Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuropediatrics ; 54(5): 351-355, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36603837

RESUMEN

OBJECTIVE: By loading transfer RNAs with their cognate amino acids, aminoacyl-tRNA synthetases (ARS) are essential for protein translation. Both cytosolic ARS1-deficiencies and mitochondrial ARS2 deficiencies can cause severe diseases. Amino acid supplementation has shown to positively influence the clinical course of four individuals with cytosolic ARS1 deficiencies. We hypothesize that this intervention could also benefit individuals with mitochondrial ARS2 deficiencies. METHODS: This study was designed as a N-of-1 trial. Daily oral L-phenylalanine supplementation was used in a 3-year-old girl with FARS2 deficiency. A period without supplementation was implemented to discriminate the effects of treatment from age-related developments and continuing physiotherapy. Treatment effects were measured through a physiotherapeutic testing battery, including movement assessment battery for children, dynamic gait index, gross motor function measure 66, and quality of life questionnaires. RESULTS: The individual showed clear improvement in all areas tested, especially in gross motor skills, movement abilities, and postural stability. In the period without supplementation, she lost newly acquired motor skills but regained these upon restarting supplementation. No adverse effects and good tolerance of treatment were observed. INTERPRETATION AND CONCLUSION: Our positive results encourage further studies both on L-phenylalanine for other individuals with FARS2 deficiency and the exploration of this treatment rationale for other ARS2 deficiencies. Additionally, treatment costs were relatively low at 1.10 €/day.


Asunto(s)
Fenilalanina-ARNt Ligasa , Niño , Femenino , Humanos , Preescolar , Fenilalanina-ARNt Ligasa/genética , Fenilalanina-ARNt Ligasa/química , Fenilalanina-ARNt Ligasa/metabolismo , Fenilalanina/metabolismo , Calidad de Vida , Mitocondrias/genética , Mitocondrias/metabolismo , ARN de Transferencia/metabolismo , Proteínas Mitocondriales/genética
2.
Int J Mol Sci ; 24(3)2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36768204

RESUMEN

Inherited deficiency in ether lipids, a subgroup of glycerophospholipids with unique biochemical and biophysical properties, evokes severe symptoms in humans resulting in a multi-organ syndrome. Mouse models with defects in ether lipid biosynthesis have widely been used to understand the pathophysiology of human disease and to study the roles of ether lipids in various cell types and tissues. However, little is known about the function of these lipids in cardiac tissue. Previous studies included case reports of cardiac defects in ether-lipid-deficient patients, but a systematic analysis of the impact of ether lipid deficiency on the mammalian heart is still missing. Here, we utilize a mouse model of complete ether lipid deficiency (Gnpat KO) to accomplish this task. Similar to a subgroup of human patients with rhizomelic chondrodysplasia punctata (RCDP), a fraction of Gnpat KO fetuses present with defects in ventricular septation, presumably evoked by a developmental delay. We did not detect any signs of cardiomyopathy but identified increased left ventricular end-systolic and end-diastolic pressure in middle-aged ether-lipid-deficient mice. By comprehensive electrocardiographic characterization, we consistently found reduced ventricular conduction velocity, as indicated by a prolonged QRS complex, as well as increased QRS and QT dispersion in the Gnpat KO group. Furthermore, a shift of the Wenckebach point to longer cycle lengths indicated depressed atrioventricular nodal function. To complement our findings in mice, we analyzed medical records and performed electrocardiography in ether-lipid-deficient human patients, which, in contrast to the murine phenotype, indicated a trend towards shortened QT intervals. Taken together, our findings demonstrate that the cardiac phenotype upon ether lipid deficiency is highly heterogeneous, and although the manifestations in the mouse model only partially match the abnormalities in human patients, the results add to our understanding of the physiological role of ether lipids and emphasize their importance for proper cardiac development and function.


