartments: An Incomplete Primer to Mechanisms of Chromosome Folding and Nuclear


Nuclear Compartments, Bodies, and Genome Organization: Their Interplay and Open Questions
Organization Relative to Nuclear Architecture Leonid Mirny and Job Dekker
Andrew S. Belmont
Uncovering the Principles of Genome Folding by Epigenetic Reprogramming in Early Animal
3D Chromatin Modeling Development
Asli Yildirim, Lorenzo Boninsegna, Yuxiang Zhan, Zhenhai Du, Ke Zhang and Wei Xie
et al.
3D or Not 3D: Shaping the Genome during Essential Roles for RNA in Shaping Nuclear
Development Organization
Juliane Glaser and Stefan Mundlos Sofia A. Quinodoz and Mitchell Guttman
The Impact of Space and Time on the Functional The Molecular and Nuclear Dynamics of
Output of the Genome X-Chromosome Inactivation
Marcelo Nollmann, Isma Bennabi, Markus Götz, et François Dossin and Edith Heard
al.
Chromatin Mechanisms Driving Cancer Structure, Maintenance, and Regulation of
Berkley Gryder, Peter C. Scacheri, Thomas Ried, et Nuclear Pore Complexes: The Gatekeepers of the
al. Eukaryotic Genome
Marcela Raices and Maximiliano A. D'Angelo
Liquid−Liquid Phase Separation in Chromatin The Nuclear Lamina
Karsten Rippe Xianrong Wong, Ashley J. Melendez-Perez and
Karen L. Reddy
Mechanical Forces in Nuclear Organization The Nuclear Pore Complex as a Transcription
Yekaterina A. Miroshnikova and Sara A. Wickström Regulator
Michael Chas Sumner and Jason Brickner
Imaging Organization of RNA Processing within Physical Nature of Chromatin in the Nucleus
the Nucleus Kazuhiro Maeshima, Shiori Iida and Sachiko
Jeetayu Biswas, Weihan Li, Robert H. Singer, et al. Tamura

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2021 Cold Spring Harbor Laboratory Press; all rights reserved
Downloaded from http://cshperspectives.cshlp.org/ at SEOUL NATL UNIV. on September 11, 2023 - Published by Cold Spring
Harbor Laboratory Press

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2021 Cold Spring Harbor Laboratory Press; all rights reserved

You might also like