Article

What Should the Cardiologist know about Lamin Disease?

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

Lamins are intermediate filament proteins able to polymerise and form an organised meshwork underlying the inner nuclear membrane in most differentiated somatic cells. Mutations in the LMNA gene, which encodes the two major lamin A and C isoforms, cause a diverse range of diseases, called laminopathies, including dilated cardiomyopathy, associated with a poor prognosis and high rate of sudden death due to conduction defect and early ventricular arrhythmia. Identification of mutations in LMNA gene in clinical practice is rapidly increasing, as well as comprehensive cardiac and genetic family screening. As a consequence, cardiologists are more and more frequently faced to difficult questions regarding optimal management of patients and relatives, especially timing for prophylactic cardioverter defibrillator. This review focuses on recent data useful for the clinician, as well as therapeutic perspectives both in human and animal models.

Disclosure:The authors have no conflicts of interest to declare.

Received:

Accepted:

Acknowledgements:Supported by research grants from EU (FP6 Euro-laminopathies grant number 018690 & FP7, INHERITANCE grant number 291924), Assistance Publique – Hôpitaux de Paris (PHRC AOM04141), the “Fondation LEDUCQ” (Genomic, epigenomic and systems dissection of mechanisms underlying dilated cardiomyopathy), the CONNY-MAEVA charitable foundation and the Association Française contre les Myopathies. The authors would like to thank Marie-Elodie Cattin for fruitful discussion and her kind help with the figure preparation.

Correspondence Details:Phillipe Charon, Centre de référence maladies cardiaques héréditaires, Hôpital Pitié-Salpêtrière, 47 boulevard de l'Hôpital, 75013 Paris, France. E: philippe.charron@psl.aphp.fr

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Most mutations of the LMNA gene affect the heart, causing a dilated cardiomyopathy, ususally with conduction defect and ventricular arrhythmia, with or without skeletal muscle involvement. Although a relatively rare disease, cardiologists should be aware of laminopathies (diseases caused by LMNA gene mutations) because of the particularly aggressive course compared with most other cardiomyopathies, and because of the benefit of early defibrillator and pace maker implantation.

What are Lamin Proteins?

Lamins are type V intermediate filament proteins that are able to polymerise and form the nuclear lamina, an organised meshwork that lies between the inner nuclear membrane and the chromatin (see Figure 1).1–6 A-type lamins, i.e. the lamin A and the lamin C, together with B-type lamins, are the constituents of the nuclear lamina and lamin A and C isoforms are both encoded by the LMNA gene via alternative splicing (see Figure 2). The LMNA gene is localised to chromosome 1q21.2-q21.3, spans approximatively 24 kb and is composed of 12 exons that encode four lamin isoforms (A, AΔ10, C and C2). The two major isoforms lamin A and C are identical for their first 566 amino acids but differ by their C-terminal domains.7 Lamin A is initially synthesised as a precursor, prelamin A, with 98 unique C-terminal amino acids. Prelamin A is farnesylated on the cysteine residue of a C-terminal CaaX box and then is endoproteolytically processed by the ZMPSTE24 protease (zinc metalloprotease Ste24 homologue) removing the last 18 amino acids to yield the mature lamin A (74 kDa). Lamin C (65 kDa) has six unique C-terminal amino acids and is not post-translationnaly modified by farnesylation. Lamin A and lamin C (henceforth referred to as lamin A/C) are expressed in terminally differentiated somatic cells but are lacking from early embryos. In contrast, B-type lamins that are encoded by different genes (LMNB1 and LMNB2) are also present in undifferentiated cells.

The central rod domain of lamins is a highly conserved alpha-helical core of approximatively 360 residues that drives the interactions between two lamin protein chains to form a coiled-coil dimer. Lamins A/C further assemble to form head-to-tail polymers that, together with B-type lamins, constitute the nuclear lamina. One of the functions of the lamina is to provide structural support to the nucleus and maintaining the mechanical integrity of cells by linking the nucleoskeleton to the cytoskeleton.8 Other studies also support a complex role of lamins in nuclear pore function, chromatin organisation, DNA replication and transcriptional regulation9,10 (see Figure 1).

Lamin A/C Mutations and Overview of the Various Phenotypes

More than 450 different mutations have been identified in the LMNA gene and they can cause a wide variety of distinct and disparate diseases involving striated muscle (dilated cardiomyopathy, skeletal myopathies), adipose tissue (lipodystrophy syndromes), peripheral nerve (Charcot-Marie-Tooth neuropathy) or multiple systems with accelerated ageing (progerias).