Asunto(s)
Éter , Plasmalógenos , Animales , Humanos , Ratones , Éteres , Éteres de Etila , Corazón , Mamíferos/metabolismo
3.
Neuropediatrics ; 52(5): 377-382, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33511595

RESUMEN

BACKGROUND: Recent research suggested an hippocalcin (HPCA)-related form of DYT2-like autosomal recessive dystonia. Two reports highlight a broad spectrum of the clinical phenotype. Here, we describe a novel HPCA gene variant in a pediatric patient and two affected relatives. METHODS: Whole exome sequencing was applied after a thorough clinical and neurological examination of the index patient and her family members. Results of neuropsychological testing were analyzed. RESULTS: Whole exome sequencing revealed a novel homozygous missense variant in the HPCA gene [c.182C>T p.(Ala61Val)] in our pediatric patient and the two affected family members. Clinically, the cases presented with dystonia, dysarthria, and jerky movements. We observed a particular cognitive profile with executive dysfunctions in our patient, which corresponds to the cognitive deficits that have been observed in the patients previously described. CONCLUSION: We present a novel genetic variant of the HPCA gene associated with autosomal recessive dystonia in a child with childhood-onset dystonia supporting its clinical features. Furthermore, we propose specific HPCA-related cognitive changes in homozygous carriers, underlining the importance of undertaking a systematic assessment of cognition in HPCA-related dystonia.


Asunto(s)
Distonía , Trastornos Distónicos , Niño , Cognición , Distonía/genética , Trastornos Distónicos/genética , Femenino , Hipocalcina/genética , Hipocalcina/metabolismo , Humanos , Mutación
4.
J Physiol ; 598(8): 1591-1609, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32003874

RESUMEN

KEY POINTS: Muscular dystrophy patients suffer from progressive degeneration of skeletal muscle fibres, sudden spontaneous falls, balance problems, as well as gait and posture abnormalities. Dystrophin- and dysferlin-deficient mice, models for different types of muscular dystrophy with different aetiology and molecular basis, were characterized to investigate if muscle spindle structure and function are impaired. The number and morphology of muscle spindles were unaltered in both dystrophic mouse lines but muscle spindle resting discharge and their responses to stretch were altered. In dystrophin-deficient muscle spindles, the expression of the paralogue utrophin was substantially upregulated, potentially compensating for the dystrophin deficiency. The results suggest that muscle spindles might contribute to the motor problems observed in patients with muscular dystrophy. ABSTRACT: Muscular dystrophies comprise a heterogeneous group of hereditary diseases characterized by progressive degeneration of extrafusal muscle fibres as well as unstable gait and frequent falls. To investigate if muscle spindle function is impaired, we analysed their number, morphology and function in wildtype mice and in murine model systems for two distinct types of muscular dystrophy with very different disease aetiology, i.e. dystrophin- and dysferlin-deficient mice. The total number and the overall structure of muscle spindles in soleus muscles of both dystrophic mouse mutants appeared unchanged. Immunohistochemical analyses of wildtype muscle spindles revealed a concentration of dystrophin and ß-dystroglycan in intrafusal fibres outside the region of contact with the sensory neuron. While utrophin was absent from the central part of intrafusal fibres of wildtype mice, it was substantially upregulated in dystrophin-deficient mice. Single-unit extracellular recordings of sensory afferents from muscle spindles of the extensor digitorum longus muscle revealed that muscle spindles from both dystrophic mouse strains have an increased resting discharge and a higher action potential firing rate during sinusoidal vibrations, particularly at low frequencies. The response to ramp-and-hold stretches appeared unaltered compared to the respective wildtype mice. We observed no exacerbated functional changes in dystrophin and dysferlin double mutant mice compared to the single mutant animals. These results show alterations in muscle spindle afferent responses in both dystrophic mouse lines, which might cause an increased muscle tone, and might contribute to the unstable gait and frequent falls observed in patients with muscular dystrophy.


Asunto(s)
Distrofias Musculares , Distrofia Muscular Animal , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Humanos , Ratones , Ratones Endogámicos mdx , Husos Musculares , Músculo Esquelético , Distrofias Musculares/genética , Utrofina
5.
Genet Med ; 21(10): 2355-2363, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30940925

RESUMEN

PURPOSE: A new syndrome with hypotonia, intellectual disability, and eye abnormalities (HIDEA) was previously described in a large consanguineous family. Linkage analysis identified the recessive disease locus, and genome sequencing yielded three candidate genes with potentially pathogenic biallelic variants: transketolase (TKT), transmembrane prolyl 4-hydroxylase (P4HTM), and ubiquitin specific peptidase 4 (USP4). However, the causative gene remained elusive. METHODS: International collaboration and exome sequencing were used to identify new patients with HIDEA and biallelic, potentially pathogenic, P4HTM variants. Segregation analysis was performed using Sanger sequencing. P4H-TM wild-type and variant constructs without the transmembrane region were overexpressed in insect cells and analyzed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blot. RESULTS: Five different homozygous or compound heterozygous pathogenic P4HTM gene variants were identified in six new and six previously published patients presenting with HIDEA. Hypoventilation, obstructive and central sleep apnea, and dysautonomia were identified as novel features associated with the phenotype. Characterization of three of the P4H-TM variants demonstrated yielding insoluble protein products and, thus, loss-of-function. CONCLUSIONS: Biallelic loss-of-function P4HTM variants were shown to cause HIDEA syndrome. Our findings enable diagnosis of the condition, and highlight the importance of assessing the need for noninvasive ventilatory support in patients.


Asunto(s)
Prolil Hidroxilasas/genética , Transcetolasa/genética , Proteasas Ubiquitina-Específicas/genética , Anomalías Múltiples/genética , Adolescente , Adulto , Niño , Preescolar , Epilepsia/genética , Exoma , Anomalías del Ojo/genética , Femenino , Humanos , Hipoventilación/genética , Discapacidad Intelectual/genética , Mutación con Pérdida de Función/genética , Masculino , Hipotonía Muscular/genética , Linaje , Fenotipo , Disautonomías Primarias/genética , Prolil Hidroxilasas/metabolismo , Síndrome , Transcetolasa/metabolismo , Secuenciación del Exoma , Adulto Joven
6.
Retina ; 39(3): 558-569, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29215532

RESUMEN

PURPOSE: To examine the involvement of the retinal pigment epithelium (RPE) in the presence of vitelliform macular lesions (VML) in Best vitelliform macular dystrophy (BVMD), autosomal recessive bestrophinopathy, and adult-onset vitelliform macular degeneration using polarization-sensitive optical coherence tomography (PS-OCT). METHODS: A total of 35 eyes of 18 patients were imaged using a PS-OCT system and blue light fundus autofluorescence imaging. Pathogenic mutations in the BEST1 gene, 3 of which were new, were detected in all patients with BVMD and autosomal recessive bestrophinopathy. RESULTS: Polarization-sensitive optical coherence tomography showed a characteristic pattern in all three diseases with nondepolarizing material in the subretinal space consistent with the yellowish VML seen on funduscopy with a visible RPE line below it. A focal RPE thickening was seen in 26 eyes under or at the edge of the VML. Retinal pigment epithelium thickness outside the VML was normal or mildly thinned in patients with BVMD and adult-onset vitelliform macular degeneration but was diffusely thinned or atrophic in patients with autosomal recessive bestrophinopathy. Patients with autosomal recessive bestrophinopathy showed sub-RPE fibrosis alongside the subretinal VML. Polarization-sensitive optical coherence tomography was more reliable in assessing the localization and the integrity of the RPE than spectral domain OCT alone. On spectral domain OCT, identification of the RPE was not possible in 19.4% of eyes. Polarization-sensitive optical coherence tomography allowed for definite identification of the location of VML in respect to the RPE in all eyes, since it provides a tissue-specific contrast. CONCLUSION: Polarization-sensitive optical coherence tomography confirms in vivo the subretinal location of VML and is useful in the assessment of RPE integrity.


Asunto(s)
Distrofia Macular Viteliforme/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios Transversales , Femenino , Angiografía con Fluoresceína/métodos , Humanos , Mácula Lútea/patología , Masculino , Persona de Mediana Edad , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica/métodos , Adulto Joven
7.
Am J Hum Genet ; 97(6): 855-61, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26581903

RESUMEN

Hereditary ataxias comprise a group of genetically heterogeneous disorders characterized by clinically variable cerebellar dysfunction and accompanied by involvement of other organ systems. The molecular underpinnings for many of these diseases are widely unknown. Previously, we discovered the disruption of Scyl1 as the molecular basis of the mouse mutant mdf, which is affected by neurogenic muscular atrophy, progressive gait ataxia with tremor, cerebellar vermis atrophy, and optic-nerve thinning. Here, we report on three human individuals, from two unrelated families, who presented with recurrent episodes of acute liver failure in early infancy and are affected by cerebellar vermis atrophy, ataxia, and peripheral neuropathy. By whole-exome sequencing, compound-heterozygous mutations within SCYL1 were identified in all affected individuals. We further show that in SCYL1-deficient human fibroblasts, the Golgi apparatus is massively enlarged, which is in line with the concept that SCYL1 regulates Golgi integrity. Thus, our findings define SCYL1 mutations as the genetic cause of a human hepatocerebellar neuropathy syndrome.


Asunto(s)
Ataxia Cerebelosa/genética , Degeneración Hepatolenticular/genética , Fallo Hepático/genética , Mutación , Enfermedades del Sistema Nervioso Periférico/genética , Factores de Transcripción/genética , Proteínas Adaptadoras del Transporte Vesicular , Adolescente , Secuencia de Bases , Ataxia Cerebelosa/patología , Proteínas de Unión al ADN , Exoma , Femenino , Expresión Génica , Degeneración Hepatolenticular/patología , Heterocigoto , Humanos , Fallo Hepático/patología , Masculino , Datos de Secuencia Molecular , Linaje , Enfermedades del Sistema Nervioso Periférico/patología , Análisis de Secuencia de ADN , Síndrome , Adulto Joven
8.
Am J Hum Genet ; 90(2): 201-16, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22265013

RESUMEN

We report on an autosomal-recessive variant of Ehlers-Danlos syndrome (EDS) characterized by severe muscle hypotonia at birth, progressive scoliosis, joint hypermobility, hyperelastic skin, myopathy, sensorineural hearing impairment, and normal pyridinoline excretion in urine. Clinically, the disorder shares many features with the kyphoscoliotic type of EDS (EDS VIA) and Ullrich congenital muscular dystrophy. Linkage analysis in a large Tyrolean kindred identified a homozygous frameshift mutation in FKBP14 in two affected individuals. Based on the cardinal clinical characteristics of the disorder, four additional individuals originating from different European countries were identified who carried either homozygous or compound heterozygous mutations in FKBP14. FKBP14 belongs to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases). ER-resident FKBPs have been suggested to act as folding catalysts by accelerating cis-trans isomerization of peptidyl-prolyl bonds and to act occasionally also as chaperones. We demonstrate that FKBP14 is localized in the endoplasmic reticulum (ER) and that deficiency of FKBP14 leads to enlarged ER cisterns in dermal fibroblasts in vivo. Furthermore, indirect immunofluorescence of FKBP14-deficient fibroblasts indicated an altered assembly of the extracellular matrix in vitro. These findings suggest that a disturbance of protein folding in the ER affecting one or more components of the extracellular matrix might cause the generalized connective tissue involvement in this disorder. FKBP14 mutation analysis should be considered in all individuals with apparent kyphoscoliotic type of EDS and normal urinary pyridinoline excretion, in particular in conjunction with sensorineural hearing impairment.


Asunto(s)
Anomalías Múltiples/genética , Síndrome de Ehlers-Danlos/genética , Mutación del Sistema de Lectura , Pérdida Auditiva/genética , Isomerasa de Peptidilprolil/genética , Adolescente , Aminoácidos/orina , Niño , Preescolar , Síndrome de Ehlers-Danlos/orina , Retículo Endoplásmico/genética , Matriz Extracelular/genética , Femenino , Fibroblastos/metabolismo , Variación Genética , Pérdida Auditiva/orina , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Pliegue de Proteína , cis-trans-Isomerasas/genética
9.
Muscle Nerve ; 52(3): 437-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26111941

RESUMEN

INTRODUCTION: Lipin 1 gene (LPIN1) mutations lead to cellular energy deficiency and cause up to 50% of the rhabdomyolysis episodes seen in pediatric patients. These episodes are associated with poor prognosis, as treatment options have been limited. We propose a novel therapeutic strategy based on prevention and early treatment of catabolism. METHODS: Five patients were diagnosed with LPIN1 mutations. They were instructed to maintain high caloric intake in situations possibly leading to catabolism such as viral infections or excessive physical activity. When an episode of rhabdomyolysis occurred, patients were treated with intravenous high-concentration glucose at first symptoms. RESULTS: The therapeutic strategies described limited the number of rhabdomyolyis episodes, and the duration of episodes was reduced from 7-10 days, as reported in the literature, to 5 days. CONCLUSION: In this small series, patients with LPIN1 mutations appear to have benefited from prevention and early treatment of catabolism.


Asunto(s)
Dietoterapia/métodos , Ingestión de Energía , Fluidoterapia/métodos , Glucosa/uso terapéutico , Rabdomiólisis/prevención & control , Edulcorantes/uso terapéutico , Anestesia General/efectos adversos , Austria , Niño , Preescolar , Femenino , Humanos , Masculino , Actividad Motora , Mutación , Fosfatidato Fosfatasa/genética , Rabdomiólisis/etiología , Rabdomiólisis/terapia , Resultado del Tratamiento , Virosis/complicaciones
10.
Am J Physiol Heart Circ Physiol ; 306(4): H564-H573, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337461

RESUMEN

Duchenne muscular dystrophy (DMD), induced by mutations in the gene encoding for the cytoskeletal protein dystrophin, is an inherited disease characterized by progressive muscle weakness. Besides the relatively well characterized skeletal muscle degenerative processes, DMD is also associated with cardiac complications. These include cardiomyopathy development and cardiac arrhythmias. The current understanding of the pathomechanisms in the heart is very limited, but recent research indicates that dysfunctional ion channels in dystrophic cardiomyocytes play a role. The aim of the present study was to characterize abnormalities in L-type calcium channel function in adult dystrophic ventricular cardiomyocytes. By using the whole cell patch-clamp technique, the properties of currents through calcium channels in ventricular cardiomyocytes isolated from the hearts of normal and dystrophic adult mice were compared. Besides the commonly used dystrophin-deficient mdx mouse model for human DMD, we also used mdx-utr mice, which are both dystrophin- and utrophin-deficient. We found that calcium channel currents were significantly increased, and channel inactivation was reduced in dystrophic cardiomyocytes. Both effects enhance the calcium influx during an action potential (AP). Whereas the AP in dystrophic mouse cardiomyocytes was nearly normal, implementation of the enhanced dystrophic calcium conductance in a computer model of a human ventricular cardiomyocyte considerably prolonged the AP. Finally, the described dystrophic calcium channel abnormalities entailed alterations in the electrocardiograms of dystrophic mice. We conclude that gain of function in cardiac L-type calcium channels may disturb the electrophysiology of the dystrophic heart and thereby cause arrhythmias.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Corazón/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Animales , Cardiomiopatías/complicaciones , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Simulación por Computador , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos mdx , Modelos Cardiovasculares , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/metabolismo , Miocitos Cardíacos/metabolismo
11.
PLoS Genet ; 7(4): e1002042, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21533183

RESUMEN

Albeit genetically highly heterogeneous, muscular dystrophies (MDs) share a convergent pathology leading to muscle wasting accompanied by proliferation of fibrous and fatty tissue, suggesting a common MD-pathomechanism. Here we show that mutations in muscular dystrophy genes (Dmd, Dysf, Capn3, Large) lead to the spontaneous formation of skeletal muscle-derived malignant tumors in mice, presenting as mixed rhabdomyo-, fibro-, and liposarcomas. Primary MD-gene defects and strain background strongly influence sarcoma incidence, latency, localization, and gender prevalence. Combined loss of dystrophin and dysferlin, as well as dystrophin and calpain-3, leads to accelerated tumor formation. Irrespective of the primary gene defects, all MD sarcomas share non-random genomic alterations including frequent losses of tumor suppressors (Cdkn2a, Nf1), amplification of oncogenes (Met, Jun), recurrent duplications of whole chromosomes 8 and 15, and DNA damage. Remarkably, these sarcoma-specific genetic lesions are already regularly present in skeletal muscles in aged MD mice even prior to sarcoma development. Accordingly, we show also that skeletal muscle from human muscular dystrophy patients is affected by gross genomic instability, represented by DNA double-strand breaks and age-related accumulation of aneusomies. These novel aspects of molecular pathologies common to muscular dystrophies and tumor biology will potentially influence the strategies to combat these diseases.


Asunto(s)
Aneuploidia , Daño del ADN , Músculo Esquelético/patología , Distrofias Musculares/genética , Sarcoma/genética , Animales , Calpaína/genética , Calpaína/metabolismo , Células Cultivadas , Hibridación Genómica Comparativa , Modelos Animales de Enfermedad , Disferlina , Distrofina/deficiencia , Distrofina/genética , Distrofina/metabolismo , Femenino , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/diagnóstico , Distrofias Musculares/metabolismo , Mutación , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo
12.
J Mol Cell Cardiol ; 53(4): 459-68, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22884543

RESUMEN

Failure of molecular chaperones to direct the correct folding of newly synthesized proteins leads to the accumulation of misfolded proteins in cells. HSPA4 is a member of the heat shock protein 110 family (HSP110) that acts as a nucleotide exchange factor of HSP70 chaperones. We found that the expression of HSPA4 is upregulated in murine hearts subjected to pressure overload and in failing human hearts. To investigate the cardiac function of HSPA4, Hspa4 knockout (KO) mice were generated and exhibited cardiac hypertrophy and fibrosis. Hspa4 KO hearts were characterized by a significant increase in heart weight/body weight ratio, elevated expression of hypertrophic and fibrotic gene markers, and concentric hypertrophy with preserved contractile function. In response to pressure overload, cardiac hypertrophy and remodeling were further aggravated in the Hspa4 KO compared to wild type (WT) mice. Cardiac hypertrophy in Hspa4 KO hearts was associated with enhanced activation of gp130-STAT3, CaMKII, and calcineurin-NFAT signaling. Protein blot and immunofluorescent analyses showed a significant accumulation of polyubiquitinated proteins in cardiac cells of Hspa4 KO mice. These results suggest that the myocardial remodeling of Hspa4 KO mice is due to accumulation of misfolded proteins resulting from impaired chaperone activity. Further analyses revealed a significant increase in cross sectional area of cardiomyocytes, and in expression levels of hypertrophic markers in cultured neonatal Hspa4 KO cardiomyocytes suggesting that the hypertrophy of mutant mice was a result of primary defects in cardiomyocytes. Gene expression profile in hearts of 3.5-week-old mice revealed a differentially expressed gene sets related to ion channels, muscle-specific contractile proteins and stress response. Taken together, our in vivo data demonstrate that Hspa4 gene ablation results in cardiac hypertrophy and fibrosis, possibly, through its role in protein quality control mechanism.


Asunto(s)
Cardiomegalia/genética , Proteínas del Choque Térmico HSP110/fisiología , Miocardio/patología , Animales , Animales Recién Nacidos , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/metabolismo , Calcineurina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/biosíntesis , Cardiomegalia/metabolismo , Cardiomegalia/patología , Células Cultivadas , Proteínas Contráctiles/genética , Receptor gp130 de Citocinas/biosíntesis , Fibrosis/genética , Proteínas del Choque Térmico HSP110/genética , Homeostasis , Humanos , Canales Iónicos/genética , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores de Transcripción NFATC/metabolismo , Pliegue de Proteína , Factor de Transcripción STAT3/biosíntesis , Transducción de Señal , Estrés Fisiológico/genética , Proteínas Ubiquitinadas/metabolismo , Remodelación Ventricular
14.
Life Sci Alliance ; 5(11)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35790299

RESUMEN

Aberrant expression of dystrophin, utrophin, dysferlin, or calpain-3 was originally identified in muscular dystrophies (MDs). Increasing evidence now indicates that these proteins might act as tumor suppressors in myogenic and non-myogenic cancers. As DNA damage and somatic aneuploidy, hallmarks of cancer, are early pathological signs in MDs, we hypothesized that a common pathway might involve the centrosome. Here, we show that dystrophin, utrophin, dysferlin, and calpain-3 are functional constituents of the centrosome. In myoblasts, lack of any of these proteins caused excess centrosomes, centrosome misorientation, nuclear abnormalities, and impaired microtubule nucleation. In dystrophin double-mutants, these defects were significantly aggravated. Moreover, we demonstrate that also in non-myogenic cells, all four MD-related proteins localize to the centrosome, including the muscle-specific full-length dystrophin isoform. Therefore, MD-related proteins might share a convergent function at the centrosome in addition to their diverse, well-established muscle-specific functions. Thus, our findings support the notion that cancer-like centrosome-related defects underlie MDs and establish a novel concept linking MDs to cancer.


Asunto(s)
Distrofias Musculares , Neoplasias , Calpaína , Centrosoma/metabolismo , Disferlina , Distrofina/genética , Humanos , Proteínas de la Membrana/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Neoplasias/genética , Utrofina
15.
Eur J Pharmacol ; 893: 173818, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33345856

RESUMEN

Ivabradine blocks hyperpolarisation-activated cyclic nucleotide-gated (HCN) channels, thereby lowering the heart rate, an action that is used clinically for the treatment of heart failure and angina pectoris. We and others have shown previously that ivabradine, in addition to its HCN channel blocking activity, also inhibits voltage-gated Na channels in vitro at concentrations that may be clinically relevant. Such action may reduce conduction velocity in cardiac atria and ventricles. Here, we explore the effect of administration of ivabradine on parameters of ventricular conduction and repolarization in the surface ECG of anesthetized mice. We found that 5 min after i.p. administration of 10 mg/kg ivabradine spontaneous heart rate had declined by ~13%, which is within the range observed in human clinical studies. At the same time a significant increase in QRS duration by ~18% was observed, suggesting a reduction in ventricular conduction velocity. During transesophageal pacing at heart rates between 100 and 220 beats/min there was no obvious rate-dependence of ivabradine-induced QRS prolongation. On the other hand, ivabradine produced substantial rate-dependent slowing of AV nodal conduction. We conclude that ivabradine prolongs conduction in the AV-node and in the ventricles in vivo.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Nodo Atrioventricular/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Ivabradina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Nodo Atrioventricular/fisiopatología , Estimulación Cardíaca Artificial , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Ratones Endogámicos C57BL , Factores de Tiempo
16.
JIMD Rep ; 46(1): 4-10, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31240148

RESUMEN

Lipin-1 is a phosphatidic acid phosphohydrolase (EC 3.1.3.4) that catalyzes the dephosphorylation of phosphatidic acid to diacylglycerol and inorganic phosphate. Deficiency of this enzyme causes potentially fatal severe, recurrent episodes of rhabdomyolysis triggered by infection. The defect has only recently been recognized so little is known about the long-term outcome in adult patients with this disorder. We report the course and outcome of a 25-year-old female patient with lipin-1 deficiency after a recent episode of rhabdomyolysis requiring intensive care admission with a peak creatine kinase of 500 000 IU/L. One-year post discharge from intensive care, the patient has residual drop foot bilaterally consistent with bilateral common peroneal neuropathies in addition to a background residual distal myopathy.

17.
Neurology ; 91(18): e1690-e1694, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30291184

RESUMEN

OBJECTIVE: To expand the clinical and genetic spectrum of nemaline myopathy 10 by a series of Austrian and German patients with a milder disease course and missense mutations in LMOD3. METHODS: We characterized the clinical features and the genetic status of 4 unrelated adolescent or adult patients with nemaline myopathy. RESULTS: The 4 patients showed a relatively mild disease course. They all have survived into adulthood, 3 of 4 have remained ambulatory, and all showed marked facial weakness. Muscle biopsy specimens gave evidence of nemaline bodies. All patients were unrelated but originated from Austria (Tyrol and Upper Austria) and Southern Germany (Bavaria). All patients carried the missense variant c.1648C>T, p.(Leu550Phe) in the LMOD3 gene, either on both alleles or in trans with another missense variant (c.1004A>G, p.Gln335Arg). Both variants were not reported previously. CONCLUSIONS: In 2014, a severe form of congenital nemaline myopathy caused by disrupting mutations in LMOD3 was identified and denoted as NEM10. Unlike the previously reported patients, who had a severe clinical picture with a substantial risk of early death, our patients showed a relatively mild disease course. As the missense variant c.1648C>T is located further downstream compared to all previously published LMOD3 mutations, it might be associated with higher protein expression compared to the reported loss-of-function mutations. The apparent clusters of 2 mild mutations in Germany and Austria in 4 unrelated families may be explained by a founder effect.


Asunto(s)
Proteínas Musculares/genética , Miopatías Nemalínicas/genética , Adolescente , Adulto , Austria , Femenino , Alemania , Humanos , Masculino , Proteínas de Microfilamentos , Mutación Missense , Fenotipo
18.
Neuromuscul Disord ; 17(2): 148-56, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17236769

RESUMEN

Limb-girdle muscular dystrophy type 2A (LGMD2A) is an autosomal recessive muscular disorder caused by mutations in the gene coding for calpain 3, a calcium-dependent protease. We developed an in vitro assay that can detect the proteolytic activity of calpain 3 in a muscle sample. This assay is based on the use of an inactive calpain 3 as a substrate for active calpain 3 molecules. A total of 79 human biopsies have been analysed using an unbiased single blind method. Results were confronted with the molecular diagnosis for confirmation. Proteolytic activity was either reduced or absent in 68% of LGMD2A biopsies. In the remaining 32%, normal proteolytic activity was found despite the presence of calpain 3 mutation(s), suggesting that other calpain 3 properties might be impaired to give rise to the LGMD2A phenotype. Our assay is easily adaptable to routine and appears to be more sensitive than common analysis by immunodetection.


Asunto(s)
Calpaína/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimología , Distrofia Muscular de Cinturas/enzimología , Animales , Western Blotting , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Heterocigoto , Humanos , Ratones , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Células 3T3 NIH , Fenotipo , Reproducibilidad de los Resultados , Bancos de Tejidos , Transfección
19.
Eur J Hum Genet ; 25(2): 262-266, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27782104

RESUMEN

The exceptionally large SYNE1 (spectrin repeat-containing nuclear envelope protein 1) gene encodes different nesprin-1 isoforms, which are differentially expressed in striated muscle and in cerebellar and cerebral neurons. Nesprin-1 isoforms can function in cytoskeletal, nuclear, and vesicle anchoring. SYNE1 variants have been associated with a spectrum of neurological and neuromuscular disease. Homozygosity mapping combined with exome sequencing identified a disease-causing nonsense mutation in the ultimate exon of full-length SYNE1 transcript in an 8-year-old boy with distal arthrogryposis and muscular hypotonia. mRNA analysis showed that the mutant transcript is expressed at wild-type levels. The variant truncates nesprin-1 isoforms for the C-terminal KASH (Klarsicht-ANC-Syne homology) domain. This is the third family with recessive arthrogryposis caused by homozygous distal-truncating SYNE1 variants. There is a SYNE1 genotype-phenotype correlation emerging, with more proximal homozygous SYNE1 variants causing recessive cerebellar ataxia of variable onset (SCAR8; ARCA-1).


Asunto(s)
Artrogriposis/genética , Codón sin Sentido , Genotipo , Debilidad Muscular/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Fenotipo , Artrogriposis/diagnóstico , Niño , Proteínas del Citoesqueleto , Homocigoto , Humanos , Masculino , Debilidad Muscular/diagnóstico , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Linaje , Síndrome
20.
J Neuropathol Exp Neurol ; 65(9): 855-65, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16957579

RESUMEN

Dysferlin is a muscle protein involved in cell membrane repair and its deficiency is associated with muscular dystrophy. We describe that dysferlin is also expressed in leaky endothelial cells. In the normal central nervous system (CNS), dysferlin is only present in endothelial cells of circumventricular organs. In the inflamed CNS of patients with multiple sclerosis (MS) or in animals with experimental autoimmune encephalomyelitis, dysferlin reactivity is induced in endothelial cells and the expression is associated with vascular leakage of serum proteins. In MS, dysferlin expression in endothelial cells is not restricted to vessels with inflammatory cuffs but is also present in noninflamed vessels. In addition, many blood vessels with perivascular inflammatory infiltrates lack dysferlin expression in inactive lesions or in the normal-appearing white matter. In vitro, dysferlin can be induced in endothelial cells by stimulation with tumor necrosis factor-alpha. Hence, dysferlin is not only a marker for leaky brain vessels, but also reveals dissociation of perivascular inflammatory infiltrates and blood-brain barrier disturbance in multiple sclerosis.


Asunto(s)
Proteínas de la Membrana/metabolismo , Esclerosis Múltiple/diagnóstico , Proteínas Musculares/metabolismo , Enfermedades Vasculares/diagnóstico , Animales , Biomarcadores/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Disferlina , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Fibrina/metabolismo , Humanos , Inmunohistoquímica/métodos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/complicaciones , Proteínas Musculares/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Enfermedades Vasculares/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA