Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JID Innov ; 4(4): 100283, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38827330

RESUMEN

The skin is a multifunctional organ, forming a barrier between the external and internal environment, thereby functioning as a safeguard against extrinsic factors. Autophagy has been implicated in epidermal differentiation and in preserving skin homeostasis. LC3-associated phagocytosis (LAP) uses some but not all components of autophagy. The Atg16l1 (Δ WD) mouse model lacks the WD40 domain required for LAP and has been widely used to study the effects of LAP deficiency and autophagy on tissue homeostasis and response to infection. In this study, the Δ WD model was used to study the relationship between LAP and skin homeostasis by determining whether LAP-deficient mice display a cutaneous phenotype. Skin histology of wild-type and Δ WD mice aged 1 year revealed minor morphological differences in the tail skin dermal layer. RT-qPCR and western blot analysis showed no differences in key keratin expression between genotypes. Skin barrier formation, assessed by dye permeation assays, demonstrated full and proper formation of the skin barrier at embryonic day 18.5 in both genotypes. Biomechanical analysis of the skin showed decreased skin elasticity in aged Δ WD but not wild-type mice. In summary, the LAP-deficient Δ WD mice displayed subtle alterations in dermal histology and age-related biomechanical changes.

2.
J Biol Chem ; 300(5): 107254, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38569934

RESUMEN

Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.


Asunto(s)
Conectina , Proteínas con Dominio LIM , Proteínas Musculares , Miocitos Cardíacos , Proteínas del Tejido Nervioso , Proteínas Nucleares , Sarcómeros , Animales , Humanos , Ratones , Ratas , Conectina/metabolismo , Conectina/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Sarcómeros/metabolismo , Factores de Transcripción
3.
Br J Pharmacol ; 2023 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-38044463

RESUMEN

BACKGROUND AND PURPOSE: Decreased aortic compliance is a precursor to numerous cardiovascular diseases. Compliance is regulated by the rigidity of the aortic wall and the vascular smooth muscle cells (VSMCs). Extracellular matrix stiffening, observed during ageing, reduces compliance. In response to increased rigidity, VSMCs generate enhanced contractile forces that result in VSMC stiffening and a further reduction in compliance. Mechanisms driving VSMC response to matrix rigidity remain poorly defined. EXPERIMENTAL APPROACH: Human aortic-VSMCs were seeded onto polyacrylamide hydrogels whose rigidity mimicked either healthy (12 kPa) or aged/diseased (72 kPa) aortae. VSMCs were treated with pharmacological agents prior to agonist stimulation to identify regulators of VSMC volume regulation. KEY RESULTS: On pliable matrices, VSMCs contracted and decreased in cell area. Meanwhile, on rigid matrices VSMCs displayed a hypertrophic-like response, increasing in area and volume. Piezo1 activation stimulated increased VSMC volume by promoting calcium ion influx and subsequent activation of PKC and aquaporin-1. Pharmacological blockade of this pathway prevented the enhanced VSMC volume response on rigid matrices whilst maintaining contractility on pliable matrices. Importantly, both piezo1 and aquaporin-1 gene expression were up-regulated during VSMC phenotypic modulation in atherosclerosis and after carotid ligation. CONCLUSIONS AND IMPLICATIONS: In response to extracellular matrix rigidity, VSMC volume is increased by a piezo1/PKC/aquaporin-1 mediated pathway. Pharmacological targeting of this pathway specifically blocks the matrix rigidity enhanced VSMC volume response, leaving VSMC contractility on healthy mimicking matrices intact. Importantly, upregulation of both piezo1 and aquaporin-1 gene expression is observed in disease relevant VSMC phenotypes.

