Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 324(6): C1223-C1235, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37125775

ABSTRACT

Dilated cardiomyopathy caused by mutations in LMNA, encoding A-type lamins (i.e., LMNA cardiomyopathy), is characterized by a left ventricle enlargement and ultimately results in poor cardiac contractility associated with conduction defects. Despite current strategies to aggressively manage the symptoms, the disorder remains a common cause of sudden death and heart failure with decreased ejection fraction. Patient care includes cardioverter defibrillator implantation but the last therapeutic option remains cardiac transplantation. A-type lamins are intermediate filaments and are the main components of the nuclear lamina, a meshwork underlying the inner nuclear membrane, which plays an essential role in both maintaining the nuclear structure and organizing the cytoskeletal structures within the cell. Cytoskeletal proteins function as scaffold to resist external mechanical stress. An increasing amount of evidence demonstrates that LMNA mutations can lead to disturbances in several structural and cytoskeletal components of the cell such as microtubules, actin cytoskeleton, and intermediate filaments. Collectively, this review focuses on the significance of these cytoskeletal modulators and emphasizes their potential therapeutic role in LMNA cardiomyopathy. Indeed, molecular tuning of cytoskeletal dynamics has been successfully used in preclinical models and provides adequate grounds for a therapeutic approach for patients with LMNA cardiomyopathy.


Subject(s)
Cardiomyopathies , Lamin Type A , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Cardiomyopathies/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Microtubules/metabolism , Mutation/genetics
2.
Nat Commun ; 13(1): 7886, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36550158

ABSTRACT

Mutations in the lamin A/C gene (LMNA) cause dilated cardiomyopathy associated with increased activity of ERK1/2 in the heart. We recently showed that ERK1/2 phosphorylates cofilin-1 on threonine 25 (phospho(T25)-cofilin-1) that in turn disassembles the actin cytoskeleton. Here, we show that in muscle cells carrying a cardiomyopathy-causing LMNA mutation, phospho(T25)-cofilin-1 binds to myocardin-related transcription factor A (MRTF-A) in the cytoplasm, thus preventing the stimulation of serum response factor (SRF) in the nucleus. Inhibiting the MRTF-A/SRF axis leads to decreased α-tubulin acetylation by reducing the expression of ATAT1 gene encoding α-tubulin acetyltransferase 1. Hence, tubulin acetylation is decreased in cardiomyocytes derived from male patients with LMNA mutations and in heart and isolated cardiomyocytes from Lmnap.H222P/H222P male mice. In Atat1 knockout mice, deficient for acetylated α-tubulin, we observe left ventricular dilation and mislocalization of Connexin 43 (Cx43) in heart. Increasing α-tubulin acetylation levels in Lmnap.H222P/H222P mice with tubastatin A treatment restores the proper localization of Cx43 and improves cardiac function. In summary, we show for the first time an actin-microtubule cytoskeletal interplay mediated by cofilin-1 and MRTF-A/SRF, promoting the dilated cardiomyopathy caused by LMNA mutations. Our findings suggest that modulating α-tubulin acetylation levels is a feasible strategy for improving cardiac function.


Subject(s)
Cardiomyopathy, Dilated , Male , Mice , Animals , Cardiomyopathy, Dilated/metabolism , Actins/metabolism , Connexin 43/genetics , Tubulin/genetics , Serum Response Factor/genetics , Lamin Type A/genetics , Lamin Type A/metabolism , Microtubules/metabolism , Myocytes, Cardiac/metabolism , Mice, Knockout , Intermediate Filament Proteins/genetics , Mutation , Actin Depolymerizing Factors/genetics
3.
Cell Rep ; 36(8): 109601, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433058

ABSTRACT

Cofilins are important for the regulation of the actin cytoskeleton, sarcomere organization, and force production. The role of cofilin-1, the non-muscle-specific isoform, in muscle function remains unclear. Mutations in LMNA encoding A-type lamins, intermediate filament proteins of the nuclear envelope, cause autosomal Emery-Dreifuss muscular dystrophy (EDMD). Here, we report increased cofilin-1 expression in LMNA mutant muscle cells caused by the inability of proteasome degradation, suggesting a protective role by ERK1/2. It is known that phosphorylated ERK1/2 directly binds to and catalyzes phosphorylation of the actin-depolymerizing factor cofilin-1 on Thr25. In vivo ectopic expression of cofilin-1, as well as its phosphorylated form on Thr25, impairs sarcomere structure and force generation. These findings present a mechanism that provides insight into the molecular pathogenesis of muscular dystrophies caused by LMNA mutations.


