Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Soc Trans ; 52(3): 1085-1098, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38716888

RESUMEN

In vivo, muscle and neuronal cells are post-mitotic, and their function is predominantly regulated by proteostasis, a multilayer molecular process that maintains a delicate balance of protein homeostasis. The ubiquitin-proteasome system (UPS) is a key regulator of proteostasis. A dysfunctional UPS is a hallmark of muscle ageing and is often impacted in neuromuscular disorders (NMDs). Malfunction of the UPS often results in aberrant protein accumulation which can lead to protein aggregation and/or mis-localization affecting its function. Deubiquitinating enzymes (DUBs) are key players in the UPS, controlling protein turnover and maintaining the free ubiquitin pool. Several mutations in DUB encoding genes are linked to human NMDs, such as ATXN3, OTUD7A, UCHL1 and USP14, whilst other NMDs are associated with dysregulation of DUB expression. USP5, USP9X and USP14 are implicated in synaptic transmission and remodeling at the neuromuscular junction. Mice lacking USP19 show increased maintenance of lean muscle mass. In this review, we highlight the involvement of DUBs in muscle physiology and NMDs, particularly in processes affecting muscle regeneration, degeneration and inflammation following muscle injury. DUBs have recently garnered much respect as promising drug targets, and their roles in muscle maturation, regeneration and degeneration may provide the framework for novel therapeutics to treat muscular disorders including NMDs, sarcopenia and cachexia.


Asunto(s)
Enzimas Desubicuitinizantes , Humanos , Animales , Enzimas Desubicuitinizantes/metabolismo , Músculo Esquelético/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/fisiopatología , Enfermedades Neuromusculares/enzimología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/genética , Ratones , Proteostasis
2.
Cytometry A ; 99(12): 1240-1249, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34089298

RESUMEN

Skeletal muscle function is inferred from the spatial arrangement of muscle fiber architecture, which corresponds to myofiber molecular and metabolic features. Myofiber features are often determined using immunofluorescence on a local sampling, typically obtained from a median region. This median region is assumed to represent the entire muscle. However, it remains largely unknown to what extent this local sampling represents the entire muscle. We present a pipeline to study the architecture of muscle fiber features over the entire muscle, including sectioning, staining, imaging to image quantification and data-driven analysis with Myofiber type were identified by the expression of myosin heavy chain (MyHC) isoforms, representing contraction properties. We reconstructed muscle architecture from consecutive cross-sections stained for laminin and MyHC isoforms. Examining the entire muscle using consecutive cross-sections is extremely laborious, we provide consideration to reduce the dataset without loosing spatial information. Data-driven analysis with over 150,000 myofibers showed spatial variations in myofiber geometric features, myofiber type, and the distribution of neuromuscular junctions over the entire muscle. We present a workflow to study histological changes over the entire muscle using high-throughput imaging, image quantification, and data-driven analysis. Our results suggest that asymmetric spatial distribution of these features over the entire muscle could impact muscle function. Therefore, instead of a single sampling from a median region, representative regions covering the entire muscle should be investigated in future studies.


Asunto(s)
Fibras Musculares Esqueléticas , Cadenas Pesadas de Miosina , Músculo Esquelético , Isoformas de Proteínas
3.
FASEB J ; 34(4): 5525-5537, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32141137

RESUMEN

Skeletal muscles control posture, mobility and strength, and influence whole-body metabolism. Muscles are built of different types of myofibers, each having specific metabolic, molecular, and contractile properties. Fiber classification is, therefore, regarded the key for understanding muscle biology, (patho-) physiology. The expression of three myosin heavy chain (MyHC) isoforms, MyHC-1, MyHC-2A, and MyHC-2X, marks myofibers in humans. Typically, myofiber classification is performed by an eye-based histological analysis. This classical approach is insufficient to capture complex fiber classes, expressing more than one MyHC-isoform. We, therefore, developed a methodological procedure for high-throughput characterization of myofibers on the basis of multiple isoforms. The mean fluorescence intensity of the three most abundant MyHC isoforms was measured per myofiber in muscle biopsies of 56 healthy elderly adults, and myofiber classes were identified using computational biology tools. Unsupervised clustering revealed the existence of six distinct myofiber clusters. A comparison with the visual assessment of myofibers using the same images showed that some of these myofiber clusters could not be detected or were frequently misclassified. The presence of these six clusters was reinforced by RNA expressions levels of sarcomeric genes. In addition, one of the clusters, expressing all three MyHC isoforms, correlated with histological measures of muscle health. To conclude, this methodological procedure enables deep characterization of the complex muscle heterogeneity. This study opens opportunities to further investigate myofiber composition in comparative studies.


