Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 33(14): 1195-1206, 2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-38621658

RESUMEN

Mutations in DNAJB6 are a well-established cause of limb girdle muscular dystrophy type D1 (LGMD D1). Patients with LGMD D1 develop progressive muscle weakness with histology showing fibre damage, autophagic vacuoles, and aggregates. Whilst there are many reports of LGMD D1 patients, the role of DNAJB6 in the muscle is still unclear. In this study, we developed a loss of function zebrafish model in order to investigate the role of Dnajb6. Using a double dnajb6a and dnajb6b mutant model, we show that loss of Dnajb6 leads to a late onset muscle weakness. Interestingly, we find that adult fish lacking Dnajb6 do not have autophagy or myofibril defects, however, they do show mitochondrial changes and damage. This study demonstrates that loss of Dnajb6 causes mitochondrial defects and suggests that this contributes to muscle weakness in LGMD D1. These findings expand our knowledge of the role of Dnajb6 in the muscle and provides a model to screen novel therapies for LGMD D1.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas del Choque Térmico HSP40 , Mitocondrias , Chaperonas Moleculares , Debilidad Muscular , Distrofia Muscular de Cinturas , Pez Cebra , Animales , Humanos , Autofagia/genética , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Mitocondrias/metabolismo , Mitocondrias/genética , Mitocondrias/patología , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Debilidad Muscular/genética , Debilidad Muscular/patología , Debilidad Muscular/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Mutación , Proteínas del Tejido Nervioso , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
Int J Mol Sci ; 24(3)2023 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-36768797

RESUMEN

Individuals homozygous for the Pi*Z allele of SERPINA1 (ZAAT) are susceptible to lung disease due to insufficient α1-antitrypsin secretion into the circulation and may develop liver disease due to compromised protein folding that leads to inclusion body formation in the endoplasmic reticulum (ER) of hepatocytes. Transgenic zebrafish expressing human ZAAT show no signs of hepatic accumulation despite displaying serum insufficiency, suggesting the defect in ZAAT secretion occurs independently of its tendency to form inclusion bodies. In this study, proteomic, transcriptomic, and biochemical analysis provided evidence of suppressed Srebp2-mediated cholesterol biosynthesis in the liver of ZAAT-expressing zebrafish. To investigate the basis for this perturbation, CRISPR/Cas9 gene editing was used to manipulate ER protein quality control factors. Mutation of erlec1 resulted in a further suppression in the cholesterol biosynthesis pathway, confirming a role for this ER lectin in targeting misfolded ZAAT for ER-associated degradation (ERAD). Mutation of the two ER mannosidase homologs enhanced ZAAT secretion without inducing hepatic accumulation. These insights into hepatic ZAAT processing suggest potential therapeutic targets to improve secretion and alleviate serum insufficiency in this form of the α1-antitrypsin disease.


Asunto(s)
Proteómica , Pez Cebra , Animales , Humanos , Animales Modificados Genéticamente , Línea Celular , Colesterol , Hígado , Pez Cebra/genética , alfa 1-Antitripsina/genética
3.
Am J Pathol ; 190(3): 554-562, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31953038

RESUMEN

BCL-2-associated athanogene 3 (BAG3) is a co-chaperone to heat shock proteins important in degrading misfolded proteins through chaperone-assisted selective autophagy. The recurrent dominant BAG3-P209L mutation results in a severe childhood-onset myofibrillar myopathy (MFM) associated with progressive muscle weakness, cardiomyopathy, and respiratory failure. Because a homozygous knock-in (KI) strain for the mP215L mutation homologous to the human P209L mutation did not have a gross phenotype, compound heterozygote knockout (KO) and KI mP215L mice were generated to establish whether further reduction in BAG3 expression would lead to a phenotype. The KI/KO mice have a significant decrease in voluntary movement compared with wild-type and KI/KI mice in the open field starting at 7 months. The KI/KI and KI/KO mice both have significantly smaller muscle fiber cross-sectional area. However, only the KI/KO mice have clear skeletal muscle histologic changes in MFM. As in patient muscle, there are increased levels of BAG3-interacting proteins, such as p62, heat shock protein B8, and αB-crystallin. The KI/KO mP215L strain is the first murine model of BAG3 myopathy that resembles the human skeletal muscle pathologic features. The results support the hypothesis that the pathologic development of MFM requires a significant decrease in BAG3 protein level and not only a gain of function caused by the dominant missense mutation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Miopatías Estructurales Congénitas/patología , Animales , Cardiomiopatías/genética , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Genes Dominantes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Mutación , Miopatías Estructurales Congénitas/genética , Fenotipo
4.
PLoS Genet ; 14(2): e1007212, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29420541