Figure 1: Schematic Presentation of Lamins Interactions

Article image

Laminopathies of the Striated Muscle

The laminopathy story began in 1999 when we identified the first mutation of the LMNA gene,11 in patients affected with autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD), characterised by a triad of early tendon contractures (elbows, Achille’s tendons, spine), muscle weakness/wasting with a predominantly humero-peroneal distribution, and dilated cardiomyopathy/conduction defect. Subsequently, LMNA mutations were identified in patients with dilated cardiomyopathy and conduction defect (DCM-CD), similar to EDMD, but with minimal or no skeletal muscle involvement.12 Shortly afterwards, LMNA mutations were reported in patients with limb-girdle muscular dystrophy type 1B (LGMD1B),13 which share the cardiac features of EDMD but muscle weakness/wasting predominantly involving pelvic and scapular girdle muscles, and no or mild tendon contractures. More recently, LMNA mutations were identified in congenital forms of muscular dystrophy (L-CMD) with onset before two years of age and evoluation towards severe respiratory insufficiency.14

Laminopathies of Other Tissues

The main entity of adipose tissue laminopathies is familial partial lipodystrophy of Dunnigan type (FPLD) characterised by abnormal distribution of subcutaneous fat (loss at extremities and accumulation in neck and face), metabolic syndrome with insulin-resistance, hypertriglyceridaemia and sometimes type 2 diabetes. Peripheral nerve laminopathy is related to a recessive form of Charcot-Marie-Tooth axonal neuropathy, characterised by distal muscle wasting/weakness and absence of osteotendinous reflexes due to axonal degeneration. Accelerated ageing laminopathies are mainly represented by Hutchinson-Gilford progeria syndrome, an extremely rare disorder with segmental premature ageing sparing the brain, with subjects dying at a mean age of 13 years from cardiovascular disease (atherosclerosis). Other rare or overlapping syndromes have been linked to LMNA mutations including mandibuloacral dysplasia (MAD), atypical Werner syndrome and restrictive dermopathy.15

Laminopathies and Genetic Heterogeneity

Apart from the large phenotypic pleiotropy there is also a large genetic variability with more than 450 different mutations identified in the LMNA gene, all published ones being available within the UMD-LMNA mutation database at www.umd.be/LMNA. All types of mutations are reported:16 missense mutation that is the most frequent mechanism (72 % of the 301 first published LMNA mutations), in-frame insertion/deletion (9 %), out-of-frame insertion/deletion (9 %), splice-site (7 %) and nonsense (5 %) mutations. Genotype-phenotype relationships are incompletely understood. However mutations that affect tissues other than striated muscle are usually related to specific amino-acid residues or specific exons (see Figure 2). By contrast, mutations related to striated muscle (68 % are missense mutations) are distributed all along the gene without clear relationships or hot spot for the various cardiac/skeletal clinical entities. Moreover, the three cardiac/skeletal clinical entities (EDMD, DCM-CD, LGMD1B) may co-exist within a same family.17,18 Interestingly, in line with previous reports,19 UMD-LMNA database analysis of patients with cardiac phenotype revealed that 33 % of the patients presenting isolated cardiac disease carry mutations leading to truncated proteins (nonsense, out-of-frame ins/del, splice-site) whereas only 8 % of the patients with cardiac and skeletal defects carry this type of mutations.

Figure 2: Distribution of LMNA Mutations in Prelamin A and Lamin C16

Article image

Cardiac Manifestations of Laminopathies

Cardiac Clinical Entities

Apart from the three main diseases initially described with LMNA mutation and cardiac expression, Emery-Dreifuss muscular dystrophy (EDMD), dilated cardiomyopathy and conduction defect (DCM-CD), limb-girdle muscular dystrophytype 1B (LGMD1B), additional entities were subsequently associated with the gene. These very few reports are related to DCM and early atrial fibrillation,20,21 left apical aneurysm without conduction defect,22 left ventricular non compaction,23 DCM and a quadriceps cardiomyopathy,24 inherited form of early onset myocardial fibrosis25 and arrhythmogenic right ventricular cardiomyopathy.26 Overlapping phenotypes have been also described as well as distinct phenotypes within a given family,17,18,21,24,27–29 so that it might be more appropriate to consider the global cardiac expression of laminopathies with its main features (dilated cardiomyopathy, conduction defect and ventricular arrhythmia), with or without skeletal muscle involvement.

Inheritance and Prevalence of LMNA Mutations

Mode of inheritance of cardiac laminopathies is autosomal dominant (50 % risk of transmission to offspring). Penetrance, or percentage of cardiac expression in mutation carriers, is not fully evaluated but appears very high and was estimated to be 100 % by the age of 60 years in one study.30 Exceptional studies reported on homozygous patients or digenism, associated with very early and severe phenotype.31–34 LMNA is one of the most frequent genes involved in dilated cardiomyopathy. In the largest study of 324 patients with DCM, prevalence of LMNA mutation was 7.5 % in familial cases and 3.6 % in sporadic cases, although the significance of some variants was unclear (in the absence of segregation in the family).35 No clinical criteria can distinguish DCM related to LMNA gene from other genetic or non genetic causes. However some clinical predictors of LMNA mutation were suggested in DCM patients: presence of skeletal muscle involvement, supraventricular arrhythmia, conduction defect, mildly dilated left ventricle, regardless of family history.36,37 Indeed, prevalence of LMNA mutations was increased to ~30 % in patients with DCM and conduction defect,37,38 but was very rare in patients with isolated DCM21,37 or isolated atrial fibrillation.39 Serum creatine kinase is increased in only part of mutation carriers (<30 %), with mild elevation (usually two-fold normal value) and is not considered as a strong predictor of LMNA mutation.12,17,36,37

Cardiac Complications

The main cardiac features were described in the princeps study about DCM and conduction defect related to LMNA mutations.12 Out of 39 patients with cardiac disease (mean age at onset 38 years, range 19 to 53) from five families, 34 patients (87 %) had atrioventricular block (AVC) or sinus node dysfunction, 23 (59 %) had atrial fibrillation or flutter and 25 (64 %) had DCM (heart transplant in six). Of note, 21 patients (54 %) were implanted with pace maker because of significant conduction defect. In this study, 20 additional relatives were mutation carriers but without cardiac abnormalities, all were younger than 30 years. Clinical features were then compiled in a meta-analysis of 299 mutations carriers from families with DCM, EDMD or LGMD1B.40 Dysrhythmia (conduction defect, supra-ventricular or ventricular arrhythmia) occurred early in life (2 children <10 years) and were highly penetrant: 74 % in those aged 20 to 30 years then 92 % of patients aged over 30 years (see Figure 3). Typical initial ECG shows a low P wave amplitude, PR interval prolongation and a normal QRS duration. Pace maker was implanted in 3 % of patients at age 10 to 20 and then increased to 44 % of patients after 30 years of age. Heart failure was reported at a later age, in 10 % in patients <30 years of age and increased progressively to 64 % of patients over 50 years. Ventricular arrhythmia was suggested as quite frequent in this meta-analysis since nearly half of sudden deaths (16 patients or 46 %) occurred in patients with a pace maker.40 Subsequent small studies or case reports observed that ventricular arrhythmia or appropriate defibrillator therapy may occur before myocardial dysfunction/DCM41,42 and sometimes as the first cardiac manifestation before conduction defect.43,44 One particular LMNA mutation (c.908- 909delCT) was suggested to be associated with a rapid progression of conduction defect and early sudden death.27

Cardiac Death

Clinical course of laminopathies is characterised by a poor prognosis and a high rate of major cardiac events. In a cohort of 105 DCM patients, cumulated survival was significantly worse in LMNA mutation carriers as compared to non carriers.36 By the age of 45 years, 55 % of mutation carriers had cardiovascular death or heart transplant as compared to 11 % in non carriers (p=0.0001 for global cumulated survival comparison). In the meta-analysis of 299 LMNA mutation carriers, cardiac death was observed in 76 patients (mean age 46 years) and sudden death was more prevalent than heart failure death (46 % of cardiac deaths versus 12 % respectively).40 Interestingly sudden death was similar in patients with isolated cardiac phenotype and in patients with cardiac and skeletal muscle phenotype.40 Recently, we reported a multicentre study that carefully examined the prognosis of 269 European LMNA mutation carriers during a median follow-up (FU) of 43 months.45 This retrospective study confirmed the high risk of ventricular arrhythmia: 18 % of patients developed sudden death, resuscitation or appropriate defibrillator therapy. In patients with defibrillator implanted at baseline, rate of appropriate therapy during FU was 13 % per year in secondary prevention and 8 % per year in primary prevention. Altogether, cardiac death due to sudden death was however lower than cardiac death due to heart failure (31 versus 47 % respectively).45

Prediction of Cardiac Death

Few studies examined the predictive role of cardiac and non-cardiac features on the prognosis of mutation carriers. Our monocentric study of 94 Italian mutation carriers during a FU of 57 months identified two independent risk factors for total cardiac events: NHYA class III to IV and highly dynamic competitive sports for ≥10 years.30 In the same study, the type of mutation within the LMNA gene (splice site mutations) and history of competitive sports were independent risk factors for sudden cardiac death (SCD). In a study of 19 mutation carriers with conduction defect requiring a pace maker, but normal left ventricle ejection fraction (LVEF), cardioverter defibrillator was implanted systematically and eight patients (42 %) received appropriate defibrillator therapy during a FU of 34 months, suggesting defibrillator implantation in the presence of significant conduction defect.42 However significant conduction defect was not found to be a risk factor in two larger cohorts of 94 and 269 patients.30,45 Electrophysiological examination and inducible ventricular tachycardia was not a risk factor for SCD in the study of 19 patients.42 Recently, the study of 269 mutation carriers specifically focussed on the prediction of SCD.45 We identified four independent and cumulative risk factors for malignant ventricular arrhythmia (SCD, resuscitation, appropriate defibrillator therapy) (see Figure 4): LVEF <45 % at the first clinical contact, non-sustained ventricular tachycardia, male gender and a specific mechanism of LMNA mutation (non missense mutation: ins-del/truncating or mutations affecting splicing). No malignant ventricular arrhythmia occurred in patients with zero or one risk factor so that defibrillator implantation is suggested in the presence of at least two of these risk factors.45

Figure 3: Age-dependent Penetrance of Major Cardiac Event in 299 LMNA Mutation Carriers40