Subject(s)
Actin Cytoskeleton/metabolism , Cofilin 1/metabolism , Destrin/metabolism , Lamin Type A/metabolism , Laminopathies/metabolism , Muscle, Striated/metabolism , Sarcomeres/metabolism , Adolescent , Adult , Animals , Cell Line , Child , Humans , Lamin Type A/genetics , Laminopathies/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Striated/pathology , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Mutation , Phosphorylation , Signal Transduction , Young Adult
4.
Hum Mol Genet ; 28(24): 4043-4052, 2019 12 15.
Article in English | MEDLINE | ID: mdl-29893868

ABSTRACT

Mutations in the lamin A/C gene (LMNA) cause an autosomal dominant inherited form of dilated cardiomyopathy associated with cardiac conduction disease (hereafter referred to as LMNA cardiomyopathy). Compared with other forms of dilated cardiomyopathy, mutations in LMNA are responsible for a more aggressive clinical course owing to a high rate of malignant ventricular arrhythmias. Gap junctions are intercellular channels that allow direct communication between neighboring cells, which are involved in electrical impulse propagation and coordinated contraction of the heart. For gap junctions to properly control electrical synchronization in the heart, connexin-based hemichannels must be correctly targeted to intercalated discs, Cx43 being the major connexin in the working myocytes. We here showed an altered distribution of Cx43 in a mouse model of LMNA cardiomyopathy. However, little is known on the molecular mechanisms of Cx43 remodeling in pathological context. We now show that microtubule cytoskeleton alteration and decreased acetylation of α-tubulin lead to remodeling of Cx43 in LMNA cardiomyopathy, which alters the correct communication between cardiomyocytes, ultimately leading to electrical conduction disturbances. Preventing or reversing this process could offer a strategy to repair damaged heart. Stabilization of microtubule cytoskeleton using Paclitaxel improved intraventricular conduction defects. These results indicate that microtubule cytoskeleton contributes to the pathogenesis of LMNA cardiomyopathy and that drugs stabilizing the microtubule may be beneficial for patients.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Connexin 43/metabolism , Lamin Type A/genetics , Paclitaxel/pharmacology , Acetylation/drug effects , Animals , Cardiac Conduction System Disease/genetics , Cardiomyopathies/pathology , Connexin 43/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/pathology , Lamin Type A/metabolism , Male , Mice , Mice, Knockout , Microtubules/metabolism , Microtubules/pathology , Mutation , Myocardium/pathology , Myocytes, Cardiac/pathology
5.
Nucleus ; 9(1): 235-248, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29578370

ABSTRACT

Mutations in LMNA, encoding A-type lamins, are responsible for laminopathies including muscular dystrophies, lipodystrophies, and premature ageing syndromes. LMNA mutations have been shown to alter nuclear structure and stiffness, binding to partners at the nuclear envelope or within the nucleoplasm, gene expression and/or prelamin A maturation. LMNA-associated lipodystrophic features, combining generalized or partial fat atrophy and metabolic alterations associated with insulin resistance, could result from altered adipocyte differentiation or from altered fat structure. Recent studies shed some light on how pathogenic A-type lamin variants could trigger lipodystrophy, metabolic complications, and precocious cardiovascular events. Alterations in adipose tissue extracellular matrix and TGF-beta signaling could initiate metabolic inflexibility. Premature senescence of vascular cells could contribute to cardiovascular complications. In affected families, metabolic alterations occur at an earlier age across generations, which could result from epigenetic deregulation induced by LMNA mutations. Novel cellular models recapitulating adipogenic developmental pathways provide scalable tools for disease modeling and therapeutic screening.


Subject(s)
Lamin Type A/genetics , Lipodystrophy , Mutation , Humans , Lamin Type A/metabolism , Lipodystrophy/drug therapy , Lipodystrophy/genetics , Lipodystrophy/metabolism
6.
Hum Mol Genet ; 26(2): 333-343, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28069793

ABSTRACT

Cardiomyopathy caused by lamin A/C gene (LMNA) mutations (hereafter referred as LMNA cardiomyopathy) is characterized by cardiac conduction abnormalities and left ventricular systolic dysfunction predisposing to heart failure. Previous cardiac transcriptional profiling of LmnaH222P/H222P mouse, a small animal model of LMNA cardiomyopathy, suggested decreased WNT/ß-catenin signalling. We confirmed decreased WNT/ß-catenin signalling in the hearts of these mice by demonstrating decreased ß-catenin and WNT proteins. This was correlated with increased expression of soluble Frizzled-related proteins that modulate the WNT/ß-catenin signalling pathway. Hearts of LmnaH222P/H222P mice also demonstrated lowered expression of the gap junction connexin 43. Activation of WNT/ß-catenin activity with 6-bromoindirubin-3'-oxime improved cardiac contractility and ameliorated intraventricular conduction defects in LmnaH222P/H222P mice, which was associated with increased expression of myocardial connexin 43. These results indicate that decreased WNT/ß-catenin contributes to the pathophysiology of LMNA cardiomyopathy and that drugs activating ß-catenin may be beneficial in affected individuals.


Subject(s)
Cardiomyopathy, Dilated/genetics , Connexin 43/genetics , Lamin Type A/genetics , beta Catenin/genetics , Animals , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/physiopathology , Connexin 43/biosynthesis , Disease Models, Animal , Gene Expression Regulation/drug effects , Glycoproteins/biosynthesis , Glycoproteins/genetics , Heart Failure/drug therapy , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Indoles/administration & dosage , Intracellular Signaling Peptides and Proteins , Mice , Mutation , Oximes/administration & dosage , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/biosynthesis
7.
J Lipid Res ; 58(1): 151-163, 2017 01.
Article in English | MEDLINE | ID: mdl-27845687

ABSTRACT

Mutations in the lamin A/C gene encoding nuclear lamins A and C (lamin A/C) cause familial partial lipodystrophy type 2 (FPLD2) and related lipodystrophy syndromes. These are mainly characterized by redistribution of adipose tissue associated with insulin resistance. Several reports suggest that alterations in the extracellular matrix of adipose tissue leading to fibrosis play a role in the pathophysiology of lipodystrophy syndromes. However, the extent of extracellular matrix alterations in FPLD2 remains unknown. We show significantly increased fibrosis and altered expression of genes encoding extracellular matrix proteins in cervical subcutaneous adipose tissue from a human subject with FLPD2. Similar extracellular matrix alterations occur in adipose tissue of transgenic mice expressing an FPLD2-causing human lamin A variant and in cultured fibroblasts from human subjects with FPLD2 and related lipodystrophies. These abnormalities are associated with increased transforming growth factor-ß signaling and defects in matrix metalloproteinase 9 activity. Our data demonstrate that lamin A/C gene mutations responsible for FPLD2 and related lipodystrophies are associated with transforming growth factor-ß activation and an extracellular matrix imbalance in adipose tissue, suggesting that targeting these alterations could be the basis of novel therapies.


Subject(s)
Adipose Tissue/metabolism , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , Matrix Metalloproteinase 9/genetics , Transforming Growth Factor beta/genetics , Adipose Tissue/pathology , Aged , Aged, 80 and over , Animals , Cell Line , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , Lipodystrophy, Familial Partial/metabolism , Lipodystrophy, Familial Partial/pathology , Male , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Knockout , Middle Aged , Mutation , Transforming Growth Factor beta/biosynthesis
8.
Hum Mol Genet ; 25(11): 2220-2233, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27131347

ABSTRACT

Cardiomyopathy caused by lamin A/C gene mutations (LMNA cardiomyopathy) is characterized by increased myocardial fibrosis, which impairs left ventricular relaxation and predisposes to heart failure, and cardiac conduction abnormalities. While we previously discovered abnormally elevated extracellular signal-regulated kinase 1/2 (ERK1/2) activities in heart in LMNA cardiomyopathy, its role on the development of myocardial fibrosis remains unclear. We now showed that transforming growth factor (TGF)-ß/Smad signaling participates in the activation of ERK1/2 signaling in LMNA cardiomyopathy. ERK1/2 acts on connective tissue growth factor (CTGF/CCN2) expression to mediate the myocardial fibrosis and left ventricular dysfunction. Studies in vivo demonstrate that inhibiting CTGF/CCN2 using a specific antibody decreases myocardial fibrosis and improves the left ventricular dysfunction. Together, these findings show that cardiac ERK1/2 activity is modulated in part by TGF-ß/Smad signaling, leading to altered activation of CTGF/CCN2 to mediate fibrosis and alter cardiac function. This identifies a novel mechanism in the development of LMNA cardiomyopathy.


Subject(s)
Cardiomyopathies/genetics , Connective Tissue Growth Factor/genetics , Fibrosis/genetics , Lamin Type A/genetics , Transforming Growth Factor beta/genetics , Animals , Cardiomyopathies/pathology , Fibrosis/pathology , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Smad Proteins/genetics , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology
9.
Nucleus ; 5(3): 260-459, 2014.
Article in English | MEDLINE | ID: mdl-24859316

ABSTRACT

We previously showed that striated muscle-selective depletion of lamina-associated polypeptide 1 (LAP1), an integral inner nuclear membrane protein, leads to profound muscular dystrophy with premature death in mice. As LAP1 is also depleted in hearts of these mice, we examined their cardiac phenotype. Striated muscle-selective LAP1 knockout mice display ventricular systolic dysfunction with abnormal induction of genes encoding cardiomyopathy related proteins. To eliminate possible confounding effects due to skeletal muscle pathology, we generated a new mouse line in which LAP1 is deleted in a cardiomyocyte-selective manner. These mice had no skeletal muscle pathology and appeared overtly normal at 20 weeks of age. However, cardiac echocardiography revealed that they developed left ventricular systolic dysfunction and cardiac gene expression analysis revealed abnormal induction of cardiomyopathy-related genes. Our results demonstrate that LAP1 expression in cardiomyocytes is required for normal left ventricular function, consistent with a report of cardiomyopathy in a human subject with mutation in the gene encoding LAP1.


Subject(s)
Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Ventricular Dysfunction, Left/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Gene Expression/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Nuclear Proteins/genetics , Ventricular Dysfunction, Left/genetics
10.
Am J Pathol ; 179(5): 2443-53, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21945321

ABSTRACT

Some LMNA mutations responsible for insulin-resistant lipodystrophic syndromes are associated with peripheral subcutaneous lipoatrophy and faciocervical fat accumulation. Their pathophysiologic characteristics are unknown. We compared histologic, immunohistologic, ultrastructural, and protein expression features of enlarged cervical subcutaneous adipose tissue (scAT) obtained during plastic surgery from four patients with LMNA p.R482W, p.R439C, or p.H506D mutations versus cervical fat from eight control subjects, buffalo humps from five patients with HIV infection treated or not with protease inhibitors, and dorsocervical lipomas from two patients with mitochondrial DNA mutations. LMNA-mutated cervical scAT and HIV-related buffalo humps were dystrophic, with an increased percentage of small adipocytes, increased fibrosis without inflammatory features, and decreased number of blood vessels, as compared with control samples. Samples from patients with LMNA mutations or protease inhibitor-based therapy demonstrated accumulation of prelamin A, altered expression of adipogenic proteins and brown fat-like features, with an increased number of mitochondria and overexpression of uncoupling protein 1 (UCP1). These features were absent in samples from control subjects and from patients with HIV not treated with protease inhibitors. Mitochondrial DNA-mutated cervical lipomas demonstrated inflammatory fibrosis with distinct mitochondrial abnormalities but neither UCP1 expression nor prelamin A accumulation. In conclusion, Enlarged cervical scAT from patients with lipodystrophy demonstrated small adipocytes, fibrosis, and decreased vessel numbers. However, only cervical fat from patients with LMNA mutations or who had received protease inhibitor therapy accumulated prelamin A and exhibited similar remodeling toward a brown-like phenotype with UCP1 overexpression and mitochondrial alterations.


Subject(s)
Adipose Tissue, Brown/pathology , Lamin Type A/genetics , Lipodystrophy/genetics , Mutation/genetics , Adipocytes/pathology , Adult , Aged , Aged, 80 and over , Female , Fibrosis/pathology , Humans , Lipodystrophy/pathology , Male , Middle Aged , Neck
11.
Int J Biochem Cell Biol ; 43(6): 862-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21392585

ABSTRACT

Adipose tissue is now recognized for its major role in the control of energy metabolism and insulin sensitivity. We review here the human lipodystrophies, that are rare conditions in which total or partial fat loss is associated with severe lipid and glucose abnormalities leading to diabetes with early cardiovascular and hepatic complications. The genetic origin of a number of human lipodystrophies has been recently unraveled, emphasizing the importance of proteins of previously unknown or unexpected functions. Major adipose functions were also illuminated when studying acquired forms of lipodystrophies linked to human immunodeficiency virus-antiretrovirals. Overall, most of the proteins or functions affected by mutations or antiretrovirals result in altered adipogenesis and insulin sensitivity, triglyceride storage and formation of the unique adipocyte lipid droplet, oxidative stress and fat remodeling. Some mutations or antiretrovirals could affect directly (peroxisome proliferator-activated receptor-γ, Akt2) or indirectly (lamin A/C, human immunodeficiency virus-protease inhibitors) adipogenesis, through the transcription factors peroxisome proliferator-activated receptor gamma-γ or sterol regulatory element binding protein 1c, and insulin signaling through Akt2 that controls adipocyte lipolysis. A number of proteins mutated in genetic lipodystrophies are involved in the control of triglyceride synthesis towards the lipid droplet (1-acylglycerol-3-phosphate-O-acyltransferase 2), or its functions (seipin, cell death-inducing DFF45-like effector C, perilipin, caveolin-1, cavin-1). Decreased triglyceride storage leads to adipocyte lipotoxicity, mitochondrial dysfunction and increased oxidative stress, which could also be induced by some thymidine analogue antiretrovirals. This results in production of inflammatory mediators and deregulated release of free fatty acids. Thus, the impaired ability of adipose tissue to safely store triglycerides inside the lipid droplet results in impaired insulin sensitivity and adverted liver, muscles and heart functions leading to early complications.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Adipose Tissue/metabolism , Lipodystrophy/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Antiretroviral Therapy, Highly Active/adverse effects , Humans , Inflammation , Insulin Resistance , Lipid Metabolism , Lipodystrophy/chemically induced , Lipodystrophy/genetics , Lipodystrophy/pathology , Oxidative Stress , PPAR gamma/genetics , PPAR gamma/metabolism , Triglycerides/metabolism
12.
J Clin Endocrinol Metab ; 96(5): E856-62, 2011 May.
Article in English | MEDLINE | ID: mdl-21346069

ABSTRACT

CONTEXT: Mutations in LMNA, encoding A-type lamins, lead to multiple laminopathies, including lipodystrophies, progeroid syndromes, and cardiomyopathies. Alterations in the prelamin-A posttranslational maturation, resulting in accumulation of farnesylated isoforms, cause human progeroid syndromes. Accumulation of mutant nonfarnesylated prelamin-A leads to cardiomyopathy or progeria in mice, but no data have been provided in humans. OBJECTIVE, DESIGN, SETTING, AND PATIENTS: We searched for LMNA mutations in seven women originating from Reunion Island who were referred for a severe lipodystrophic syndrome. Clinical, molecular, genealogical, and cellular studies were performed in probands and relatives. RESULTS: The seven probands showed a severe partial lipodystrophic syndrome with diabetes and/or acanthosis nigricans, liver steatosis, hypertriglyceridemia, and low serum leptin and adiponectin levels. Three probands also had severe cardiac rhythm and conduction disturbances. We identified in all probands a homozygous LMNA p.T655fsX49 mutation leading to expression of a mutated prelamin-A with 48 aberrant C-terminal amino acids, preventing its physiological posttranslational farnesylation and maturation. Genealogical and haplotype analyses were consistent with a founder mutation transmitted from a common ancestor in the 17th century. In probands' cultured fibroblasts, mutated prelamin-A was associated with typical laminopathic nuclear dysmorphies, increased oxidative stress, and premature senescence. Heterozygous relatives were asymptomatic or partially affected, in favor of a codominant transmission of the disease with incomplete penetrance in heterozygotes. CONCLUSIONS: We reveal that a homozygous mutation of prelamin-A preventing its farnesylation leads to a severe lipodystrophic laminopathy in humans, which can be associated with cardiac conduction disturbances, stressing the pathogenicity of nonfarnesylated prelamin-A in human laminopathies.


Subject(s)
Lipodystrophy/blood , Lipodystrophy/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Prenylation/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , Acanthosis Nigricans/genetics , Adiponectin/blood , Adult , Arrhythmias, Cardiac/genetics , Cellular Senescence/genetics , Diabetes Mellitus/genetics , Fatty Liver/genetics , Female , Fibroblasts/ultrastructure , Founder Effect , Humans , Hypertriglyceridemia/genetics , Lamin Type A/genetics , Leptin/blood , Middle Aged , Mutation/genetics , Mutation/physiology , Oxidative Stress/physiology , Phenotype , Young Adult
13.
N Engl J Med ; 364(8): 740-8, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21345103

ABSTRACT

Perilipin is the most abundant adipocyte-specific protein that coats lipid droplets, and it is required for optimal lipid incorporation and release from the droplet. We identified two heterozygous frameshift mutations in the perilipin gene (PLIN1) in three families with partial lipodystrophy, severe dyslipidemia, and insulin-resistant diabetes. Subcutaneous fat from the patients was characterized by smaller-than-normal adipocytes, macrophage infiltration, and fibrosis. In contrast to wild-type perilipin, mutant forms of the protein failed to increase triglyceride accumulation when expressed heterologously in preadipocytes. These findings define a novel dominant form of inherited lipodystrophy and highlight the serious metabolic consequences of a primary defect in the formation of lipid droplets in adipose tissue.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Frameshift Mutation , Hypertriglyceridemia/genetics , Lipodystrophy, Familial Partial/genetics , Phosphoproteins/deficiency , Phosphoproteins/genetics , Acanthosis Nigricans/genetics , Adult , Carrier Proteins , Female , Genes, Dominant , Heterozygote , Humans , Insulin Resistance/genetics , Middle Aged , Pedigree , Perilipin-1
14.
J Cell Mol Med ; 13(5): 959-71, 2009 May.
Article in English | MEDLINE | ID: mdl-19220582

ABSTRACT

Dunnigan-type familial partial lipodystrophy (FPLD) is a laminopathy characterized by an aberrant fat distribution and a metabolic syndrome for which oxidative stress has recently been suggested as one of the disease-causing mechanisms. In a family affected with FPLD, we identified a heterozygous missense mutation c.1315C>T in the LMNA gene leading to the p.R439C substitution. Cultured patient fibroblasts do not show any prelamin A accumulation and reveal honeycomb-like lamin A/C formations in a significant percentage of nuclei. The mutation affects a region in the C-terminal globular domain of lamins A and C, different from the FPLD-related hot spot. Here, the introduction of an extra cysteine allows for the formation of disulphide-mediated lamin A/C oligomers. This oligomerization affects the interaction properties of the C-terminal domain with DNA as shown by gel retardation assays and causes a DNA-interaction pattern that is distinct from the classical R482W FPLD mutant. Particularly, whereas the R482W mutation decreases the binding efficiency of the C-terminal domain to DNA, the R439C mutation increases it. Electron spin resonance spectroscopy studies show significantly higher levels of reactive oxygen species (ROS) upon induction of oxidative stress in R439C patient fibroblasts compared to healthy controls. This increased sensitivity to oxidative stress seems independent of the oligomerization and enhanced DNA binding typical for R439C, as both the R439C and R482W mutants show a similar and significant increase in ROS upon induction of oxidative stress by H2O2.


Subject(s)
Lamin Type A/physiology , Lipodystrophy, Familial Partial/metabolism , Mutation, Missense , Nuclear Proteins/metabolism , Oxidative Stress , Protein Precursors/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Fibroblasts/metabolism , Genetic Predisposition to Disease , Humans , Hydrogen Peroxide/pharmacology , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , Multiprotein Complexes , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...