RESUMEN

The lack of a mutant phenotype in homozygous mutant individuals' due to compensatory gene expression triggered upstream of protein function has been identified as genetic compensation. Whilst this intriguing process has been recognized in zebrafish, the presence of homozygous loss of function mutations in healthy human individuals suggests that compensation may not be restricted to this model. Loss of skeletal α-actin results in nemaline myopathy and we have previously shown that the pathological symptoms of the disease and reduction in muscle performance are recapitulated in a zebrafish antisense morpholino knockdown model. Here we reveal that a genetic actc1b mutant exhibits mild muscle defects and is unaffected by injection of the actc1b targeting morpholino. We further show that the milder phenotype results from a compensatory transcriptional upregulation of an actin paralogue providing a novel approach to be explored for the treatment of actin myopathy. Our findings provide further evidence that genetic compensation may influence the penetrance of disease-causing mutations.


Asunto(s)
Actinas/genética , Compensación de Dosificación (Genética)/fisiología , Músculo Esquelético/patología , Mutación , Miopatías Nemalínicas/genética , Animales , Animales Modificados Genéticamente , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Músculo Esquelético/metabolismo , Miopatías Nemalínicas/patología , Penetrancia , Fenotipo , Isoformas de Proteínas/genética , Pez Cebra/embriología , Pez Cebra/genética
5.
Proc Natl Acad Sci U S A ; 115(34): E8077-E8085, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30076230

RESUMEN

Dravet syndrome is a catastrophic, pharmacoresistant epileptic encephalopathy. Disease onset occurs in the first year of life, followed by developmental delay with cognitive and behavioral dysfunction and substantially elevated risk of premature death. The majority of affected individuals harbor a loss-of-function mutation in one allele of SCN1A, which encodes the voltage-gated sodium channel NaV1.1. Brain NaV1.1 is primarily localized to fast-spiking inhibitory interneurons; thus the mechanism of epileptogenesis in Dravet syndrome is hypothesized to be reduced inhibitory neurotransmission leading to brain hyperexcitability. We show that selective activation of NaV1.1 by venom peptide Hm1a restores the function of inhibitory interneurons from Dravet syndrome mice without affecting the firing of excitatory neurons. Intracerebroventricular infusion of Hm1a rescues Dravet syndrome mice from seizures and premature death. This precision medicine approach, which specifically targets the molecular deficit in Dravet syndrome, presents an opportunity for treatment of this intractable epilepsy.


Asunto(s)
Epilepsias Mioclónicas/tratamiento farmacológico , Interneuronas/metabolismo , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Venenos de Araña/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Células CHO , Cricetulus , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Epilepsias Mioclónicas/patología , Células HEK293 , Humanos , Interneuronas/patología , Ratones , Ratones Mutantes , Canal de Sodio Activado por Voltaje NAV1.1/genética
6.
Bull Environ Contam Toxicol ; 104(2): 228-234, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31760444

RESUMEN

Nuisance algal infestations are increasing globally in distribution and frequency. Copper-based algaecides are routinely applied to control these infestations, though there is an ever-present concern of risks to non-target species. This research evaluated risks associated with a commonly applied chelated copper algaecide (Captain® XTR; SePRO Corporation) to a sentinel non-target species (Daphnia magna) and further assessed alteration of the exposure and toxicity when a nuisance mat-forming cyanobacterium, Lyngbya wollei, was present in exposures. Aqueous copper concentrations in treatments with algae significantly decreased within 1 h after treatment and averaged 57.5% of nominal amended Cu through the experiment duration. The 48 h LC50 values were 371 µg Cu/L with no algae present in exposures and increased significantly to 531 µg Cu/L when L. wollei was simultaneously exposed. This research provides information on the short-term fate of copper and hazard assessment by incorporating targeted binding ligands, as present in operational treatments.