Article image

Pathophysiology

Understanding of how mutations in a gene, LMNA, encoding ubiquitously expressed proteins, the lamin A/C, could give rise to defects specifically affecting the myocardium, remain so far an unsolved challenge. Numerous studies have tried to solve this mechanistic puzzle using tissues and/or cells available from mutated patients or various cellular and animal models that have been created in an attempt to phenocopy the human disease context.46

Lamin A/C expression explored in various explanted heart tissues from patients carrying nonsense LMNA mutations revealed reduced lamin A/C level, i.e. haplo-insufficiency in the cardiomyocyte nuclei.17,37 This is due to either the degradation of the mutated mRNA carrying a premature stop codon via the non sense mediated decay pathway or to the degradation of the corresponding truncated lamin A/C by the proteasome.47 As for missense mutations, the lamin A/C expression levels may vary from normal to reduced, underlying in those cases dominant negative effect of the mutant protein.37 Recently, we analyzed the quantitative gene expression of LMNA in 311 patients with DCM and observed that LMNA mRNA expression from peripheral blood, and myocardium, was reduced in DCM patients with LMNA mutation (p<0.001).48 Therefore, reduced mRNA expression level in blood might be a novel potential biomarker predictive for cardiac laminopathies. Ultrastructure analyses of explanted hearts carrying different mutations revealed focal disruptions and bleb formation of the cardiomyocyte nuclear membranes.37 These structural nuclear abnormalities are associated with altered chromosomal positioning and abnormal gene expression.49 The pathogenicity of several missense LMNA variants has been explored and desmonstrated abnormal intranuclear aggregates when overexpressed in heterologous context such as Hela cells50 or C2C12 myoblasts.39

Figure 3: Age-dependent Penetrance of Major Cardiac Event in 299 LMNA Mutation Carriers40

Article image

To go further into the pathophysiological mechanisms, numerous murine models have been created.46 Mice lacking lamin A/C (Lmna-/-) display muscular dystrophy, dilated cardiomyopathy, signs of axonal neuropathy, reduction of adipose tissue and die by eight weeks of age.51–53 The heterozygous Lmna+/- mice, expressing 50 % of lamin A/C, develop arrhythymias and conduction defects by 10 weeks of age, in link with apoptosis of the conduction tissue, develop mild ventricle dilation by one year of age and 20 % of them die after eight months of age.6,54 Investigation of mechanotransduction pathways revealed abnormal desmin network and defective force transmission

in both Lmna-/- and Lmna+/- mice, associated in the latter with reduced hypertophic response after transaortic contstriction.52,55 The KI-LmnaH222P, knock-in mice reproducing the LMNA p.His222Pro mutation identified in EDMD patients, developed muscular dystrophy and DCM-CD similar to the human condition,56 at the homozygous state. Microarray analysis of cardiac gene expression performed at the incipience of the cardiac defects, desmonstrated abnormal activation of various branchs of the MAP kinase cascade (ERK1/2, JNK, p38α) and of AKT signalling pathway, linking the mutation to contractile dysfunction and myocardial fibrosis.57–59 Another knock-in model was created to study DCM-CD, the KI-LmnaN195K, reproducing a DCM-CD related LMNA mutation. Homozygous mice developed DCM-CD with minimal or no muscular dystrophy and died at three months due to arrhythmia. The transcription factor Hf1b/Sp4 and the connexin 40 and 43 were misexpressed/mislocalised in mutant hearts. As for knock-out mice, desmin displayed abnormal organisation.60

Altogether, these observations suggest that LMNA mutations may cause cardiomyopathy by haploinsufficiency and/or dominant negative effect, with disrupting the internal organisation of the cardiomyocyte and/or altering the gene expression of transciptional factors and proteins involved in various signalling pathways, all essential to normal cardiac development, ageing and function.

Perspective for Therapeutics

Even if the pathophysiology of cardiac laminopathies remains to be fully elucidated, the analyses of the mouse models that phenocopy the human disease allowed to test several therapeutical approaches. The first approach tested has been pharmacological. Inhibitors of different branchs of the MAPK and AKT pathways in the KI-LmnaH222P mice not only delay the onset but also slow down the progression of the cardiac disease.57,61–64 As some MAPK inhibitors have been tested in humans during clinical trials for other indications, their efficacy and safety in laminopathy deserved further investigations. Calcium sensitiser also gives beneficial effects on the contractile function and lead to increased survival of the mice.65

Another possible approach is gene therapy. Indeed, combination of stem cell-based approaches with gene-editing technologies has been recently reported for laminopathy and represents an attractive therapeutical strategy.66 Homologous recombination-based gene correction of multiple LMNA mutations using helper-dependent adenoviral vectors (HDAdVs) demonstrated a highly efficient and safe method for correcting mutations in human induced pluripotent stem cells (hIPSC).66 The fast advancing field of hIPSC67 will certainly lead to innovative therapeutic approaches in the near future.

Finally, improvement in therapeutic management might come from very early treatment with drugs already used in human in heart failure. We designed the Pre-clinical mutation carriers from families with dilated cardiomyopathy and ACE inhibitors (PRECARDIA) trial that is a multicentre randomised double blind trial (perindopril versus placebo) proposed to participants without significant systolic dysfunction but who are mutation carriers from families with dilated cardiomyopathy (whatever the underlying gene) in order to delay or prevent the systolic dysfunction. The enrollment of participants is underway.68

Key Messages for the Cardiologist

Identification of mutations in LMNA gene in clinical practice is rapidly increasing so that cardiologists are more and more frequently faced to difficult questions regarding optimal management of patients and relatives. The following section briefly summarises the management we propose, that reflects our personal view and is based on available data and our experience.

When Should the Cardiologist Suspect a Laminopathy?

The diagnosis should be suspected in a patient with DCM and conduction defect (atrioventricular block or sinus node dysfunction) or DCM and skeletal muscle abnormality (muscle weakness/wasting, tendon contractures, increased creatine kinase level) or DCM preceded (few years before) by surpa-ventricular or ventricular arrhythmia, whatever the familial context (familial or sporadic form).

How Can we Confirm a Laminopathy?

The diagnosis, when suspected because of the above criteria, should be confirmed by genetic testing with the analysis of the LMNA gene. When conventional LMNA direct sequence analysis does not identify a mutation in suggestive pedigrees, alternative strategies such as multiplex ligation-dependent probe amplification might be discussed to detect large gene rearrangements.69 Differential diagnoses include other genes such as SCN5A, desmin, DMPK (Steinert), dystrophin and desmosomal genes.

What is the Clinical Course of a Laminopathy?

The disease is associated with a poor prognosis, related to heart failure and to sudden cardiac death (caused by conduction defect or ventricular arrhythmia). First cardiac expression includes conduction defect (AVB type 1) or supra-ventricular arrhythmia, ususally between 20 and 30 years of age. DCM is common between 30 and 50 years of age. Ventricular arrhythmia can occur at various stage of the disease. Skeletal muscle weakness or wasting can be absent or can occur at a late stage. A subject can develop only part of the features and the cardiac expression or chronology may be different among relatives within a given family.

What is the Screening that can be Proposed to the Patients and the Relatives?

The diagnosis in a patient should lead to regular cardiac examination including electrocardiogram (ECG), echocardiography, Holter-ECG and exercise testing. Skeletal muscle examination and creatine kinase dosage are also warranted. Cardiac examination, or predictive genetic testing, is proposed to all first degree-relatives within the family (from 10–12 years of age).70

What is the Therapeutic that should Currently be Proposed?

Competitive sport should be discouraged in all LMNA mutation carriers, whatever the stage of the disease and the presence of cardiac abnormalities. Myocardial systolic dysfunction, supra-ventricular arrhythmia and conduction defect should be regularly screened for and lead to non-specific management (no specific indication for pace maker implantation for example) except careful use of drugs with negative chronotropic effect in the absence of a pace maker. In contrast, there is a high and early risk of ventricular arrhythmia. Cardiovertor defibrillator can be proposed in the presence of two criteria among the four following: LVEF <45 %, non sustained ventricular tachycardia, male gender and non-missense LMNA mutation. If only male gender and non-missense LMNA mutation are present, the situation is however not sufficient to propose a defibrillator. Although based on lesser evidence, it seems also possible to propose defibrillator implantation in the presence of significant conduction defect that requires a pace maker implantation.

References

  1. Broers J, Ramaekers F, Bonne G, et al. The nuclear lamins: laminopathies and their role in premature ageing. Physiological Reviews 2006;86:967–1008.
    Crossref | PubMed
  2. Capell BC, Collins FS. Human laminopathies: nuclei gone genetically awry. Nat Rev Genet 2006;7:940–52.
    Crossref | PubMed
  3. Davies BS, Fong LG, Yang SH, et al. The posttranslational processing of prelamin A and disease. Annu Rev Genomics Hum Genet 2009;10:153–74.
    Crossref | PubMed
  4. Kennedy BK, Barbie DA, Classon M, et al. Nuclear organization of DNA replication in primary mammalian cells. Genes Dev 2000;14:2855–68.
    Crossref | PubMed
  5. Wilson KL, Foisner R. Lamin-binding Proteins. Cold Spring Harb Perspect Biol 2010;2:a000554.
    Crossref | PubMed
  6. Wolf CM, Wang L, Alcalai R, et al. Lamin A/C haploinsufficiency causes dilated cardiomyopathy and apoptosis-triggered cardiac conduction system disease. J Mol Cell Cardiol 2008;44:293–303.
    Crossref | PubMed
  7. Lin F, Worman HJ. Structural organization of the human gene encoding nuclear lamin A and nuclear lamin C. J Biol Chem 1993;268:16321–6.
    PubMed
  8. Crisp M, Liu Q, Roux K, et al. Coupling of the nucleus and cytoplasm: role of the LINC complex. J Cell Biol 2006;172:41–53.
    Crossref | PubMed
  9. Dechat T, Pfleghaar K, Sengupta K, et al. Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. Genes Dev 2008;22:832–53.
    Crossref | PubMed
  10. Vlcek S, Dechat T, Foisner R. Nuclear envelope and nuclear matrix: interactions and dynamics. Cell Mol Life Sci 2001;58:1758–65.
    Crossref | PubMed
  11. Bonne G, Di Barletta MR, Varnous S, et al. Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy. Nature Genet 1999;21:285–8.
    Crossref | PubMed
  12. Fatkin D, MacRae C, Sasaki T, et al. Missense mutations in the rod domain of tle lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease. N Engl J Med 1999;341:1715–24.
    Crossref | PubMed
  13. Muchir A, Bonne G, van der Kooi AJ, et al. Identification of mutations in the gene encoding lamins A/C in autosomal dominant limb girdle muscular dystrophy with atrioventricular conduction disturbances (LGMD1B). Hum Mol Genet 2000;9:1453–9.
    Crossref | PubMed
  14. Quijano-Roy S, Mbieleu B, Bonnemann CG, et al. De novo lmna mutations cause a new form of congenital muscular dystrophy. Ann Neurol 2008;64:177–86.
    Crossref | PubMed
  15. Worman HJ, Bonne G. "Laminopathies": a wide spectrum of human diseases. Exp Cell Res 2007;313:2121–33.
    Crossref | PubMed
  16. Bertrand AT, Chikhaoui K, Ben Yaou RB, Bonne G. Clinical and genetic heterogeneity in laminopathies. Biochem Soc Trans 2011;39:1687–92.
    Crossref | PubMed
  17. Bécane H-M, Bonne G, Varnous S, et al. High incidence of sudden death with conduction system and myocardial disease due to lamins A and C gene mutation. Pacing Clin Electrophysiol 2000;23:1661–6.
    Crossref | PubMed
  18. Brodsky GL, Muntoni F, Miocic S, et al. Lamin A/C gene mutation associated with dilated cardiomyopathy with variable skeletal muscle involvement. Circulation 2000;101:473–6.
    Crossref | PubMed
  19. Benedetti S, Menditto I, Degano M, et al. Phenotypic clustering of lamin A/C mutations in neuromuscular patients. Neurology 2007;69:1285–92.
    Crossref | PubMed
  20. Otomo J, Kure S, Shiba T, et al. Electrophysiological and histopathological characteristics of progressive atrioventricular block accompanied by familial dilated cardiomyopathy caused by a novel mutation of lamin A/C gene. J Cardiovasc Electrophysiol 2005;16:137–45.
    Crossref | PubMed
  21. Sebillon P, Bouchier C, Bidot LD, et al. Expanding the phenotype of LMNA mutations in dilated cardiomyopathy and functional consequences of these mutations. J Med Genet 2003;40:560–7.
    Crossref | PubMed
  22. Forissier JF, Bonne G, Bouchier C, et al. Apical left ventricular aneurysm without atrio-ventricular block due to a lamin A/C gene mutation. Eur J Heart Fail 2003;5:821–5.
    Crossref | PubMed
  23. Hermida-Prieto M, Monserrat L, Castro-Beiras A, et al. Familial dilated cardiomyopathy and isolated left ventricular noncompaction associated with lamin A/C gene mutations. Am J Cardiol 2004;94:50–4.
    Crossref | PubMed
  24. Charniot JC, Pascal C, Bouchier C, et al. Functional consequences of an LMNA mutation associated with a new cardiac and non-cardiac phenotype. Hum Mutat 2003;21:473–81.
    Crossref | PubMed
  25. van Tintelen JP, Tio RA, Kerstjens-Frederikse WS, et al. Severe myocardial fibrosis caused by a deletion of the 5' end of the lamin A/C gene. J Am Coll Cardiol 2007;49:2430–9.
    Crossref | PubMed
  26. Quarta G, Syrris P, Ashworth M, et al. Mutations in the Lamin A/C gene mimic arrhythmogenic right ventricular cardiomyopathy. Eur Heart J 2012;33:1128–36.
    Crossref | PubMed
  27. Antoniades L, Eftychiou C, Kyriakides T, et al. Malignant mutation in the lamin A/C gene causing progressive conduction system disease and early sudden death in a family with mild form of limb-girdle muscular dystrophy. J Interv Card Electrophysiol 2007;19:1–7.
    Crossref | PubMed
  28. Ben Yaou R, Becane HM, Demay L, et al. Autosomal dominant limb-girdle muscular dystrophy associated with conduction defects (LGMD1B): a description of 8 new families with the LMNA gene mutations. Rev Neurol (Paris) 2005;161:42–54.
    Crossref | PubMed
  29. Carboni N, Mura M, Marrosu G, et al. Muscle MRI findings in patients with an apparently exclusive cardiac phenotype due to a novel LMNA gene mutation. Neuromuscul Disord 2008:18:291–8.
    Crossref | PubMed
  30. Pasotti M, Klersy C, Pilotto A, et al. Long-term outcome and risk stratification in dilated cardiolaminopathies. J Am Coll Cardiol 2008;52:1250–60.
    Crossref | PubMed
  31. di Barletta MR, Ricci E, Galluzzi G, et al. Different mutations in the LMNA gene cause autosomal dominant and autosomal recessive Emery-Dreifuss muscular dystrophy. Am J Hum Genet 2000;66:1407–12.
    Crossref | PubMed
  32. Jimenez-Escrig A, Gobernado I, Garcia-Villanueva M, Sanchez-Herranz A. Autosomal recessive Emery-Dreifuss muscular dystrophy caused by a novel mutation (R225Q) in the lamin A/C gene identified by exome sequencing. Muscle Nerve 2012;45:605–10.
    Crossref | PubMed
  33. Muntoni F, Bonne G, Goldfarb LG, et al. Disease severity in dominant Emery Dreifuss is increased by mutations in both emerin and desmin proteins. Brain 2006;129:1260–8.
    Crossref | PubMed
  34. van Engelen BG, Muchir A, Hutchison CJ, et al. The lethal phenotype of a homozygous nonsense mutation in the lamin A/C gene. Neurology 2005;64:374–6.
    Crossref | PubMed
  35. Parks SB, Kushner JD, Nauman D, et al. Lamin A/C mutation analysis in a cohort of 324 unrelated patients with idiopathic or familial dilated cardiomyopathy. Am Heart J 2008;156:161–9.
    Crossref | PubMed
  36. Taylor MR, Fain PR, Sinagra G, et al. Natural history of dilated cardiomyopathy due to lamin A/C gene mutations. J Am Coll Cardiol 2003;41:771–80.
    Crossref | PubMed
  37. Arbustini E, Pilotto A, Repetto A, et al. Autosomal dominant dilated cardiomyopathy with atrioventricular block: a lamin A/C defect-related disease. J Am Coll Cardiol 2002;39:981–90.
    Crossref | PubMed
  38. van Tintelen JP, Hofstra RM, Katerberg H, et al. High yield of LMNA mutations in patients with dilated cardiomyopathy and/or conduction disease referred to cardiogenetics outpatient clinics. Am Heart J 2007;154:1130–9.
    Crossref | PubMed
  39. Saj M, Dabrowski R, Labib S, et al. Variants of the lamin A/C (LMNA) gene in non-valvular atrial fibrillation patients: a possible pathogenic role of the Thr528Met mutation. Mol Diagn Ther 2012;16:99–107.
    PubMed
  40. van Berlo JH, de Voogt WG, van der Kooi AJ, et al. Meta-analysis of clinical characteristics of 299 carriers of LMNA gene mutations: do lamin A/C mutations portend a high risk of sudden death? J Mol Med 2005;83:79–83.
    Crossref | PubMed
  41. Hookana E, Junttila MJ, Sarkioja T, et al. Cardiac arrest and left ventricular fibrosis in a Finnish family with the lamin A/C mutation. J Cardiovasc Electrophysiol 2008;19:743–47.
    Crossref | PubMed
  42. Meune C, Van Berlo JH, Anselme F, et al. Primary prevention of sudden death in patients with lamin A/C gene mutations. N Engl J Med 2006;354:209–10.
    Crossref | PubMed
  43. Fernandez X, Dumont CA, Monserrat L, et al. Sudden death in a patient with lamin A/C gene mutation and near normal left ventricular systolic function. Int J Cardiol 2008;126:136–7.
    Crossref | PubMed
  44. Ehlermann P, Lehrke S, Papavassiliu T, et al. Sudden cardiac death in a patient with lamin A/C mutation in the absence of dilated cardiomyopathy or conduction disease. Clin Res Cardiol 2011;100:547–51.
    Crossref | PubMed
  45. van Rijsingen IA, Arbustini E, Elliott PM, et al. Risk factors for malignant ventricular arrhythmias in lamin A/C mutation carriers a European cohort study. J Am Coll Cardiol 2012;59:493–500.
    Crossref | PubMed
  46. Stewart CL, Kozlov S, Fong LG, Young SG. Mouse models of the laminopathies. Exp Cell Res 2007;313:2144–56.
    Crossref | PubMed
  47. Geiger SK, Bar H, Ehlermann P, et al. Incomplete nonsensemediated decay of mutant lamin A/C mRNA provokes dilated cardiomyopathy and ventricular tachycardia. J Mol Med (Berl) 2008;86:281–9.
    Crossref | PubMed
  48. Narula N, Favalli V, Tarantino P, et al. Quantitative expression of the mutated Lamin A/C gene in patients with cardiolaminopathies. J Am Coll Cardiol 2012; in press.
