Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
BMC Cell Biol ; 17(1): 26, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27350129

RESUMEN

BACKGROUND: Nesprin-1-giant (1008kD) is a protein of the outer nuclear membrane that links nuclei to the actin cytoskeleton via amino-terminal calponin homology domains. The short nesprin-1 isoform, nesprin-1-α2, is present only in skeletal and cardiac muscle and several pathogenic mutations occur within it, but the functions of this short isoform without calponin homology domains are unclear. The aim of this study was to determine mRNA levels and protein localization of nesprin-1-α2 at different stages of muscle development in order to shed light on its functions. RESULTS: mRNA levels of all known nesprin-1 isoforms with a KASH domain were determined by quantitative PCR. The mRNA for the 111 kD muscle-specific short isoform, nesprin-1-α2, was not detected in pre-differentiation human myoblasts but was present at significant levels in multinucleate myotubes. We developed a monoclonal antibody against the unique amino-terminal sequence of nesprin-1-α2, enabling specific immunolocalization for the first time. Nesprin-1-α2 protein was undetectable in pre-differentiation myoblasts but appeared at the nuclear rim in post-mitotic, multinucleate myotubes and reached its highest levels in fetal muscle. In muscle from a Duchenne muscular dystrophy biopsy, nesprin-1-α2 protein was detected mainly in regenerating fibres expressing neonatal myosin. Nesprin-1-giant was present at all developmental stages, but was also highest in fetal and regenerating fibres. In fetal muscle, both isoforms were present in the cytoplasm, as well as at the nuclear rim. A pathogenic early stop codon (E7854X) in nesprin-1 caused reduced mRNA levels and loss of protein levels of both nesprin-1-giant and (unexpectedly) nesprin-1-α2, but did not affect myogenesis in vitro. CONCLUSIONS: Nesprin-1-α2 mRNA and protein expression is switched on during myogenesis, alongside other known markers of muscle differentiation. The results show that nesprin-1-α2 is dynamically controlled and may be involved in some process occurring during early myofibre formation, such as re-positioning of nuclei.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Feto/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Desarrollo de Músculos , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Regeneración , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Portadoras/genética , Núcleo Celular/metabolismo , Células Cultivadas , Niño , Preescolar , Proteínas del Citoesqueleto , Femenino , Humanos , Recién Nacido , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Mutación/genética , Mioblastos/metabolismo , Proteínas del Tejido Nervioso , Péptidos/metabolismo , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN , Adulto Joven
3.
Hum Mol Genet ; 23(6): 1551-62, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24179176

RESUMEN

Myotonic dystrophy (DM) is a multi-system neuromuscular disorder for which there is no treatment. We have developed a medium throughput phenotypic assay, based on the identification of nuclear foci in DM patient cell lines using in situ hybridization and high-content imaging to screen for potentially useful therapeutic compounds. A series of further assays based on molecular features of DM have also been employed. Two compounds that reduce and/or remove nuclear foci have been identified, Ro 31-8220 and chromomycin A3. Ro 31-8220 is a PKC inhibitor, previously shown to affect the hyperphosphorylation of CELF1 and ameliorate the cardiac phenotype in a DM1 mouse model. We show that the same compound eliminates nuclear foci, reduces MBNL1 protein in the nucleus, affects ATP2A1 alternative splicing and reduces steady-state levels of CELF1 protein. We demonstrate that this effect is independent of PKC activity and conclude that this compound may be acting on alternative kinase targets within DM pathophysiology. Understanding the activity profile for this compound is key for the development of targeted therapeutics in the treatment of DM.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Cromomicina A3/farmacología , Indoles/farmacología , Distrofia Miotónica/patología , Proteínas de Unión al ARN/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Empalme Alternativo , Animales , Proteínas CELF1 , Núcleo Celular/patología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Biblioteca de Péptidos , Proteínas de Unión al ARN/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transducción de Señal/efectos de los fármacos , Pez Cebra
4.
Hum Mol Genet ; 20(12): 2406-21, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21478199