Asunto(s)
Biología Computacional/métodos , Fibras Musculares Esqueléticas/clasificación , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Cadenas Pesadas de Miosina/metabolismo , Femenino , Humanos , Masculino , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo
4.
Genomics ; 112(5): 3157-3165, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32479991

RESUMEN

Identifying genes involved in functional differences between similar tissues from expression profiles is challenging, because the expected differences in expression levels are small. To exemplify this challenge, we studied the expression profiles of two skeletal muscles, deltoid and biceps, in healthy individuals. We provide a series of guides and recommendations for the analysis of this type of studies. These include how to account for batch effects and inter-individual differences to optimize the detection of gene signatures associated with tissue function. We provide guidance on the selection of optimal settings for constructing gene co-expression networks through parameter sweeps of settings and calculation of the overlap with an established knowledge network. Our main recommendation is to use a combination of the data-driven approaches, such as differential gene expression analysis and gene co-expression network analysis, and hypothesis-driven approaches, such as gene set connectivity analysis. Accordingly, we detected differences in metabolic gene expression between deltoid and biceps that were supported by both data- and hypothesis-driven approaches. Finally, we provide a bioinformatic framework that support the biological interpretation of expression profiles from related tissues from this combination of approaches, which is available at github.com/tabbassidaloii/AnalysisFrameworkSimilarTissues.


Asunto(s)
Perfilación de la Expresión Génica , Músculo Esquelético/metabolismo , Aerobiosis , Músculo Deltoides/metabolismo , Redes Reguladoras de Genes , Humanos , Bases del Conocimiento , Mitocondrias Musculares/metabolismo
5.
FASEB J ; 33(3): 4046-4053, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30485132

RESUMEN

Contractile properties of myofibers are dictated by the abundance of myosin heavy chain (MyHC) isoforms. MyHC composition designates muscle function, and its alterations could unravel differential muscle involvement in muscular dystrophies and aging. Current analyses are limited to visual assessments in which myofibers expressing multiple MyHC isoforms are prone to misclassification. As a result, complex patterns and subtle alterations are unidentified. We developed a high-throughput, data-driven myofiber analysis to quantitatively describe the variations in myofibers across the muscle. We investigated alterations in myofiber composition between genotypes, 2 muscles, and 2 age groups. We show that this analysis facilitates the discovery of complex myofiber compositions and its dependency on age, muscle type, and genetic conditions.-Raz, V., Raz, Y., van de Vijver, D., Bindellini, D., van Putten, M., van den Akker, E. B. High-throughput data-driven analysis of myofiber composition reveals muscle-specific disease and age-associated patterns.


Asunto(s)
Envejecimiento/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/metabolismo , Cadenas Pesadas de Miosina/genética , Envejecimiento/genética , Envejecimiento/patología , Animales , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/clasificación , Fibras Musculares Esqueléticas/patología , Distrofias Musculares/genética , Distrofias Musculares/patología , Cadenas Pesadas de Miosina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
6.
Int J Mol Sci ; 21(17)2020 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-32842713

RESUMEN

Small non-coding microRNAs (miRNAs) are involved in the regulation of mRNA stability. Their features, including high stability and secretion to biofluids, make them attractive as potential biomarkers for diverse pathologies. This is the first study reporting miRNA as potential biomarkers for oculopharyngeal muscular dystrophy (OPMD), an adult-onset myopathy. We hypothesized that miRNA that is differentially expressed in affected muscles from OPMD patients is secreted to biofluids and those miRNAs could be used as biomarkers for OPMD. We first identified candidate miRNAs from OPMD-affected muscles and from muscles from an OPMD mouse model using RNA sequencing. We then compared the OPMD-deregulated miRNAs to the literature and, subsequently, we selected a few candidates for expression studies in serum and saliva biofluids using qRT-PCR. We identified 126 miRNAs OPMD-deregulated in human muscles, but 36 deregulated miRNAs in mice only (pFDR < 0.05). Only 15 OPMD-deregulated miRNAs overlapped between the in humans and mouse studies. The majority of the OPMD-deregulated miRNAs showed opposite deregulation direction compared with known muscular dystrophies miRNAs (myoMirs), which are associated. In contrast, similar dysregulation direction was found for 13 miRNAs that are common between OPMD and aging muscles. A significant age-association (p < 0.05) was found for 17 OPMD-deregulated miRNAs (13.4%), whereas in controls, only six miRNAs (1.4%) showed a significant age-association, suggesting that miRNA expression in OPMD is highly age-associated. miRNA expression in biofluids revealed that OPMD-associated deregulation in saliva was similar to that in muscles, but not in serum. The same as in muscle, miRNA expression levels in saliva were also found to be associated with age (p < 0.05). Moreover, the majority of OPMD-miRNAs were found to be associated with dysphagia as an initial symptom. We suggest that levels of specific miRNAs in saliva can mark muscle degeneration in general and dysphagia in OPMD.


Asunto(s)
MicroARNs/genética , Distrofia Muscular Oculofaríngea/genética , Saliva/fisiología , Adulto , Factores de Edad , Anciano , Animales , Biomarcadores , Estudios de Casos y Controles , Trastornos de Deglución/genética , Modelos Animales de Enfermedad , Expresión Génica , Humanos , MicroARNs/análisis , MicroARNs/sangre , Músculo Esquelético/fisiopatología , Distrofia Muscular Oculofaríngea/etiología , Análisis de Secuencia de ARN
7.
Am J Hum Genet ; 98(5): 1020-1029, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27153398

RESUMEN

Facioscapulohumeral dystrophy (FSHD) is associated with somatic chromatin relaxation of the D4Z4 repeat array and derepression of the D4Z4-encoded DUX4 retrogene coding for a germline transcription factor. Somatic DUX4 derepression is caused either by a 1-10 unit repeat-array contraction (FSHD1) or by mutations in SMCHD1, which encodes a chromatin repressor that binds to D4Z4 (FSHD2). Here, we show that heterozygous mutations in DNA methyltransferase 3B (DNMT3B) are a likely cause of D4Z4 derepression associated with low levels of DUX4 expression from the D4Z4 repeat and increased penetrance of FSHD. Recessive mutations in DNMT3B were previously shown to cause immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome. This study suggests that transcription of DUX4 in somatic cells is modified by variations in its epigenetic state and provides a basis for understanding the reduced penetrance of FSHD within families.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Represión Epigenética/genética , Distrofia Muscular Facioescapulohumeral/genética , Mutación/genética , Penetrancia , Secuencias Repetidas en Tándem/genética , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Niño , Preescolar , Cromatina/genética , ADN (Citosina-5-)-Metiltransferasas/química , Metilación de ADN , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Linaje , Conformación Proteica , Homología de Secuencia de Aminoácido , ADN Metiltransferasa 3B
8.
FASEB J ; 32(3): 1579-1590, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29141996

RESUMEN

Adult muscles have a vast adaptation capacity, enabling function switches in response to altered conditions. During intensive physical activity, disease, or aging, adult skeletal muscles change and adjust their functions. The competence to adjust varies among muscles. Muscle-specific molecular mechanisms in healthy and normal conditions could designate changes in physiologic and pathologic conditions. We generated deep mRNA-sequencing data in adult fast and slow mouse muscles, and applying paired analysis, we identified that the muscle-specific signatures are composed of half of the muscle transcriptome. The fast muscles showed a more compact gene network that is concordant with homogenous myofiber typing, compared with the pattern in the slow muscle. The muscle-specific mRNA landscape did not correlate with alternative spicing, alternative polyadenylation, or the expression of muscle transcription factor gene networks. However, we found significant correlation between the differentially expressed noncoding RNAs, microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) and their target genes. More than 25% of the genes expressed in a muscle-specific fashion were found to be targets of muscle-specific miRNAs and lncRNAs. We suggest that muscle-specific miRNAs and lncRNAs contribute to the establishment of muscle-specific transcriptomes in adult muscles.-Raz, V., Riaz, M., Tatum, Z., Kielbasa, S. M., 't Hoen, P. A. C. The distinct transcriptomes of slow and fast adult muscles are delineated by noncoding RNAs.


Asunto(s)
Redes Reguladoras de Genes/fisiología , MicroARNs/biosíntesis , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , ARN Largo no Codificante/biosíntesis , Transcriptoma/fisiología , Animales , Masculino , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética
9.
PLoS Genet ; 12(5): e1006031, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27152426

RESUMEN

Poly(A) Binding Protein Nuclear 1 (PABPN1) is a multifunctional regulator of mRNA processing, and its expression levels specifically decline in aging muscles. An expansion mutation in PABPN1 is the genetic cause of oculopharyngeal muscle dystrophy (OPMD), a late onset and rare myopathy. Moreover, reduced PABPN1 expression correlates with symptom manifestation in OPMD. PABPN1 regulates alternative polyadenylation site (PAS) utilization. However, the impact of PAS utilization on cell and tissue function is poorly understood. We hypothesized that altered PABPN1 expression levels is an underlying cause of muscle wasting. To test this, we stably down-regulated PABPN1 in mouse tibialis anterior (TA) muscles by localized injection of adeno-associated viruses expressing shRNA to PABPN1 (shPab). We found that a mild reduction in PABPN1 levels causes muscle pathology including myofiber atrophy, thickening of extracellular matrix and myofiber-type transition. Moreover, reduced PABPN1 levels caused a consistent decline in distal PAS utilization in the 3'-UTR of a subset of OPMD-dysregulated genes. This alternative PAS utilization led to up-regulation of Atrogin-1, a key muscle atrophy regulator, but down regulation of proteasomal genes. Additionally reduced PABPN1 levels caused a reduction in proteasomal activity, and transition in MyHC isotope expression pattern in myofibers. We suggest that PABPN1-mediated alternative PAS utilization plays a central role in aging-associated muscle wasting.


Asunto(s)
Envejecimiento/genética , Proteínas Musculares/biosíntesis , Distrofia Muscular Oculofaríngea/genética , Proteína I de Unión a Poli(A)/genética , Proteínas Ligasas SKP Cullina F-box/biosíntesis , Envejecimiento/patología , Animales , Dependovirus/genética , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/metabolismo , Distrofia Muscular Oculofaríngea/patología , Proteína I de Unión a Poli(A)/biosíntesis , ARN Mensajero/biosíntesis , Proteínas Ligasas SKP Cullina F-box/genética
10.
Hum Mol Genet ; 25(19): 4282-4287, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27506982

RESUMEN

Molecular biomarkers emerge as an accurate diagnostic tool, but are scarce for myopathies. Lack of outcome measures sensitive to disease onset and symptom severity hamper evaluation of therapeutic developments. Cytokines are circulating immunogenic molecules, and their potential as biomarkers has been exploited in the last decade. Cytokines are released from many tissues, including skeletal muscles, but their application to monitor muscle pathology is sparse. We report that the cytokine functional group is altered in the transcriptome of oculopharyngeal muscular dystrophy (OPMD). OPMD is a dominant, late-onset myopathy, caused by an alanine-expansion mutation in the gene encoding for poly(A) binding protein nuclear 1 (expPABPN1). Here, we investigated the hypothesis that cytokines could mark OPMD disease state. We determined cytokines levels the vastus lateralis muscle from genetically confirmed expPABPN1 carriers at a symptomatic or a presymptomatic stage. We identified cytokine-related genes candidates from a transcriptome study in a mouse overexpressing exp PABPN1 Six cytokines were found to be consistently down-regulated in OPMD vastus lateralis muscles. Expression levels of these cytokines were highly correlated in controls, but this correlation pattern was disrupted in OPMD. The levels of these 6 cytokines were not altered in expPABPN1 carriers at a pre-symptomatic stage, suggesting that this group of cytokines is a potential biomarker for muscle weakness in OPMD. Correlation pattern of expression levels could be a novel measurer for disease state.


Asunto(s)
Citocinas/biosíntesis , Distrofia Muscular Oculofaríngea/genética , Proteína I de Unión a Poli(A)/genética , Transcriptoma/genética , Animales , Biomarcadores , Citocinas/genética , Regulación de la Expresión Génica , Heterocigoto , Humanos , Ratones , Distrofia Muscular Oculofaríngea/diagnóstico , Distrofia Muscular Oculofaríngea/patología , Patología Molecular , Proteína I de Unión a Poli(A)/biosíntesis , Músculo Cuádriceps/patología
11.
Am J Pathol ; 184(4): 1119-1131, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24486325

RESUMEN

Alanine expansion mutations in poly(A)-binding protein nuclear 1 (PABPN1) cause muscle weakness in the late-onset disorder oculopharyngeal muscular dystrophy. In affected muscles, expanded PABPN1 forms nuclear aggregates, depleting levels of soluble PABPN1 and inducing a genome-wide shift from distal to proximal polyadenylation site usage. PABPN1 protein accumulation is regulated by the ubiquitin proteasome system, which is highly dysregulated in oculopharyngeal muscular dystrophy. We show that ARIH2 E3-ligase regulates PABPN1 protein accumulation and aggregation. Levels of ARIH2 mRNA are regulated by PABPN1 via proximal polyadenylation site usage. We demonstrate that masking the proximal polyadenylation site in ARIH2 3' untranslated region by antisense oligonucleotides elevates the expression of ARIH2 and PABPN1 and restores myogenic defects that are induced by ARIH2 or PABPN1 down-regulation in cell culture. In vivo ARIH2 mRNA levels significantly decrease from midlife in vastus lateralis muscles and highly correlate with muscle degeneration. We suggest that the expression of both genes is maintained by a feed-forward loop between mRNA stability regulated by PABPN1 and protein turnover regulated by ARIH2.


Asunto(s)
Envejecimiento/patología , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/metabolismo , Proteína I de Unión a Poli(A)/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Envejecimiento/fisiología , Animales , Western Blotting , Línea Celular , Regulación de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína I de Unión a Poli(A)/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ubiquitina-Proteína Ligasas/genética
12.
BMC Genomics ; 15: 132, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24524210

RESUMEN

BACKGROUND: Genome-wide expression profiles are altered during biological aging and can describe molecular regulation of tissue degeneration. Age-regulated mRNA expression trends from cross-sectional studies could describe how aging progresses. We developed a novel statistical methodology to identify age-regulated expression trends in cross-sectional datasets. RESULTS: We studied six cross-sectional RNA expression profiles from different human tissues. Our methodology, capable of overcoming technical and genetic background differences, identified an age-regulation in four of the tissues. For the identification of expression trends, five regression models were compared and the quadratic model was found as the most suitable for this study. After k-means clustering of the age-associated probes, expression trends were found to change at two major age-positions in brain cortex and in Vastus lateralis muscles. The first age-position was found to occur during the fifth decade and a later one during the eighth decade. In kidney cortex, however, only one age-position was identified correlating with a late age-position. Functional mapping of genes at each age-position suggests that calcium homeostasis and lipid metabolisms are initially affected and subsequently, in elderly mitochondria, apoptosis and hormonal signaling pathways are affected. CONCLUSIONS: Our results suggest that age-associated temporal changes in human tissues progress at distinct age-positions, which differ between tissues and in their molecular composition.


Asunto(s)
Envejecimiento , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Encéfalo/metabolismo , Análisis por Conglomerados , Estudios Transversales , Humanos , Riñón/metabolismo , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Transcriptoma
13.
Nucleic Acids Res ; 40(18): 9089-101, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22772983

RESUMEN

The choice for a polyadenylation site determines the length of the 3'-untranslated region (3'-UTRs) of an mRNA. Inclusion or exclusion of regulatory sequences in the 3'-UTR may ultimately affect gene expression levels. Poly(A) binding protein nuclear 1 (PABPN1) is involved in polyadenylation of pre-mRNAs. An alanine repeat expansion in PABPN1 (exp-PABPN1) causes oculopharyngeal muscular dystrophy (OPMD). We hypothesized that previously observed disturbed gene expression patterns in OPMD muscles may have been the result of an effect of PABPN1 on alternative polyadenylation, influencing mRNA stability, localization and translation. A single molecule polyadenylation site sequencing method was developed to explore polyadenylation site usage on a genome-wide level in mice overexpressing exp-PABPN1. We identified 2012 transcripts with altered polyadenylation site usage. In the far majority, more proximal alternative polyadenylation sites were used, resulting in shorter 3'-UTRs. 3'-UTR shortening was generally associated with increased expression. Similar changes in polyadenylation site usage were observed after knockdown or overexpression of expanded but not wild-type PABPN1 in cultured myogenic cells. Our data indicate that PABPN1 is important for polyadenylation site selection and that reduced availability of functional PABPN1 in OPMD muscles results in use of alternative polyadenylation sites, leading to large-scale deregulation of gene expression.


Asunto(s)
Regiones no Traducidas 3' , Proteína I de Unión a Poli(A)/metabolismo , Poliadenilación , Animales , Línea Celular , Humanos , Ratones , Músculo Esquelético/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ARN
14.
Bio Protoc ; 14(2): e4922, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38268975

RESUMEN

Capillary density in skeletal muscles is key to estimate exercise capacity in healthy individuals, athletes, and those with muscle-related pathologies. Here, we present a step-by-step, high-throughput semi-automated method for quantifying capillary density from whole human skeletal muscle cross-sections, in areas of the muscle occupied by myofibers. We provide a detailed protocol for immunofluorescence staining, image acquisition, processing, and quantification. Image processing is performed in ImageJ, and data analysis is conducted in R. The provided protocol allows high-throughput quantification of capillary density. Key features • This protocol builds upon the method and results described in Abbassi-Daloii et al. (2023b). • It includes step-by-step details on image acquisition and image processing of the entire muscle section. • It enables high-throughput and semi-automated image quantification of capillary density. • It provides a robust analysis for determining capillary density over the entire muscle cross section.

15.
HGG Adv ; 5(2): 100269, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38213032

RESUMEN

Alternative polyadenylation (APA) at the 3' UTR of transcripts contributes to the cell transcriptome. APA is suppressed by the nuclear RNA-binding protein PABPN1. Aging-associated reduced PABPN1 levels in skeletal muscles lead to muscle wasting. Muscle weakness in oculopharyngeal muscular dystrophy (OPMD) is caused by short alanine expansion in PABPN1 exon1. The expanded PABPN1 forms nuclear aggregates, an OPMD hallmark. Whether the expanded PABPN1 affects APA and how it contributes to muscle pathology is unresolved. To investigate these questions, we developed a procedure including RNA library preparation and a simple pipeline calculating the APA-shift ratio as a readout for PABPN1 activity. Comparing APA-shift results to previously published PAS utilization and APA-shift results, we validated this procedure. The procedure was then applied on the OPMD cell model and on RNA from OPMD muscles. APA-shift was genome-wide in the mouse OPMD model, primarily affecting muscle transcripts. In OPMD individuals, APA-shift was enriched with muscle transcripts. In an OPMD cell model APA-shift was not significant. APA-shift correlated with reduced expression levels of a subset of PABPN1 isoforms, whereas the expression of the expanded PABPN1 did not correlate with APA-shift. PABPN1 activity is not affected by the expression of expanded PABPN1, but rather by reduced PABPN1 expression levels. In muscles, PABPN1 activity initially affects muscle transcripts. We suggest that muscle weakness in OPMD is caused by PABPN1 loss-of-function leading to APA-shift that primarily affects in muscle transcripts.


Asunto(s)
Distrofia Muscular Oculofaríngea , Animales , Ratones , Modelos Animales de Enfermedad , Debilidad Muscular/genética , Músculo Esquelético/metabolismo , Distrofia Muscular Oculofaríngea/genética , Poliadenilación/genética , ARN/metabolismo
16.
Development ; 137(4): 597-606, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20110325

RESUMEN

Dark-grown dicotyledonous seedlings form a hook-like structure at the top of the hypocotyl, which is controlled by the hormones auxin and ethylene. Hook formation is dependent on an auxin signal gradient, whereas hook exaggeration is part of the triple response provoked by ethylene in dark-grown Arabidopsis seedlings. Several other hormones and light are also known to be involved in hook development, but the molecular mechanisms that lead to the initial installation of an auxin gradient are still poorly understood. In this study, we aimed to unravel the cross-talk between auxin and ethylene in the apical hook. Auxin measurements, the expression pattern of the auxin reporter DR5::GUS and the localization of auxin biosynthesis enzymes and influx carriers collectively indicate the necessity for auxin biosynthesis and efficient auxin translocation from the cotyledons and meristem into the hypocotyl in order to support proper hook development. Auxin accumulation in the meristem and cotyledons and in the hypocotyl is increased approximately 2-fold upon treatment with ethylene. In addition, a strong ethylene signal leads to enhanced auxin biosynthesis at the inner side of the hook. Finally, mutant analysis demonstrates that the auxin influx carrier LAX3 is indispensable for proper hook formation, whereas the auxin influx carrier AUX1 is involved in the hook exaggeration phenotype induced by ethylene.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Ácidos Indolacéticos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Etilenos/farmacología , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Hipocótilo/crecimiento & desarrollo , Hipocótilo/metabolismo , Proteínas de Transporte de Membrana/genética , Meristema/efectos de los fármacos , Meristema/crecimiento & desarrollo , Meristema/metabolismo , Mutación , Fenotipo , Plantas Modificadas Genéticamente , Plantones/efectos de los fármacos , Plantones/crecimiento & desarrollo , Plantones/metabolismo
17.
Development ; 137(4): 607-17, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20110326

RESUMEN

The apical hook of dark-grown Arabidopsis seedlings is a simple structure that develops soon after germination to protect the meristem tissues during emergence through the soil and that opens upon exposure to light. Differential growth at the apical hook proceeds in three sequential steps that are regulated by multiple hormones, principally auxin and ethylene. We show that the progress of the apical hook through these developmental phases depends on the dynamic, asymmetric distribution of auxin, which is regulated by auxin efflux carriers of the PIN family. Several PIN proteins exhibited specific, partially overlapping spatial and temporal expression patterns, and their subcellular localization suggested auxin fluxes during hook development. Genetic manipulation of individual PIN activities interfered with different stages of hook development, implying that specific combinations of PIN genes are required for progress of the apical hook through the developmental phases. Furthermore, ethylene might modulate apical hook development by prolonging the formation phase and strongly suppressing the maintenance phase. This ethylene effect is in part mediated by regulation of PIN-dependent auxin efflux and auxin signaling.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Ácidos Indolacéticos/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Etilenos/farmacología , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Meristema/efectos de los fármacos , Meristema/crecimiento & desarrollo , Meristema/metabolismo , Modelos Biológicos , Mutación , Plantas Modificadas Genéticamente , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Plantones/efectos de los fármacos , Plantones/crecimiento & desarrollo , Plantones/metabolismo
18.
BMC Neurol ; 13: 70, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23815790

RESUMEN

BACKGROUND: Muscle fibrosis characterizes degenerated muscles in muscular dystrophies and in late onset myopathies. Fibrotic muscles often exhibit thickening of the extracellular matrix (ECM). The molecular regulation of this process is not fully understood. In oculopharyngeal muscular dystrophy (OPMD), an expansion of an alanine tract at the N-terminus of poly(A)-binding protein nuclear 1 (PABPN1) causes muscle symptoms. OPMD patient muscle degeneration initiates after midlife, while at an earlier age carriers of alanine expansion mutant PABPN1 (expPABPN1) are clinically pre-symptomatic. OPMD is characterized by fibrosis in skeletal muscles but the causative molecular mechanisms are not fully understood. METHODS: We studied the molecular processes that are involved in OPMD pathology using cross-species mRNA expression profiles in muscles from patients and model systems. We identified significant dysregulation of the ECM functional group, among which the procollagen C-endopeptidase enhancer 1 gene (PCOLCE) was consistently down-regulated across species. We investigated PCOLCE subcellular localization in OPMD muscle samples and OPMD model systems to investigate any functional relevance of PCOLCE down-regulation in this disease. RESULTS: We found that muscle degeneration in OPMD is associated with PCOLCE down-regulation. In addition to its known presence at the ECM, we also found PCOLCE within the nucleus of muscle cells. PCOLCE sub-cellular localization changes during myoblast cell fusion and is disrupted in cells expressing mutant expPABPN1. Our results show that PCOLCE binds to soluble PABPN1 and co-localizes with aggregated PABPN1 with a preference for the mutant protein. In muscle biopsies from OPMD patients we find that extracellular PCOLCE is depleted with its concomitant enrichment within the nuclear compartment. CONCLUSIONS: PCOLCE regulates collagen processing at the ECM. Depletion of extracellular PCOLCE is associated with the expression of expPABPN1 in OPMD patient muscles. PCOLCE is also localized within the nucleus where it binds to PABPN1, suggesting that PCOLCE shuttles between the ECM and the nucleus. PCOLCE preferentially binds to expPABPN1. Nuclear-localized PCOLCE is enriched in muscle cells expressing expPABPN1. We suggest that nuclear entrapment of PCOLCE and its extracellular depletion represents a novel molecular mechanism in late-onset muscle fibrosis.


Asunto(s)
Regulación hacia Abajo/genética , Proteínas de la Matriz Extracelular/deficiencia , Glicoproteínas/deficiencia , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/patología , Factores de Edad , Alanina/genética , Animales , Nucléolo Celular/metabolismo , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofia Muscular Oculofaríngea/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Mioblastos/patología , Proteína I de Unión a Poli(A)/genética , Proteína I de Unión a Poli(A)/metabolismo , Transfección
19.
STAR Protoc ; 4(1): 102075, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36853713

RESUMEN

Skeletal muscles are composed of different myofiber types characterized by the expression of myosin heavy chain isoforms, which can be affected by physical activity, aging, and pathological conditions. Here, we present a step-by-step high-throughput semi-automated approach for performing myofiber type quantification of entire human or mouse muscle tissue sections, including immunofluorescence staining, image acquisition, processing, and quantification. For complete details on the use and execution of this protocol, please refer to Abbassi-Daloii et al. (2022).1.


Asunto(s)
Envejecimiento , Músculo Esquelético , Ratones , Animales , Humanos , Envejecimiento/genética , Envejecimiento/metabolismo
20.
Metabolites ; 13(6)2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37367926

RESUMEN

The identification of metabolomic biomarkers relies on the analysis of large cohorts of patients compared to healthy controls followed by the validation of markers in an independent sample set. Indeed, circulating biomarkers should be causally linked to pathology to ensure that changes in the marker precede changes in the disease. However, this approach becomes unfeasible in rare diseases due to the paucity of samples, necessitating the development of new methods for biomarker identification. The present study describes a novel approach that combines samples from both mouse models and human patients to identify biomarkers of OPMD. We initially identified a pathology-specific metabolic fingerprint in murine dystrophic muscle. This metabolic fingerprint was then translated into (paired) murine serum samples and then to human plasma samples. This study identified a panel of nine candidate biomarkers that could predict muscle pathology with a sensitivity of 74.3% and specificity of 100% in a random forest model. These findings demonstrate that the proposed approach can identify biomarkers with good predictive performance and a higher degree of confidence in their relevance to pathology than markers identified in a small cohort of human samples alone. Therefore, this approach has a high potential utility for identifying circulating biomarkers in rare diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA