Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and...

9
Please cite this article in press as: Gesson K, et al. Lamina-associated polypeptide (LAP)2 and nucleoplasmic lamins in adult stem cell regulation and disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcdb.2013.12.009 ARTICLE IN PRESS G Model YSCDB-1485; No. of Pages 9 Seminars in Cell & Developmental Biology xxx (2014) xxx–xxx Contents lists available at ScienceDirect Seminars in Cell & Developmental Biology jo ur nal homep age: www.elsevier.com/locate/semcdb Review Lamina-associated polypeptide (LAP)2 and nucleoplasmic lamins in adult stem cell regulation and disease Kevin Gesson 1 , Sandra Vidak 1 , Roland Foisner Max F. Perutz Laboratories, Department of Medical Biochemistry, Medical University Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria a r t i c l e i n f o Article history: Available online xxx Keywords: Adult stem cells Lamins Laminopathies Nuclear envelope Nuclear envelopathies Progeria Self-renewal a b s t r a c t A-type lamins are components of the lamina network at the nuclear envelope, which mediates nuclear stiffness and anchors chromatin to the nuclear periphery. However, A-type lamins are also found in the nuclear interior. Here we review the roles of the chromatin-associated, nucleoplasmic LEM protein, lamina-associated polypeptide 2 (LAP2) in the regulation of A-type lamins in the nuclear interior. The lamin A/C–LAP2 complex may be involved in the regulation of the retinoblastoma protein-mediated pathway and other signaling pathways balancing proliferation and differentiation, and in the stabilization of higher-order chromatin organization throughout the nucleus. Loss of LAP2 in mice leads to selective depletion of the nucleoplasmic A-type lamin pool, promotes the proliferative stem cell phenotype of tissue progenitor cells, and delays stem cell differentiation. These findings support the hypothesis that LAP2 and nucleoplasmic lamins are regulators of adult stem cell function and tissue homeostasis. Finally, we discuss potential implications of this concept for defining the molecular disease mechanisms of lamin- linked diseases such as muscular dystrophy and premature aging syndromes. © 2014 The Authors. Published by Elsevier Ltd. All rights reserved. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 2. Interplay between A-type lamins and LAP2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3. Role of A-type lamins in disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.1. The mechanical model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.2. The gene regulation model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 3.3. The stem cell model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 4. Functions of nucleoplasmic lamin A/C–LAP2 complexes and their link to disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 4.1. Nucleoplasmic LAP2–lamin A/C complexes in proliferation and differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 4.2. Lamins in stem cell regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 4.3. Impaired proliferation/differentiation pathways in laminopathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 5. Lamins in chromatin organization and implication for laminopathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 5.1. The role of the nuclear lamina in chromatin organization and gene expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 5.2. A-type lamins and chromatin regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00 Abbreviations: ASC, (somatic) adult stem cell; BAF, barrier-to-autointegration factor; Dam, DNA adenine methyltransferase; DCM, dilated cardiomyopathy; EDMD, Emery Dreifuss muscular dystrophy; ESC, embryonic stem cell; FPLD, familial partial lipodystrophy; INM, inner nuclear membrane; HGPS, Hutchinson–Gilford Progeria Syndrome; iPS, induced pluripotent stem cell; LAD, lamina-associated domain; LAP, lamina-associated polypeptide; LEM, LAP2-Emerin-Man1; LRD, lamin rich domain; MDPSC, muscle-derived stem/progenitor cells; MSC, mesenchymal stem cell; NE, nuclear envelope; pRb, retinoblastoma protein. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited. Corresponding author at: Max F. Perutz Laboratories, Medical University Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria. Tel.: +43 1 427761680. E-mail address: [email protected] (R. Foisner). 1 These authors contributed equally. 1084-9521/$ see front matter © 2014 The Authors. Published by Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.semcdb.2013.12.009

Transcript of Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and...

Page 1: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

G

Y

R

La

KM

a

AA

KALLNNPS

C

ESM

a

1h

ARTICLE IN PRESS Model

SCDB-1485; No. of Pages 9

Seminars in Cell & Developmental Biology xxx (2014) xxx– xxx

Contents lists available at ScienceDirect

Seminars in Cell & Developmental Biology

jo ur nal homep age: www.elsev ier .com/ locate /semcdb

eview

amina-associated polypeptide (LAP)2� and nucleoplasmic lamins indult stem cell regulation and disease�

evin Gesson1, Sandra Vidak1, Roland Foisner ∗

ax F. Perutz Laboratories, Department of Medical Biochemistry, Medical University Vienna, Dr. Bohr-Gasse 9, 1030 Vienna, Austria

r t i c l e i n f o

rticle history:vailable online xxx

eywords:dult stem cellsaminsaminopathies

a b s t r a c t

A-type lamins are components of the lamina network at the nuclear envelope, which mediates nuclearstiffness and anchors chromatin to the nuclear periphery. However, A-type lamins are also found inthe nuclear interior. Here we review the roles of the chromatin-associated, nucleoplasmic LEM protein,lamina-associated polypeptide 2� (LAP2�) in the regulation of A-type lamins in the nuclear interior. Thelamin A/C–LAP2� complex may be involved in the regulation of the retinoblastoma protein-mediatedpathway and other signaling pathways balancing proliferation and differentiation, and in the stabilization

uclear envelopeuclear envelopathiesrogeriaelf-renewal

of higher-order chromatin organization throughout the nucleus. Loss of LAP2� in mice leads to selectivedepletion of the nucleoplasmic A-type lamin pool, promotes the proliferative stem cell phenotype oftissue progenitor cells, and delays stem cell differentiation. These findings support the hypothesis thatLAP2� and nucleoplasmic lamins are regulators of adult stem cell function and tissue homeostasis. Finally,we discuss potential implications of this concept for defining the molecular disease mechanisms of lamin-linked diseases such as muscular dystrophy and premature aging syndromes.

© 2014 The Authors. Published by Elsevier Ltd. All rights reserved.

ontents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 002. Interplay between A-type lamins and LAP2� . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003. Role of A-type lamins in disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

3.1. The mechanical model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.2. The gene regulation model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 003.3. The stem cell model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

4. Functions of nucleoplasmic lamin A/C–LAP2� complexes and their link to disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 004.1. Nucleoplasmic LAP2�–lamin A/C complexes in proliferation and differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 004.2. Lamins in stem cell regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 004.3. Impaired proliferation/differentiation pathways in laminopathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

5. Lamins in chromatin organization and implication for laminopathies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 005.1. The role of the nuclear lamina in chromatin organization and gene expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

5.2. A-type lamins and chromatin regulation . . . . . . . . . . . . . . . . . . . . . . . . . . .Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Abbreviations: ASC, (somatic) adult stem cell; BAF, barrier-to-autointegration factomery Dreifuss muscular dystrophy; ESC, embryonic stem cell; FPLD, familial partial liyndrome; iPS, induced pluripotent stem cell; LAD, lamina-associated domain; LAP, lamDPSC, muscle-derived stem/progenitor cells; MSC, mesenchymal stem cell; NE, nuclear

� This is an open-access article distributed under the terms of the Creative Commons Atny medium, provided the original author and source are credited.∗ Corresponding author at: Max F. Perutz Laboratories, Medical University Vienna, Dr. B

E-mail address: [email protected] (R. Foisner).1 These authors contributed equally.

084-9521/$ – see front matter © 2014 The Authors. Published by Elsevier Ltd. All rights ttp://dx.doi.org/10.1016/j.semcdb.2013.12.009

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

r; Dam, DNA adenine methyltransferase; DCM, dilated cardiomyopathy; EDMD,podystrophy; INM, inner nuclear membrane; HGPS, Hutchinson–Gilford Progeria

ina-associated polypeptide; LEM, LAP2-Emerin-Man1; LRD, lamin rich domain; envelope; pRb, retinoblastoma protein.tribution License, which permits unrestricted use, distribution and reproduction in

ohr-Gasse 9, 1030 Vienna, Austria. Tel.: +43 1 427761680.

reserved.

Page 2: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ING Model

Y

2 evelo

1

cv[ntVbfTLealilfttasmlantpnncnptAh

2

v[[mn[lLCoadtmlbmft

bisam

ARTICLESCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & D

. Introduction

The nuclear lamina is a proteinaceous network in metazoanells that underlies the inner nuclear membrane (INM) and pro-ides mechanical stability for the nuclear envelope (NE) (Fig. 1)1–3]. It also fulfills a plethora of functions in chromatin orga-ization, gene expression and signaling during development andissue maintenance [4–10]. The lamina network is formed by type

intermediate filaments, the lamins [11–13], and a large num-er of lamin-binding proteins of the INM [14,15]. Structurally andunctionally, lamins are grouped into A- and B-type lamins [16].he main B-type lamins, lamin B1 and lamin B2 are encoded byMNB1 and LMNB2, respectively, and at least one B-type lamin isxpressed in most cells throughout development. A-type laminsre encoded by the LMNA gene, giving rise to two major isoforms,amin A and C, which are expressed later in development andn a differentiation-dependent manner [17]. Importantly, B-typeamins are processed post-translationally to yield a C-terminallyarnesylated mature protein that is tightly associated with the INMhrough its hydrophobic farnesyl group. In contrast, newly syn-hesized pre-lamin A is also farnesylated during processing, but in

final maturation step a C-terminal peptide, including the farne-yl group, is proteolytically cleaved, producing a non-farnesylatedature lamin A [18–20]. Therefore, unlike B-type lamins, A-type

amins are less tightly linked to the INM and the lamina and arelso found in a more mobile and dynamic pool throughout theucleoplasm [21–24]. However, the regulation and specific func-ions of this dynamic, nucleoplasmic pool of A-type lamins are stilloorly understood. Recent studies revealed evidence for excitingovel functions of this nucleoplasmic lamin pool in chromatin orga-ization, cell signaling and cell cycle control in adult tissue stemells (ASCs). In this review we discuss the potential functions ofucleoplasmic A-type lamins in fine-tuning the balance betweenroliferation and differentiation of ASCs, which is of crucial impor-ance for tissue homeostasis. We also discuss how nucleoplasmic-type lamins may affect the regulation of stem cell activity andow these functions may be altered in lamin-linked diseases.

. Interplay between A-type lamins and LAP2�

Lamina-associated polypeptide 2 � (LAP2�) is one of six spliceariants of the mammalian LAP2 gene (originally termed TMPO)25–28]. All LAP2 isoforms share the first 187 N-terminal residues29] harboring the LAP2-Emerin-MAN1 (LEM)-domain [30], which

ediates interaction with DNA in a sequence-independent man-er via the adaptor protein barrier-to-autointegration factor (BAF)31]. The common N-terminal LAP2 domain also contains a LEM-ike motif enabling direct interaction with DNA [30,31]. Thus, allAP2 proteins interact with chromatin by several mechanisms. The-terminal domain of LAP2� differs considerably from that of thether LAP2 isoforms. Whereas most LAP2 isoforms, such as LAP2�,re stably anchored in the INM via a C-terminal transmembraneomain, LAP2� is a non-membrane protein uniformly distributedhroughout the nucleoplasm [32]. Furthermore, whereas the LAP2

embrane proteins primarily bind B-type lamins at the nuclearamina [33], LAP2�’s unique C-terminal tail mediates exclusiveinding to A-type lamins [22,24] and contains an additional chro-osome association domain [34,35], as well as an interaction site

or the cell cycle and differentiation regulator, retinoblastoma pro-ein (pRb) [36,37].

The specific interaction of A-type lamins and LAP2� haseen extensively studied by several means, including co-

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

mmunoprecipitation, cell cycle-dependent co-localization analy-es and a proximity based biotin ligase assay in mammalian cells,s well as by in vitro solid phase overlay and pull-down experi-ents [22,32,38,39]. These studies revealed direct interaction of

PRESSpmental Biology xxx (2014) xxx– xxx

lamins A/C and LAP2� via their C-terminal tails [22] and a dynamicassociation of the proteins during the cell cycle. The nucleoplas-mic lamin A/C–LAP2� complexes exist in G1 and early S-phase ofproliferating cells but are absent during mitosis [32,40]. Intrigu-ingly, LAP2� appears to be a crucial factor for the regulation andstabilization of the nucleoplasmic pool of lamin A/C and its local-ization in the nuclear interior (Fig. 1). In cells and epithelial tissuesderived from LAP2�-deficient mice, A-type lamins localize exclu-sively to the nuclear lamina and are absent from the nuclearinterior. Re-expression of full length LAP2�, but not of a laminbinding-defective LAP2� mutant, into LAP2�-deficient cells res-cues the nucleoplasmic pool of lamin A/C [24]. Furthermore, lossof the nucleoplasmic pool of A-type lamins during myoblast differ-entiation correlates with the downregulation of LAP2� [41].

Therefore, LAP2� is a master regulator of the nucleoplasmiclamin A/C pool, but the mechanisms by which LAP2� affects nucle-oplasmic lamins remain elusive. In G1 phase of the cell cycle,nucleoplasmic A-type lamins may originate from lamin complexesdisassembled in the preceding mitosis, or may represent newlysynthesized pre-lamin A, which may interact with LAP2� in thenucleoplasm only transiently, before they assemble into the nuclearlamina. The most intriguing scenario, however, is that A-typelamins are dynamically exchanged between the peripheral and thenucleopasmic pool, depending on post-translational modificationsand/or the interaction of LAP2� and other factors.

3. Role of A-type lamins in disease

In 1999, Bonne et al. described the first mutation in the LMNAgene linked to autosomal dominant Emery Dreifuss muscular dys-trophy (EDMD) [42]. Since then about 400 disease-linked mutationswere identified in A-type lamins and in several lamin-binding pro-teins of the nuclear envelope.

These mutations cause a variety of diseases, collectively termedprimary laminopathies for lamin A/C-linked diseases and nuclearenvelopathies for diseases linked to nuclear envelope proteins Theyaffect different tissues (striated muscle, heart, fat, bone, skin, orneuronal tissues) in isolation or in various combinations, or causepremature aging diseases, e.g., Hutchinson–Gilford Progeria Syn-drome (HGPS) [43–47].

Also a mutation in LAP2 ̨ has been linked to dilated cardiomy-opathy (DCM) [39], the pathological features of which resemblethose of lamin A-linked DCM. Interestingly, this DCM-causingLAP2� mutation, which leads to a single amino acid exchange inthe C-terminal lamin A/C-binding domain of LAP2� was shown toimpair LAP2�’s interaction with lamin A/C in vitro [39].

Most disease-causing mutations in the LMNA gene are heterozy-gous single point mutations in LMNA found throughout the gene,leading to the expression of mutant lamin A/C variants with a sin-gle amino acid exchange. In contrast, the majority of mutationslinked to HGPS introduce a cryptic splice site in exon 11 of LMNA,causing incorrect splicing and generation of a slightly smaller pre-lamin A variant (called progerin) that cannot be cleaved in the finalstep of post-translational processing and therefore remains perma-nently farnesylated [48]. Given that A-type lamins are expressedin nearly every differentiated cell, the tissue-specific phenotypesof many laminopathies are surprising, and the molecular pathwaysleading to the different pathological phenotypes are still not under-stood. Several non-mutually exclusive disease mechanisms havebeen proposed to explain the tissue-specific aspects and variabilityof laminopathic phenotypes [49,50].

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

3.1. The mechanical model

LMNA mutations may disrupt the stability or assembly of laminnetworks, rendering the nucleus more fragile and less resistant

Page 3: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ARTICLE IN PRESSG Model

YSCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & Developmental Biology xxx (2014) xxx– xxx 3

F al A-tr

tcsamp[lidngcaciaemE

3

a[iwcsmuatsWa

ig. 1. LAP2� facilitates translocation of A-type lamins to the nucleoplasm. Peripheregulate chromatin organization.

o mechanical stress, ultimately leading to structural damage andell death in mechanically stressed tissues [51]. This model isupported by reports that lamin A/C-deficient fibroblasts, as wells cells derived from several laminopathy patients have abnor-ally shaped nuclei [52–54], and skeletal muscle from EDMD

atients and mouse disease models exhibit fragmented nuclei55,56]. Biomechanical studies showed that, unlike B-type lamins,amins A/C are the primary contributors to nuclear mechan-cs [54,57]. Accordingly, lamin A/C-deficient fibroblasts showecreased nuclear and cytoskeletal mechanical stiffness, increaseduclear fragility and impaired activation of mechanosensitiveenes [58–61]. Mutations in A-type lamins can also disrupt nucleo-ytoskeletal coupling, leading to a disturbance of nuclear anchoragend impaired ability to transmit intracellular forces between theytoskeleton and nuclear interior [51,62]. Therefore, the mechan-cal model may best describe muscular-dystrophy laminopathies,s muscles are exposed to high physical forces. For instance, cellsxpressing Familial Partial Lipodystrophy (FPLD)-linked lamin A/Cutants have normal nuclear stiffness, while mutations linked to

DMD and DCM result in a loss of nuclear stability [51].

.2. The gene regulation model

This model proposes that mutations in A-type lamins or theirssociated proteins cause dysregulation of tissue-specific genes46]. The altered regulation of genes may be caused by thempairment of heterochromatin formation and epigenetic path-

ays found in many laminopathic cells and in Lmna−/− mouseells [63–65]. Lamins can also affect signaling and gene expres-ion by direct interactions with transcription factors and signalingolecules. In particular, signaling pathways involved in the reg-

lation of proliferation and differentiation have been found to beffected in laminopathies, including pRb, mitogen activated pro-

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

ein kinase (MAPK), Notch, transforming growth factor � (TGF-�),terol response element binding protein-1 (SREBP-1), NF-�B and

nt/�-catenin pathways [4,10]. In support of this notion, cellsnd tissues derived from EDMD and DCM mouse models and

ype lamins and nucleoplasmic A-type lamins, alone or in complex with LAP2�, may

patients show upregulated MAPK signaling [66], and HGPS patientcells present defective Wnt-, Notch- and pRb signaling [67–69]. Inaddition, in mouse models for progeria, NF-�B signaling was con-stitutively hyperactivated, leading to upregulation of inflammatorycytokines [70]. Lamin A/C-deficient cells show impaired activationof mechanosensitive genes (Egr1, lex1 and Mlk1) and decreased NF-�B signaling [59,60,71], thus potentially linking the mechanical andgene regulation disease models.

3.3. The stem cell model

At the cellular level, this model proposes that mutations inLMNA result in proliferation and differentiation defects, which maywell be directly linked to the mechanic and/or gene regulationdefects mentioned above. This model is based on findings thatA-type lamins interact functionally with two important regula-tors of G1 to S phase cell cycle progression, pRb and cyclin D3[37,72,73] (see Section 4.1). Human HGPS fibroblasts show rapidgrowth at early passages, but undergo premature senescence athigher passage numbers [74], and murine progeria fibroblasts alsoundergo premature cellular senescence [75]. Both, mesenchymalstem cells expressing progerin [76] and epidermal stem cells in skinof progeria mice [67] display impaired proliferation and/or differ-entiation. Loss of wild-type lamin A/C or the expression of EDMDlamin A/C mutants compromise myoblast differentiation [77,78],and overexpression of wild-type lamin A or FPLD-linked lamin Amutants affect adipocyte differentiation [79]. Lamins A/C appearto be involved also in osteoblast differentiation, as knock-downof lamin A/C caused impaired osteoblastogenesis and acceleratedosteoclastogenesis in human bone marrow stromal cells [80,81].

4. Functions of nucleoplasmic lamin A/C–LAP2� complexesand their link to disease

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

To date, only a handful of studies have addressed the potentialfunctions of nucleoplasmic A-type lamins as opposed to those oflamins at the nuclear periphery. However, many of the described

Page 4: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ING Model

Y

4 evelo

fpelfoibf

sbndpo

4a

vtcIitodasfs

plplpaispiAm

Asrptimlddwhll[abfip

ARTICLESCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & D

unctions of lamins, which intrinsically have been linked to theeripheral lamina, may partly or predominantly require their pres-nce within the nucleus. It is plausible to assume that, similar to theamina, nucleoplasmic lamin A/C complexes can serve as a scaffoldor signaling molecules [4,82] and may contribute to chromatinrganization and epigenetic regulation of genes in the nuclearnterior [83]. They may even contribute to nuclear mechanicsy stabilizing an intranuclear meshwork that absorbs mechanicalorces evenly like a sponge [60].

As in classical lamin A/C knock-out experiments or by expres-ion of mutant versions of lamin A/C both lamin pools are likely toe affected, it is difficult to distinguish between peripheral versusucleoplasmic lamin functions. Hence, the LAP2� knock-out mice,isplaying significantly reduced levels of nucleoplasmic lamins A/C,rovide an experimental system to selectively study the functionsf A-type lamins in the nuclear interior [24].

.1. Nucleoplasmic LAP2˛–lamin A/C complexes in proliferationnd differentiation

Both LAP2� [36,37] and lamins A and C [84,85] bind pRb initro and in vivo. pRb is a major cell cycle regulator that represseshe activity of E2F transcription factors and thereby inhibits cellycle progression in a phosphorylation-dependent manner [86].n the absence of mitogenic signals or upon differentiation, pRbs hypo-phosphorylated, binds to E2F and inhibits E2F target generanscription, allowing cells to exit the cell cycle. In the presencef mitogenic signals, pRb is heavily phosphorylated by cyclin-ependent kinases, causing release from E2F transcription factors,ctivation of E2F-dependent transcription and cell cycle progres-ion. This basic pRb cell proliferation-regulating cycle is subject tourther control by many additional pathways and feedback loops,ome of which may also include nucleoplasmic A-type lamins.

Several mechanisms have been proposed to explain how nucleo-lasmic lamin A/C and LAP2� affect pRb function (Fig. 2): (i) A-type

amins may stabilize pRb protein, since pRb was degraded via theroteosomal pathway in lamin A/C-deficient cells [72]. (ii) A-type

amins may provide a scaffold for efficient dephosphorylation ofRb by PP2A protein phosphatase upon TGFß-induced cell cyclerrest [87]. (iii) Interaction of pRb with A-type lamins may keep pRbn its active (repressive) hypo-phosphorylated state. Upon growthtimulation, ERK kinase translocates to the nucleus and may com-ete with pRb for binding to lamins A/C, causing release of pRb and

ts efficient phosphorylation by cyclin-dependent kinases [88]. (iv) complex of LAP2�, lamin A/C and hypo-phosphorylated pRb [37]ay be involved in efficient E2F target gene repression [36].In accordance with these mechanisms, nucleoplasmic lamins

/C and LAP2� were found to negatively affect cell cycle progres-ion and thus enhance cell cycle arrest in tissue progenitor cells ofegenerating tissues. Overexpression of LAP2� in cultured murinere-adipocytes drives cells into cell cycle exit and initiates differen-iation in the absence of hormones [36]. In contrast, loss of LAP2�mpairs efficient cell cycle exit by contact inhibition in primary

urine fibroblasts [24]. LAP2�-deficient myoblasts express higherevels of stemness factors compared to wild-type cells and showelayed differentiation in vivo [89]. Correspondingly, in LAP2�-eficient mice, the number of proliferating tissue progenitor cellsas significantly increased in skin, colon, skeletal muscle, and in theematopoietic system [24,40,89,90]. Furthermore, loss of LAP2� in

amin A/C-deficient mice, which lack wild-type lamin A but expressow levels of a lamin A �8–11 variant in some cells and tissues91], prolonged life span of double mutant mice from 30 to 70 days

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

nd partially rescued the muscle growth phenotype [92], proba-ly by promoting proliferation of muscle progenitor cells. Thesendings suggest that nucleoplasmic lamin A/C–LAP2� complexesermit and/or promote differentiation of tissue progenitor cells and

PRESSpmental Biology xxx (2014) xxx– xxx

may thus be involved in tissue homeostasis by controlling the bal-ance between proliferation and differentiation of adult stem cellsas described in the following section.

4.2. Lamins in stem cell regulation

A-type lamins are absent or expressed at very low levels inundifferentiated embryonic stem cells (ESCs), and are upregulatedonly during cell differentiation [93,94]. Furthermore, upon repro-gramming of somatic fibroblasts to induced pluripotent stem (iPS)cells, lamin A levels are vastly decreased. Knockdown of laminA during reprogramming facilitates iPS cell generation, whereasoverexpression inhibits the induction of pluripotency and drivesdifferentiation [95].

While A-type lamins may be less important for initiation of EScell differentiation, they may have important regulatory roles insomatic (adult) stem cells (ASCs). These are tissue-specific stemcells which serve as a clonogenic, self-renewing reservoir with thecapability to differentiate into multiple cell lineages and are respon-sible for maintaining tissue homeostasis in adult organism byreplenishing dying and non-functional cells [96]. Adult stem cellsinclude hematopoietic and mesenchymal stem cells (MSCs). MSCsdifferentiate to committed precursor cells important for the regen-eration of muscle, heart, bone, adipose, nerve and skin tissue, allof which are severely affected in different laminopathies. There isevidence that A-type lamins are important for regulating the main-tenance and differentiation of both MSCs and tissue progenitorcells by influencing key signaling pathways [49,97]. Downregu-lation of A-type lamins or expression of HGPS lamin A variantin MSCs affects osteogenic, chondrogenic and adipogenic differ-entiation [76,81,98]. Furthermore, in two different HGPS mousemodels, epidermal stem cells were depleted causing an inflam-matory response [67,99]. Muscle-derived stem/progenitor cells(MDPSCs) from progeria mice also displayed defective proliferationand differentiation [100]. Interestingly, intraperitoneal administra-tion of MDPSCs derived from young wild-type mice to progeroidmice leads to significant extension of lifespan, suggesting thatimpaired MDPSC function in progeria mice results in a reduced lifeexpectancy.

Altogether, the findings that lamins alter adult stem cell functionled to the hypothesis that at least part of the phenotypes observedin laminopathies are due to defects in stem cell-mediated tissueregeneration [49,101]. Increased turnover and abnormal differenti-ation of adult stem cells in laminopathies may also deplete the stemcell pool. The stem cell defect, coupled with a potentially increasedmechanical sensitivity, could result in an inefficient repair of dam-aged tissues in HGPS and other laminopathies [101–103].

4.3. Impaired proliferation/differentiation pathways inlaminopathies

Many mutations linked to laminopathies affect the localizationof A-type lamins, either increasing or decreasing the nucleoplasmicpool of A-type lamins or leading to aggregation of mutant laminsin the nucleoplasm [104–107]. Therefore, it is conceivable that atleast some of the molecular defects underlying laminopathies arelinked to a misregulation of the functions of nucleoplasmic laminsA/C. Based on the role of nucleoplasmic lamins A/C and LAP2� inpRb-mediated cell cycle control (Fig. 2), it is tempting to speculatethat mutations in these proteins affect pRb-mediated pathways andderail the balance between proliferation/self-renewal and differ-entiation of tissue progenitor cells. EDMD- or HGPS-linked lamin A

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

mutants impair phosphorylation of pRb [41,108], which may leadto premature cell cycle exit and senescence and to the inhibitionof differentiation [109,110]. In addition, pRb is downregulated inthe Zinc metalloproteinase Ste24 homolog (Zmpste24)−/− progeria

Page 5: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ARTICLE IN PRESSG Model

YSCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & Developmental Biology xxx (2014) xxx– xxx 5

F by sed r LAP2

mfii[

faptMipmtom

itplsa

ig. 2. A-type lamins and LAP2� affect the cell cycle-regulating functions of pRbifferentiation of adult stem cells. Disease linked perturbations of lamins A/C and/o

ouse model [111], and genome-wide expression analysis identi-ed the lamin A-pRb signaling network as a major pathway affected

n HGPS [69]. Also the abnormal pRb localization in laminopathies112] could further contribute to pRb dysregulation.

Besides its role in cell cycle control, pRb has well-establishedunctions in the differentiation of muscle, adipose tissue, bone,nd epidermis, all of which are affected in laminopathies. TheRb/MyoD pathway is the master regulator of myogenesis in skele-al muscle. pRb interacts with the myogenic transcription factor

yoD, subsequently activating MyoD-target genes and therebynitiating myoblast differentiation [113]. Thus, defects of the pRbathway in laminopathies may not only affect cell cycle exit, butay also impair pRb’s role in differentiation [103]. In line with

his model, Lmna-deficient skeletal myocytes express lower levelsf MyoD protein and consequently exhibit impaired MyoD/Rb-ediated in vitro myogenesis [77,78].Besides pRb pathways, other differentiation-mediating signal-

ng pathways were shown to be affected in mutant cells andissues. The aberrant differentiation of MSCs ectopically expressing

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

rogerin was linked to increased Notch signaling [76]. Wild-typeamin A associates with the Notch co-activator SKIP, therebycavenging SKIP and reducing Notch-dependent transcriptionalctivity. Progerin has reduced affinity for SKIP leading to an

veral mechanisms (for details see text) balancing proliferation/self-renewal and� may result in an imbalance between these two cell fates.

increase in SKIP availability and activation of Notch downstreameffectors. Moreover, the Wnt/�-catenin pathway, which is knownto promote stem cell proliferation in stem-cell niches of theintestine, bone marrow, brain, and epidermis, was found to beattenuated in HGPS mouse models, altering extracellular matrixproduction [68].

5. Lamins in chromatin organization and implication forlaminopathies

Chromatin is non-randomly organized in the nucleus throughformation of chromosome territories [114] and associations withthe NE/nuclear lamina and possibly other structural components inthe nucleus [7,115,8]. A-type lamins have long been proposed to beinvolved in the spatial organization of chromatin due to their abilityto interact with DNA and core histones [11]. Similarly, several laminbinding proteins, including Lamin B Receptor (LBR) and the LEMproteins [15,30] have been shown to interact with chromatin. How-

ptide (LAP)2˛ and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

ever, only recently a few studies revealed two redundant pathwaystethering chromatin to the periphery, an LBR-mediated anchor-age (probably involving B-type lamins) and an A-type lamin-LEMprotein-dependent mechanism [116,117].

Page 6: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ING Model

Y

6 evelo

5g

mgtwotsHit[asitntlEagvtcsnf

trastdpumot

5

lel(brSc“icfgattflo

ARTICLESCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & D

.1. The role of the nuclear lamina in chromatin organization andene expression

The implementation of the DamID method using a DNA adenineethyltransferase (Dam)-lamin B1 fusion protein led to the first

enome-wide map of in vivo nuclear lamina–chromatin interac-ions [118,119]. The identified lamina-associated domains (LADs)ere shown to be large-scale, yet sharply confined genomic regions

f 0.1–10 Mb in size, which have transcriptional repressive fea-ures and represent gene-poor and heterochromatic regions withignificant enrichments of repressive histone marks (H3K27me3,3K9me3) [120]. This led to the concept that the nuclear periphery

s an overall transcriptionally repressive environment as opposedo the transcriptionally permissive conditions in the nucleoplasm83]. This model was supported by experiments showing thatrtificial tethering of genomic loci to the NE leads, at least inome cases, to their silencing [121–123]. Furthermore, genome-ntegrated arrays containing tissue-specific promoters were foundo localize at the periphery in embryos and translocated to theuclear center upon differentiation-dependent promoter activa-ion [124]. Additionally, a lamin B1-DamID approach trackingamina–chromatin interactions during differentiation of murineSCs to neuronal precursor cells and to terminally differentiatedstrocytes showed that previously stably NE-associated genes orene clusters detach from the NE and subsequently become acti-ated during differentiation [125]. These experiments indicatedhat the NE not only anchors heterochromatin, but may also activelyontribute to the generation of a heterochromatic, transcriptionallyilent environment. However, detachment from the NE per se doesot necessarily trigger immediate activation, but may poise genes

or later activation during terminal differentiation.Opposing the view that NE–chromatin interactions are stable in

he sense that they are inherited from mother to daughter cells, aecent study showed that in a given cell only 30% of all LADs aressociated with the NE and are stochastically reshuffled after mito-is [126]. It remains to be investigated whether LADs containingissue-specific genes are more specifically tethered to the NE duringifferentiation. Also, the “signature” (i.e., the epigenetic and geneticrofile) of chromatin mediating its association at the NE is poorlynderstood. A few recent studies have identified the heterochro-atic histone mark, H3K9me2 [126,127], A/T rich sequences [128],

r GAGA motifs [129] as important determinants for chromatin–NEethering.

.2. A-type lamins and chromatin regulation

Considering the dual location of A-type lamins at the nuclearamina and in the nucleoplasm, as opposed to the exclusive periph-ral localization of B-type lamins, it is conceivable that A-typeamins may also interact with chromatin in the nuclear interiorFig. 1). At the NE, certain chromatin attachment regions maye common for both A- and B-type lamins, while other genomicegions may be exclusive to one or the other. In support of this,himi et al. found that A- and B-type lamins form distinct, but inter-onnected, networks at the nuclear lamina [130]. While the termLAD” was originally coined for lamin B1–chromatin interactions,t appears to be extendable toward the association of lamin A withhromosomes at the NE. Genomic DamID maps for lamin A–Damusion proteins in human and murine cells are very similar to theenomic lamin B1 DamID maps, and constitutive NE–chromatinssociations (cLADs) are highly conserved across species and cellypes [128]. In addition to regions generally not associated with

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

he lamina (constitutive inter-LADs), certain regions were found toacultatively interact with A- and B-type lamins in the course ofineage commitment, and do so with a potential preference for oner the other lamin type [128].

PRESSpmental Biology xxx (2014) xxx– xxx

A recent study by Kubben et al. [131] shed light on the in vivochromatin interactions of wildtype versus mutant lamin A linkedto HGPS (progerin). In a genome-wide analysis of promoter inter-actions, they showed a preference of A-type lamins for binding topromoters of silent or lowly expressed genes preferentially locatedat the nuclear periphery. Progerin–promoter interactions over-lapped to a large extent with that of wild-type lamin A, but a fewprogerin-specific (inherently silent) gene promoters were iden-tified. This extensive overlap of wildtype lamin A and progerinpromoter interactions is surprising, given that progerin appears tohave reduced binding affinity to DNA and H3K27 trimethylated his-tones [132]. While the former study used overexpressed lamin Aand progerin, genome interaction mapping of endogenous laminA and progerin revealed global changes in the heterochromaticmark H3K27me3, which appeared to be lost in gene-poor regionsin the mutant cells and appeared stronger in gene-rich regions,resulting in the detachment from the nuclear lamina of gene-poorheterochromatic regions [133].

Another recent study on genome-wide lamin A/C-promoterassociation during adipogenic differentiation identified lamin-richdomains (LRDs) throughout the genome in a manner consistentwith lamin A/C–promoter interactions not being restricted to thenuclear periphery [134]. Complementary to previous findings, thisstudy showed that binding of lamin A/C fine-tunes target generegulation depending on the local chromatin environment in spe-cific promoter subregions. The results indicate that lamin A/Cassociation with promoters per se does not inhibit transcription,but additionally requires certain repressive histone marks in sub-promoter regions; conversely, loss of lamin A/C from promoters isa prerequisite, but not sufficient for transcriptional activation.

LAP2� is a potential candidate for mediating the interaction ofnucleoplasmic lamins A/C with chromatin. LAP2� possesses a LEMand LEM-like domain [30], and thereby binds to chromatin in asequence – independent manner. In human cells, LAP2� was foundto interact with genomic DNA in a highly dynamic manner and toaffect the chromatin-binding behavior of the high-mobility groupN protein 5 (HMGN5) [135]. Downregulation of LAP2� led to theredistribution of HMGN5-targeted chromatin sites. Based on thesefindings it is tempting to speculate that LAP2� could also affect thechromatin interaction of other proteins in the nuclear interior, suchas nucleoplasmic lamin A/C.

In conclusion, A-type lamins not only ensure structural integrityof (metazoan) nuclei, but also maintain the balance between dif-ferentiation and proliferation. In part, these activities may rely onlamin A/C-binding proteins, such as LAP2�. The potential effects oflaminopathy-linked lamin mutants on chromatin organization mayalter gene expression by destabilization of higher-order chromatinstructure and/or interference with the binding of A-type lamins toother proteins. These perturbations may particularly affect stemcell homeostasis.

Acknowledgements

We thank Thomas Dechat, MFPL, for valuable discussions andWolfgang J. Schneider for carefully reading the manuscript. Work inthe authors’ laboratory was supported by grants from the AustrianScience Fund (FWF P22043-B12 and P23805-B2) to RF. KG and SVare members of doctoral program funded by Austrian Science Fund(FWF, DK W1220).

References

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

[1] Burke B, Stewart CL. The nuclear lamins: flexibility in function. Nat Rev MolCell Biol 2013;14:13–24.

[2] Dahl KN, Ribeiro AJ, Lammerding J. Nuclear shape, mechanics, and mechan-otransduction. Circ Res 2008;102:1307–18.

Page 7: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ING Model

Y

evelo

ARTICLESCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & D

[3] Wilson KL, Berk JM. The nuclear envelope at a glance. J Cell Sci2010;123:1973–8.

[4] Andres V, Gonzalez JM. Role of A-type lamins in signaling, transcription, andchromatin organization. J Cell Biol 2009;187:945–57.

[5] Dauer WT, Worman HJ. The nuclear envelope as a signaling node in develop-ment and disease. Dev Cell 2009;17:626–38.

[6] Meister P, Mango SE, Gasser SM. Locking the genome: nuclear organizationand cell fate. Curr Opin Genet Dev 2011;21:167–74.

[7] Peric-Hupkes D, van Steensel B. Role of the nuclear lamina in genomeorganization and gene expression. Cold Spring Harb Symp Quant Biol2010;75:517–24.

[8] Simon DN, Wilson KL. The nucleoskeleton as a genome-associated dynamic‘network of networks’. Nat Rev Mol Cell Biol 2011;12:695–708.

[9] Young SG, Jung HJ, Coffinier C, Fong LG. Understanding the roles of nuclear A-and B-type lamins in brain development. J Biol Chem 2012;287:16103–10.

[10] Zuela N, Bar DZ, Gruenbaum Y. Lamins in development, tissue maintenanceand stress. EMBO Rep 2012;13:1070–8.

[11] Dechat T, Pfleghaar K, Sengupta K, Shimi T, Shumaker DK, Solimando L, et al.Nuclear lamins: major factors in the structural organization and function ofthe nucleus and chromatin. Genes Dev 2008;22:832–53.

[12] Dittmer TA, Misteli T. The lamin protein family. Genome Biol 2011;12:222.[13] Ho CY, Lammerding J. Lamins at a glance. J Cell Sci 2012;125:2087–93.[14] Korfali N, Wilkie GS, Swanson SK, Srsen V, de Las Heras J, Batrakou DG,

et al. The nuclear envelope proteome differs notably between tissues. Nucleus2012;3:552–64.

[15] Wilson KL, Foisner R. Lamin-binding proteins. Cold Spring Harb Perspect Biol2010;2:a000554.

[16] Butin-Israeli V, Adam SA, Goldman AE, Goldman RD. Nuclear lamin functionsand disease. Trends Genet 2012;28:464–71.

[17] Dechat T, Adam SA, Taimen P, Shimi T, Goldman RD. Nuclear lamins. ColdSpring Harb Perspect Biol 2010;2:a000547.

[18] Davies BS, Coffinier C, Yang SH, Barnes 2nd RH, Jung HJ, Young SG, et al.Investigating the purpose of prelamin A processing. Nucleus 2011;2:4–9.

[19] Rusinol AE, Sinensky MS. Farnesylated lamins, progeroid syndromes and far-nesyl transferase inhibitors. J Cell Sci 2006;119:3265–72.

[20] Sinensky M, Fantle K, Trujillo M, McLain T, Kupfer A, Dalton M. The processingpathway of prelamin A. J Cell Sci 1994;107(Pt 1):61–7.

[21] Dechat T, Gesson K, Foisner R. Lamina-independent lamins in the nuclearinterior serve important functions. Cold Spring Harb Symp Quant Biol2010;75:533–43.

[22] Dechat T, Korbei B, Vaughan OA, Vlcek S, Hutchison CJ, Foisner R. Lamina-associated polypeptide 2alpha binds intranuclear A-type lamins. J Cell Sci2000;113(Pt 19):3473–84.

[23] Moir RD, Yoon M, Khuon S, Goldman RD. Nuclear lamins A and B1: differentpathways of assembly during nuclear envelope formation in living cells. J CellBiol 2000;151:1155–68.

[24] Naetar N, Korbei B, Kozlov S, Kerenyi MA, Dorner D, Kral R, et al.Loss of nucleoplasmic LAP2alpha–lamin A complexes causes erythroidand epidermal progenitor hyperproliferation. Nat Cell Biol 2008;10:1341–8.

[25] Berger R, Theodor L, Shoham J, Gokkel E, Brok-Simoni F, Avraham KB, et al.The characterization and localization of the mouse thymopoietin/lamina-associated polypeptide 2 gene and its alternatively spliced products. GenomeRes 1996;6:361–70.

[26] Dechat T, Gotzmann J, Stockinger A, Harris CA, Talle MA, SiekierkaJJ, et al. Detergent-salt resistance of LAP2alpha in interphase nucleiand phosphorylation-dependent association with chromosomes early innuclear assembly implies functions in nuclear structure dynamics. EMBO J1998;17:4887–902.

[27] Furukawa K, Pante N, Aebi U, Gerace L. Cloning of a cDNA for lamina-associated polypeptide 2 (LAP2) and identification of regions that specifytargeting to the nuclear envelope. EMBO J 1995;14:1626–36.

[28] Harris CA, Andryuk PJ, Cline S, Chan HK, Natarajan A, Siekierka JJ, et al. Threedistinct human thymopoietins are derived from alternatively spliced mRNAs.Proc Natl Acad Sci USA 1994;91:6283–7.

[29] Dechat T, Vlcek S, Foisner R. Review: lamina-associated polypeptide 2isoforms and related proteins in cell cycle-dependent nuclear structuredynamics. J Struct Biol 2000;129:335–45.

[30] Brachner A, Foisner R. Evolvement of LEM proteins as chromatin tethers atthe nuclear periphery. Biochem Soc Trans 2011;39:1735–41.

[31] Cai M, Huang Y, Ghirlando R, Wilson KL, Craigie R, Clore GM. Solution struc-ture of the constant region of nuclear envelope protein LAP2 reveals twoLEM-domain structures: one binds BAF and the other binds DNA. EMBO J2001;20:4399–407.

[32] Dechat T, Gajewski A, Korbei B, Gerlich D, Daigle N, Haraguchi T, et al.LAP2alpha and BAF transiently localize to telomeres and specific regions onchromatin during nuclear assembly. J Cell Sci 2004;117:6117–28.

[33] Foisner R, Gerace L. Integral membrane proteins of the nuclear envelopeinteract with lamins and chromosomes, and binding is modulated by mitoticphosphorylation. Cell 1993;73:1267–79.

[34] Gajewski A, Csaszar E, Foisner R. A phosphorylation cluster in the chromatin-

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

binding region regulates chromosome association of LAP2alpha. J Biol Chem2004;279:35813–21.

[35] Vlcek S, Just H, Dechat T, Foisner R. Functional diversity of LAP2alpha andLAP2beta in postmitotic chromosome association is caused by an alpha-specific nuclear targeting domain. EMBO J 1999;18:6370–84.

PRESSpmental Biology xxx (2014) xxx– xxx 7

[36] Dorner D, Vlcek S, Foeger N, Gajewski A, Makolm C, Gotzmann J,et al. Lamina-associated polypeptide 2alpha regulates cell cycle progres-sion and differentiation via the retinoblastoma-E2F pathway. J Cell Biol2006;173:83–93.

[37] Markiewicz E, Dechat T, Foisner R, Quinlan RA, Hutchison CJ. Lamin A/C bind-ing protein LAP2alpha is required for nuclear anchorage of retinoblastomaprotein. Mol Biol Cell 2002;13:4401–13.

[38] Roux KJ, Kim DI, Raida M, Burke B. A promiscuous biotin ligase fusion proteinidentifies proximal and interacting proteins in mammalian cells. J Cell Biol2012;196:801–10.

[39] Taylor MR, Slavov D, Gajewski A, Vlcek S, Ku L, Fain PR, et al. Thymopoi-etin (lamina-associated polypeptide 2) gene mutation associated with dilatedcardiomyopathy. Hum Mutat 2005;26:566–74.

[40] Naetar N, Foisner R. Lamin complexes in the nuclear interior control progen-itor cell proliferation and tissue homeostasis. Cell Cycle 2009;8:1488–93.

[41] Markiewicz E, Ledran M, Hutchison CJ. Remodelling of the nuclear lamina andnucleoskeleton is required for skeletal muscle differentiation in vitro. J CellSci 2005;118:409–20.

[42] Bonne G, Di Barletta MR, Varnous S, Becane HM, Hammouda EH, Merlini L,et al. Mutations in the gene encoding lamin A/C cause autosomal dominantEmery–Dreifuss muscular dystrophy. Nat Genet 1999;21:285–8.

[43] Capell BC, Erdos MR, Madigan JP, Fiordalisi JJ, Varga R, Conneely KN, et al.Inhibiting farnesylation of progerin prevents the characteristic nuclear bleb-bing of Hutchinson–Gilford progeria syndrome. Proc Natl Acad Sci USA2005;102:12879–84.

[44] Mendez-Lopez I, Worman HJ. Inner nuclear membrane proteins: impact onhuman disease. Chromosoma 2012;121:153–67.

[45] Schreiber KH, Kennedy BK. When lamins go bad: nuclear structure and dis-ease. Cell 2013;152:1365–75.

[46] Worman HJ. Nuclear lamins and laminopathies. J Pathol 2012;226:316–25.[47] Worman HJ, Bonne G. “Laminopathies”: a wide spectrum of human diseases.

Exp Cell Res 2007;313:2121–33.[48] Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, et al. Recur-

rent de novo point mutations in lamin A cause Hutchinson–Gilford progeriasyndrome. Nature 2003;423:293–8.

[49] Gotzmann J, Foisner R. A-type lamin complexes and regenerative potential:a step towards understanding laminopathic diseases? Histochem Cell Biol2006;125:33–41.

[50] Mattout A, Dechat T, Adam SA, Goldman RD, Gruenbaum Y. Nuclear lamins,diseases and aging. Curr Opin Cell Biol 2006;18:335–41.

[51] Zwerger M, Jaalouk DE, Lombardi ML, Isermann P, Mauermann M, Dialynas G,et al. Myopathic lamin mutations impair nuclear stability in cells and tissueand disrupt nucleo-cytoskeletal coupling. Hum Mol Genet 2013;22:2335–49.

[52] Favreau C, Dubosclard E, Ostlund C, Vigouroux C, Capeau J, Wehnert M, et al.Expression of lamin A mutated in the carboxyl-terminal tail generates anaberrant nuclear phenotype similar to that observed in cells from patientswith Dunnigan-type partial lipodystrophy and Emery–Dreifuss muscular dys-trophy. Exp Cell Res 2003;282:14–23.

[53] Goldman RD, Shumaker DK, Erdos MR, Eriksson M, Goldman AE, Gordon LB,et al. Accumulation of mutant lamin A causes progressive changes in nucleararchitecture in Hutchinson–Gilford progeria syndrome. Proc Natl Acad SciUSA 2004;101:8963–8.

[54] Lammerding J, Fong LG, Ji JY, Reue K, Stewart CL, Young SG, et al. LaminsA and C but not lamin B1 regulate nuclear mechanics. J Biol Chem2006;281:25768–80.

[55] Arimura T, Helbling-Leclerc A, Massart C, Varnous S, Niel F, Lacene E, et al.Mouse model carrying H222P-Lmna mutation develops muscular dystrophyand dilated cardiomyopathy similar to human striated muscle laminopathies.Hum Mol Genet 2005;14:155–69.

[56] Fidzianska A, Hausmanowa-Petrusewicz I. Architectural abnormalities inmuscle nuclei. Ultrastructural differences between X-linked and autosomaldominant forms of EDMD. J Neurol Sci 2003;210:47–51.

[57] Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PC, Pinter J, et al. Nuclearlamin-A scales with tissue stiffness and enhances matrix-directed differenti-ation. Science 2013;341:1240104.

[58] De Vos WH, Houben F, Kamps M, Malhas A, Verheyen F, Cox J, et al. Repet-itive disruptions of the nuclear envelope invoke temporary loss of cellularcompartmentalization in laminopathies. Hum Mol Genet 2011;20:4175–86.

[59] Ho CY, Jaalouk DE, Vartiainen MK, Lammerding J. Lamin A/C andemerin regulate MKL1-SRF activity by modulating actin dynamics. Nature2013;497:507–11.

[60] Lammerding J, Schulze PC, Takahashi T, Kozlov S, Sullivan T, Kamm RD, et al.Lamin A/C deficiency causes defective nuclear mechanics and mechanotrans-duction. J Clin Invest 2004;113:370–8.

[61] Verstraeten VL, Ji JY, Cummings KS, Lee RT, Lammerding J. Increasedmechanosensitivity and nuclear stiffness in Hutchinson–Gilford progeriacells: effects of farnesyltransferase inhibitors. Aging Cell 2008;7:383–93.

[62] Broers JL, Peeters EA, Kuijpers HJ, Endert J, Bouten CV, Oomens CW, et al.Decreased mechanical stiffness in LMNA−/− cells is caused by defec-tive nucleo-cytoskeletal integrity: implications for the development oflaminopathies. Hum Mol Genet 2004;13:2567–80.

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

[63] Dechat T, Adam SA, Goldman RD. Nuclear lamins and chromatin: when struc-ture meets function. Adv Enzyme Regul 2009;49:157–66.

[64] Nikolova V, Leimena C, McMahon AC, Tan JC, Chandar S, Jogia D, et al. Defectsin nuclear structure and function promote dilated cardiomyopathy in laminA/C-deficient mice. J Clin Invest 2004;113:357–69.

Page 8: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ING Model

Y

8 evelo

ARTICLESCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & D

[65] Shumaker DK, Dechat T, Kohlmaier A, Adam SA, Bozovsky MR, Erdos MR, et al.Mutant nuclear lamin A leads to progressive alterations of epigenetic controlin premature aging. Proc Natl Acad Sci USA 2006;103:8703–8.

[66] Muchir A, Pavlidis P, Decostre V, Herron AJ, Arimura T, Bonne G, et al.Activation of MAPK pathways links LMNA mutations to cardiomyopathy inEmery–Dreifuss muscular dystrophy. J Clin Invest 2007;117:1282–93.

[67] Espada J, Varela I, Flores I, Ugalde AP, Cadinanos J, Pendas AM, et al. Nuclearenvelope defects cause stem cell dysfunction in premature-aging mice. J CellBiol 2008;181:27–35.

[68] Hernandez L, Roux KJ, Wong ESM, Mounkes LC, Mutalif R, Navasankari R, et al.Functional coupling between the extracellular matrix and nuclear lamina byWnt signaling in progeria. Dev Cell 2010;19:413–25.

[69] Marji J, O’Donoghue SI, McClintock D, Satagopam VP, Schneider R, RatnerD, et al. Defective lamin A-Rb signaling in Hutchinson–Gilford proge-ria syndrome and reversal by farnesyltransferase inhibition. PLoS ONE2010;5:e11132.

[70] Osorio FG, Lopez-Otin C, Freije JM. NF-kB in premature aging. Aging (Albany,NY) 2012;4:726–7.

[71] Cupesi M, Yoshioka J, Gannon J, Kudinova A, Stewart CL, Lammerding J.Attenuated hypertrophic response to pressure overload in a lamin A/C hap-loinsufficiency mouse. J Mol Cell Cardiol 2010;48:1290–7.

[72] Johnson BR, Nitta RT, Frock RL, Mounkes L, Barbie DA, Stewart CL, et al. A-typelamins regulate retinoblastoma protein function by promoting subnuclearlocalization and preventing proteasomal degradation. Proc Natl Acad Sci USA2004;101:9677–82.

[73] Mariappan I, Parnaik VK. Sequestration of pRb by cyclin D3 causes intranu-clear reorganization of lamin A/C during muscle cell differentiation. Mol BiolCell 2005;16:1948–60.

[74] Bridger JM, Kill IR. Aging of Hutchinson–Gilford progeria syndrome fibroblastsis characterised by hyperproliferation and increased apoptosis. Exp Gerontol2004;39:717–24.

[75] Mounkes LC, Kozlov S, Hernandez L, Sullivan T, Stewart CL. A progeroidsyndrome in mice is caused by defects in A-type lamins. Nature2003;423:298–301.

[76] Scaffidi P, Misteli T. Lamin A-dependent misregulation of adult stem cellsassociated with accelerated ageing. Nat Cell Biol 2008;10:452–9.

[77] Bakay M, Wang ZY, Melcon G, Schiltz L, Xuan JH, Zhao P, et al. Nuclear envelopedystrophies show a transcriptional fingerprint suggesting disruption of Rb-MyoD pathways in muscle regeneration. Brain 2006;129:996–1013.

[78] Frock RL, Kudlow BA, Evans AM, Jameson SA, Hauschka SD, Kennedy BK, et al.Lamin A/C and emerin are critical for skeletal muscle satellite cell differenti-ation. Genes Dev 2006;20:486–500.

[79] Boguslavsky RL, Stewart CL, Worman HJ. Nuclear lamin A inhibits adipocytedifferentiation: implications for Dunnigan-type familial partial lipodystro-phy. Hum Mol Genet 2006;15:653–63.

[80] Akter R, Rivas D, Geneau G, Drissi H, Duque G. Effect of lamin A/C knock-down on osteoblast differentiation and function. J Bone Miner Res 2009;24:283–93.

[81] Rauner M, Sipos W, Goettsch C, Wutzl A, Foisner R, Pietschmann P, et al.Inhibition of lamin A/C attenuates osteoblast differentiation and enhancesRANKL-dependent osteoclastogenesis. J Bone Miner Res 2009;24:78–86.

[82] Heessen S, Fornerod M. The inner nuclear envelope as a transcription factorresting place. EMBO Rep 2007;8:914–9.

[83] Kind J, van Steensel B. Genome–nuclear lamina interactions and gene regu-lation. Curr Opin Cell Biol 2010;22:320–5.

[84] Mancini MA, Shan B, Nickerson JA, Penman S, Lee WH. The retinoblastomagene product is a cell cycle-dependent, nuclear matrix-associated protein.Proc Natl Acad Sci USA 1994;91:418–22.

[85] Ozaki T, Saijo M, Murakami K, Enomoto H, Taya Y, Sakiyama S. Complexformation between lamin A and the retinoblastoma gene product: identi-fication of the domain on lamin A required for its interaction. Oncogene1994;9:2649–53.

[86] Weinberg RA. The retinoblastoma protein and cell cycle control. Cell1995;81:323–30.

[87] Van Berlo JH, Voncken JW, Kubben N, Broers JL, Duisters R, van Leeuwen RE,et al. A-type lamins are essential for TGF-beta1 induced PP2A to dephospho-rylate transcription factors. Hum Mol Genet 2005;14:2839–49.

[88] Rodriguez J, Calvo F, Gonzalez JM, Casar B, Andres V, Crespo P. ERK1/2 MAPkinases promote cell cycle entry by rapid, kinase-independent disruption ofretinoblastoma–lamin A complexes. J Cell Biol 2010;191:967–79.

[89] Gotic I, Schmidt WM, Biadasiewicz K, Leschnik M, Spilka R, Braun J, et al.Loss of LAP2 alpha delays satellite cell differentiation and affects postnatalfiber-type determination. Stem Cells 2010;28:480–8.

[90] Gotic I, Foisner R. Multiple novel functions of lamina associated polypeptide2alpha in striated muscle. Nucleus 2010;1:397–401.

[91] Jahn D, Schramm S, Schnolzer M, Heilmann CJ, de Koster CG, Schutz W, et al.A truncated lamin A in the Lmna −/− mouse line: implications for the under-standing of laminopathies. Nucleus 2012;3:463–74.

[92] Cohen TV, Gnocchi VF, Cohen JE, Phadke A, Liu H, Ellis JA, et al. Defective skele-tal muscle growth in lamin A/C-deficient mice is rescued by loss of Lap2alpha.Hum Mol Genet 2013;22:2852–69.

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

[93] Constantinescu D, Gray HL, Sammak PJ, Schatten GP, Csoka AB. Lamin A/Cexpression is a marker of mouse and human embryonic stem cell differenti-ation. Stem Cells 2006;24:177–85.

[94] Eckersley-Maslin MA, Bergmann JH, Lazar Z, Spector DL. Lamin A/C isexpressed in pluripotent mouse embryonic stem cells. Nucleus 2013;4:53–60.

PRESSpmental Biology xxx (2014) xxx– xxx

[95] Zuo B, Yang J, Wang F, Wang L, Yin Y, Dan J, et al. Influences of lamin A levelson induction of pluripotent stem cells. Biol Open 2012;1:1118–27.

[96] Wagers AJ, Weissman IL. Plasticity of adult stem cells. Cell 2004;116:639–48.[97] Pekovic V, Hutchison CJ. Adult stem cell maintenance and tissue regeneration

in the ageing context: the role for A-type lamins as intrinsic modulators ofageing in adult stem cells and their niches. J Anat 2008;213:5–25.

[98] Mateos J, De la Fuente A, Lesende-Rodriguez I, Fernandez-Pernas P, ArufeMC, Blanco FJ. Lamin A deregulation in human mesenchymal stem cells pro-motes an impairment in their chondrogenic potential and imbalance in theirresponse to oxidative stress. Stem Cell Res 2013;11:1137–48.

[99] Rosengardten Y, McKenna T, Grochova D, Eriksson M. Stem cell depletion inHutchinson–Gilford progeria syndrome. Aging Cell 2011;10:1011–20.

[100] Lavasani M, Lu A, Thompson SD, Robbins PD, Huard J, Niedernhofer LJ.Isolation of muscle-derived stem/progenitor cells based on adhesion char-acteristics to collagen-coated surfaces. Methods Mol Biol 2013;976:53–65.

[101] Meshorer E, Gruenbaum Y. Rejuvenating premature aging. Nat Med2008;14:713–5.

[102] Halaschek-Wiener J, Brooks-Wilson A. Progeria of stem cells: stem cellexhaustion in Hutchinson–Gilford progeria syndrome. J Gerontol A Biol SciMed Sci 2007;62:3–8.

[103] Prokocimer M, Davidovich M, Nissim-Rafinia M, Wiesel-Motiuk N, Bar DZ,Barkan R, et al. Nuclear lamins: key regulators of nuclear structure and activ-ities. J Cell Mol Med 2009;13:1059–85.

[104] Gilchrist S, Gilbert N, Perry P, Ostlund C, Worman HJ, Bickmore WA. Alteredprotein dynamics of disease-associated lamin A mutants. BMC Cell Biol2004;5:46.

[105] Markiewicz E, Venables R, Mauricio Alvarez R, Quinlan R, Dorobek M,Hausmanowa-Petrucewicz I, et al. Increased solubility of lamins and redistri-bution of lamin C in X-linked Emery–Dreifuss muscular dystrophy fibroblasts.J Struct Biol 2002;140:241–53.

[106] Muchir A, Medioni J, Laluc M, Massart C, Arimura T, van der Kooi AJ, et al.Nuclear envelope alterations in fibroblasts from patients with muscular dys-trophy, cardiomyopathy, and partial lipodystrophy carrying lamin A/C genemutations. Muscle Nerve 2004;30:444–50.

[107] Wiesel N, Mattout A, Melcer S, Melamed-Book N, Herrmann H, Medalia O,et al. Laminopathic mutations interfere with the assembly, localization, anddynamics of nuclear lamins. Proc Natl Acad Sci USA 2008;105:180–5.

[108] Dechat T, Shimi T, Adam SA, Rusinol AE, Andres DA, Spielmann HP, et al.Alterations in mitosis and cell cycle progression caused by a mutant lamin Aknown to accelerate human aging. Proc Natl Acad Sci USA 2007;104:4955–60.

[109] Favreau C, Higuet D, Courvalin JC, Buendia B. Expression of a mutant lamin Athat causes Emery–Dreifuss muscular dystrophy inhibits in vitro differentia-tion of C2C12 myoblasts. Mol Cell Biol 2004;24:1481–92.

[110] Kandert S, Wehnert M, Muller CR, Buendia B, Dabauvalle MC. Impairednuclear functions lead to increased senescence and inefficient differentiationin human myoblasts with a dominant p.R545C mutation in the LMNA gene.Eur J Cell Biol 2009;88:593–608.

[111] Varela I, Cadinanos J, Pendas AM, Gutierrez-Fernandez A, Folgueras AR,Sanchez LM, et al. Accelerated ageing in mice deficient in Zmpste24 proteaseis linked to p53 signalling activation. Nature 2005;437:564–8.

[112] Meaburn KJ, Cabuy E, Bonne G, Levy N, Morris GE, Novelli G, et al. Primarylaminopathy fibroblasts display altered genome organization and apoptosis.Aging Cell 2007;6:139–53.

[113] De Falco G, Comes F, Simone C. pRb: master of differentiation. Coupling irre-versible cell cycle withdrawal with induction of muscle-specific transcription.Oncogene 2006;25:5244–9.

[114] Cremer T, Cremer M. Chromosome territories. Cold Spring Harb Perspect Biol2010;2:a003889.

[115] Misteli T. Beyond the sequence: cellular organization of genome function. Cell2007;128:787–800.

[116] Clowney EJ, LeGros MA, Mosley CP, Clowney FG, Markenskoff-PapadimitriouEC, Myllys M, et al. Nuclear aggregation of olfactory receptor genes governstheir monogenic expression. Cell 2012;151:724–37.

[117] Solovei I, Wang AS, Thanisch K, Schmidt CS, Krebs S, Zwerger M, et al. LBRand lamin A/C sequentially tether peripheral heterochromatin and inverselyregulate differentiation. Cell 2013;152:584–98.

[118] Greil F, Moorman C, van Steensel B. DamID: mapping of in vivoprotein–genome interactions using tethered DNA adenine methyltransferase.Methods Enzymol 2006;410:342–59.

[119] Vogel MJ, Peric-Hupkes D, van Steensel B. Detection of in vivo protein–DNAinteractions using DamID in mammalian cells. Nat Protoc 2007;2:1467–78.

[120] Guelen L, Pagie L, Brasset E, Meuleman W, Faza MB, Talhout W, et al. Domainorganization of human chromosomes revealed by mapping of nuclear laminainteractions. Nature 2008;453:948–51.

[121] Finlan LE, Sproul D, Thomson I, Boyle S, Kerr E, Perry P, et al. Recruitmentto the nuclear periphery can alter expression of genes in human cells. PLoSGenet 2008;4:e1000039.

[122] Kumaran RI, Spector DL. A genetic locus targeted to the nuclear periph-ery in living cells maintains its transcriptional competence. J Cell Biol2008;180:51–65.

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

[123] Reddy KL, Zullo JM, Bertolino E, Singh H. Transcriptional repression mediatedby repositioning of genes to the nuclear lamina. Nature 2008;452:243–7.

[124] Meister P, Towbin BD, Pike BL, Ponti A, Gasser SM. The spatial dynam-ics of tissue-specific promoters during C. elegans development. Genes Dev2010;24:766–82.

Page 9: Lamina-associated polypeptide (LAP)2α and nucleoplasmic lamins in adult stem cell regulation and disease

ING Model

Y

evelo

[

[

[

[

[

[

ARTICLESCDB-1485; No. of Pages 9

K. Gesson et al. / Seminars in Cell & D

125] Peric-Hupkes D, Meuleman W, Pagie L, Bruggeman SW, Solovei I, BrugmanW, et al. Molecular maps of the reorganization of genome–nuclear laminainteractions during differentiation. Mol Cell 2010;38:603–13.

126] Kind J, Pagie L, Ortabozkoyun H, Boyle S, de Vries SS, Janssen H, et al. Single-celldynamics of genome–nuclear lamina interactions. Cell 2013;153:178–92.

127] Towbin BD, Gonzalez-Aguilera C, Sack R, Gaidatzis D, Kalck V, Meister P,et al. Step-wise methylation of histone H3K9 positions heterochromatin atthe nuclear periphery. Cell 2012;150:934–47.

128] Meuleman W, Peric-Hupkes D, Kind J, Beaudry JB, Pagie L, Kellis M, et al.Constitutive nuclear lamina–genome interactions are highly conserved andassociated with A/T-rich sequence. Genome Res 2013;23:270–80.

129] Zullo JM, Demarco IA, Pique-Regi R, Gaffney DJ, Epstein CB, Spooner CJ, et al.

Please cite this article in press as: Gesson K, et al. Lamina-associated polypeand disease. Semin Cell Dev Biol (2014), http://dx.doi.org/10.1016/j.semcd

DNA sequence-dependent compartmentalization and silencing of chromatinat the nuclear lamina. Cell 2012;149:1474–87.

130] Shimi T, Pfleghaar K, Kojima S, Pack CG, Solovei I, Goldman AE, et al. The A-and B-type nuclear lamin networks: microdomains involved in chromatinorganization and transcription. Genes Dev 2008;22:3409–21.

PRESSpmental Biology xxx (2014) xxx– xxx 9

[131] Kubben N, Adriaens M, Meuleman W, Voncken JW, van Steensel B, Misteli T.Mapping of lamin A- and progerin-interacting genome regions. Chromosoma2012;121:447–64.

[132] Bruston F, Delbarre E, Ostlund C, Worman HJ, Buendia B, Duband-Goulet I.Loss of a DNA binding site within the tail of prelamin A contributes to alteredheterochromatin anchorage by progerin. FEBS Lett 2010;584:2999–3004.

[133] McCord RP, Nazario-Toole A, Zhang H, Chines PS, Zhan Y, Erdos MR,et al. Correlated alterations in genome organization, histone methylation,and DNA-lamin A/C interactions in Hutchinson–Gilford progeria syndrome.Genome Res 2013;23:260–9.

[134] Lund E, Oldenburg AR, Delbarre E, Freberg CT, Duband-Goulet I, Eskeland R,et al. Lamin A/C–promoter interactions specify chromatin state-dependent

ptide (LAP)2� and nucleoplasmic lamins in adult stem cell regulationb.2013.12.009

transcription outcomes. Genome Res 2013.[135] Zhang S, Schones DE, Malicet C, Rochman M, Zhou M, Foisner R, et al.

High mobility group protein N5 (HMGN5) and lamina-associated polypep-tide 2alpha (LAP2alpha) interact and reciprocally affect their genome-widechromatin organization. J Biol Chem 2013;288:18104–9.