  49. Mewborn SK, Puckelwartz MJ, Abuisneineh F, et al. Altered chromosomal positioning, compaction, and gene expression with a lamin A/C gene mutation. PLoS One 2011;5:e14342.
    Crossref | PubMed
  50. Ostlund C, Bonne G, Schwartz K, Worman HJ. Properties of lamin A mutants found in Emery-Dreifuss muscular dystrophy, cardiomyopathy and Dunnigan-type partial lipodystrophy. J Cell Sci 2001;114:4435–45.
    PubMed
  51. De Sandre-Giovannoli A, Chaouch M, Kozlov S, et al. Homozygous defects in LMNA, encoding lamin A/C nuclear-envelope proteins, cause autosomal recessive axonal neuropathy in human (Charcot-Marie-Tooth Disorder Type 2) and mouse. Am J Hum Genet 2002;70:726–36.
    Crossref | PubMed
  52. Nikolova V, Leimena C, McMahon AC, et al. Defects in nuclear structure and function promote dilated cardiomyopathy in lamin A/C-deficient mice. J Clin Invest 2004;113:357–69.
    Crossref | PubMed
  53. Sullivan T, Escalante-Alcalde D, Bhatt H, et al. Loss of A-type Lamin Expression Compromises Nuclear Envelope Integrity Leading to Muscular Dystrophy. J Cell Biol 1999;147:913–20.
    Crossref | PubMed
  54. Chandar S, Yeo LS, Leimena C, et al. Effects of mechanical stress and carvedilol in lamin A/C-deficient dilated cardiomyopathy. Circ Res 2010;106:573–82.
    Crossref | PubMed
  55. Cupesi M, Yoshioka J, Gannon J, et al. Attenuated hypertrophic response to pressure overload in a lamin A/C haploinsufficiency mouse. J Mol Cell Cardiol 2010;48:1290–7.
    Crossref | PubMed
  56. Arimura T, Helbling-Leclerc A, Massart C, et al. Mouse model carrying H222P-Lmna mutation develops muscular dystrophy and dilated cardiomyopathy similar to human striated muscle laminopathies. Hum Mol Genet 2005;14:155–69.
    Crossref | PubMed
  57. Choi J, Muchir A, Wu W, et al. Temsirolimus activates autophagy and ameliorates cardiomyopathy caused by lamin A/C gene mutation. Sci Transl Med 2012; in press.
    Crossref | PubMed
  58. Muchir A, Pavlidis P, Decostre V, et al. Activation of MAPK pathways links LMNA mutations to cardiomyopathy in Emery-Dreifuss muscular dystrophy. J Clin Invest 2007;117:1282–93.
    Crossref | PubMed
  59. Muchir A, Wu W, Choi JC, et al. Abnormal p38alpha mitogen-activated protein kinase signaling in dilated cardiomyopathy caused by lamin A/C gene mutation. Hum Mol Genet 2012; in press.
    Crossref | PubMed
  60. Mounkes LC, Kozlov SV, Rottman JN, Stewart CL. Expression of a LMNA-N195K variant of A-type lamins results in cardiac conduction defects and death in mice. Hum Mol Genet 2005;14:2167–80.
    Crossref | PubMed
  61. Muchir A, Reilly SA, Wu W, et al. Treatment with selumetinib preserves cardiac function and improves survival in cardiomyopathy caused by mutation in the lamin A/C gene. Cardiovasc Res 2012;93:311–9.
    Crossref | PubMed
  62. Muchir A, Shan J, Bonne G, et al. Inhibition of extracellular signal-regulated kinase signaling to prevent cardiomyopathy caused by mutation in the gene encoding A-type lamins. Hum Mol Genet 2009;18:241–7.
    Crossref | PubMed
  63. Wu W, Muchir A, Shan J, et al. Mitogen-activated protein kinase inhibitors improve heart function and prevent fibrosis in cardiomyopathy caused by mutation in lamin A/C gene. Circulation 2011;123:53–61.
    Crossref | PubMed
  64. Wu W, Shan J, Bonne G, et al. Pharmacological inhibition of c-Jun N-terminal kinase signaling prevents cardiomyopathy caused by mutation in LMNA gene. Biochim Biophys Acta 2010;1802:632–8.
    Crossref | PubMed
  65. Arimura T, Sato R, Machida N, et al. Improvement of left ventricular dysfunction and of survival prognosis of dilated cardiomyopathy by administration of calcium sensitizer SCH00013 in a mouse model. J Am Coll Cardiol 2010;55:1503–5.
    Crossref | PubMed
  66. Liu GH, Suzuki K, Qu J, et al. Targeted gene correction of laminopathy-associated LMNA mutations in patient-specific iPSCs. Cell Stem Cell 2011;8:688–94.
    Crossref | PubMed
  67. Tiscornia G, Vivas EL, Belmonte JC. Diseases in a dish: modeling human genetic disorders using induced pluripotent cells. Nat Med 2011;17:1570–6.
    Crossref | PubMed
  68. PREclinical Mutation CARriers From Families With DIlated Cardiomyopathy and ACE Inhibitors trial, (PRECARDIA) ClinicalTrial.Gov number: 01583114. http://clinicaltrials.gov/ct2/show/NCT01583114 (20 August 2012).
  69. Marsman RF, Bardai A, Postma AV, et al. A complex double deletion in LMNA underlies progressive cardiac conduction disease, atrial arrhythmias, and sudden death. Circ Cardiovasc Genet 2011;4(3):280–7.
    Crossref | PubMed
  70. Charron P, Arad M, Arbustini E, et al. Genetic counselling and testing in cardiomyopathies: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2010;31:2715–26.
    Crossref | PubMed