RESUMEN

Apolipoprotein E (apoE) is a 34 kDa glycoprotein with three distinct isoforms in the human population (apoE2, apoE3 and apoE4) known to play a major role in differentially influencing risk to, as well as outcome from, disease and injury in the central nervous system. In general, the apoE4 allele is associated with poorer outcomes after disease or injury, whereas apoE3 is associated with better responses. The extent to which different apoE isoforms influence degenerative and regenerative events in the peripheral nervous system (PNS) is still to be established, and the mechanisms through which apoE exerts its isoform-specific effects remain unclear. Here, we have investigated isoform-specific effects of human apoE on the mouse PNS. Experiments in mice ubiquitously expressing human apoE3 or human apoE4 on a null mouse apoE background revealed that apoE4 expression significantly disrupted peripheral nerve regeneration and subsequent neuromuscular junction re-innervation following nerve injury compared with apoE3, with no observable effects on normal development, maturation or Wallerian degeneration. Proteomic isobaric tag for relative and absolute quantitation (iTRAQ) screens comparing healthy and regenerating peripheral nerves from mice expressing apoE3 or apoE4 revealed significant differences in networks of proteins regulating cellular outgrowth and regeneration (myosin/actin proteins), as well as differences in expression levels of proteins involved in regulating the blood-nerve barrier (including orosomucoid 1). Taken together, these findings have identified isoform-specific roles for apoE in determining the protein composition of peripheral nerve as well as regulating nerve regeneration pathways in vivo.


Asunto(s)
Apolipoproteínas E/metabolismo , Regeneración Nerviosa/fisiología , Sistema Nervioso Periférico/fisiología , Isoformas de Proteínas/metabolismo , Animales , Apolipoproteínas E/genética , Axones/metabolismo , Axones/ultraestructura , Western Blotting , Electrofisiología , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Orosomucoide/metabolismo , Sistema Nervioso Periférico/lesiones , Isoformas de Proteínas/genética , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem
5.
Biochem Biophys Res Commun ; 438(4): 624-7, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23939045

RESUMEN

Reduced levels of SMN (survival-of-motor-neurons) protein are the cause of spinal muscular atrophy, an inherited disorder characterised by loss of motor neurons in early childhood. SMN associates with more than eight other proteins to form an RNA-binding complex involved in assembly of the spliceosome. Two monoclonal antibodies (mAbs), MANSMA1 and MANSMA12, have been widely-used in studies of SMN function and their precise binding sites on SMN have now been identified using a phage-displayed peptide library. The amino-acid residues in SMN required for antibody binding are the same as the five most important contact residues for interaction with gemin2. MANSMA12 immuno-precipitated SMN and gemin2 from HeLa cell extracts as efficiently as mAbs against other SMN epitopes or against gemin2. We explain this by showing that SMN exists as large multimeric complexes. This SMN epitope is highly-conserved and identical in human and mouse. To explain the vigorous immune response when mice are immunised with recombinant SMN alone, we suggest this region is masked by gemin2, or a related protein, throughout development, preventing its recognition as a "self-antigen". The epitope for a third mAb, MANSMA3, has been located to eight amino-acids in the proline-rich domain of SMN.


Asunto(s)
Proteínas del Complejo SMN/química , Proteínas del Complejo SMN/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Sitios de Unión , Mapeo Epitopo , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Proteínas del Complejo SMN/inmunología
6.
J Biol Chem ; 286(44): 38427-38438, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21900255

RESUMEN

To understand the role of the splice regulator muscleblind 1 (MBNL1) in the development of RNA splice defects in myotonic dystrophy I (DM1), we purified RNA-independent MBNL1 complexes from normal human myoblasts and examined the behavior of these complexes in DM1 myoblasts. Antibodies recognizing MBNL1 variants (MBNL1(CUG)), which can sequester in the toxic CUG RNA foci that develop in DM1 nuclei, were used to purify MBNL1(CUG) complexes from normal myoblasts. In normal myoblasts, MBNL1(CUG) bind 10 proteins involved in remodeling ribonucleoprotein complexes including hnRNP H, H2, H3, F, A2/B1, K, L, DDX5, DDX17, and DHX9. Of these proteins, only MBNL1(CUG) colocalizes extensively with DM1 CUG foci (>80% of foci) with its partners being present in <10% of foci. Importantly, the stoichiometry of MBNL1(CUG) complexes is altered in DM1 myoblasts, demonstrating an increase in the steady state levels of nine of its partner proteins. These changes are recapitulated by the expression of expanded CUG repeat RNA in Cos7 cells. Altered stoichiometry of MBNL1(CUG) complexes results from aberrant protein synthesis or stability and is unlinked to PKCα function. Modeling these changes in normal myoblasts demonstrates that increased levels of hnRNP H, H2, H3, F, and DDX5 independently dysregulate splicing in overlapping RNA subsets. Thus expression of expanded CUG repeats alters the stoichiometry of MBNL1(CUG) complexes to allow both the reinforcement and expansion of RNA processing defects.


Asunto(s)
Distrofia Miotónica/metabolismo , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Inmunoprecipitación , Espectrometría de Masas/métodos , Músculo Esquelético/metabolismo , Distrofia Miotónica/genética , Mapeo de Interacción de Proteínas/métodos , ARN Interferente Pequeño/metabolismo , Fracciones Subcelulares
7.
J Biol Chem ; 286(18): 16435-46, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21454535

RESUMEN

Muscleblind-like-1 (MBNL1) is a splicing regulatory factor controlling the fetal-to-adult alternative splicing transitions during vertebrate muscle development. Its capture by nuclear CUG expansions is one major cause for type 1 myotonic dystrophy (DM1). Alternative splicing produces MBNL1 isoforms that differ by the presence or absence of the exonic regions 3, 5, and 7. To understand better their respective roles and the consequences of the deregulation of their expression in DM1, here we studied the respective roles of MBNL1 alternative and constitutive exons. By combining genetics, molecular and cellular approaches, we found that (i) the exon 5 and 6 regions are both needed to control the nuclear localization of MBNL1; (ii) the exon 3 region strongly enhances the affinity of MBNL1 for its pre-mRNA target sites; (iii) the exon 3 and 6 regions are both required for the splicing regulatory activity, and this function is not enhanced by an exclusive nuclear localization of MBNL1; and finally (iv) the exon 7 region enhances MBNL1-MBNL1 dimerization properties. Consequently, the abnormally high inclusion of the exon 5 and 7 regions in DM1 is expected to enhance the potential of MBNL1 of being sequestered with nuclear CUG expansions, which provides new insight into DM1 pathophysiology.


Asunto(s)
Empalme Alternativo , Núcleo Celular/metabolismo , Exones , Multimerización de Proteína , Proteínas de Unión al ARN/metabolismo , Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Núcleo Celular/patología , Células HeLa , Humanos , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Distrofia Miotónica/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/genética , Expansión de Repetición de Trinucleótido
8.
Biochem Biophys Res Commun ; 424(2): 354-7, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22771323

RESUMEN

Mutations in the gene encoding fukutin protein cause Fukuyama muscular dystrophy, a severe congenital disorder that occurs mainly in Japan. A major consequence of the mutation is reduced glycosylation of alpha-dystroglycan, which is also a feature of other forms of congenital and limb-girdle muscular dystrophy. Immunodetection of endogenous fukutin in cells and tissues has been difficult and this has hampered progress in understanding fukutin function and disease pathogenesis. Using a new panel of monoclonal antibodies which bind to different defined sites on the fukutin molecule, we now show that fukutin has the predicted size for a protein without extensive glycosylation and is present at the Golgi apparatus at very low levels. These antibodies should enable more rapid future progress in understanding the molecular function of fukutin.


Asunto(s)
Anticuerpos Monoclonales , Proteínas de la Membrana/análisis , Síndrome de Walker-Warburg/diagnóstico , Secuencia de Aminoácidos , Animales , Mapeo Epitopo , Glicosilación , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Hibridomas , Epítopos Inmunodominantes/análisis , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Biblioteca de Péptidos , Síndrome de Walker-Warburg/genética , Síndrome de Walker-Warburg/metabolismo
9.
PLoS Genet ; 5(5): e1000482, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19436705

RESUMEN

Myotonic dystrophy type 1 (DM1) is associated with one of the most highly unstable CTG*CAG repeat expansions. The formation of further repeat expansions in transgenic mice carrying expanded CTG*CAG tracts requires the mismatch repair (MMR) proteins MSH2 and MSH3, forming the MutSbeta complex. It has been proposed that binding of MutSbeta to CAG hairpins blocks its ATPase activity compromising hairpin repair, thereby causing expansions. This would suggest that binding, but not ATP hydrolysis, by MutSbeta is critical for trinucleotide expansions. However, it is unknown if the MSH2 ATPase activity is dispensible for instability. To get insight into the mechanism by which MSH2 generates trinucleotide expansions, we crossed DM1 transgenic mice carrying a highly unstable >(CTG)(300) repeat tract with mice carrying the G674A mutation in the MSH2 ATPase domain. This mutation impairs MSH2 ATPase activity and ablates base-base MMR, but does not affect the ability of MSH2 (associated with MSH6) to bind DNA mismatches. We found that the ATPase domain mutation of MSH2 strongly affects the formation of CTG expansions and leads instead to transmitted contractions, similar to a Msh2-null or Msh3-null deficiency. While a decrease in MSH2 protein level was observed in tissues from Msh2(G674) mice, the dramatic reduction of expansions suggests that the expansion-biased trinucleotide repeat instability requires a functional MSH2 ATPase domain and probably a functional MMR system.


Asunto(s)
Inestabilidad Genómica , Proteína 2 Homóloga a MutS/química , Proteína 2 Homóloga a MutS/genética , Mutación Puntual , Expansión de Repetición de Trinucleótido , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Secuencia de Bases , Encéfalo/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Cartilla de ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína 2 Homóloga a MutS/deficiencia , Proteína 2 Homóloga a MutS/metabolismo , Proteína 3 Homóloga de MutS , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Proteína Quinasa de Distrofia Miotónica , Ovario/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Proteínas/genética , Proteínas/metabolismo , Testículo/metabolismo
10.
Hum Mutat ; 32(4): 389-96, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21309037

RESUMEN

Mutations in the mismatch repair gene MSH2 underlie hereditary nonpolyposis colorectal cancer (Lynch syndrome). Whereas disruptive mutations are overtly pathogenic, the implications of missense mutations found in sporadic colorectal cancer patients or in suspected Lynch syndrome families are often unknown. Adequate genetic counseling of mutation carriers requires phenotypic characterization of the variant allele. We present a novel approach to functionally characterize MSH2 missense mutations. Our approach involves introduction of the mutation into the endogenous gene of murine embryonic stem cells (ESC) by oligonucleotide-directed gene modification, a technique we recently developed in our lab. Subsequently, the mismatch repair capacity of mutant ESC is determined using a set of validated functional assays. We have evaluated four clinically relevant MSH2 variants and found one to completely lack mismatch repair capacity while three behaved as wild-type MSH2 and can therefore be considered as polymorphisms. Our approach contributes to an adequate risk assessment of mismatch repair missense mutations. We have also shown that oligonucleotide-directed gene modification provides a straightforward approach to recreate allelic variants in the endogenous gene in murine ESC. This approach can be extended to other hereditary conditions.


Asunto(s)
Células Madre Embrionarias/metabolismo , Variación Genética , Proteína 2 Homóloga a MutS/genética , Alelos , Animales , Células Madre Embrionarias/citología , Humanos , Ratones , Inestabilidad de Microsatélites
11.
J Cell Biochem ; 112(6): 1612-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21344488

RESUMEN

The mismatch repair protein, MSH3, together with MSH2, forms the MutSß heterodimer which recognizes and repairs base pair mismatches and larger insertion/deletion loops in DNA. Lack of specific antibodies against mouse MSH3 has hampered studies of its expression and localization. Mouse MSH3 is not immunogenic in normal mice. This problem was overcome by immunizing msh3-knockout mice and generating a panel of ten monoclonal antibodies, two of which localize MSH3 specifically in cultured mouse cells and bind to an epitope containing amino-acids 33-37. The panel also includes two antibodies that recognise both mouse and human MSH3 and bind to a conserved epitope containing amino-acids 187-194. The mouse MSH3-specific antibodies show that MSH3 is a nuclear protein with a finely-granular nucleoplasmic distribution, largely absent from areas of condensed heterochromatin. Specificity of the localization was demonstrated by absence of immunostaining in a cell line from the msh3-knockout mouse. Furthermore, we show for the first time that stress treatment of mouse cells with ethanol or hydrogen peroxide caused the re-distribution of MSH3 into nuclear bodies containing the proliferating cell nuclear antigen (PCNA), a known binding partner of MutSß.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas/metabolismo , Animales , Anticuerpos Monoclonales , Western Blotting , Línea Celular , Núcleo Celular/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Células HeLa , Humanos , Hibridomas , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Noqueados , Proteína 3 Homóloga de MutS , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas/genética
12.
Biochem Biophys Res Commun ; 412(2): 291-5, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21820406

RESUMEN

The nuclear envelope-associated cytoskeletal protein, nesprin-2, is encoded by a large gene containing several internal promoters that produce shorter isoforms. In a study of Ntera-2 teratocarcinoma cells, a novel isoform, nesprin-2-epsilon, was found to be the major mRNA and protein product of the nesprin-2 gene. Its existence was predicted by bioinformatic analysis, but this is the first direct demonstration of both the mRNA and the 120 kDa protein which is located at the nuclear envelope. In a panel of 21 adult and foetal human tissues, the nesprin-2-epsilon mRNA was strongly expressed in ovary but was a minor isoform elsewhere. The expression pattern suggests a possible link with very early development and a likely physiological role in ovary.


Asunto(s)
Proteínas de Microfilamentos/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Ovario/metabolismo , Línea Celular Tumoral , Femenino , Expresión Génica , Humanos , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Ovario/crecimiento & desarrollo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética
13.
Nat Med ; 9(8): 1009-14, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12847521

RESUMEN

As a target for gene therapy, Duchenne muscular dystrophy (DMD) presents many obstacles but also an unparalleled prospect for correction by alternative splicing. The majority of mutations in the dystrophin gene occur in the region encoding the spectrin-like central rod domain, which is largely dispensable. Thus, splicing around mutations can generate a shortened but in-frame transcript, permitting translation of a partially functional dystrophin protein. We have tested this idea in vivo in the mdx dystrophic mouse (carrying a mutation in exon 23 of the dystrophin gene) by combining a potent transfection protocol with a 2-O-methylated phosphorothioated antisense oligoribonucleotide (2OMeAO) designed to promote skipping of the mutated exon*. The treated mice show persistent production of dystrophin at normal levels in large numbers of muscle fibers and show functional improvement of the treated muscle. Repeated administration enhances dystrophin expression without eliciting immune responses. Our data establishes the realistic practicality of an approach that is applicable, in principle, to a majority of cases of severe dystrophinopathy.


Asunto(s)
Distrofina/genética , Exones , Terapia Genética/métodos , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Mutación , Animales , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Oligonucleótidos Antisentido/metabolismo , Empalme del ARN
14.
Dev Dyn ; 239(3): 998-1009, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20108321

RESUMEN

Nesprins are a family of nuclear transmembrane proteins anchored via Sun proteins to the nuclear membrane. Analysis of nesprins during human muscle development revealed an increase in nesprin-1-giant during early myogenesis in vitro. During the transition from immature to mature muscle fibres in vivo, nesprin-2 partly replaced nesprin-1 at the nuclear envelope and short nesprin isoforms became dominant. Sun1 and Sun2 proteins remained unchanged during this fibre maturation. In emerin-negative skin fibroblasts, nesprin-2-giant was relocated from the nuclear envelope to the cytoplasm, not to the endoplasmic reticulum, while nesprin-1 remained at the nuclear envelope. In emerin-negative keratinocytes lacking nesprin-1, nesprin-2 remained at the nuclear envelope. HeLa cell nuclear envelopes lacked nesprin-1, which was the dominant form in myoblasts, while a novel 130-kD nesprin-2 isoform dominated Ntera-2 cells. The results suggest the possibility of isoform-specific and tissue-specific roles for nesprins in nuclear positioning.


Asunto(s)
Proteínas de Microfilamentos/química , Músculos/embriología , Proteínas del Tejido Nervioso/química , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Animales , Anticuerpos Monoclonales/química , Núcleo Celular/metabolismo , Proteínas del Citoesqueleto , Fibroblastos/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas , Proteínas de Unión a Telómeros/metabolismo
15.
J Proteome Res ; 9(8): 4228-33, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20568814

RESUMEN

Valproate is commonly used as an anticonvulsant and mood stabilizer, but its long-term side-effects can include bone loss. As a histone deacetylase (HDAC) inhibitor, valproate has also been considered for treatment of spinal muscular atrophy (SMA). Using iTRAQ labeling technology, followed by two-dimensional liquid chromatography and mass spectrometry analysis, a quantitative comparison of the proteome of an SMA cell line, with and without valproate treatment, was performed. The most striking change was a reduction in collagens I and VI, while over 1000 other proteins remained unchanged. The collagen I alpha-chain precursor was also reduced by more than 50% suggesting that valproate affects collagen I synthesis. The collagen-binding glycoprotein, osteonectin (SPARC, BM-40) was one of the few other proteins that were significantly reduced by valproate treatment. Collagen I is the main protein component of bone matrix and osteonectin has a major role in bone development, so the results suggest a possible molecular mechanism for bone loss following long-term exposure to valproate. SMA patients may already suffer bone weakness as a result of SMN1 gene deletion, so further bone loss would be undesirable.


Asunto(s)
Enfermedades Óseas Metabólicas/inducido químicamente , Colágeno/metabolismo , Inhibidores de Histona Desacetilasas/efectos adversos , Atrofia Muscular Espinal/tratamiento farmacológico , Osteonectina/metabolismo , Proteómica/métodos , Ácido Valproico/efectos adversos , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Espectrometría de Masas , Piel/citología
16.
J Proteome Res ; 9(1): 556-63, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19928837

RESUMEN

Understanding networks of interacting proteins is a major goal in cell biology. The survival of motor neurons protein (SMN) interacts, directly or indirectly, with a large number of other proteins and reduced levels of SMN cause the inherited disorder spinal muscular atrophy (SMA). Some SMN interactions are stable and stoichiometric, such as those with gemins, while others are expected to be transient and substoichiometric, such as the functional interaction of SMN with coilin in Cajal bodies. This study set out to determine whether novel components of the extensive SMN interactome can be identified by a proteomic approach. SMN complexes were immuno-precipitated from HeLa nuclear extracts, using anti-SMN monoclonal antibody attached to magnetic beads, digested with trypsin, separated by capillary-liquid chromatography and analyzed by MALDI TOF/TOF mass spectrometry. One-hundred and one proteins were detected with a p value of <0.05, SMN, gemins and U snRNPs being the dominant "hits". Sixty-nine of these were rejected after MALDI analysis of two control pull-downs using antibodies against unrelated nuclear proteins. The proteins found only in anti-SMN pulldowns were either known SMN partners, and/or contained dimethylated RG domains involved in direct interaction with the SMN tudor domain, or they were known binding partners of such direct SMN interactors. Myb-binding protein 1a, identified as a novel candidate, is a mainly nucleolar protein of unknown function but it partially colocalized with SMN in Cajal bodies in HeLa cell nucleoplasm and, like SMN, was reduced in cells from an SMA patient.


Asunto(s)
Núcleo Celular/química , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteoma/análisis , Proteínas del Complejo SMN/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN , Células HeLa , Humanos , Inmunohistoquímica , Inmunoprecipitación , Atrofia Muscular Espinal/metabolismo , Proteínas Nucleares/química , Proteínas de Transporte Nucleocitoplasmático/química , Proteínas de Unión al ARN , Ratas , Ribonucleoproteínas/metabolismo , Proteínas del Complejo SMN/química , Empalmosomas/metabolismo , Factores de Transcripción
17.
Hum Mol Genet ; 17(8): 1063-75, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18178576

RESUMEN

Spinal muscular atrophy (SMA) is caused by loss of the survival motor neuron gene (SMN1) and retention of the SMN2 gene. The copy number of SMN2 affects the amount of SMN protein produced and the severity of the SMA phenotype. While loss of mouse Smn is embryonic lethal, two copies of SMN2 prevents this embryonic lethality resulting in a mouse with severe SMA that dies 5 days after birth. Here we show that expression of full-length SMN under the prion promoter (PrP) rescues severe SMA mice. The PrP results in high levels of SMN in neurons at embryonic day 15. Mice homozygous for PrP-SMN with two copies of SMN2 and lacking mouse Smn survive for an average of 210 days and lumbar motor neuron root counts in these mice were normal. Expression of SMN solely in skeletal muscle using the human skeletal actin (HSA) promoter resulted in no improvement of the SMA phenotype or extension of survival. One HSA line displaying nerve expression of SMN did affect the SMA phenotype with mice living for an average of 160 days. Thus, we conclude that expression of full-length SMN in neurons can correct the severe SMA phenotype in mice. Furthermore, a small increase of SMN in neurons has a substantial impact on survival of SMA mice while high SMN levels in mature skeletal muscle alone has no impact.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Atrofia Muscular Espinal/genética , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Animales , Embrión de Mamíferos/metabolismo , Dosificación de Gen , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Neuronas , Regiones Promotoras Genéticas , Proteínas del Complejo SMN , Análisis de Supervivencia , Proteína 1 para la Supervivencia de la Neurona Motora , Proteína 2 para la Supervivencia de la Neurona Motora
18.
Biochem Soc Trans ; 38(Pt 1): 278-80, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20074074

RESUMEN

The giant isoforms of nesprins 1 and 2 are emerging as important players in cellular organization, particularly in the positioning of nuclei, and possibly other organelles, within the cytoplasm. The experimental evidence suggests that nesprins also occur at the inner nuclear membrane, where they interact with the nuclear lamina. In this paper, we consider whether this is consistent with current ideas about nesprin anchorage and about mechanisms for nuclear import of membrane proteins.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Proteínas del Citoesqueleto , Humanos , Membrana Nuclear/ultraestructura
19.
Am J Pathol ; 174(1): 216-27, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19095965

RESUMEN

In myotonic dystrophy, muscleblind-like protein 1 (MBNL1) protein binds specifically to expanded CUG or CCUG repeats, which accumulate as discrete nuclear foci, and this is thought to prevent its function in the regulation of alternative splicing of pre-mRNAs. There is strong evidence for the role of the MBNL1 gene in disease pathology, but the roles of two related genes, MBNL2 and MBNL3, are less clear. Using new monoclonal antibodies specific for each of the three gene products, we found that MBNL2 decreased during human fetal development and myoblast culture, while MBNL1 was unchanged. In Duchenne muscular dystrophy muscle, MBNL2 was elevated in immature, regenerating fibres compared with mature fibres, supporting some developmental role for MBNL2. MBNL3 was found only in C2C12 mouse myoblasts. Both MBNL1 and MBNL2 were partially sequestered by nuclear foci of expanded repeats in adult muscle and cultured cells from myotonic dystrophy patients. In adult muscle nucleoplasm, both proteins were reduced in myotonic dystrophy type 1 compared with an age-matched control. In normal human myoblast cultures, MBNL1 and MBNL2 always co-distributed but their distribution could change rapidly from nucleoplasmic to cytoplasmic. Functional differences between MBNL1 and MBNL2 have not yet been found and may prove quite subtle. The dominance of MBNL1 in mature, striated muscle would explain why ablation of the mouse mbnl1 gene alone is sufficient to cause a myotonic dystrophy.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofia Miotónica/metabolismo , Proteínas de Unión al ARN/metabolismo , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Western Blotting , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma , Electroforesis en Gel de Poliacrilamida , Feto , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Persona de Mediana Edad , Músculo Esquelético/embriología , Mioblastos/citología , Mioblastos/metabolismo , Transporte de Proteínas/fisiología , ARN Interferente Pequeño , Transfección
20.
Neuromuscul Disord ; 30(6): 443-456, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32522500

RESUMEN

Emery-Dreifuss muscular dystrophy (EDMD) is a rare genetic disorder characterised by the early development of muscle contractures, progressive muscle weakness, and heart abnormalities. The latter may result in serious complications, or in severe cases, sudden death. Currently, there are very few effective treatment options available for EDMD and so there is a high clinical need for new therapies. Various genetic mutations have been identified in the development and causation of EDMD, each encoding proteins that are components of the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex, which spans the nuclear envelope and serves to connect the nuclear lamina to the cytoskeleton. Within this review, we examine how mutations in the genes encoding these proteins, including lamins A/C, emerin, nesprins 1/2, FHL1, and SUN1/2 lead to muscle cell differentiation and development pathway defects. Further work to identify conserved molecular pathways downstream of these defective proteins may reveal potential targets for therapy design.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/genética , Células Musculares/fisiología , Proteínas Musculares/genética , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/fisiopatología , Proteínas Nucleares/genética , Transducción de Señal/genética , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA