Diseases of the Nuclear Envelope

19
Diseases of the Nuclear Envelope Howard J. Worman, Cecilia O ¨ stlund, and Yuexia Wang Department of Medicine and Department of Pathologyand Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032 Correspondence: [email protected] In the past decade, a wide range of fascinating monogenic diseases have been linked to mutations in the LMNA gene, which encodes the A-type nuclear lamins, intermediate fila- ment proteins of the nuclear envelope. These diseases include dilated cardiomyopathy with variable muscular dystrophy, Dunnigan-type familial partial lipodystrophy, a Charcot- Marie-Tooth type 2 disease, mandibuloacral dysplasia, and Hutchinson-Gilford progeria syndrome. Several diseases are also caused by mutations in genes encoding B-type lamins and proteins that associate with the nuclear lamina. Studies of these so-called laminopathies or nuclear envelopathies, some of which phenocopy common human disorders, are provid- ing clues about functions of the nuclear envelope and insights into disease pathogenesis and human aging. M utations in LMNA encoding the A-type lamins cause a group of human disorders often collectively called laminopathies. The ma- jor A-type lamins, lamin A and lamin C, arise by alternative splicing of the LMNA pre-mRNA and are expressed in virtually all differentiated somatic cells. Although the A-type lamins are widely expressed, LMNA mutations are respon- sible for at least a dozen different clinically defined disorders with tissue-selective abnor- malities. Mutations in genes encoding B-type lamins and lamin-associated proteins, most of which are similarly expressed in almost all so- matic cells, also cause tissue-selective diseases. Research on the laminopathies has provided novel clues about nuclear envelope function. Recent studies have begun to shed light on how alterations in the nuclear envelope could explain disease pathogenesis. Along with basic research on nuclear structure, the nuclear lam- ins, and lamina-associated proteins, clinical re- search on the laminopathies will contribute to a complete understanding of the functions of the nuclear envelope in normal physiology and in human pathology. LMNA: ONE GENE, MANY DISEASES George Beadle and Edward Tatum (Beadle and Tatum 1941) proposed what became known as the “one gene-one enzyme” hypothesis and was later modified to the “one gene-one- polypeptide” hypothesis. The premise under- lying this hypothesis was that genes act through the production of polypeptides, with each gene producing a single polypeptide functioning in Editors: Tom Misteli and David Spector Additional Perspectives on The Nucleus available at www.cshperspectives.org Copyright # 2010 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a000760 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 1 on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/ Downloaded from

Transcript of Diseases of the Nuclear Envelope

Page 1: Diseases of the Nuclear Envelope

Diseases of the Nuclear Envelope

Howard J. Worman, Cecilia Ostlund, and Yuexia Wang

Department of Medicine and Department of Pathology and Cell Biology, College of Physiciansand Surgeons, Columbia University, New York, New York 10032

Correspondence: [email protected]

In the past decade, a wide range of fascinating monogenic diseases have been linked tomutations in the LMNA gene, which encodes the A-type nuclear lamins, intermediate fila-ment proteins of the nuclear envelope. These diseases include dilated cardiomyopathywith variable muscular dystrophy, Dunnigan-type familial partial lipodystrophy, a Charcot-Marie-Tooth type 2 disease, mandibuloacral dysplasia, and Hutchinson-Gilford progeriasyndrome. Several diseases are also caused by mutations in genes encoding B-type laminsand proteins that associate with the nuclear lamina. Studies of these so-called laminopathiesor nuclear envelopathies, some of which phenocopy common human disorders, are provid-ing clues about functions of the nuclear envelope and insights into disease pathogenesis andhuman aging.

Mutations in LMNA encoding the A-typelamins cause a group of human disorders

often collectively called laminopathies. The ma-jor A-type lamins, lamin A and lamin C, arise byalternative splicing of the LMNA pre-mRNAand are expressed in virtually all differentiatedsomatic cells. Although the A-type lamins arewidely expressed, LMNA mutations are respon-sible for at least a dozen different clinicallydefined disorders with tissue-selective abnor-malities. Mutations in genes encoding B-typelamins and lamin-associated proteins, most ofwhich are similarly expressed in almost all so-matic cells, also cause tissue-selective diseases.

Research on the laminopathies has providednovel clues about nuclear envelope function.Recent studies have begun to shed light onhow alterations in the nuclear envelope could

explain disease pathogenesis. Along with basicresearch on nuclear structure, the nuclear lam-ins, and lamina-associated proteins, clinical re-search on the laminopathies will contribute to acomplete understanding of the functions of thenuclear envelope in normal physiology and inhuman pathology.

LMNA: ONE GENE, MANY DISEASES

George Beadle and Edward Tatum (Beadle andTatum 1941) proposed what became knownas the “one gene-one enzyme” hypothesis andwas later modified to the “one gene-one-polypeptide” hypothesis. The premise under-lying this hypothesis was that genes act throughthe production of polypeptides, with each geneproducing a single polypeptide functioning in

Editors: Tom Misteli and David Spector

Additional Perspectives on The Nucleus available at www.cshperspectives.org

Copyright # 2010 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a000760

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

1

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 2: Diseases of the Nuclear Envelope

a particular step in a metabolic pathway or othercellular process. A corollary of this hypothesis,which formed the foundation of most earlystudies using positional cloning to identifydisease genes, was the “one gene-one disease”principle. We now know that this is not correctand perhaps the best example that disproves thisprinciple is LMNA.

LMNA encoding the A-type lamins wascharacterized in 1993 and subsequently map-ped to chromosome 1q21.2-q21.3 (Lin andWorman 1993; Wydner et al. 1996). The firsthuman disease identified by positional cloningto be caused by LMNA mutations was auto-somal dominant Emery-Dreifuss musculardystrophy (Bonne et al. 1999). Rare compoundheterozygous mutations in LMNA causingrecessively inherited Emery-Dreifuss musculardystrophy were described shortly thereafter(Raffaele di Barletta et al. 2000). Patients withEmery-Dreifuss muscular dystrophy classicallyhave early contractures of the elbows, Achillestendons, and posterior neck, rigidity of thespine, slowly progressive muscle weakness inthe upper arms and lower legs, and dilatedcardiomyopathy with an early onset atrio-ventricular conduction block (Emery 2000;Muchir and Worman 2007).

Soon after LMNA mutations were shown tocause Emery-Dreifuss muscular dystrophy,mutations in this gene were shown to causeother dominantly inherited diseases affectingprimarily striated muscle, including dilated car-diomyopathy 1A (Fatkin et al. 1999) and limbgirdle muscular dystrophy type 1B (Muchiret al. 2000). Like Emery-Dreifuss musculardystrophy, these conditions have a predominantdilated cardiomyopathy with early onset atrio-ventricular conduction block. In dilated cardio-myopathy 1A, skeletal muscle is minimallyaffected or unaffected. In limb-girdle musculardystrophy, the distribution of skeletal muscleinvolvement is primarily around the shouldersand hips with sparing of the distal extremities.Most subjects with limb-girdle musculardystrophy do not have joint contractures char-acteristic of classical Emery-Dreifuss musculardystrophy. It was originally proposed thatEmery-Dreifuss muscular dystrophy, dilated

cardiomyopathy 1A, and limb-girdle musculardystrophy type 1B resulted from different muta-tions in LMNA. However, the phenotypic vari-ability is most likely because of the influenceof modifier genes or environmental factors.This is suggested by intrafamilial variabilityand phenotypic overlap in patients withLMNA mutations and muscle disease (Bonneet al. 2000). Within a single family, one affectedindividual can be diagnosed with isolated cardi-omyopathy, another with Emery-Dreifuss mus-cular dystrophy, and others with limb-girdlemuscular dystrophy (Brodsky et al. 2000). Basedon the combined phenotypic and genetic data,dilated cardiomyopathy with variable skeletalmuscle involvement, a phrase used by Brodskyet al. (2000), is a very appropriate descriptorof the striated muscle disease caused byLMNA mutations.

Although most LMNA mutations causingmuscle disorders present during childhoodor early adulthood, rare subjects present withcongenital muscular dystrophy (Quijano-Royet al. 2008). Congenital muscular dystrophyhas an earlier onset and more severe phenotypethan the later-onset muscle disorders causedby LMNA mutations. Most cases of LMNA-associated congenital muscular dystrophy arecaused by de novo mutations but cases ofgerminal mosaicism have also been identified(Makri et al. 2009). Although some of theLMNA mutations causing congenital musculardystrophy appear to be unique, others havebeen reported in patients with the later-onsetmyopathies. A unique LMNA splice site muta-tion has also been associated with a heart-handsyndrome, which is characterized by the asso-ciation of congenital cardiac disease and limbdeformities (Renou et al. 2008).

After LMNA mutations were shown to causestriated muscle diseases, a surprising discoverywas made regarding another monogenic diseaseaffecting different tissues. In 1998, the geneticlocus for Dunnigan-type familial partial lipo-dystrophy had been mapped using positionalcloning to chromosome 1q21-22 (Jacksonet al. 1998; Peters et al. 1998). Lipodystrophiesare a group of disorders characterized by theabsence or reduction of subcutaneous adipose

H.J. Worman, C. Ostlund, and Y. Wang

2 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 3: Diseases of the Nuclear Envelope

tissue. Patients with Dunnigan-type familialpartial lipodystrophy, a dominantly inheriteddisorder, are born with normal fat distributionbut after the onset of puberty there is regionalloss of fat from the extremities associated withinsulin resistance and frequently diabetes melli-tus (Dunnigan et al. 1974). Knowing that thegenetic locus for this disease was at chromo-some 1q21-22, Cao and Hegele (2000) hypothe-sized that the analogy between the regionalmuscle wasting in autosomal dominant Emery-Dreifuss muscular dystrophy and the regionaladipocyte degeneration in this disease madeLMNA a candidate gene. They identified a novelmissense mutation in exon 8 leading to a R482Qamino-acid substitution, which cosegregatedwith the lipodystrophy phenotype in five Cana-dian families. At around the same time, the twogroups that had mapped the disease to chromo-some 1q21-22 performed finer mapping andidentified the LMNA R482Q and other muta-tions in exon 8 leading to amino-acid substitu-tions (Shackleton et al. 2000; Speckman et al.2000). Missense mutations in exon 11 ofLMNA leading to R582H and R584H amino-acid substitutions in lamin A, but not laminC, were further identified in some atypical cases(Speckman et al. 2000; Vigouroux et al. 2000).Subsequently, there have been a few reports ofpatients with other LMNA mutations withatypical lipodystrophy syndromes, sometimesin combination with muscle abnormalities(Vigouroux and Capeau 2005).

By 2000, the positional cloners had clearlyshown that mutations in LMNA cause two quitedifferent diseases: dilated cardiomyopathy withvariable muscular dystrophy and partial lipo-dystrophy. However, the situation soon becamemore complicated when just a couple of yearslater De Sandre-Giovannoli et al. (2002) per-formed homozygosity mapping in inbredAlgerian families with an autosomal recessiveform of Charcot-Marie-Tooth disease type 2,linked it to chromosome 1q21.2-q21.3, andidentified a LMNA mutation leading to theR298C amino-acid substitution. Subjects withCharcot-Marie-Tooth disease type 2 diseases,including the subtype caused by LMNA muta-tion, have slight or absent reduction of nerve

conduction velocities, loss of large myelinatedfibers, and axonal degeneration (Chaouchet al. 2003). Affected individuals with theR298C mutation have variable severity andprogression of disease, suggesting that modifiergenes influence the phenotype of peripheralneuropathy caused by LMNA mutation (Taziret al. 2004). Later in 2002, Novelli et al. (2002)hypothesized that LMNA mutations mightcause mandibuloacral dysplasia, a rare auto-somal recessive disorder in which subjects havean undersized jaw, underdeveloped clavicles,other congenital bone abnormalities, and partiallipodystrophy. Theystudied five consanguineousItalian families and identified a homozygousLMNA missense mutation causing a R527Hamino-acid substitution that was shared by allaffected patients. Subsequent subjects havebeen described with homozygous LMNA muta-tions causing R527C or A529V amino-acidsubstitutions (Agarwal et al. 2008; Garg et al.2005). A compound heterozygous subject forthe LMNA R527H and a V440M mutationwith some features of mandibuloacral dysplasia,lack of muscle strength, and decreased muscletone has also been reported (Lombardi et al.2007).

Hutchinson-Gilford progeria syndrome,first described over a century ago, is a rare dis-ease with features of accelerated or prematureaging (Hutchinson 1886; Gilford 1904;McKusick 1952; DeBusk 1972). Individualswith this autosomal dominant sporadic syn-drome generally die in the second decade of lifefrom myocardial infarction or stroke (DeBusk1972; Merideth et al. 2008). Other prominentphenotypic features are sclerotic skin, joint con-tractures, prominent eyes, an undersized jaw,decreased subcutaneous fat, alopecia, skindimpling and mottling, prominent vasculaturein the skin, fingertip tufting, and growth impair-ment (Merideth et al. 2008). In 2003, FrancisCollins and colleagues localized the responsiblegene to chromosome 1q by observing two casesin which this chromosomal region was from thesame parent and one case with a six-megabasepaternal interstitial deletion (Eriksson et al.2003). They then showed that 18 out of 20classical cases of Hutchinson-Gilford progeria

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 3

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 4: Diseases of the Nuclear Envelope

had a de novo G608G (nucleotide 1824 C.T)mutation within exon 11 of LMNA and anothercase with a G608S (nucleotide 1822 G.A)mutation (Eriksson et al. 2003), a finding thatwas simultaneously reported by De Sandre-Giovannoli et al. (2003) and then confirmedby Cao and Hegele (2003). These mutations ac-tivate a cryptic splice donor site resulting in thesynthesis of a protein with 50 amino acidsdeleted near the carboxyl terminus of prelaminA. This truncated prelamin A variant is notappropriately processed to lamin A (see below).Other LMNA missense mutations not generat-ing abnormal RNA splicing within exon 11have also been reported in variant progeroidsyndromes (Chen et al. 2003; Csoka et al. 2004;Verstraeten et al. 2006). Mandibuloacral dyspla-sia caused by LMNA mutations, as discussedpreviously, also has progeroid features.

In summary, genetic studies since the late1990s have shown that mutations in LMNAcause about a dozen clinical disorders with dif-ferent names (Table 1). These can more broadly

be classified into diseases affecting predomi-nantly (1) striated muscle, (2) adipose tissue,(3) peripheral nerve, or (4) multiple tissuesresulting in progeroid phenotypes. Thesemostly tissue-selective disorders occur eventhough A-type lamins are intermediate filamentprotein components of the nuclear lamina invirtually all differentiated somatic cells.

OTHER LAMINOPATHIES/NUCLEARENVELOPATHIES

Monogenic diseases resulting from mutationsin genes encoding B-type lamins and proteinsthat are directly or indirectly associated withthe nuclear lamina are also sometimes referredto as laminopathies or nuclear envelopathies(Table 2). Emery-Dreifuss muscular dystrophywas shown to be inherited in an X-linked man-ner years before autosomal inheritance wasdescribed (Emery and Dreifuss 1966). In itsclassical presentation, the X-linked inheriteddisease phenocopies the autosomal form. In1994, Daniella Toniolo and colleagues reportedthat the gene responsible for X-linked Emery-Dreifuss muscular dystrophy encoded a mono-topic transmembrane protein expressed in virt-ually all cells that the authors named emerin(Bione et al. 1994). They extended their initialfindings to more patients (Bione et al. 1995).Soon after, emerin was shown to be a proteinof the inner nuclear membrane (Manilal et al.1996; Nagano et al. 1996). Emerin was furthershown to depend on A-type lamins for its local-ization to the nuclear envelope and to directlyinteract with lamins (Clements et al. 2000;Fairley et al. 1999; Sullivan et al. 1999). The clin-ical spectrum of disease resulting from muta-tions in EMD encoding emerin is actuallywider than the classical Emery-Dreifuss pheno-type and includes a limb-girdle muscular dys-trophy, cardiomyopathy with minimal muscleor joint involvement, and various intermittentforms (Astejada et al. 2007).

Lamin B receptor is an integral protein ofthe inner nuclear membrane that binds toB-type lamins (Worman et al. 1988). It has abasically charged, nucleoplasmic, amino-termi-nal domain that binds to lamins, DNA, and

Table 1. Mutations in LMNA cause several distinctclinical diseases predominantly affecting striatedmuscle, adipose, and peripheral nerve, or give aprogeria phenotype

Striated MuscleAutosomal dominant (and rarely recessive)Emery-Dreifuss muscular dystrophyCardiomyopathy dilated 1ALimb-girdle muscular dystrophy type 1BCongenital muscular dystrophy“Heart-hand” syndrome

Adipose TissueDunnigan-type familial partial lipodystrophyLipoatrophy with diabetes and other features of

insulin resistanceAtypical lipodystrophy syndromesMandibuloacral dysplasia�

Peripheral NerveCharcot-Marie-Tooth disease type 2B1

Progeria PhenotypeHutchinson-Gilford progeria syndromeAtypical Werner SyndromeVariant progeroid disordersMandibuloacral Dysplasia�

�Mandibuloacral dysplasia has features of

lipodystrophy and progeria

H.J. Worman, C. Ostlund, and Y. Wang

4 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 5: Diseases of the Nuclear Envelope

chromatin proteins followed by a stretch ofeight putative transmembrane segments thathas high sequence similarity to sterol reductases(Worman et al. 1990; Ye and Worman 1994,1996; Holmer et al. 1998). Heterozygous muta-tions in LBR that lead to reduced expressionof the protein cause Pelger-Huet anomaly, abenign autosomal dominant disorder charac-terized by abnormal nuclear shape and chro-matin organization in blood neutrophils(Hoffmann et al. 2002). In contrast, homozy-gous LBR mutations lead to severe develop-mental abnormalities or are lethal in utero(Hoffmann et al. 2002; Oosterwijk et al. 2003).In one case, a homozygous mutation leadingto production of a truncated protein lackingthe carboxyl-terminal 82 amino acids wasreported to cause hydrops-ectopic calcifica-tion-“moth-eaten” or Greenberg skeletal dys-plasia, a lethal disorder, which was associatedwith loss of detectable sterol D14-reductaseactivity (Waterham et al. 2003). It appears thatdepending on the amount of expression andthe affected functional domains of the protein,the phenotypes resulting from mutations inLBR can range from a benign alteration in neu-trophil nuclear morphology to death in utero.

MAN1 is an integral inner nuclear mem-brane protein with two transmembrane seg-ments and two nucleoplasmic domains (Linet al. 2000). The nucleoplasmic domain preced-ing the first transmembrane segment has been

reported to bind to A-type lamins, B-type lam-ins, and emerin (Mansharamani and Wilson,2005). The nucleoplasmic domain followingthe second transmembrane segment binds toregulatory-Smads and DNA (Hellemans et al.2004; Lin et al. 2005; Pan et al. 2005; Caputoet al. 2006). Heterozygous loss-of-function mu-tations in LEMD3 encoding MAN1 cause osteo-poikilosis, Buschke-Ollendorff syndrome, andnonsporadic melorheostosis, sclerosing bonedysplasias that sometimes have hyperprolifera-tive skin abnormalities. These phenotypes arelikely associated with enhanced transforminggrowth factor-b and bone morphogenic proteinsignaling, the effects of which are mediated byregulatory-Smads.

SYNE1 encodes nesprin-1, a protein withseveral isoforms that arise by alternative RNAsplicing. Depending on their size, nesprin-1 iso-form may localize to the inner or outer nuclearmembrane. Larger nesprin-1 isoforms localizeto the outer nuclear membrane and interactin the perinuclear space with Sun proteins, in-tegral proteins of the inner nuclear membranethat bind to lamins, forming a complex con-necting the nucleus to the cytoskeleton (Crispet al. 2006). Homozygous mutations in SYNE1have been shown to cause an autosomal reces-sive cerebellar ataxia specifically affectinga part of the brain (Gros-Louis et al. 2007). Inone large family, however, a SYNE1 mutationwas shown to cosegregate with autosomal

Table 2. Diseases caused by mutations in genes encoding B-type lamins or proteins associated with the nuclearlamina

Gene Protein Disease

EMD emerin cardiomyopathy with muscular dystrophyLBR lamin B receptor Pelger-Huet anomaly (heterozygous)

Greenberg skeletal dysplasia (homozygous)LEMD3 MAN1 Sclerosing bone dysplasiasSYNE1 nesprin-1 cerebellar ataxiaTMPO lamina-associated

polypeptide 2cardiomyopathy�

TOR1A torsinA DYT1 dystoniaLMNB1 lamin B1 adult-onset autosomal dominant leukodystrophyLMNB2 lamin B2 acquired partial lipodystrophyZMPSTE24 prelamin A endoprotease restrictive dermopathy and progeroid disorders�Reported in two affected individuals in a single family without gene sequencing from unaffected individuals in the

same family.

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 5

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 6: Diseases of the Nuclear Envelope

recessive arthrogryposis, a disease characterizedby bilateral clubfoot, decreased fetal move-ments, and delayed motor milestones with pro-gressive motor decline after the first decade(Attali et al. 2009).

Mutations in genes encoding other proteinsthat interact directly or indirectly with laminsalso cause tissue-selective human diseases.Polymorphisms in the gene encoding lamina-associated polypeptide 2 have been identifiedin two individuals with cardiomyopathy in asingle family; however, sequencing of the genein unaffected family members was not reported(Taylor et al. 2005). DYT1 dystonia is a centralnervous system movement disorder in whichsustained muscle contractions lead to twistingand repetitive movements or abnormal pos-tures; it is caused by an in-frame deletion inTOR1A encoding torsinA that leads to loss ofa glutamic acid residue (DE302/3) from theprotein (Ozelius et al. 1997). TorsinA is anAAAþ ATPase of the endoplasmic reticulumthat interacts with the luminal domain oflamina-associated polypeptide 1, an integral in-ner nuclear membrane protein that interactswith lamins (Goodchild and Dauer 2005). ThetorsinA DE302/3 variant concentrates in theperinuclear space relative to the bulk endo-plasmic reticulum (Gonzalez-Alegre and Paulson2004; Goodchild and Dauer 2004; Naismithet al. 2004), where it appears to selectivelydisrupt the structure of the nuclear envelopesof neurons (Goodchild et al. 2005).

Mutations in genes other than LMNA butdirectly affecting lamins also cause diseases.ZMPSTE24 is an endoprotease responsible forthe processing of prelamin A to lamin A (see later).Loss-of-function mutations in ZMPSTE24cause autosomal recessive restrictive dermop-athy, a neonatal lethal progeroid disorder(Navarro et al. 2005). Compound heterozygousmutations in ZMPSTE24 also cause progeroiddisorders with some cases being diagnosedas mandibuloacral dysplasia (Agarwal et al.2003; Shackleton et al. 2005). Duplication ofLMNB1 encoding lamin B1 leading to increasedexpression of the protein causes adult onsetautosomal dominant leukodystrophy, a slowlyprogressive neurological disorder characterized

by symmetrical widespread myelin loss in thecentral nervous system (Padiath et al. 2006).Heterozygous mutations or polymorphisms inLMNB2 encoding lamin B2 have been alsoreported in patients with acquired partial lipo-dystrophy (Hegele et al. 2006).

Overall, a number of human disorders havebeen described that are caused by mutationsin genes encoding lamins and associatednuclear envelope proteins. This number is likelyto continue to grow over time. Achalsia-Addi-sonianism-alacrima syndrome, familial atrialfibrillation, infantile bilateral striatal necrosis,and infection-triggered acute necrotizing ence-phalopathy have also been shown to result frommutations in genes encoding proteins of the nu-clear pore complex, a major component of thenuclear envelope that mediates nucleocytoplas-mic transport (Tullio-Pelet et al. 2000; Cron-shaw and Matunis 2003; Basel-Vanagaite et al.2006; Zhang et al. 2008; Neilson et al. 2009).

NUCLEAR ENVELOPE FUNCTION ANDDISEASE PATHOGENESIS

Studies of laminopathies have provided insightsinto novel functions of the nuclear envelope.Perhaps most significantly, these studiesstrongly suggest that the intermediate filamentnuclear lamina, although serving as a structuralsupport for the nuclear membranes, must haveadditional functions. Because very different dis-ease phenotypes can result from alterations inlamins, the nuclear lamina likely has cell-typeand tissue-selective properties.

Like all intermediate filament proteins,A-type lamins have a relatively small headdomain, a conserved a-helical rod domain,and a tail domain. Lamins A and C are identicalfor the first 566 amino acids, sharing the head,rod, and first portion of the tail domain, withlamin C having six unique carboxy-terminalamino acids and prelamin A having 98 uniquecarboxy-terminal residues. Examination of thepredominant genotype-phenotype correlationsfor alterations in lamin A structure stronglysuggests that different domains of the proteinshave different tissue-selective functions (Fig. 1).Most mutations that cause striated muscle

H.J. Worman, C. Ostlund, and Y. Wang

6 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 7: Diseases of the Nuclear Envelope

diseases lead to amino-acid substitutions, smalldeletions, splice site alterations, or truncationsthroughout lamins A and C. Approximately90% of the mutations that cause Dunnigan-typefamilial partial lipodystrophy generate amino-acid substitutions within an immunoglobulin-type fold in the tails of lamins A and C. Mostmutations causing peripheral neuropathy leadto the R298C substitution in the rod domainand most mutations causing mandibuloacraldysplasia generate amino-acid substitutionsat or very near amino-acid residue 527 in the

immunoglobulin-type fold. Classical Hutchinson-Gilford progeria syndrome is caused by mu-tations within exon 11 of LMNA, leading toan in frame deletion of 50 amino acids fromthe tail of prelamin A.

The distribution of LMNA mutations caus-ing striated muscle diseases suggests that theselead to a defect in overall lamina structure incells. Homozygous Lmna knockout mice devel-op regional skeletal and cardiac muscle ab-normalities within the first 2 months of lifeand their heterozygous littermates develop

Head domain

Rod domain

Tail domain lg-like fold

CaaX

ZMPSTE24cleavage site

HGPS

Δ

MAD

CMT

Myopathies FPLD

Figure 1. Schematic diagram of a prelamin A molecule with mutations causing laminopathies indicated. Mutationscausing Dunnigan-type famililal partial lipodystrophy (FPLD, dark blue asterisks), Charcot-Marie-Tooth disease(CMT, orange asterisk), mandibuloacral dysplasia (MAD, light blue asterisks), or Hutchinson-Gilford progeriasyndrome (HGPS, purple asterisk) are found in specific areas of the molecule, whereas mutations causingmyopathies (red asterisks) are found throughout. Mutation data are from Leiden Muscular Dystrophy pages(http://www.dmd.nl/lmna_home.html). Photographs are reproduced with permission from Elsevier(Chaouch et al. 2003; Emery 2000; Novelli et al. 2002), Macmillan Publishers Ltd (Peters et al. 1998; Towbinand Bowles 2002), and the American Association for the Advancement of Science (De Sandre-Giovannoliet al. 2003).

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 7

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 8: Diseases of the Nuclear Envelope

cardiomyopathy with conduction block atmuch older ages (Sullivan et al. 1999; Wolfet al. 2008). Heterozygous mutations leadingto significant lamin truncations in humansalso cause striated muscle disease (Bonne et al.1999; Jakobs et al. 2001; MacLeod et al. 2003).Expression of missense lamin A variants thatcause muscle disease disrupts the structure ofthe nuclear lamina and leads to morphologicalalterations similar to those in cells from whichA-type lamins are depleted (Ostlund et al.2001; Raharjo et al. 2001). Transgenic expres-sion of a lamin A variant encoded by a LMNAmutation that causes cardiomyopathy and mus-cular dystrophy in humans alters lamina struc-ture and induces severe heart disease in mice(Wang et al. 2006). These observations suggestthat muscle disease results either from a partialloss of A-type lamins or expression of variantsthat “dominantly interfere” with the overallstructure and function of the nuclear lamina.

It remains generally accepted that onefunction of the lamina is to provide structuralsupport to the nuclear envelope. One hypothe-sis for the pathogenesis of striated muscle dis-ease is that a defective lamina fails to properlycarry out this support function. Fibroblastsfrom Lmna knockout mice, transfected cellsthat express lamin A variants and fibroblastsfrom human subjects with LMNA mutationsand muscle diseases, all have abnormal nuclearmorphology at the light microscopy level(Sullivan et al. 1999; Ostlund et al. 2001; Raharjoet al. 2001; Muchir et al. 2004). Fibroblastsfrom Lmna knockout mice also have decreasedmechanical stiffness (Broers et al. 2004;Lammerding et al. 2004; Lee et al. 2007). Abnor-malities in the nuclear lamina could even poten-tially affect cytoskeleton functions, as thelamina is connected to cytoplasmic actin viathe nesprin isoforms and Sun proteins thatspan the perinuclear space (Crisp et al. 2006).Depletion of A-type lamins indeed disruptscytoskeletal processes such as cellular migra-tion and nuclear positioning (Lee et al. 2007;Houben et al. 2009).

In hearts and to a lesser extent in skeletalmuscle from Lmna H222P knockin mice, amodel of Emery-Dreifuss muscular dystrophy,

there is abnormal activation of the MAP kinasesERK1/2 and JNK (Muchir et al. 2007b). TheseMAP kinases are activated by mechanical stressin cardiomyocytes (Baines and Molkentin2005). Chronically increased ERK and JNK acti-vation is detrimental to hearts and treatment ofLmna H222P knockin mice with an inhibitor ofERK signaling prevents development of cardio-myopathy (Muchir et al. 2009). Altered nuclearenvelope elasticity is also caused by loss ofemerin, which binds to A-type lamins, and thiscould contribute to increased nuclear fragilityin humans subjects with mutations in EDMand striated muscle disease (Rowat et al. 2006).ERK is also abnormally activated in heartsof mice lacking emerin (Muchir et al. 2007a).Structural alterations in the nuclear envelopeand connected cytoskeleton resulting fromLMNA or EDM mutations may therefore makecells such as cardiomyocytes highly susceptibleto damage by recurrent mechanical stress, lead-ing to the activation of stress-response pathwaysthat are further detrimental to cells over time.

Data from subjects with Dunnigan-typefamilial partial lipodystrophy suggest that theimmunoglobulin-type fold in the tail ofA-type lamins has specific functions in adiposecells. LMNA mutations responsible for approx-imately 90% of cases lead to amino-acid sub-stitutions that decrease the positive charge of asolvent-exposed surface on the immunoglob-ulin-type fold but are not predicted to alterthe three-dimensional structure of lamins(Dhe-Paganon et al. 2002; Krimm et al.2002). However, they affect the ability of theimmunoglobulin-type fold of A-type laminsto bind to DNA (Stierle et al. 2003) and couldpotentially alter binding of a protein importantin adipocyte differentiation or survival. Thesemutations have in fact been shown to decreasean interaction between lamin A and SREBP1(Lloyd et al. 2002); however, the physiologicalconsequences of this interaction remain to beshown. LMNA mutations that cause Dunnigan-type familial partial lipodystrophy may resultin a “gain of function,” as overexpression ofeither lamin A with a causative amino-acidchange or wild-type lamin A both block differ-entiation of preadipocytes in adipocytes in vitro

H.J. Worman, C. Ostlund, and Y. Wang

8 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 9: Diseases of the Nuclear Envelope

(Boguslavsky et al. 2006) and deficiency ofA-type lamins does not cause lipodystrophy inmice (Cutler et al. 2002). The immunoglobulin-type fold of A-type lamins may thereforenegatively regulate adipocyte differentiation,or survival “gain of function” may cause partiallipodystrophy. Mandibuloacral dysplasia, whichhas partial lipodystrophy as a predominant fea-ture, also results from amino-acid substitutionsin the immunoglobulin-type fold; however,there are other abnormalities affecting differenttissues in this autosomal recessive diseaserequiring inheritance of two mutant LMNAalleles.

Perhaps the best example of a laminopathyproviding insights into nuclear envelope pro-tein function is what progeroid disorders havetaught us about the need for proper prelaminA processing. Since the early 1980s, it has beenrecognized that lamin A is synthesized as a pre-cursor molecule prelamin A (Gerace et al.1984). Prelamin A contains a CaaX motif at itscarboxyl terminus, a sequence known to initiatethree sequential chemical reactions (Clarke1992; Zhang and Casey 1996; Young et al.2005). In the first reaction, the cysteine of thecarboxy-terminal CaaX motif is farnesylated

by protein farnesyltransferase. In the secondreaction, the –aaX is clipped off. In the third,the farnesylcysteine is methylated by isoprenyl-cysteine carboxyl methyltransferase. The role ofthese three reactions in prelamin A processing isshown in Figure 2.

Initially, the groups of Klaus Weber andMichael Sinensky showed that processing ofprelamin A resulted from cleavage of an isopre-nylated, specifically farnesylated, polypeptide15 amino acids away from the carboxy-terminalcysteine (Weber et al. 1989; Beck et al. 1990;Sinensky et al. 1994). Sinensky’s group thencharacterized a farnesylation-dependent pre-lamin A endoprotease activity in cells (Kilicet al. 1997). In 2002, the groups of StephenYoung and Carlos Lopez-Otın reported thatmice deficient in zinc metalloproteinaseZMPSTE24 were defective in the processingof prelamin A to lamin A (Bergo et al. 2002;Pendas et al. 2002). In 2005, Sinensky’s groupconfirmed in vitro using recombinantZMPSTE24 that this endoprotease clips the–aaX from prelamin A and catalyzes the secondcleavage that removes the remaining 15 carbox-yl-terminal amino acids (Corrigan et al. 2005).Removal of the –aaX from prelamin A is likely

RD WT HGPSCaaX CaaX

CaaX

COCH3

C

CaaX

COCH3

C

CaaX

CaaX

COCH3

C

Farnesyltransferase Farnesyltransferase Farnesyltransferase

Isoprenylcysteine carboxylmethyltransferase

Isoprenylcysteine carboxylmethyltransferase

Isoprenylcysteine carboxylmethyltransferase

RCE1 ZMPSTE24 or RCE1 ZMPSTE24 or RCE1

ZMPSTE24

Mature lamin A

Figure 2. Processing of prelamin A to mature lamin A in wild-type (WT) cells occurs in several steps, described inthe text (middle column). In restrictive dermopathy (RD), the ZMPSTE24 enzyme is nonfunctioning, resultingin accumulation of farnesylated prelamin A (left column). In Hutchinson-Gilford progeria syndrome (HGPS),the second cleavage site for ZMPSTE24 is deleted, resulting in accumulation of a truncated form of farnesylatedprelamin A (right column).

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 9

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 10: Diseases of the Nuclear Envelope

redundantly catalyzed by the enzyme RCE1(Young et al. 2005).

The LMNA mutation causing Hutchinson-Gilford progeria syndrome that activates a cryp-tic splice site in exon 11 leads to an in framedeletion of 50 amino acids that contains thesecond ZMPSTE24 endoproteolytic site inprelamin A (De Sandre-Giovannoli et al. 2003;Eriksson et al. 2003). As a result, a farnesylated,truncated prelamin A variant that cannot beproperly processed accumulates in cells (Fig. 2).Loss of ZMPSTE24 activity leads to accumula-tion of unprocessed, farnesylated prelamin A(Fig. 2), which causes restrictive dermopathyand other progeroid disorders that have clinicaloverlap with Hutchinson-Gilford progeria syn-drome. Zmpste24 knockout mice have proge-roid features that overlap with mice having atargeted knockin mutation of Lmna that causesHutchinson-Gilford progeria syndrome (Yanget al. 2006). Blocking farnesylation of the trun-cated prelamin A or unprocessed prelamin A inthese mice with chemical inhibitors improvesthe mouse phenotypes (Fong et al. 2006; Yanget al. 2006; Varela et al. 2008). Similarly, hetero-zygosity for Lmna deficiency eliminates theprogeria-like phenotypes in Zmpste24 knockoutmice (Fong et al. 2004). These results indicatethat accumulation of farnesylated prelamin Aor the truncated variant plays a key role in thepathogenesis of the progeroid phenotype.

It is less clear how accumulation of farnesy-lated prelamin A polypeptides lead to progeriaphenotypes. Cultured cells expressing theseproteins have microscopic abnormalities innuclear morphology and blocking proteinfarnesyltransferase activity significantly reversesthese abnormalities (Eriksson et al. 2003; DeSandre-Giovannoli et al. 2003; Bridger andKill 2004; Goldman et al. 2004; Paradisi et al.2005; Yang et al. 2005; Capell et al. 2005; Glynnand Glover 2005; Mallampalli et al. 2005; Tothet al. 2005; Young et al. 2005; Wang et al. 2008).These nuclear morphological abnormalities areassociated with reduced deformability of thelamina and increased stiffness of the nucleus(Dahl et al. 2006; Verstraeten et al. 2008). How-ever, alterations in nuclear morphology andnuclear mechanics are not unique to progeroid

syndromes and occur as a result of LMNAmutations that cause other laminopathies. Ex-pression of farnesylated prelamin A or the trun-cated variant in Hutchinson-Gilford progeriasyndrome perturb DNA damage response andrepair, leading to genomic instability (Liu et al.2005), but inhibition of protein farnesylationdoes not appear to reduce DNA double-strandbreaks or damage checkpoint signaling (Liuet al. 2006). Cells accumulating these isopreny-lated A-type lamins also have altered signalingpathways involved in regulating stem cell behav-ior (Espada et al. 2008; Scaffidi and Misteli2008).

Although accumulation of isoprenylatedprelamin A polypeptides is important in proge-ria pathogenesis, genetic studies show that it isnot the entire explanation. Some atypical pro-geroid disorders resulting from LMNA muta-tions are not associated with accumulation ofprelamin A (Verstraeten et al. 2006). Further-more, Yang et al. (2008) elegantly showed thatexpression in mice of a nonfarnesylated variantof the truncated prelamin A in Hutchinson-Gilford progeria syndrome nonetheless causesa progeroid-like phenotype, albeit less severethan that in mice expressing the farnesylatedprotein. Hence, alterations in A-type laminsother than accumulation of isoprenylated formscan lead to the same cellular defects that give riseto progeroid phenotypes.

CONCLUDING REMARKS

Are studies of the rare monogenic laminopa-thies relevant to common human diseases andphysiological aging? Some data suggest thatthis might indeed be the case. As patients withfamilial dilated cardiomyopathies have beenscreened for genetic causes at some medicalcenters, LMNA mutations have been shown tobe responsible for approximately ten percentof all cases and a third with atrioventricular con-duction block (Arbustini et al. 2002; Taylor et al.2003; van Tintelen et al. 2007; Parks et al. 2008).Compared with other dilated cardiomyo-pathies, those caused by LMNA mutations areassociated with the rapid development of heartfailure, early life-threatening arrhythmias, and

H.J. Worman, C. Ostlund, and Y. Wang

10 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 11: Diseases of the Nuclear Envelope

sudden death (Becane et al. 2000; Taylor et al.2003; van Berlo et al. 2005; Pasotti et al. 2008).Hence, screening for LMNA mutations as partof the clinical routine could provide informationthat leads to early placement of a pacemakerand an implantable cardioverter defibrillator toprevent sudden death (Meune et al. 2006).

Given that mutations in genes encoding nu-clear envelope proteins cause rare monogenicdiseases, it is possible that polymorphic variantsof the same genes predispose or contributequantitatively to the development of commondiseases. As LMNA mutations cause rare lipo-dystrophy disorders, several groups have exam-ined if LMNA polymorphisms contribute to thedevelopment of common disorders. Althoughnot completely conclusive, some studies suggestthat polymorphic variations in LMNA may pre-dispose to insulin resistance, diabetes mellitus,and metabolic syndrome (Duesing et al. 2008;Steinle et al. 2004; Wegner et al. 2007; Owenet al. 2007; Mesa et al. 2007; Murase et al.2002). Hence, subtle alterations in A-type lam-ins may contribute to the pathogenesis of dis-eases that are endemic in the developed world.One could similarly hypothesize that poly-morphisms in genes encoding other nuclearenvelope proteins could contribute to othercommon diseases. For example, subtle altera-tions in MAN1, loss of function of which causessclerosing bone dysplasias, could hypotheticallycontribute to the development of osteoporosis.

The involvement of A-type lamins in thepathogenesis of progeroid syndromes hasraised interest about their role in physiologicalaging. The abnormal RNA splicing occur-ring as a result of the LMNA mutations thatcause Hutchinson-Gilford progeria syndrometakes place at very low levels in normal cells(McClintock et al. 2007; Scaffidi and Misteli2006). One study has shown that the truncatedprelamin A that results from this RNA splicingevent accumulates in dermal fibroblasts andkeratinocytes in older individuals (McClintocket al. 2007). Fibroblasts from older normal sub-jects have also been reported to show defectssimilar to those in cells from subjects withHutchinson-Gilford progeria syndrome, suchas abnormal nuclear morphology, increased

DNA damage, and changes in histone modifica-tions (Scaffidi and Misteli 2006). These findingssupport a hypothesis that low-level expressionof the truncated prelamin A generated as a resultof the LMNA mutation that causes Hutchinson-Gilford progeria syndrome may contribute toaspects of physiological aging.

In closing, a word of caution is warrantedabout extrapolating data from laminopathiesto common diseases. For example, childrenwith Hutchinson-Gilford progeria syndromehave normal cognitive and other brain func-tions, whereas central nervous system degenera-tion is a major feature of normal human aging.Elevated blood levels of total cholesterol,C-reactive protein, and low density lipoproteindo not appear to contribute to the acceler-ated vascular occlusive disease in Hutchinson-Gilford progeria syndrome (Gordon et al. 2005)and affected vessels show pathological featuresthat are unusual for typical atherosclerosis(Stehbens et al. 1999). Hence, Hutchinson-Gilford progeria syndrome may not be a per-fectly accurate model to understand some ofthe major complications of physiological humanaging. Similarly, other laminopathies that sharephenotypic features with common humandisorders may have different underlying patho-genic mechanisms. Nonetheless, research onthis fascinating group of rare diseases is clearlyproviding clues about fundamental functions ofthe nuclear envelope as well as relevant insightsinto cellular processes that must be at least partlyinvolved in certain aspects of common diseasesand aging.

ACKNOWLEDGMENTS

The authors were supported by grants fromthe National Institutes of Health (AR048997,NS059352, and AG025240) and a grant fromthe Muscular Dystrophy Association (MDA4287)to H.J. Worman.

REFERENCES

Agarwal AK, Fryns JP, Auchus RJ, Garg A. 2003. Zinc metal-loproteinase, ZMPSTE24, is mutated in mandibuloacraldysplasia. Hum Mol Genet 12: 1995–2001.

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 11

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 12: Diseases of the Nuclear Envelope

Agarwal AK, Kazachkova I, Ten S, Garg A. 2008. Severemandibuloacral dysplasia-associated lipodystrophy andprogeria in a young girl with a novel homozygousArg527Cys LMNA mutation. J Clin Endocrinol Metab93: 4617–4623.

Arbustini E, Pilotto A, Repetto A, Grasso M, Negri A,Diegoli M, Campana C, Scelsi L, Baldini E, Gavazzi A,Tavazzi L. 2002. Autosomal dominant dilated cardiomy-opathy with atrioventricular block: A lamin A/Cdefect-related disease. J Am Coll Cardiol 39: 981–990.

Astejada MN, Goto K, Nagano A, Ura S, Noguchi S, NonakaI, Nishino I, Hayashi YK. 2007. Emerinopathy and lamin-opathy clinical, pathological and molecular features ofmuscular dystrophy with nuclear envelopathy in Japan.Acta Myol 26: 159–164.

Attali R, Warwar N, Israel A, Gurt I, McNally E, PuckelwartzM, Glick B, Nevo Y, Ben-Neriah Z, Melki J. 2009. Muta-tion of SYNE-1, encoding an essential component of thenuclear lamina, is responsible for autosomal recessivearthrogryposis. Hum Mol Genet 18: 3462–3469.

Baines CP, Molkentin JD. 2005. STRESS signaling pathwaysthat modulate cardiac myocyte apoptosis. J Mol CellCardiol 38: 47–62.

Basel-Vanagaite L, Muncher L, Straussberg R, Pasmanik-Chor M, Yahav M, Rainshtein L, Walsh CA, Magal N,Taub E, Drasinover V, et al. 2006. Mutated nup62 causesautosomal recessive infantile bilateral striatal necrosis.Ann Neurol 60: 214–222.

Beadle GW, Tatum EL. 1941. Genetic control of biochemicalreactions in Neurospora. Proc Natl Acad Sci 27: 499–506.

Becane HM, Bonne G, Varnous S, Muchir A, Ortega V,Hammouda EH, Urtizberea JA, Lavergne T, Fardeau M,Eymard B, et al. 2000. High incidence of sudden deathwith conduction system and myocardial disease due tolamins A and C gene mutation. Pacing Clin Electrophysiol23: 1661–1666.

Beck LA, Hosick TJ, Sinensky M. 1990. Isoprenylation is re-quired for the processing of the lamin A precursor. J CellBiol 110: 1489–1499.

Bergo MO, Gavino B, Ross J, Schmidt WK, Hong C, KendallLV, Mohr A, Meta M, Genant H, Jiang Y, et al. 2002.Zmpste24 deficiency in mice causes spontaneous bonefractures, muscle weakness, and a prelamin A processingdefect. Proc Natl Acad Sci 99: 13049–13054.

Bione S, Maestrini E, Rivella S, Mancini M, Regis S, RomeoG, Toniolo D. 1994. Identification of a novel X-linkedgene responsible for Emery-Dreifuss muscular dystrophy.Nat Genet 8: 323–327.

Bione S, Small K, Aksmanovic VM, D’Urso M, CiccodicolaA, Merlini L, Morandi L, Kress W, Yates JR, Warren ST,et al. 1995. Identification of new mutations in the Emery-Dreifuss muscular dystrophy gene and evidence forgenetic heterogeneity of the disease. Hum Mol Genet 4:1859–1863.

Boguslavsky RL, Stewart CL, Worman HJ. 2006. Nuclearlamin A inhibits adipocyte differentiation: Implicationsfor Dunnigan-type familial partial lipodystrophy. HumMol Genet 15: 653–663.

Bonne G, Di Barletta MR, Varnous S, Becane HM, Hammou-da EH, Merlini L, Muntoni F, Greenberg CR, Gary F,Urtizberea JA, et al. 1999. Mutations in the gene encoding

lamin A/C cause autosomal dominant Emery-Dreifussmuscular dystrophy. Nat Genet 21: 285–288.

Bonne G, Mercuri E, Muchir A, Urtizberea A, Becane HM,Recan D, Merlini L, Wehnert M, Boor R, Reuner U,et al. 2000. Clinical and molecular genetic spectrum ofautosomal dominant Emery-Dreifuss muscular dystro-phy due to mutations of the lamin A/C gene. Ann Neurol48: 170–180.

Bridger JM, Kill IR. 2004. Aging of Hutchinson–Gilfordprogeria syndrome fibroblasts is characterised by hyper-proliferation and increased apoptosis. Exp Gerontol 39:717–724.

Brodsky GL, Muntoni F, Miocic S, Sinagra G, Sewry C,Mestroni L. 2000. Lamin A/C gene mutation associatedwith dilated cardiomyopathy with variable skeletalmuscle involvement. Circulation 101: 473–476.

Broers JL, Peeters EA, Kuijpers HJ, Endert J, Bouten CV,Oomens CW, Baaijens FP, Ramaekers FC. 2004.Decreased mechanical stiffness in LMNA-/- cells iscaused by defective nucleo-cytoskeletal integrity: Impli-cations for the development of laminopathies. HumMol Genet 13: 2567–2580.

Cao H, Hegele RA. 2000. Nuclear lamin A/C R482Qmutation in Canadian kindreds with Dunnigan-typefamilial partial lipodystrophy. Hum Mol Genet 9:109–112.

Cao H, Hegele RA. 2003. LMNA is mutated in Hutchinson-Gilford progeria (MIM 176670) but not in Wiedemann-Rautenstrauch progeroid syndrome (MIM 264090).J Hum Genet 48: 271–274.

Capell BC, Erdos MR, Madigan JP, Fiordalisi JJ, Varga R,Conneely KN, Gordon LB, Der CJ, Cox AD, Collins FS.2005. Inhibiting farnesylation of progerin preventsthe characteristic nuclear blebbing of Hutchinson–Gilford progeria syndrome. Proc Natl Acad Sci 102:12879–12884.

Caputo S, Couprie J, Duband-Goulet I, Konde E, Lin F,Braud S, Gondry M, Gilquin B, Worman HJ, Zinn-JustinS. 2006. The carboxyl-terminal nucleoplasmic region ofMAN1 exhibits a DNA binding winged helix domain. JBiol Chem 281: 18208–18215.

Chaouch M, Allal Y, De Sandre-Giovannoli A, Vallat JM,Amer-el-Khedoud A, Kassouri N, Chaouch A, SindouP, Hammadouche T, Tazir M, et al. 2003. The pheno-typic manifestations of autosomal recessive axonalCharcot-Marie-Tooth due to a mutation in Lamin A/Cgene. Neuromuscul Disord 13: 60–67.

Chen L, Lee L, Kudlow BA, Dos Santos HG, Sletvold O,Shafeghati Y, Botha EG, Garg A, Hanson NB, MartinGM, et al. 2003. LMNA mutations in atypical Werner’ssyndrome. Lancet 362: 440–445.

Clarke S. 1992. Protein isoprenylation and methylation atcarboxyl-terminal cysteine residues. Annu Rev Biochem61: 355–386.

Clements L, Manilal S, Love DR, Morris GE. 2000. Directinteraction between emerin and lamin A. Biochem Bio-phys Res Commun 267: 709–714.

Corrigan DP, Kuszczak D, Rusinol AE, Thewke DP, HrycynaCA, Michaelis S, Sinensky MS. 2005. Prelamin Aendoproteolytic processing in vitro by recombinantZmpste24. Biochem J 387: 129–138.

H.J. Worman, C. Ostlund, and Y. Wang

12 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 13: Diseases of the Nuclear Envelope

Crisp M, Liu Q, Roux K, Rattner JB, Shanahan C, Burke B,Stahl PD, Hodzic D. 2006. Coupling of the nucleus andcytoplasm: Role of the LINC complex. J Cell Biol 172:41–53.

Cronshaw JM, Matunis MJ. 2003. The nuclear pore complexprotein ALADIN is mislocalized in triple A syndrome.Proc Natl Acad Sci 100: 5823–5827.

Csoka AB, Cao H, Sammak PJ, Constantinescu D, SchattenGP, Hegele RA. 2004. Novel lamin A/C gene (LMNA)mutations in atypical progeroid syndromes. J Med Genet41: 304–308.

Cutler DA, Sullivan T, Marcus-Samuels B, Stewart CL,Reitman ML. 2002. Characterization of adiposity andmetabolism in Lmna-deficient mice. Biochem BiophysRes Commun 291: 522–527.

Dahl KN, Scaffidi P, Islam MF, Yodh AG, Wilson KL, MisteliT. 2006. Distinct structural and mechanical propertiesof the nuclear lamina in Hutchinson-Gilford progeriasyndrome. Proc Natl Acad Sci 103: 10271–10276.

DeBusk FL. 1972. The Hutchinson-Gilford progeriasyndrome. J Pediat 80: 697–724.

De Sandre-Giovannoli A, Bernard R, Cau P, Navarro C,Amiel J, Boccaccio I, Lyonnet S, Stewart CL, MunnichA, Le Merrer M, et al. 2003. Lamin a truncation inHutchinson-Gilford progeria. Science 300: 2055.

De Sandre-Giovannoli A, Chaouch M, Kozlov S, Vallat JM,Tazir M, Kassouri N, Szepetowski P, Hammadouche T,Vandenberghe A, Stewart CL, et al. 2002. Homozygousdefects in LMNA, encoding lamin A/C nuclear-envelopeproteins, cause autosomal recessive axonal neuropathyin human (Charcot-Marie-Tooth disorder type 2) andmouse. Am J Hum Genet 70: 726–736.

Dhe-Paganon S, Werner ED, Chi YI, Shoelson SE. 2002.Structure of the globular tail of nuclear lamin. J BiolChem 277: 17381–17384.

Duesing K, Charpentier G, Marre M, Tichet J, Hercberg S,Froguel P, Gibson F. 2008. Evaluating the associationof common LMNA variants with type 2 diabetes andquantitative metabolic phenotypes in French Europids.Diabetologia 51: 76–81.

Dunnigan MG, Cochrane MA, Kelly A, Scott JW. 1974.Familial lipoatrophic diabetes with dominant transmis-sion. A new syndrome. Q J Med 43: 33–48.

Emery AE. 2000. Emery-Dreifuss muscular dystrophy—a 40year retrospective. Neuromuscul Disord 10: 228–232.

Emery AE, Dreifuss FE. 1966. Unusual type of benignx-linked muscular dystrophy. J Neurol Neurosurg Psychia-try 29: 338–342.

Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J,Scott L, Erdos MR, Robbins CM, Moses TY, Berglund P,et al. 2003. Recurrent de novo point mutations in laminA cause Hutchinson-Gilford progeria syndrome. Nature423: 293–298.

Espada J, Varela I, Flores I, Ugalde AP, Cadinanos J, PendasAM, Stewart CL, Tryggvason K, Blasco MA, Freije JM,et al. 2008. Nuclear envelope defects cause stem cell dys-function in premature-aging mice. J Cell Biol 181: 27–35.

Fairley EA, Kendrick-Jones J, Ellis JA. 1999. The Emery-Dreifuss muscular dystrophy phenotype arises from aber-rant targeting and binding of emerin at the inner nuclearmembrane. J Cell Sci 112: 2571–2582.

Fatkin D, MacRae C, Sasaki T, Wolff MR, Porcu M,Frenneaux M, Atherton J, Vidaillet HJ Jr, Spudich S, DeGirolami U, et al. 1999. Missense mutations in the roddomain of the lamin A/C gene as causes of dilated cardi-omyopathy and conduction-system disease. N Engl J Med341: 1715–1724.

Fong LG, Frost D, Meta M, Qiao X, Yang SH, Coffinier C,Young SG. 2006. A protein farnesyltransferase inhibitorameliorates disease in a mouse model of progeria. Science311: 1621–1623.

Fong LG, Ng JK, Meta M, Cote N, Yang SH, Stewart CL,Sullivan T, Burghardt A, Majumdar S, Reue K, et al.2004. Heterozygosity for Lmna deficiency eliminatesthe progeria-like phenotypes in Zmpste24-deficientmice. Proc Natl Acad Sci 101: 18111–18116.

Garg A, Cogulu O, Ozkinay F, Onay H, Agarwal AK. 2005. Anovel homozygous Ala529Val LMNA mutation inTurkish patients with mandibuloacral dysplasia. J ClinEndocrinol Metab 90: 5259–5264.

Gerace L, Comeau C, Benson M. 1984. Organization andmodulation of nuclear lamina structure. J Cell Sci Suppl1: 137–160.

Gilford H. 1904. Ateleiosis and progeria: Continuous youthand premature old age. Brit Med J 2: 914–918.

Glynn MW, Glover TW. 2005. Incomplete processing of mu-tant lamin A in Hutchinson–Gilford progeria leads tonuclear abnormalities, which are reversed by farnesyl-transferase inhibition. Hum Mol Genet 14: 2959–2969.

Goldman RD, Shumaker DK, Erdos MR, Eriksson M,Goldman AE, Gordon LB, Gruenbaum Y, Khuon S,Mendez M, Varga R, et al. 2004. Accumulation of mutantlamin A causes progressive changes in nuclear archi-tecture in Hutchinson-Gilford progeria syndrome. ProcNatl Acad Sci 101: 8963–8968.

Gonzalez-Alegre P, Paulson HL. 2004. Aberrant cellular be-havior of mutant torsinA implicates nuclear envelopedysfunction in DYT1 dystonia. J Neurosci 24: 2593–2601.

Goodchild RE, Dauer WT. 2004. Mislocalization to the nu-clear envelope: An effect of the dystonia-causing torsinAmutation. Proc Natl Acad Sci 101: 847–852.

Goodchild RE, Dauer WT. 2005. The AAAþ protein torsinAinteracts with a conserved domain present in LAP1 and anovel ER protein. J Cell Biol 168: 855–862.

Goodchild RE, Kim CE, Dauer WT. 2005. Loss of thedystonia-associated protein torsinA selectively disruptsthe neuronal nuclear envelope. Neuron 48: 923–932.

Gordon LB, Harten IA, Patti ME, Lichtenstein AH. 2005.Reduced adiponectin and HDL cholesterol withoutelevated C-reactive protein: Clues to the biology of pre-mature atherosclerosis in Hutchinson-Gilford ProgeriaSyndrome. J Pediatr 146: 336–341.

Gros-Louis F, Dupre N, Dion P, Fox MA, Laurent S,Verreault S, Sanes JR, Bouchard JP, Rouleau GA. 2007.Mutations in SYNE1 lead to a newly discovered form ofautosomal recessive cerebellar ataxia. Nat Genet 39:80–85.

Hegele RA, Cao H, Liu DM, Costain GA, Charlton-Menys V,Rodger NW, Durrington PN. 2006. Sequencing of thereannotated LMNB2 gene reveals novel mutations inpatients with acquired partial lipodystrophy. Am J HumGenet 79: 383–389.

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 13

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 14: Diseases of the Nuclear Envelope

Hellemans J, Preobrazhenska O, Willaert A, Debeer P,Verdonk PC, Costa T, Janssens K, Menten B, Van RoyN, Vermeulen SJ, et al. 2004. Loss-of-function mutationsin LEMD3 result in osteopoikilosis, Buschke-Ollendorffsyndrome and melorheostosis. Nat Genet 36: 1213–1238.

Hoffmann K, Dreger CK, Olins AL, Olins DE, Shultz LD,Lucke B, Karl H, Kaps R, Muller D, Vaya A, et al. 2002.Mutations in the gene encoding the lamin B receptor pro-duce an altered nuclear morphology in granulocytes(Pelger-Huet anomaly). Nat Genet 31: 410–414.

Holmer L, Pezhman A, Worman HJ. 1998. The humanlamin B receptor/sterol reductase multigene family.Genomics 54: 469–476.

Houben F, Willems CH, Declercq IL, Hochstenbach K,Kamps MA, Snoeckx LH, Ramaekers FC, Broers JL.2009. Disturbed nuclear orientation and cellular migra-tion in A-type lamin deficient cells. Biochim BiophysActa 1793: 312–324.

Hutchinson J. 1886. Case of congenital absence of hair, withatrophic condition of the skin and its appendages, in aboy whose mother had been almost wholly bald from alo-pecia areata from the age of six. Lancet I: 923.

Jackson SN, Pinkney J, Bargiotta A, Veal CD, Howlett TA,McNally PG, Corral R, Johnson A, Trembath RC. 1998.A defect in the regional deposition of adipose tissue (par-tial lipodystrophy) is encoded by a gene at chromosome1q. Am J Hum Genet 63: 534–540.

Jakobs PM, Hanson EL, Crispell KA, Toy W, Keegan H,Schilling K, Icenogle TB, Litt M, Hershberger RE. 2001.Novel lamin A/C mutations in two families with dilatedcardiomyopathy and conduction system disease. J CardFail 7: 249–256.

Kilic F, Dalton MB, Burrell SK, Mayer JP, Patterson SD,Sinensky M. 1997. In vitro assay and characterization ofthe farnesylation-dependent prelamin A endoprotease.J Biol Chem 272: 5298–5304.

Krimm I, Ostlund C, Gilquin B, Couprie J, Hossenlopp P,Mornon JP, Bonne G, Courvalin JC, Worman HJ,Zinn-Justin S. 2002. The Ig-like structure of theC-terminal domain of lamin A/C, mutated in musculardystrophies, cardiomyopathy, and partial lipodystrophy.Structure 10: 811–823.

Lammerding J, Schulze PC, Takahashi T, Kozlov S, SullivanT, Kamm RD, Stewart CL, Lee RT. 2004. Lamin A/C de-ficiency causes defective nuclear mechanics and mecha-notransduction. J Clin Invest 113: 370–378.

Lee JS, Hale CM, Panorchan P, Khatau SB, George JP, TsengY, Stewart CL, Hodzic D, Wirtz D. 2007. Nuclear laminA/C deficiency induces defects in cell mechanics, polar-ization, and migration. Biophys J 93: 2542–2552.

Lin F, Worman HJ. 1993. Structural organization of the hu-man gene encoding nuclear lamin A and nuclear lamin C.J Biol Chem 268: 16321–16326.

Lin F, Blake DL, Callebaut I, Skerjanc IS, Holmer L,McBurney MW, Paulin-Levasseur M, Worman HJ.2000. MAN1, an inner nuclear membrane protein thatshares the LEM domain with lamina-associated poly-peptide 2 and emerin. J Biol Chem 275: 4840–4847.

Lin F, Morrison JM, Wu W, Worman HJ. 2005. MAN1, anintegral protein of the inner nuclear membrane, bindsSmad2 and Smad3 and antagonizes transforming growthfactor-b signaling. Hum Mol Genet 14: 437–445.

Liu Y, Rusinol A, Sinensky M, Wang Y, Zou Y. 2006. DNAdamage responses in progeroid syndromes arise fromdefective maturation of prelamin A. J Cell Sci 119:4644–4649.

Liu B, Wang J, Chan KM, Tjia WM, Deng W, Guan X, HuangJD, Li KM, Chau PY, Chen DJ, et al. 2005. Genomic insta-bility in laminopathy-based premature aging. Nat Med11: 780–785.

Lloyd DJ, Trembath RC, Shackleton S. 2002. A novel interac-tion between lamin A and SREBP 1: Implications forpartial lipodystrophy and other laminopathies. HumMol Genet 11: 769–777.

Lombardi F, Gullotta F, Columbaro M, Filareto A, D’AdamoM, Vielle A, Guglielmi V, Nardone AM, Azzolini V, Gros-so E, et al. 2007. Compound heterozygosity for muta-tions in LMNA in a patient with a myopathic andlipodystrophic mandibuloacral dysplasia type A pheno-type. J Clin Endocrinol Metab 92: 4467–4471.

MacLeod HM, Culley MR, Huber JM, McNally EM. 2003.Lamin A/C truncation in dilated cardiomyopathy withconduction disease. BMC Med Genet 4: 4.

Makri S, Clarke NF, Richard P, Maugenre S, Demay L, BonneG, Guicheney P. 2009. Germinal mosaicism for LMNAmimics autosomal recessive congenital muscular dystro-phy. Neuromuscul Disord 19: 26–28.

Mallampalli MP, Huyer G, Bendale P, Gelb MH, Michaelis S.2005. Inhibiting farnesylation reverses the nuclear mor-phology defect in a HeLa cell model for Hutchinson–Gilford progeria syndrome. Proc Natl Acad Sci 102:14416–14421.

Manilal S, Nguyen TM, Sewry CA, Morris GE. 1996. TheEmery-Dreifuss muscular dystrophy protein, emerin,is a nuclear membrane protein. Hum Mol Genet 5:801–808.

Mansharamani M, Wilson KL. 2005. Direct binding of nu-clear membrane protein MAN1 to emerin in vitro andtwo modes of binding to barrier-to-autointegrationfactor. J Biol Chem 280: 13863–13870.

McClintock D, Ratner D, Lokuge M, Owens DM, GordonLB, Collins FS, Djabali K. 2007. The mutant form of lam-in A that causes Hutchinson-Gilford progeria is a bio-marker of cellular aging in human skin. PLoS One 2:e1269.

McKusick VA. 1952. The clinical observations of Jona-than Hutchinson. Am J Syph Gonorrhea Vener Dis 36:101–126.

Merideth MA, Gordon LB, Clauss S, Sachdev V, Smith AC,Perry MB, Brewer CC, Zalewski C, Kim HJ, Solomon B,et al. 2008. Phenotype and course of Hutchinson-Gilfordprogeria syndrome. N Engl J Med 358: 592–604.

Mesa JL, Loos RJ, Franks PW, Ong KK, Luan J, O’Rahilly S,Wareham NJ, Barroso I. 2007. Lamin A/C polymor-phisms, type 2 diabetes, and the metabolic syndrome:Case-control and quantitative trait studies. Diabetes 56:884–889.

Meune C, Van Berlo JH, Anselme F, Bonne G, Pinto YM,Duboc D. 2006. Primary prevention of sudden death inpatients with lamin A/C gene mutations. N Engl J Med354: 209–210.

Muchir A, Bonne G, van der Kooi AJ, van Meegen M, Baas F,Bolhuis PA, de Visser M, Schwartz K. 2000. Identificationof mutations in the gene encoding lamins A/C in

H.J. Worman, C. Ostlund, and Y. Wang

14 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 15: Diseases of the Nuclear Envelope

autosomal dominant limb girdle muscular dystrophy withatrioventricular conduction disturbances (LGMD1B).Hum Mol Genet 9: 1453–1459.

Muchir A, Medioni J, Laluc M, Massart C, Arimura T, vander Kooi AJ, Desguerre I, Mayer M, Ferrer X, Briault S,et al. 2004. Nuclear envelope alterations in fibroblastsfrom patients with muscular dystrophy, cardiomyopathy,and partial lipodystrophy carrying lamin A/C gene mu-tations. Muscle Nerve 30: 444–450.

Muchir A, Worman HJ. 2007. Emery-Dreifuss musculardystrophy. Curr Neurol Neurosci Rep 7: 78–83.

Muchir A, Pavlidis P, Bonne G, Hayashi YK, Worman HJ.2007a. Activation of MAPK in hearts of EMD nullmice: Similarities between mouse models of X-linkedand autosomal dominant Emery Dreifuss muscular dys-trophy. Hum Mol Genet 16: 1884–1895.

Muchir A, Pavlidis P, Decostre V, Herron AJ, Arimura T,Bonne G, Worman HJ. 2007b. Activation of MAPK path-ways links LMNA mutations to cardiomyopathy inEmery-Dreifuss muscular dystrophy. J Clin Invest 117:1282–1293.

Muchir A, Shan J, Bonne G, Lehnart SE, Worman HJ. 2009.Inhibition of extracellular signal-regulated kinase signal-ing to prevent cardiomyopathy caused by mutation in thegene encoding A-type lamins. Hum Mol Genet 18:241–247.

Murase Y, Yagi K, Katsuda Y, Asano A, Koizumi J, MabuchiH. 2002. An LMNA variant is associated with dyslipide-mia and insulin resistance in the Japanese. Metabolism51: 1017–1021.

Nagano A, Koga R, Ogawa M, Kurano Y, Kawada J, Okada R,Hayashi YK, Tsukahara T, Arahata K. 1996. Emerin defi-ciency at the nuclear membrane in patients with Emery-Dreifuss muscular dystrophy. Nat Genet 12: 254–259.

Naismith TV, Heuser JE, Breakefield XO, Hanson PI. 2004.TorsinA in the nuclear envelope. Proc Natl Acad Sci101: 7612–7617.

Navarro CL, Cadinanos J, De Sandre-Giovannoli A, BernardR, Courrier S, Boccaccio I, Boyer A, Kleijer WJ, Wagner A,Giuliano F, et al. 2005. Loss of ZMPSTE24 (FACE-1)causes autosomal recessive restrictive dermopathy andaccumulation of Lamin A precursors. Hum Mol Genet14: 1503–1513.

Neilson DE, Adams MD, Orr CM, Schelling DK, Eiben RM,Kerr DS, Anderson J, Bassuk AG, Bye AM, Childs AM,et al. 2009. Infection-triggered familial or recurrent casesof acute necrotizing encephalopathy caused by mutationsin a component of the nuclear pore, RANBP2. Am J HumGenet 84: 44–51.

Novelli G, Muchir A, Sangiuolo F, Helbling-Leclerc A,D’Apice MR, Massart C, Capon F, Sbraccia P, FedericiM, Lauro R, et al. 2002. Mandibuloacral dysplasia iscaused by a mutation in LMNA-encoding lamin A/C.Am J Hum Genet 71: 426–431.

Oosterwijk JC, Mansour S, van Noort G, Waterham HR,Hall CM, Hennekam RC. 2003. Congenital abnormalitiesreported in Pelger-Huet homozygosity as compared toGreenberg/HEM dysplasia: highly variable expressionof allelic phenotypes. J Med Genet 40: 937–941.

Ostlund C, Bonne G, Schwartz K, Worman HJ. 2001. Prop-erties of lamin A mutants found in Emery-Dreifuss

muscular dystrophy, cardiomyopathy and Dunnigan-type partial lipodystrophy. J Cell Sci 114: 4435–4445.

Owen KR, Groves CJ, Hanson RL, Knowler WC, ShuldinerAR, Elbein SC, Mitchell BD, Froguel P, Ng MC, ChanJC, et al. 2007. Common variation in the LMNA gene(encoding lamin A/C) and type 2 diabetes: Associationanalyses in 9,518 subjects. Diabetes 56: 879–883.

Ozelius LJ, Hewett JW, Page CE, Bressman SB, Kramer PL,Shalish C, de Leon D, Brin MF, Raymond D, Corey DP,et al. 1997. The early-onset torsion dystonia gene(DYT1) encodes an ATP-binding protein. Nat Genet 17:40–48.

Padiath QS, Saigoh K, Schiffmann R, Asahara H, Yamada T,Koeppen A, Hogan K, Ptacek LJ, Fu YH. 2006. Lamin B1duplications cause autosomal dominant leukodystrophy.Nat Genet 38: 1114–1123.

Pan D, Estevez-Salmeron LD, Stroschein SL, Zhu X, He J,Zhou S, Luo K. 2005. The integral inner nuclear mem-brane protein MAN1 physically interacts with theR-Smad proteins to repress signaling by the transforminggrowth factor-{b} superfamily of cytokines. J Biol Chem280: 15992–16001.

Paradisi M, McClintock D, Boguslavsky RL, Pedicelli C,Worman HJ, Djabali K. 2005. Dermal fibroblasts inHutchinson–Gilford progeria syndrome with the laminA G608G mutation have dysmorphic nuclei and arehypersensitive to heat stress. BMC Cell Biol 6: 27.

Parks SB, Kushner JD, Nauman D, Burgess D, Ludwigsen S,Peterson A, Li D, Jakobs P, Litt M, Porter CB, et al. 2008.Lamin A/C mutation analysis in a cohort of 324 unre-lated patients with idiopathic or familial dilated cardio-myopathy. Am Heart J 156: 161–169.

Pasotti M, Klersy C, Pilotto A, Marziliano N, Rapezzi C,Serio A, Mannarino S, Gambarin F, Favalli V, Grasso M,et al. 2008. Long-term outcome and risk stratificationin dilated cardiolaminopathies. J Am Coll Cardiol 52:1250–1260.

Pendas AM, Zhou Z, Cadinanos J, Freije JM, Wang J,Hultenby K, Astudillo A, Wernerson A, Rodrıguez F,Tryggvason K, et al. 2002. Defective prelamin A process-ing and muscular and adipocyte alterations in Zmpste24metalloproteinase-deficient mice. Nat Genet 31: 94–99.

Peters JM, Barnes R, Bennett L, Gitomer WM, BowcockAM, Garg A. 1998. Localization of the gene for familialpartial lipodystrophy (Dunnigan variety) to chromo-some 1q21-22. Nat Genet 18: 292–295.

Quijano-Roy S, Mbieleu B, Bonnemann CG, Jeannet PY,Colomer J, Clarke NF, Cuisset JM, Roper H, De MeirleirL, D’Amico A, et al. 2008. De novo LMNA mutationscause a new form of congenital muscular dystrophy.Ann Neurol 64: 177–186.

Raharjo WH, Enarson P, Sullivan T, Stewart CL, Burke B.2001. Nuclear envelope defects associated with LMNAmutations cause dilated cardiomyopathy and Emery-Dreifuss muscular dystrophy. J Cell Sci 114: 4447–4457.

Raffaele di Barletta M, Ricci E, Galluzzi G, Tonali P, Mora M,Morandi L, Romorini A, Voit T, Orstavik KH, Merlini L,et al. 2000. Different mutations in the LMNA gene causeautosomal dominant and autosomal recessive Emery-Dreifuss muscular dystrophy. Am J Hum Genet 66:1407–1412.

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 15

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 16: Diseases of the Nuclear Envelope

Renou L, Stora S, Yaou RB, Volk M, Sinkovec M, Demay L,Richard P, Peterlin B, Bonne G. 2008. Heart-hand syn-drome of Slovenian type: A new kind of laminopathy. JMed Genet 45: 666–671.

Rowat AC, Lammerding J, Ipsen JH. 2006. Mechanical prop-erties of the cell nucleus and the effect of emerin defi-ciency. Biophys J 91: 4649–4664.

Scaffidi P, Misteli T. 2006. Lamin A-dependent nuclear de-fects in human aging. Science 312: 1059–1063.

Scaffidi P, Misteli T. 2008. Lamin A-dependent misregula-tion of adult stem cells associated with accelerated ageing.Nat Cell Biol 10: 452–459.

Shackleton S, Lloyd DJ, Jackson SN, Evans R, NiermeijerMF, Singh BM, Schmidt H, Brabant G, Kumar S, Dur-rington PN, et al. 2000. LMNA, encoding lamin A/C, ismutated in partial lipodystrophy. Nat Genet 24: 153–156.

Shackleton S, Smallwood DT, Clayton P, Wilson LC, AgarwalAK, Garg A, Trembath RC. 2005. Compound heterozy-gous ZMPSTE24 mutations reduce prelamin A process-ing and result in a severe progeroid phenotype. J MedGenet 42: e36.

Sinensky M, Fantle K, Trujillo M, McLain T, Kupfer A,Dalton M. 1994. The processing pathway of prelaminA. J Cell Sci 107: 61–67.

Speckman RA, Garg A, Du F, Bennett L, Veile R, Arioglu E,Taylor SI, Lovett M, Bowcock AM. 2000. Mutational andhaplotype analyses of families with familial partial lipo-dystrophy (Dunnigan variety) reveal recurrent missensemutations in the globular C-terminal domain of laminA/C. Am J Hum Genet 66: 1192–1198.

Stehbens WE, Wakefield SJ, Gilbert-Barness E, Olson RE,Ackerman J. 1999. Histological and ultrastructural fea-tures of atherosclerosis in progeria. Cardiovasc Pathol 8:29–39.

Steinle NI, Kazlauskaite R, Imumorin IG, Hsueh WC, PollinTI, O’Connell JR, Mitchell BD, Shuldiner AR. 2004.Variation in the lamin A/C gene: Associations with meta-bolic syndrome. Arterioscler Thromb Vasc Biol 24:1708–1713.

Stierle V, Couprie J, Ostlund C, Krimm I, Zinn-Justin S,Hossenlopp P, Worman HJ, Courvalin JC, Duband-Gou-let I. 2003. The carboxyl-terminal region common tolamins A and C contains a DNA binding domain. Bio-chemistry 42: 4819–4828.

Sullivan T, Escalante-Alcalde D, Bhatt H, Anver M, Bhat N,Nagashima K, Stewart CL, Burke B. 1999. Loss of A-typelamin expression compromises nuclear envelope integ-rity leading to muscular dystrophy. J Cell Biol 147:913–920.

Taylor MR, Fain PR, Sinagra G, Robinson ML, RobertsonAD, Carniel E, Di Lenarda A, Bohlmeyer TJ, FergusonDA, Brodsky GL, et al. 2003. Natural history of dilatedcardiomyopathy due to lamin A/C gene mutations.J Am Coll Cardiol 41: 771–780.

Taylor MR, Slavov D, Gajewski A, Vlcek S, Ku L, Fain PR,Carniel E, Di Lenarda A, Sinagra G, Boucek MM, et al.Familial Cardiomyopathy Registry Research Group.2005. Thymopoietin (lamina-associated polypeptide 2)gene mutation associated with dilated cardiomyopathy.Hum Mutat 26: 566–574.

Tazir M, Azzedine H, Assami S, Sindou P, Nouioua S,Zemmouri R, Hamadouche T, Chaouch M, Feingold J,

Vallat JM, et al. 2004. Phenotypic variability in autosomalrecessive axonal Charcot-Marie-Tooth disease due to theR298C mutation in lamin A/C. Brain 127: 154–163.

Toth JI, Yang SH, Qiao X, Beigneux AP, Gelb MH, MoulsonCL, Miner JH, Young SG, Fong LG. 2005. Blocking pro-tein farnesyl transferase improves nuclear shape in fibro-blasts from humans with progeroid syndromes. Proc NatlAcad Sci 102: 12873–12878.

Towbin JA, Bowles NE. 2002. The failing heart. Nature 415:227–233.

Tullio-Pelet A, Salomon R, Hadj-Rabia S, Mugnier C, deLaet MH, Chaouachi B, Bakiri F, Brottier P, Cattolico L,Penet C, et al. 2000. Mutant WD-repeat protein intriple-A syndrome. Nat Genet 26: 332–335.

van Berlo JH, de Voogt WG, van der Kooi AJ, van TintelenJP, Bonne G, Yaou RB, Duboc D, Rossenbacker T, Heid-buchel H, de Visser M, et al. 2005. Meta-analysis of clin-ical characteristics of 299 carriers of LMNA genemutations: Do lamin A/C mutations portend a highrisk of sudden death? J Mol Med 83: 79–83.

van Tintelen JP, Hofstra RM, Katerberg H, Rossenbacker T,Wiesfeld AC, du Marchie Sarvaas GJ, Wilde AA, van Lan-gen IM, Nannenberg EA, van der Kooi AJ, et al. 2007.High yield of LMNA mutations in patients with di-lated cardiomyopathy and/or conduction disease re-ferred to cardiogenetics outpatient clinics. Am Heart J154: 1130–1139.

Varela I, Pereira S, Ugalde AP, Navarro CL, Suarez MF, Cau P,Cadinanos J, Osorio FG, Foray N, Cobo J, et al. 2008.Combined treatment with statins and aminobisphospho-nates extends longevity in a mouse model of humanpremature aging. Nat Med 14: 767–772.

Verstraeten VL, Broers JLV, Steensel MAM, Zinn-Justin S,Ramaekers FCS, Steijlen PM, Kamps M, Kuijpers HJH,Merckx D, Smeets HJM, et al. 2006. Compound hetero-zygosity for mutations in LMNA causes a progeria syn-drome without prelamin A accumulation. Hum MolGenet 15: 2509–2522.

Verstraeten VL, Ji JY, Cummings KS, Lee RT, Lammerding J.2008. Increased mechanosensitivity and nuclear stiffnessin Hutchinson-Gilford progeria cells: Effects of farnesyl-transferase inhibitors. Aging Cell 7: 383–393.

Vigouroux C, Capeau J. 2005. A-type lamin-linked lipody-strophies. Novartis Found Symp 264: 166–177.

Vigouroux C, Magre J, Vantyghem MC, Bourut C, LascolsO, Shackleton S, Lloyd DJ, Guerci B, Padova G, ValensiP, et al. 2000. Lamin A/C gene: Sex-determined expres-sion of mutations in Dunnigan-type familial partial lip-odystrophy and absence of coding mutations incongenital and acquired generalized lipoatrophy. Diabe-tes 49: 1958–1962.

Wang Y, Herron AJ, Worman HJ. 2006. Pathology and nu-clear abnormalities in hearts of transgenic mice express-ing M371K lamin A encoded by an LMNA mutationcausing Emery-Dreifuss muscular dystrophy. Hum MolGenet 15: 2479–2489.

Wang Y, Panteleyev AA, Owens DM, Djabali K, Stewart CL,Worman HJ. 2008. Epidermal expression of the truncatedprelamin A causing Hutchinson-Gilford progeria syn-drome: Effects on keratinocytes, hair and skin. HumMol Genet 17: 2357–2369.

H.J. Worman, C. Ostlund, and Y. Wang

16 Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 17: Diseases of the Nuclear Envelope

Waterham HR, Koster J, Mooyer P, Noort Gv G, Kelley RI,Wilcox WR, Wanders RJ, Hennekam RC, OosterwijkJC. 2003. Autosomal recessive HEM/Greenberg skeletaldysplasia is caused by 3 b-hydroxysterol d14-reductasedeficiency due to mutations in the lamin B receptorgene. Am J Hum Genet 72: 1013–1017.

Weber K, Plessmann U, Traub P. 1989. Maturation of nuclearlamin A involves a specific carboxy-terminal trimming,which removes the polyisoprenylation site from the pre-cursor; implications for the structure of the nuclear lam-ina. FEBS Lett 257: 411–414.

Wegner L, Andersen G, Sparsø T, Grarup N, Glumer C,Borch-Johnsen K, Jørgensen T, Hansen T, Pedersen O.2007. Common variation in LMNA increases susceptibil-ity to type 2 diabetes and associates with elevated fastingglycemia and estimates of body fat and height in the gen-eral population: Studies of 7,495 Danish whites. Diabetes56: 694–698.

Wolf CM, Wang L, Alcalai R, Pizard A, Burgon PG,Ahmad F, Sherwood M, Branco DM, Wakimoto H,Fishman GI, et al. 2008. Lamin A/C haploinsuffi-ciency causes dilated cardiomyopathy and apoptosis-triggered cardiac conduction system disease. J MolCell Cardiol 44: 293–303.

Worman HJ, Evans CD, Blobel G. 1990. The lamin B recep-tor of the nuclear envelope inner membrane: A polytopicprotein with eight potential transmembrane domains. JCell Biol 111: 1535–1542.

Worman HJ, Yuan J, Blobel G, Georgatos SD. 1988. A laminB receptor in the nuclear envelope. Proc Natl Acad Sci 85:8531–8534.

Wydner KL, McNeil JA, Lin F, Worman HJ, Lawrence JB.1996. Chromosomal assignment of human nuclear enve-lope protein genes LMNA, LMNB1, and LBR by fluores-cence in situ hybridization. Genomics 32: 474–478.

Yang SH, Andres DA, Spielmann HP, Young SG, Fong LG.2008. Progerin elicits disease phenotypes of progeria inmice whether or not it is farnesylated. J Clin Invest 118:3291–3300.

Yang SH, Bergo MO, Toth JI, Qiao X, Hu Y, Sandoval S, MetaM, Bendale P, Gelb MH, Young SG, et al. 2005. Blockingprotein farnesyltransferase improves nuclear blebbing inmouse fibroblasts with a targeted Hutchinson–Gilfordprogeria syndrome mutation. Proc Natl Acad Sci 102:10291–10296.

Yang SH, Meta M, Qiao X, Frost D, Bauch J, Coffinier C,Majumdar S, Bergo MO, Young SG, Fong LG. 2006. A far-nesyltransferase inhibitor improves disease phenotypesin mice with a Hutchinson-Gilford progeria syndromemutation. J Clin Invest 116: 2115–2121.

Ye Q, Worman HJ. 1994. Primary structure analysis and lam-in B and DNA binding of human LBR, an integral proteinof the nuclear envelope inner membrane. J Biol Chem269: 11306–11311.

Ye Q, Worman HJ. 1996. Interaction between an integralprotein of the nuclear envelope inner membrane and hu-man chromodomain proteins homologous to DrosophilaHP1. J Biol Chem 271: 14653–14656.

Young SG, Fong LG, Michaelis S. 2005. Prelamin A,Zmpste24, misshapen cell nuclei, and progeria—Newevidence suggesting that protein farnesylation could beimportant for disease pathogenesis. J Lipid Res 46:2531–2558.

Zhang FL, Casey PJ. 1996. Protein prenylation: Molecularmechanisms and functional consequences. Annu RevBiochem 65: 241–269.

Zhang X, Chen S, Yoo S, Chakrabarti S, Zhang T, Ke T,Oberti C, Yong SL, Fang F, Li L, et al. 2008. Mutationin nuclear pore component NUP155 leads to atrial fibri-llation and early sudden cardiac death. Cell 135:1017–1027.

Laminopathies and Aging

Cite this article as Cold Spring Harb Perspect Biol 2010;2:a000760 17

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 18: Diseases of the Nuclear Envelope

2010; doi: 10.1101/cshperspect.a000760Cold Spring Harb Perspect Biol  Howard J. Worman, Cecilia Östlund and Yuexia Wang Diseases of the Nuclear Envelope

Subject Collection The Nucleus

Transcription Factor DynamicsFeiyue Lu and Timothée Lionnet Expression

The Stochastic Genome and Its Role in Gene

Christopher H. Bohrer and Daniel R. Larson

Organization: Their Interplay and Open QuestionsMechanisms of Chromosome Folding and Nuclear

Leonid Mirny and Job DekkerMembrane ProteinsThe Diverse Cellular Functions of Inner Nuclear

Sumit Pawar and Ulrike Kutay

Organization Relative to Nuclear ArchitectureNuclear Compartments, Bodies, and Genome Nuclear Compartments: An Incomplete Primer to

Andrew S. Belmont

The Nuclear Lamina

Karen L. ReddyXianrong Wong, Ashley J. Melendez-Perez and

OrganizationEssential Roles for RNA in Shaping Nuclear

Sofia A. Quinodoz and Mitchell Guttman3D Chromatin ModelingUncovering the Principles of Genome Folding by

et al.Asli Yildirim, Lorenzo Boninsegna, Yuxiang Zhan,

DevelopmentEpigenetic Reprogramming in Early Animal

Zhenhai Du, Ke Zhang and Wei Xie

Viruses in the NucleusBojana Lucic, Ines J. de Castro and Marina Lusic

Eukaryotic GenomeNuclear Pore Complexes: The Gatekeepers of the Structure, Maintenance, and Regulation of

Marcela Raices and Maximiliano A. D'Angelo

X-Chromosome InactivationThe Molecular and Nuclear Dynamics of

François Dossin and Edith Heard

Development3D or Not 3D: Shaping the Genome during

Juliane Glaser and Stefan MundlosOutput of the GenomeThe Impact of Space and Time on the Functional

al.Marcelo Nollmann, Isma Bennabi, Markus Götz, et

Mammalian DNA Replication TimingAthanasios E. Vouzas and David M. Gilbert

Chromatin Mechanisms Driving Cancer

al.Berkley Gryder, Peter C. Scacheri, Thomas Ried, et

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

Copyright © 2010 Cold Spring Harbor Laboratory Press; all rights reserved

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Page 19: Diseases of the Nuclear Envelope

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

Copyright © 2010 Cold Spring Harbor Laboratory Press; all rights reserved

on October 28, 2021 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from