4.
Front Pharmacol ; 13: 836710, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35153800

RESUMEN

Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aortic wall and normally exist in a quiescent, contractile phenotype where actomyosin-derived contractile forces maintain vascular tone. However, VSMCs are not terminally differentiated and can dedifferentiate into a proliferative, synthetic phenotype. Actomyosin force generation is essential for the function of both phenotypes. Whilst much is already known about the mechanisms of VSMC actomyosin force generation, existing assays are either low throughput and time consuming, or qualitative and inconsistent. In this study, we use polyacrylamide hydrogels, tuned to mimic the physiological stiffness of the aortic wall, in a VSMC contractility assay. Isolated VSMC area decreases following stimulation with the contractile agonists angiotensin II or carbachol. Importantly, the angiotensin II induced reduction in cell area correlated with increased traction stress generation. Inhibition of actomyosin activity using blebbistatin or Y-27632 prevented angiotensin II mediated changes in VSMC morphology, suggesting that changes in VSMC morphology and actomyosin activity are core components of the contractile response. Furthermore, we show that microtubule stability is an essential regulator of isolated VSMC contractility. Treatment with either colchicine or paclitaxel uncoupled the morphological and/or traction stress responses of angiotensin II stimulated VSMCs. Our findings support the tensegrity model of cellular mechanics and we demonstrate that microtubules act to balance actomyosin-derived traction stress generation and regulate the morphological responses of VSMCs.

5.
Biophys Rev ; 13(5): 757-768, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34745374

RESUMEN

Arterial smooth muscle cells (ASMCs), the predominant cell type within the arterial wall, detect and respond to external mechanical forces. These forces can be derived from blood flow (i.e. pressure and stretch) or from the supporting extracellular matrix (i.e. stiffness and topography). The healthy arterial wall is elastic, allowing the artery to change shape in response to changes in blood pressure, a property known as arterial compliance. As we age, the mechanical forces applied to ASMCs change; blood pressure and arterial wall rigidity increase and result in a reduction in arterial compliance. These changes in mechanical environment enhance ASMC contractility and promote disease-associated changes in ASMC phenotype. For mechanical stimuli to programme ASMCs, forces must influence the cell's load-bearing apparatus, the cytoskeleton. Comprised of an interconnected network of actin filaments, microtubules and intermediate filaments, each cytoskeletal component has distinct mechanical properties that enable ASMCs to respond to changes within the mechanical environment whilst maintaining cell integrity. In this review, we discuss how mechanically driven cytoskeletal reorganisation programmes ASMC function and phenotypic switching.

6.
Biochem Soc Trans ; 46(3): 669-681, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29784648

RESUMEN

Nesprins (nuclear envelope spectrin repeat proteins) are a family of multi-isomeric scaffolding proteins. Nesprins form the LInker of Nucleoskeleton-and-Cytoskeleton (LINC) complex with SUN (Sad1p/UNC84) domain-containing proteins at the nuclear envelope, in association with lamin A/C and emerin, linking the nucleoskeleton to the cytoskeleton. The LINC complex serves as both a physical linker between the nuclear lamina and the cytoskeleton and a mechanosensor. The LINC complex has a broad range of functions and is involved in maintaining nuclear architecture, nuclear positioning and migration, and also modulating gene expression. Over 80 disease-related variants have been identified in SYNE-1/2 (nesprin-1/2) genes, which result in muscular or central nervous system disorders including autosomal dominant Emery-Dreifuss muscular dystrophy, dilated cardiomyopathy and autosomal recessive cerebellar ataxia type 1. To date, 17 different nesprin mouse lines have been established to mimic these nesprin-related human diseases, which have provided valuable insights into the roles of nesprin and its scaffold LINC complex in a tissue-specific manner. In this review, we summarise the existing nesprin mouse models, compare their phenotypes and discuss the potential mechanisms underlying nesprin-associated diseases.


Asunto(s)
Modelos Animales de Enfermedad , Cardiopatías/fisiopatología , Enfermedades Musculares/fisiopatología , Proteínas del Tejido Nervioso/fisiología , Membrana Nuclear/fisiología , Animales , Cardiopatías/genética , Humanos , Ratones , Enfermedades Musculares/genética , Proteínas del Tejido Nervioso/genética , Fenotipo
7.
Stem Cell Reports ; 9(2): 681-696, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28757161

RESUMEN

Adventitial progenitor cells, including SCA-1+ and mesenchymal stem cells, are believed to be important in vascular remodeling. It has been shown that SCA-1+ progenitor cells are involved in neointimal hyperplasia of vein grafts, but little is known concerning their involvement in hyperlipidemia-induced atherosclerosis. We employed single-cell sequencing technology on primary adventitial mouse SCA-1+ cells from wild-type and atherosclerotic-prone (ApoE-deficient) mice and found that a group of genes controlling cell migration and matrix protein degradation was highly altered. Adventitial progenitors from ApoE-deficient mice displayed an augmented migratory potential both in vitro and in vivo. This increased migratory ability was mimicked by lipid loading to SCA-1+ cells. Furthermore, we show that lipid loading increased miRNA-29b expression and induced sirtuin-1 and matrix metalloproteinase-9 levels to promote cell migration. These results provide direct evidence that blood cholesterol levels influence vascular progenitor cell function, which could be a potential target cell for treatment of vascular disease.


Asunto(s)
Ataxina-1/genética , Movimiento Celular/genética , Hiperlipidemias/etiología , Hiperlipidemias/metabolismo , Células Madre/metabolismo , Animales , Apolipoproteínas E/deficiencia , Ataxina-1/metabolismo , Aterosclerosis/sangre , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores , Diferenciación Celular/genética , LDL-Colesterol/metabolismo , Biología Computacional/métodos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Hiperlipidemias/sangre , Inmunohistoquímica , Mediadores de Inflamación/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Células Madre/citología
8.
Hum Mol Genet ; 26(12): 2258-2276, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28398466

RESUMEN

Nesprins-1 and -2 are highly expressed in skeletal and cardiac muscle and together with SUN (Sad1p/UNC84)-domain containing proteins and lamin A/C form the LInker of Nucleoskeleton-and-Cytoskeleton (LINC) bridging complex at the nuclear envelope (NE). Mutations in nesprin-1/2 have previously been found in patients with autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD) as well as dilated cardiomyopathy (DCM). In this study, three novel rare variants (R8272Q, S8381C and N8406K) in the C-terminus of the SYNE1 gene (nesprin-1) were identified in seven DCM patients by mutation screening. Expression of these mutants caused nuclear morphology defects and reduced lamin A/C and SUN2 staining at the NE. GST pull-down indicated that nesprin-1/lamin/SUN interactions were disrupted. Nesprin-1 mutations were also associated with augmented activation of the ERK pathway in vitro and in hearts in vivo. During C2C12 muscle cell differentiation, nesprin-1 levels are increased concomitantly with kinesin light chain (KLC-1/2) and immunoprecipitation and GST pull-down showed that these proteins interacted via a recently identified LEWD domain in the C-terminus of nesprin-1. Expression of nesprin-1 mutants in C2C12 cells caused defects in myoblast differentiation and fusion associated with dysregulation of myogenic transcription factors and disruption of the nesprin-1 and KLC-1/2 interaction at the outer nuclear membrane. Expression of nesprin-1α2 WT and mutants in zebrafish embryos caused heart developmental defects that varied in severity. These findings support a role for nesprin-1 in myogenesis and muscle disease, and uncover a novel mechanism whereby disruption of the LINC complex may contribute to the pathogenesis of DCM.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Técnicas de Cultivo de Célula , Proteínas del Citoesqueleto , Citoesqueleto/metabolismo , Humanos , Cinesinas , Lamina Tipo A/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Distrofia Muscular de Emery-Dreifuss/genética , Mutación , Membrana Nuclear/metabolismo , Pez Cebra/genética
9.
Cells ; 5(4)2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27854297

RESUMEN

Vascular smooth muscle cell (VSMC) motility is essential during both physiological and pathological vessel remodeling. Although ageing has emerged as a major risk factor in the development of cardiovascular disease, our understanding of the impact of ageing on VSMC motility remains limited. Prelamin A accumulation is known to drive VSMC ageing and we show that presenescent VSMCs, that have accumulated prelamin A, display increased focal adhesion dynamics, augmented migrational velocity/persistence and attenuated Rac1 activity. Importantly, prelamin A accumulation in proliferative VSMCs, induced by depletion of the prelamin A processing enzyme FACE1, recapitulated the focal adhesion, migrational persistence and Rac1 phenotypes observed in presenescent VSMCs. Moreover, lamin A/C-depleted VSMCs also display reduced Rac1 activity, suggesting that prelamin A influences Rac1 activity by interfering with lamin A/C function at the nuclear envelope. Taken together, these data demonstrate that lamin A/C maintains Rac1 activity in VSMCs and prelamin A disrupts lamin A/C function to reduce Rac1 activity and induce migrational persistence during VSMC ageing.

10.
Nucleus ; 7(5): 498-511, 2016 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-27676213

RESUMEN

The accumulation of prelamin A is linked to disruption of cellular homeostasis, tissue degeneration and aging. Its expression is implicated in compromised genome stability and increased levels of DNA damage, but to date there is no complete explanation for how prelamin A exerts its toxic effects. As the nuclear lamina is important for DNA replication we wanted to investigate the relationship between prelamin A expression and DNA replication fork stability. In this study we report that the expression of prelamin A in U2OS cells induced both mono-ubiquitination of proliferating cell nuclear antigen (PCNA) and subsequent induction of Pol η, two hallmarks of DNA replication fork stalling. Immunofluorescence microscopy revealed that cells expressing prelamin A presented with high levels of colocalisation between PCNA and γH2AX, indicating collapse of stalled DNA replication forks into DNA double-strand breaks. Subsequent protein-protein interaction assays showed prelamin A interacted with PCNA and that its presence mitigated interactions between PCNA and the mature nuclear lamina. Thus, we propose that the cytotoxicity of prelamin A arises in part, from it actively competing against mature lamin A to bind PCNA and that this destabilises DNA replication to induce fork stalling which in turn contributes to genomic instability.


Asunto(s)
Replicación del ADN , Lamina Tipo A/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Línea Celular Tumoral , Daño del ADN , Dimerización , Histonas/metabolismo , Humanos , Unión Proteica , Transporte de Proteínas , Timina/metabolismo , Ubiquitinación
11.
Aging Cell ; 15(6): 1039-1050, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27464478

RESUMEN

The nuclear lamina is essential for the proper structure and organization of the nucleus. Deregulation of A-type lamins can compromise genomic stability, alter chromatin organization and cause premature vascular aging. Here, we show that accumulation of the lamin A precursor, prelamin A, inhibits 53BP1 recruitment to sites of DNA damage and increases basal levels of DNA damage in aged vascular smooth muscle cells. We identify that this genome instability arises through defective nuclear import of 53BP1 as a consequence of abnormal topological arrangement of nucleoporin NUP153. We show for the first time that this nucleoporin is important for the nuclear localization of Ran and that the deregulated Ran gradient is likely to be compromising the nuclear import of 53BP1. Importantly, many of the defects associated with prelamin A expression were significantly reduced upon treatment with Remodelin, a small molecule recently reported to reverse deficiencies associated with abnormal nuclear lamina.

12.
Exp Cell Res ; 345(2): 168-79, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27321956

RESUMEN

The spatial compartmentalisation of biochemical signalling pathways is essential for cell function. Nesprins are a multi-isomeric family of proteins that have emerged as signalling scaffolds, herein, we investigate the localisation and function of novel nesprin-2 N-terminal variants. We show that these nesprin-2 variants display cell specific distribution and reside in both the cytoplasm and nucleus. Immunofluorescence microscopy revealed that nesprin-2 N-terminal variants colocalised with ß-catenin at cell-cell junctions in U2OS cells. Calcium switch assays demonstrated that nesprin-2 and ß-catenin are lost from cell-cell junctions in low calcium conditions whereas emerin localisation at the NE remained unaltered, furthermore, an N-terminal fragment of nesprin-2 was sufficient for cell-cell junction localisation and interacted with ß-catenin. Disruption of these N-terminal nesprin-2 variants, using siRNA depletion resulted in loss of ß-catenin from cell-cell junctions, nuclear accumulation of active ß-catenin and augmented ß-catenin transcriptional activity. Importantly, we show that U2OS cells lack nesprin-2 giant, suggesting that the N-terminal nesprin-2 variants regulate ß-catenin signalling independently of the NE. Together, these data identify N-terminal nesprin-2 variants as novel regulators of ß-catenin signalling that tether ß-catenin to cell-cell contacts to inhibit ß-catenin transcriptional activity.


Asunto(s)
Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Humanos , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , Reproducibilidad de los Resultados , Transcripción Genética
13.
Methods Mol Biol ; 1411: 233-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27147046

RESUMEN

Matrix-derived mechanical cues influence cell proliferation, motility, and differentiation. Recent findings clearly demonstrate that the nuclear envelope (NE) adapts and remodels in response to mechanical signals, including matrix stiffness, yet a plethora of studies have been performed on tissue culture plastic or glass that have a similar stiffness to cortical bone. Using methods that allow modulation of matrix stiffness will provide further insight into the role of the NE in physiological conditions and the impact of changes in stiffness observed during ageing and disease on cellular function. In this chapter, we describe the polyacrylamide hydrogel system, which allows fabrication of hydrogels with variable stiffness to better mimic the environment experienced by cells in most tissues of the body.


Asunto(s)
Resinas Acrílicas , Matriz Extracelular/metabolismo , Hidrogeles , Membrana Nuclear/metabolismo , Fibroblastos/metabolismo , Mecanotransducción Celular , Microscopía de Fuerza Atómica/métodos , Miocitos del Músculo Liso/metabolismo
14.
Biochem Soc Trans ; 39(6): 1780-5, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22103525

RESUMEN

Accumulation of DNA damage is a major driving force of normal cellular aging and has recently been demonstrated to hasten the development of vascular diseases such as atherosclerosis. VSMCs (vascular smooth muscle cells) are essential for vessel wall integrity and repair, and maintenance of their proliferative capacity is essential for vascular health. The signalling pathways that determine VSMC aging remain poorly defined; however, recent evidence implicates persistent DNA damage and the A-type nuclear lamins as key regulators of this process. In the present review, we discuss the importance of the nuclear lamina in the spatial organization of nuclear signalling events, including the DNA-damage response. In particular, we focus on the evidence suggesting that prelamin A accumulation interferes with nuclear spatial compartmentalization by disrupting chromatin organization and DNA-damage repair pathways to promote VSMC aging and senescence.


Asunto(s)
Senescencia Celular , Daño del ADN , Reparación del ADN , Miocitos del Músculo Liso/patología , Lámina Nuclear/metabolismo , Animales , Humanos , Lamina Tipo A , Proteínas Nucleares/metabolismo , Precursores de Proteínas/metabolismo
15.
Curr Opin Cell Biol ; 23(1): 47-54, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21177090

RESUMEN

Like other spectrin repeat proteins, nesprins co-ordinate and maintain cellular architecture by linking membranous organelles to the cytoskeleton. However nuclear envelope (NE) nesprins, uniquely hardwire the nuclear lamina to the cytoskeleton and molecular motors. Emerging evidence suggests that nesprins also form a continuous network linking the plasma membrane to the NE that potentially translates mechanical stimuli into nuclear reorganisation. Surprisingly, this network is also essential for cytoskeletal organisation and its disruption has dramatic effects on nuclear migration, centrosomal positioning, focal adhesion maturation and cell motility. Herein we review recent advances in our understanding of how nesprins couple to various filamentous systems within the cell and emphasise the importance of both KASH and KASH-less nesprin isoforms in these interactions.


Asunto(s)
Núcleo Celular/metabolismo , Citoesqueleto/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Actinas/metabolismo , Animales , Humanos , Microtúbulos/metabolismo
16.
Circulation ; 121(20): 2200-10, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20458013

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome is a rare inherited disorder of premature aging caused by mutations in LMNA or Zmpste24 that disrupt nuclear lamin A processing, leading to the accumulation of prelamin A. Patients develop severe premature arteriosclerosis characterized by vascular smooth muscle cell (VSMC) calcification and attrition. METHODS AND RESULTS: To determine whether defective lamin A processing is associated with vascular aging in the normal population, we examined the profile of lamin A expression in normal and aged VSMCs. In vitro, aged VSMCs rapidly accumulated prelamin A coincidently with nuclear morphology defects, and these defects were reversible by treatment with farnesylation inhibitors and statins. In human arteries, prelamin A accumulation was not observed in young healthy vessels but was prevalent in medial VSMCs from aged individuals and in atherosclerotic lesions, where it often colocalized with senescent and degenerate VSMCs. Prelamin A accumulation correlated with downregulation of the lamin A processing enzyme Zmpste24/FACE1, and FACE1 mRNA and protein levels were reduced in response to oxidative stress. Small interfering RNA knockdown of FACE1 reiterated the prelamin A-induced nuclear morphology defects characteristic of aged VSMCs, and overexpression of prelamin A accelerated VSMC senescence. We show that prelamin A acts to disrupt mitosis and induce DNA damage in VSMCs, leading to mitotic failure, genomic instability, and premature senescence. CONCLUSIONS: This study shows that prelamin A is a novel biomarker of VSMC aging and disease that acts to accelerate senescence. It therefore represents a novel target to ameliorate the effects of age-induced vascular dysfunction.


Asunto(s)
Envejecimiento/fisiología , Vasos Sanguíneos/fisiología , Senescencia Celular/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Proteínas Nucleares/metabolismo , Precursores de Proteínas/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/metabolismo , División Celular/fisiología , Núcleo Celular/ultraestructura , Células Cultivadas , Daño del ADN , Regulación hacia Abajo , Humanos , Lamina Tipo A/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mitosis , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Estrés Oxidativo/fisiología , ARN Mensajero/metabolismo , Factores de Tiempo , Regulación hacia Arriba
17.
J Biol Chem ; 285(2): 1311-20, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19861416

RESUMEN

Nuclear and cytoplasmic scaffold proteins have been shown to be essential for temporal and spatial organization, as well as the fidelity, of MAPK signaling pathways. In this study we show that nesprin-2 is a novel extracellular signal-regulated MAPK1 and 2 (ERK1/2) scaffold protein that serves to regulate nuclear signaling by tethering these kinases at promyelocytic leukemia protein nuclear bodies (PML NBs). Using immunofluorescence microscopy, GST pull-down and immunoprecipitation, we show that nesprin-2, ERK1/2, and PML colocalize and bind to form a nuclear complex. Interference of nesprin-2 function, by either siRNA-mediated knockdown or overexpression of a dominant negative nesprin-2 fragment, augmented ERK1/2 nuclear signaling shown by increased SP1 activity and ELK1 phosphorylation. The functional outcome of nesprin-2 disruption and the resultant sustained ERK1/2 signal was increased proliferation. Importantly, these activities were not induced by previously identified nuclear envelope (NE)-targeted nesprin-2 isoforms but rather were mediated by novel nuclear isoforms that lacked the KASH domain. Taken together, this study suggests that nesprin-2 is a novel intranuclear scaffold, essential for nuclear ERK1/2 signaling fidelity and cell cycle progression.


Asunto(s)
Ciclo Celular/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de Microfilamentos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular , Humanos , Proteínas de Microfilamentos/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/genética , Membrana Nuclear/genética , Proteínas Nucleares/genética , Fosforilación/fisiología , Proteína de la Leucemia Promielocítica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína/fisiología , ARN Interferente Pequeño/genética , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
18.
Hum Mol Genet ; 16(23): 2816-33, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17761684

RESUMEN

Emery-Dreifuss muscular dystrophy (EDMD) is a heterogeneous late-onset disease involving skeletal muscle wasting and heart defects caused, in a minority of cases, by mutations in either of two genes encoding the inner nuclear membrane (INM) proteins, emerin and lamins A/C. Nesprin-1 and -2 are multi-isomeric, spectrin-repeat proteins that bind both emerin and lamins A/C and form a network in muscle linking the nucleoskeleton to the INM, the outer nuclear membrane, membraneous organelles, the sarcomere and the actin cytoskeleton. Thus, disruptions in nesprin/lamin/emerin interactions might play a role in the muscle-specific pathogenesis of EDMD. Screening for DNA variations in the genes encoding nesprin-1 (SYNE1) and nesprin-2 (SYNE2) in 190 probands with EDMD or EDMD-like phenotypes identified four heterozygous missense mutations. Fibroblasts from these patients exhibited nuclear morphology defects and specific patterns of emerin and SUN2 mislocalization. In addition, diminished nuclear envelope localization of nesprins and impaired nesprin/emerin/lamin binding interactions were common features of all EDMD patient fibroblasts. siRNA knockdown of nesprin-1 or -2 in normal fibroblasts reproduced the nuclear morphological changes and mislocalization of emerin and SUN2 observed in patient fibroblasts. Taken together, these data suggest that EDMD may be caused, in part, by uncoupling of the nucleoskeleton and cytoskeleton because of perturbed nesprin/emerin/lamin interactions.


Asunto(s)
Proteínas de Microfilamentos/genética , Distrofia Muscular de Emery-Dreifuss/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Proteínas del Citoesqueleto , ADN/genética , Análisis Mutacional de ADN , Femenino , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Heterocigoto , Humanos , Laminas/genética , Laminas/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Distrofia Muscular de Emery-Dreifuss/etiología , Distrofia Muscular de Emery-Dreifuss/metabolismo , Mutación Missense , Proteínas del Tejido Nervioso/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Linaje , ARN Interferente Pequeño/genética , Homología de Secuencia de Aminoácido
19.
Expert Rev Mol Med ; 7(11): 1-15, 2005 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-15953398

RESUMEN

Nesprins are a recently discovered family of ubiquitously expressed intracellular proteins. Through alternative transcriptional initiation, termination and splicing, two genes - nesprin-1 and nesprin-2 (also known as syne-1 and syne-2) - give rise to many protein isoforms that vary markedly in size. The largest of these isoforms comprise a C-terminal transmembrane domain (the KLS domain) linked by a spectrin-repeat rod domain to an N-terminal paired, actin-binding, calponin-homology domain. This structure suggests that they are well suited to orchestrate signalling between cell membranes and the cytoskeleton. Other isoforms have variable lengths of this rod domain linked to either end of the protein. Smaller isoforms with the KLS domain are localised at the inner nuclear membrane, where they bind lamin A/C and emerin. Larger nesprin isoforms link the outer nuclear membrane with intracellular organelles and the actin cytoskeleton and are thought to regulate nuclear anchorage and organelle migration. Thus, nesprins might have a variety of fundamental roles in cells, particularly muscle cells where they are highly expressed. We speculate that nesprin mutations might contribute to a broad range of human disease syndromes, including laminopathies.


Asunto(s)
Proteínas Nucleares/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Lamina Tipo A/metabolismo , Células Musculares/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína
20.
J Cell Sci ; 118(Pt 4): 673-87, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15671068

RESUMEN

Nesprin-2 is a multi-isomeric, modular protein composed of variable numbers of spectrin-repeats linked to a C-terminal transmembrane domain and/or to N-terminal paired calponin homology (CH) domains. The smaller isoforms of nesprin-2 co-localize with and bind lamin A and emerin at the inner nuclear envelope (NE). In SW-13 cells, which lack lamin A/C, nesprin-2 epitopes and emerin were both mislocalized and formed aggregates in the endoplasmic reticulum (ER). The larger isoforms and other CH-domain-containing isoforms co-localize with heterochromatin within the nucleus and are also present at the outer NE and in multiple cytoplasmic compartments. Nesprin-2 isoforms relocalize during in vitro muscle differentiation of C2C12 myoblasts to the sarcomere of myotubes. Immunogold electron microscopy using antibodies specific for three different epitopes detected nesprin-2 isoforms at multiple locations including intranuclear foci, both membranes of the NE, mitochondria, sarcomeric structures and plasma membrane foci. In adult skeletal muscle, confocal immunolocalization studies demonstrated that nesprin-2 epitopes were present at the Z-line and were also associated with the sarcoplasmic reticulum (SR) in close apposition to SERCA2. These data suggest that nesprin-2 isoforms form a linking network between organelles and the actin cytoskeleton and thus may be important for maintaining sub-cellular spatial organisation. Moreover, its association at the NE with lamin and emerin, the genes mutated in Emery-Dreifuss muscular dystrophy, suggests a mechanism to explain how disruption of the NE leads to muscle dysfunction.


Asunto(s)
Laminas/metabolismo , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Timopoyetinas/metabolismo , Citoesqueleto de Actina/química , Empalme Alternativo , Sitios de Unión , Fraccionamiento Celular , Humanos , Inmunoprecipitación , Proteínas de Microfilamentos , Músculo Esquelético/química , Músculo Esquelético/ultraestructura , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Isoformas de Proteínas/análisis , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...