Asunto(s)
Cobre/metabolismo , Cianobacterias/metabolismo , Exposición a Riesgos Ambientales/prevención & control , Herbicidas/metabolismo , Contaminantes Químicos del Agua/metabolismo , Animales , Biodegradación Ambiental , Biomasa , Cobre/toxicidad , Daphnia/metabolismo , Exposición a Riesgos Ambientales/efectos adversos , Herbicidas/toxicidad , Contaminantes Químicos del Agua/toxicidad
7.
Dev Biol ; 438(2): 69-83, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29608877

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is a highly heritable neurodevelopmental disorder of childhood. It is primarily characterised by high levels of activity, inattention, and impulsivity, and has strong negative impacts on academic functioning. Children with ADHD show a reduction in volume, and hypoactivity, in a range of brain regions. The underlying mechanisms behind these phenotypes are unknown, however, variants in several genes with known roles in neurodevelopment are associated with ADHD. In this review we discuss how these ADHD associated genes contribute to neurodevelopment, and how variants in these genes could give rise to the neurological phenotypes seen in ADHD.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/etiología , Encéfalo/fisiopatología , Humanos , Conducta Impulsiva , Trastornos del Neurodesarrollo/genética , Neuronas/fisiología , Sinapsis/genética , Sinapsis/fisiología
8.
Ecol Lett ; 22(3): 518-526, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30618178

RESUMEN

Temperature often affects maternal investment in offspring. Across and within species, mothers in colder environments generally produce larger offspring than mothers in warmer environments, but the underlying drivers of this relationship remain unresolved. We formally evaluated the ubiquity of the temperature-offspring size relationship and found strong support for a negative relationship across a wide variety of ectotherms. We then tested an explanation for this relationship that formally links life-history and metabolic theories. We estimated the costs of development across temperatures using a series of laboratory experiments on model organisms, and a meta-analysis across 72 species of ectotherms spanning five phyla. We found that both metabolic and developmental rates increase with temperature, but developmental rate is more temperature sensitive than metabolic rate, such that the overall costs of development decrease with temperature. Hence, within a species' natural temperature range, development at relatively cooler temperatures requires mothers to produce larger, better provisioned offspring.


Asunto(s)
Tamaño Corporal , Madres , Temperatura , Adaptación Fisiológica , Animales , Femenino
9.
Am J Hum Genet ; 99(5): 1086-1105, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27745833

RESUMEN

This study establishes PYROXD1 variants as a cause of early-onset myopathy and uses biospecimens and cell lines, yeast, and zebrafish models to elucidate the fundamental role of PYROXD1 in skeletal muscle. Exome sequencing identified recessive variants in PYROXD1 in nine probands from five families. Affected individuals presented in infancy or childhood with slowly progressive proximal and distal weakness, facial weakness, nasal speech, swallowing difficulties, and normal to moderately elevated creatine kinase. Distinctive histopathology showed abundant internalized nuclei, myofibrillar disorganization, desmin-positive inclusions, and thickened Z-bands. PYROXD1 is a nuclear-cytoplasmic pyridine nucleotide-disulphide reductase (PNDR). PNDRs are flavoproteins (FAD-binding) and catalyze pyridine-nucleotide-dependent (NAD/NADH) reduction of thiol residues in other proteins. Complementation experiments in yeast lacking glutathione reductase glr1 show that human PYROXD1 has reductase activity that is strongly impaired by the disease-associated missense mutations. Immunolocalization studies in human muscle and zebrafish myofibers demonstrate that PYROXD1 localizes to the nucleus and to striated sarcomeric compartments. Zebrafish with ryroxD1 knock-down recapitulate features of PYROXD1 myopathy with sarcomeric disorganization, myofibrillar aggregates, and marked swimming defect. We characterize variants in the oxidoreductase PYROXD1 as a cause of early-onset myopathy with distinctive histopathology and introduce altered redox regulation as a primary cause of congenital muscle disease.


Asunto(s)
Núcleo Celular/genética , Miopatías Distales/genética , Variación Genética , Miopatías Estructurales Congénitas/genética , Oxidorreductasas/genética , Secuencia de Aminoácidos , Animales , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Estudios de Cohortes , Creatina Quinasa/genética , Creatina Quinasa/metabolismo , Citoplasma/metabolismo , Miopatías Distales/patología , Proteína 4 Similar a ELAV/genética , Proteína 4 Similar a ELAV/metabolismo , Femenino , Flavoproteínas/metabolismo , Eliminación de Gen , Estudio de Asociación del Genoma Completo , Glutatión Reductasa/genética , Glutatión Reductasa/metabolismo , Células HEK293 , Humanos , Masculino , Músculo Esquelético/patología , Mutación Missense , Miopatías Estructurales Congénitas/patología , Oxidorreductasas/metabolismo , Linaje , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Pez Cebra/genética
10.
Biol Chem ; 400(12): 1603-1616, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31091192

RESUMEN

In human α1-antitrypsin deficiency, homozygous carriers of the Z (E324K) mutation in the gene SERPINA1 have insufficient circulating α1-antitrypsin and are predisposed to emphysema. Misfolding and accumulation of the mutant protein in hepatocytes also causes endoplasmic reticulum stress and underpins long-term liver damage. Here, we describe transgenic zebrafish (Danio rerio) expressing the wildtype or the Z mutant form of human α1-antitrypsin in hepatocytes. As observed in afflicted humans, and in rodent models, about 80% less α1-antitrypsin is evident in the circulation of zebrafish expressing the Z mutant. Although these zebrafish also show signs of liver stress, they do not accumulate α1-antitrypsin in hepatocytes. This new zebrafish model will provide useful insights into understanding and treatment of α1-antitrypsin deficiency.


Asunto(s)
Hepatocitos/metabolismo , Modelos Animales , Deficiencia de alfa 1-Antitripsina/metabolismo , alfa 1-Antitripsina/metabolismo , Animales , Células CHO , Línea Celular , Cricetulus , Humanos , Mutación , Pez Cebra , alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/genética
11.
Hum Mol Genet ; 25(11): 2131-2142, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26969713

RESUMEN

Myofibrillar myopathy is a progressive muscle disease characterized by the disintegration of muscle fibers and formation of protein aggregates. Causative mutations have been identified in nine genes encoding Z-disk proteins, including the actin binding protein filamin C (FLNC). To investigate the mechanism of disease in FLNCW2710X myopathy we overexpressed fluorescently tagged FLNC or FLNCW2710X in zebrafish. Expression of FLNCW2710X causes formation of protein aggregates but surprisingly, our studies reveal that the mutant protein localizes correctly to the Z-disk and is capable of rescuing the fiber disintegration phenotype that results from FLNC knockdown. This demonstrates that the functions necessary for muscle integrity are not impaired, and suggests that it is the formation of protein aggregates and subsequent sequestration of FLNC away from the Z-disk that results in myofibrillar disintegration. Similar to those found in patients, the aggregates in FLNCW2710X expressing fish contain the co-chaperone BAG3. FLNC is a target of the BAG3-mediated chaperone assisted selective autophagy (CASA) pathway and therefore we investigated its role, and the role of autophagy in general, in clearing protein aggregates. We reveal that despite BAG3 recruitment to the aggregates they are not degraded via CASA. Additionally, recruitment of BAG3 is sufficient to block alternative autophagy pathways which would otherwise clear the aggregates. This blockage can be relieved by reducing BAG3 levels or by stimulating autophagy. This study therefore identifies both BAG3 reduction and autophagy promotion as potential therapies for FLNCW2710X myofibrillar myopathy, and identifies protein insufficiency due to sequestration, compounded by impaired autophagy, as the cause.


Asunto(s)
Autofagia/genética , Filaminas/genética , Fibras Musculares Esqueléticas/patología , Miopatías Estructurales Congénitas/genética , Animales , Regulación de la Expresión Génica , Humanos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/patología , Mutación , Miofibrillas/genética , Miofibrillas/patología , Fenotipo , Agregación Patológica de Proteínas/genética , Sarcómeros/genética , Sarcómeros/patología , Pez Cebra/genética
12.
Hum Mol Genet ; 25(13): 2776-2788, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27206985

RESUMEN

Filamin c (FLNc) is a large dimeric actin-binding protein located at premyofibrils, myofibrillar Z-discs and myofibrillar attachment sites of striated muscle cells, where it is involved in mechanical stabilization, mechanosensation and intracellular signaling. Mutations in the gene encoding FLNc give rise to skeletal muscle diseases and cardiomyopathies. Here, we demonstrate by fluorescence recovery after photobleaching that a large fraction of FLNc is highly mobile in cultured neonatal mouse cardiomyocytes and in cardiac and skeletal muscles of live transgenic zebrafish embryos. Analysis of cardiomyocytes from Xirp1 and Xirp2 deficient animals indicates that both Xin actin-binding repeat-containing proteins stabilize FLNc selectively in premyofibrils. Using a novel assay to analyze myofibrillar microdamage and subsequent repair in cultured contracting cardiomyocytes by live cell imaging, we demonstrate that repair of damaged myofibrils is achieved within only 4 h, even in the absence of de novo protein synthesis. FLNc is immediately recruited to these sarcomeric lesions together with its binding partner aciculin and precedes detectable assembly of filamentous actin and recruitment of other myofibrillar proteins. These data disclose an unprecedented degree of flexibility of the almost crystalline contractile machinery and imply FLNc as a dynamic signaling hub, rather than a primarily structural protein. Our myofibrillar damage/repair model illustrates how (cardio)myocytes are kept functional in their mechanically and metabolically strained environment. Our results help to better understand the pathomechanisms and pathophysiology of early stages of FLNc-related myofibrillar myopathy and skeletal and cardiac diseases preceding pathological protein aggregation.


Asunto(s)
Filaminas/genética , Filaminas/metabolismo , Miofibrillas/patología , Actinas/metabolismo , Animales , Técnicas de Cultivo de Célula , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Filaminas/fisiología , Humanos , Ratones , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Mutación , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Unión Proteica
13.
BMC Genomics ; 18(1): 259, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28347272

RESUMEN

BACKGROUND: Computational identification of non-coding RNAs (ncRNAs) is a challenging problem. We describe a genome-wide analysis using Bayesian segmentation to identify intronic elements highly conserved between three evolutionarily distant vertebrate species: human, mouse and zebrafish. We investigate the extent to which these elements include ncRNAs (or conserved domains of ncRNAs) and regulatory sequences. RESULTS: We identified 655 deeply conserved intronic sequences in a genome-wide analysis. We also performed a pathway-focussed analysis on genes involved in muscle development, detecting 27 intronic elements, of which 22 were not detected in the genome-wide analysis. At least 87% of the genome-wide and 70% of the pathway-focussed elements have existing annotations indicative of conserved RNA secondary structure. The expression of 26 of the pathway-focused elements was examined using RT-PCR, providing confirmation that they include expressed ncRNAs. Consistent with previous studies, these elements are significantly over-represented in the introns of transcription factors. CONCLUSIONS: This study demonstrates a novel, highly effective, Bayesian approach to identifying conserved non-coding sequences. Our results complement previous findings that these sequences are enriched in transcription factors. However, in contrast to previous studies which suggest the majority of conserved sequences are regulatory factor binding sites, the majority of conserved sequences identified using our approach contain evidence of conserved RNA secondary structures, and our laboratory results suggest most are expressed. Functional roles at DNA and RNA levels are not mutually exclusive, and many of our elements possess evidence of both. Moreover, ncRNAs play roles in transcriptional and post-transcriptional regulation, and this may contribute to the over-representation of these elements in introns of transcription factors. We attribute the higher sensitivity of the pathway-focussed analysis compared to the genome-wide analysis to improved alignment quality, suggesting that enhanced genomic alignments may reveal many more conserved intronic sequences.


Asunto(s)
Genoma , ARN no Traducido/metabolismo , Animales , Teorema de Bayes , Sitios de Unión , Secuencia Conservada , Humanos , Intrones , Ratones , Desarrollo de Músculos/genética , Conformación de Ácido Nucleico , ARN no Traducido/química , ARN no Traducido/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Interfaz Usuario-Computador , Pez Cebra/genética
14.
Biotechnol Bioeng ; 114(9): 2142-2148, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28475237

RESUMEN

The hearts of adult zebrafish (Danio rerio) are capable of complete regeneration in vivo even after major injury, making this species of particular interest for understanding the growth and differentiation processes required for cardiac tissue engineering. To date, little research has been carried out on in vitro culture of adult zebrafish cardiac cells. In this work, progenitor-rich cardiospheres suitable for cardiomyocyte differentiation and myocardial regeneration were produced from adult zebrafish hearts. The cardiospheres contained a mixed population of c-kit+ and Mef2c+ cells; proliferative peripheral cells of possible mesenchymal lineage were also observed. Cellular outgrowth from cardiac explants and cardiospheres was enhanced significantly using conditioned medium harvested from cultures of a rainbow trout cell line, suggesting that fish-specific trophic factors are required for zebrafish cardiac cell expansion. Three-dimensional culture of zebrafish heart cells in fibrous polyglycolic acid (PGA) scaffolds was carried out under dynamic fluid flow conditions. High levels of cell viability and cardiomyocyte differentiation were maintained within the scaffolds. Expression of cardiac troponin T, a marker of differentiated cardiomyocytes, increased during the first 7 days of scaffold culture; after 15 days, premature disintegration of the biodegradable scaffolds led to cell detachment and a decline in differentiation status. This work expands our technical capabilities for three-dimensional zebrafish cardiac cell culture with potential applications in tissue engineering, drug and toxicology screening, and ontogeny research. Biotechnol. Bioeng. 2017;114: 2142-2148. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Técnicas de Cultivo de Órganos/instrumentación , Esferoides Celulares/citología , Células Madre/citología , Andamios del Tejido , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Diseño de Equipo , Análisis de Falla de Equipo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Técnicas de Cultivo de Órganos/métodos , Esferoides Celulares/fisiología , Células Madre/fisiología , Pez Cebra/anatomía & histología , Pez Cebra/fisiología
15.
Am J Hum Genet ; 93(1): 6-18, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23746549

RESUMEN

Nemaline myopathy (NEM) is a common congenital myopathy. At the very severe end of the NEM clinical spectrum are genetically unresolved cases of autosomal-recessive fetal akinesia sequence. We studied a multinational cohort of 143 severe-NEM-affected families lacking genetic diagnosis. We performed whole-exome sequencing of six families and targeted gene sequencing of additional families. We identified 19 mutations in KLHL40 (kelch-like family member 40) in 28 apparently unrelated NEM kindreds of various ethnicities. Accounting for up to 28% of the tested individuals in the Japanese cohort, KLHL40 mutations were found to be the most common cause of this severe form of NEM. Clinical features of affected individuals were severe and distinctive and included fetal akinesia or hypokinesia and contractures, fractures, respiratory failure, and swallowing difficulties at birth. Molecular modeling suggested that the missense substitutions would destabilize the protein. Protein studies showed that KLHL40 is a striated-muscle-specific protein that is absent in KLHL40-associated NEM skeletal muscle. In zebrafish, klhl40a and klhl40b expression is largely confined to the myotome and skeletal muscle, and knockdown of these isoforms results in disruption of muscle structure and loss of movement. We identified KLHL40 mutations as a frequent cause of severe autosomal-recessive NEM and showed that it plays a key role in muscle development and function. Screening of KLHL40 should be a priority in individuals who are affected by autosomal-recessive NEM and who present with prenatal symptoms and/or contractures and in all Japanese individuals with severe NEM.


Asunto(s)
Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Mutación Missense , Miopatías Nemalínicas/genética , Sustitución de Aminoácidos , Animales , Pueblo Asiatico/genética , Estudios de Cohortes , Mutación del Sistema de Lectura , Genes Recesivos , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Proteínas Musculares/genética , Miopatías Nemalínicas/etnología , Miopatías Nemalínicas/patología , Linaje , Polimorfismo de Nucleótido Simple , Índice de Severidad de la Enfermedad , Pez Cebra/genética
16.
Acta Neuropathol ; 130(3): 389-406, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25931053

RESUMEN

Nemaline myopathy is characterized by muscle weakness and the presence of rod-like (nemaline) bodies. The genetic etiology of nemaline myopathy is becoming increasingly understood with mutations in ten genes now known to cause the disease. Despite this, the mechanism by which skeletal muscle weakness occurs remains elusive, with previous studies showing no correlation between the frequency of nemaline bodies and disease severity. To investigate the formation of nemaline bodies and their role in pathogenesis, we generated overexpression and loss-of-function zebrafish models for skeletal muscle α-actin (ACTA1) and nebulin (NEB). We identify three distinct types of nemaline bodies and visualize their formation in vivo, demonstrating these nemaline bodies not only exhibit different subcellular origins, but also have distinct pathological consequences within the skeletal muscle. One subtype is highly dynamic and upon breakdown leads to the accumulation of cytoplasmic actin contributing to muscle weakness. Examination of a Neb-deficient model suggests this mechanism may be common in nemaline myopathy. Another subtype results from a reduction of actin and forms a more stable cytoplasmic body. In contrast, the final type originates at the Z-disk and is associated with myofibrillar disorganization. Analysis of zebrafish and muscle biopsies from ACTA1 nemaline myopathy patients demonstrates that nemaline bodies also possess a different protein signature. In addition, we show that the ACTA1(D286G) mutation causes impaired actin incorporation and localization in the sarcomere. Together these data provide a novel examination of nemaline body origins and dynamics in vivo and identifies pathological changes that correlate with muscle weakness.


Asunto(s)
Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miopatías Nemalínicas/patología , Miopatías Nemalínicas/fisiopatología , Actinina/genética , Actinina/metabolismo , Actinas/metabolismo , Animales , Animales Modificados Genéticamente , Citoplasma/metabolismo , Citoplasma/patología , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Morfolinos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Debilidad Muscular/patología , Debilidad Muscular/fisiopatología , Mutación , Fenotipo , Sarcómeros/metabolismo , Sarcómeros/patología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
Nature ; 457(7229): 603-7, 2009 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19092805

RESUMEN

Cancer stem cells are remarkably similar to normal stem cells: both self-renew, are multipotent and express common surface markers, for example, prominin 1 (PROM1, also called CD133). What remains unclear is whether cancer stem cells are the direct progeny of mutated stem cells or more mature cells that reacquire stem cell properties during tumour formation. Answering this question will require knowledge of whether normal stem cells are susceptible to cancer-causing mutations; however, this has proved difficult to test because the identity of most adult tissue stem cells is not known. Here, using an inducible Cre, nuclear LacZ reporter allele knocked into the Prom1 locus (Prom1(C-L)), we show that Prom1 is expressed in a variety of developing and adult tissues. Lineage-tracing studies of adult Prom1(+/C-L) mice containing the Rosa26-YFP reporter allele showed that Prom1(+) cells are located at the base of crypts in the small intestine, co-express Lgr5 (ref. 2), generate the entire intestinal epithelium, and are therefore the small intestinal stem cell. Prom1 was reported recently to mark cancer stem cells of human intestinal tumours that arise frequently as a consequence of aberrant wingless (Wnt) signalling. Activation of endogenous Wnt signalling in Prom1(+/C-L) mice containing a Cre-dependent mutant allele of beta-catenin (Ctnnb1(lox(ex3))) resulted in a gross disruption of crypt architecture and a disproportionate expansion of Prom1(+) cells at the crypt base. Lineage tracing demonstrated that the progeny of these cells replaced the mucosa of the entire small intestine with neoplastic tissue that was characterized by focal high-grade intraepithelial neoplasia and crypt adenoma formation. Although all neoplastic cells arose from Prom1(+) cells in these mice, only 7% of tumour cells retained Prom1 expression. Our data indicate that Prom1 marks stem cells in the adult small intestine that are susceptible to transformation into tumours retaining a fraction of mutant Prom1(+) tumour cells.


Asunto(s)
Antígenos CD/metabolismo , Linaje de la Célula , Transformación Celular Neoplásica , Glicoproteínas/metabolismo , Intestino Delgado/citología , Células Madre Neoplásicas/metabolismo , Péptidos/metabolismo , Células Madre/metabolismo , Células Madre/patología , Antígeno AC133 , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Antígenos CD/análisis , Antígenos CD/genética , Biomarcadores/análisis , Biomarcadores/metabolismo , Células Cultivadas , Genes Reporteros/genética , Glicoproteínas/análisis , Glicoproteínas/genética , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Intestino Delgado/patología , Ratones , Mutación , Trasplante de Neoplasias , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/patología , Péptidos/análisis , Péptidos/genética , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Trasplante Heterólogo , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
18.
Hum Mol Genet ; 21(18): 4073-83, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22706277

RESUMEN

Myofibrillar myopathies are a group of muscle disorders characterized by the disintegration of skeletal muscle fibers and formation of sarcomeric protein aggregates. All the proteins known to be involved in myofibrillar myopathies localize to a region of the sarcomere known as the Z-disk, the site at which defects are first observed. Given the common cellular phenotype observed in this group of disorders, it is thought that there is a common mechanism of pathology. Mutations in filamin C, which has several proposed roles in the development and function of skeletal muscle, can result in filamin-related myofibrillar myopathy. The lack of a suitable animal model system has limited investigation into the mechanism of pathology in this disease and the role of filamin C in muscle development. Here, we characterize stretched out (sot), a zebrafish filamin Cb mutant, together with targeted knockdown of zebrafish filamin Ca, revealing fiber dissolution and formation of protein aggregates strikingly similar to those seen in filamin-related myofibrillar myopathies. Through knockdown of both zebrafish filamin C homologues, we demonstrate that filamin C is not required for fiber specification and that fiber damage is a consequence of muscle activity. The remarkable similarities in the myopathology between our models and filamin-related myofibrillar myopathy makes them suitable for the study of these diseases and provides unique opportunities for the investigation of the function of filamin C in muscle and development of therapies.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Microfilamentos/genética , Distrofias Musculares/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Tipificación del Cuerpo/genética , Codón sin Sentido , Análisis Mutacional de ADN , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Filaminas , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Humanos , Funciones de Verosimilitud , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/fisiología , Contracción Muscular , Fibras Musculares de Contracción Lenta/metabolismo , Fibras Musculares de Contracción Lenta/patología , Distrofias Musculares/embriología , Mutagénesis , Miosinas/metabolismo , Fenotipo , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología
19.
Acta Neuropathol ; 128(6): 821-33, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25273835

RESUMEN

Mutations in the co-chaperone Bcl2-associated athanogene 3 (BAG3) can cause myofibrillar myopathy (MFM), a childhood-onset progressive muscle disease, characterized by the formation of protein aggregates and myofibrillar disintegration. In contrast to other MFM-causing proteins, BAG3 has no direct structural role, but regulates autophagy and the degradation of misfolded proteins. To investigate the mechanism of disease in BAG3-related MFM, we expressed wild-type BAG3 or the dominant MFM-causing BAG3 (BAG3(P209L)) in zebrafish. Expression of the mutant protein results in the formation of aggregates that contain wild-type BAG3. Through the stimulation and inhibition of autophagy, we tested the prevailing hypothesis that impaired autophagic function is responsible for the formation of protein aggregates. Contrary to the existing theory, our studies reveal that inhibition of autophagy is not sufficient to induce protein aggregation. Expression of the mutant protein, however, did not induce myofibrillar disintegration and we therefore examined the effect of knocking down Bag3 function. Loss of Bag3 resulted in myofibrillar disintegration, but not in the formation of protein aggregates. Remarkably, BAG3(P209L) is able to rescue the myofibrillar disintegration phenotype, further demonstrating that its function is not impaired. Together, our knockdown and overexpression experiments identify a mechanism whereby BAG3(P209L) aggregates form, gradually reducing the pool of available BAG3, which eventually results in BAG3 insufficiency and myofibrillar disintegration. This mechanism is consistent with the childhood onset and progressive nature of MFM and suggests that reducing aggregation through enhanced degradation or inhibition of nucleation would be an effective therapy for this disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Miopatías Estructurales Congénitas/fisiopatología , Proteínas de Pez Cebra/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Animales Modificados Genéticamente , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/fisiología , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Corazón/fisiopatología , Humanos , Contracción Muscular/fisiología , Músculo Esquelético/fisiopatología , Mutación , Miofibrillas/fisiología , Agregado de Proteínas/fisiología , Sarcómeros/metabolismo , Temperatura , Pez Cebra , Proteínas de Pez Cebra/genética
20.
Nat Rev Genet ; 9(8): 632-46, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18636072

RESUMEN

The molecular, genetic and cellular bases for skeletal muscle growth and regeneration have been recently documented in a number of vertebrate species. These studies highlight the role of transient subcompartments of the early somite as a source of distinct waves of myogenic precursors. Individual myogenic progenitor populations undergo a complex series of cell rearrangements and specification events in different regions of the body, all of which are controlled by distinct gene regulatory networks. Collectively, these studies have opened a window into the morphogenetic and molecular bases of the different phases of vertebrate myogenesis, from embryo to adult.


Asunto(s)
Desarrollo de Músculos/genética , Músculos/embriología , Vertebrados/genética , Animales , Embrión de Mamíferos , Embrión no Mamífero , Cabeza/embriología , Corazón/embriología , Corazón/fisiología , Humanos , Modelos Biológicos , Especificidad de Órganos/genética , ARN Mensajero/metabolismo , ARN Mensajero/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA