Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36835433

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal late-onset motor neuron disease characterized by the loss of the upper and lower motor neurons. Our understanding of the molecular basis of ALS pathology remains elusive, complicating the development of efficient treatment. Gene-set analyses of genome-wide data have offered insight into the biological processes and pathways of complex diseases and can suggest new hypotheses regarding causal mechanisms. Our aim in this study was to identify and explore biological pathways and other gene sets having genomic association to ALS. Two cohorts of genomic data from the dbGaP repository were combined: (a) the largest available ALS individual-level genotype dataset (N = 12,319), and (b) a similarly sized control cohort (N = 13,210). Following comprehensive quality control pipelines, imputation and meta-analysis, we assembled a large European descent ALS-control cohort of 9244 ALS cases and 12,795 healthy controls represented by genetic variants of 19,242 genes. Multi-marker analysis of genomic annotation (MAGMA) gene-set analysis was applied to an extensive collection of 31,454 gene sets from the molecular signatures database (MSigDB). Statistically significant associations were observed for gene sets related to immune response, apoptosis, lipid metabolism, neuron differentiation, muscle cell function, synaptic plasticity and development. We also report novel interactions between gene sets, suggestive of mechanistic overlaps. A manual meta-categorization and enrichment mapping approach is used to explore the overlap of gene membership between significant gene sets, revealing a number of shared mechanisms.


Asunto(s)
Esclerosis Amiotrófica Lateral , Humanos , Esclerosis Amiotrófica Lateral/genética , Estudio de Asociación del Genoma Completo , Genotipo , Neuronas Motoras
2.
Eur J Neurol ; 29(3): 707-714, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34748676

RESUMEN

BACKGROUND AND PURPOSE: This study evaluates the incidence, prevalence and survival trends of motor neurone disease (MND) in Northern Ireland from 2015 to 2019. METHODS: A capture-recapture analysis was performed using five independent data sources. Incidence and prevalence rates were standardized to the European Standard Population. Survival outcomes were analysed using Kaplan-Meier curves and Cox regression analysis. RESULTS: Amongst 254 total cases of MND, capture-recapture analysis estimated three missing cases (case ascertainment 98.8%). Age standardized incidence of captured cases was 3.12 per 100,000 (2.73, 3.50) and standardized prevalence ranged from 9.45 to 6.49 per 100,000 from 2015 to 2019. Standardized incidence and prevalence rates in 2006 were 1.4 and 3.3 per 100,000 respectively. Of identified cases, 133 (52.4%) were male; 94.5% had amyotrophic lateral sclerosis; median age of onset was 67 years; median time to diagnosis was 12 months (95% confidence interval 11.2, 12.8); survival from diagnosis was 12 months (95% confidence interval 10.6, 15.4); 25 (9.8%) reported a family history of MND or frontotemporal dementia; and a known MND-associated genetic mutation was identified in 7.9% of total cases, of which the most common was C9orf72 (5.7% of all patients). Factors associated with improved survival were younger age at onset, longer time to diagnosis, attendance at regional MND clinic, and initial neurology presentation as outpatient (all p < 0.001). CONCLUSION: The incidence and prevalence of MND in Northern Ireland has increased over the last 10 years, in line with increasing rates reported from other European countries. Improved survival was associated with younger age at onset, longer time to diagnosis, attendance at a regional MND clinic and outpatient presentation to a Neurology Department.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedad de la Neurona Motora , Anciano , Esclerosis Amiotrófica Lateral/diagnóstico , Esclerosis Amiotrófica Lateral/epidemiología , Humanos , Incidencia , Masculino , Enfermedad de la Neurona Motora/epidemiología , Irlanda del Norte/epidemiología , Prevalencia
3.
Ann Neurol ; 88(2): 332-347, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32403198

RESUMEN

OBJECTIVE: A hitherto undescribed phenotype of early onset muscular dystrophy associated with sensorineural hearing loss and primary ovarian insufficiency was initially identified in 2 siblings and in subsequent patients with a similar constellation of findings. The goal of this study was to understand the genetic and molecular etiology of this condition. METHODS: We applied whole exome sequencing (WES) superimposed on shared haplotype regions to identify the initial biallelic variants in GGPS1 followed by GGPS1 Sanger sequencing or WES in 5 additional families with the same phenotype. Molecular modeling, biochemical analysis, laser membrane injury assay, and the generation of a Y259C knock-in mouse were done. RESULTS: A total of 11 patients in 6 families carrying 5 different biallelic pathogenic variants in specific domains of GGPS1 were identified. GGPS1 encodes geranylgeranyl diphosphate synthase in the mevalonate/isoprenoid pathway, which catalyzes the synthesis of geranylgeranyl pyrophosphate, the lipid precursor of geranylgeranylated proteins including small guanosine triphosphatases. In addition to proximal weakness, all but one patient presented with congenital sensorineural hearing loss, and all postpubertal females had primary ovarian insufficiency. Muscle histology was dystrophic, with ultrastructural evidence of autophagic material and large mitochondria in the most severe cases. There was delayed membrane healing after laser injury in patient-derived myogenic cells, and a knock-in mouse of one of the mutations (Y259C) resulted in prenatal lethality. INTERPRETATION: The identification of specific GGPS1 mutations defines the cause of a unique form of muscular dystrophy with hearing loss and ovarian insufficiency and points to a novel pathway for this clinical constellation. ANN NEUROL 2020;88:332-347.


Asunto(s)
Dimetilaliltranstransferasa/genética , Farnesiltransferasa/genética , Geraniltranstransferasa/genética , Pérdida Auditiva/genética , Distrofias Musculares/genética , Mutación/genética , Insuficiencia Ovárica Primaria/genética , Adolescente , Adulto , Animales , Femenino , Técnicas de Sustitución del Gen/métodos , Pérdida Auditiva/diagnóstico por imagen , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofias Musculares/diagnóstico por imagen , Linaje , Insuficiencia Ovárica Primaria/diagnóstico por imagen , Estructura Secundaria de Proteína , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos , Adulto Joven
4.
J Infect Dis ; 220(4): 710-719, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31001627

RESUMEN

BACKGROUND: Necrotizing soft tissue infections (NSTIs) caused by group A Streptococcus (GAS) and occasionally by Staphylococcus aureus (SA) frequently involve the deep fascia and often lead to muscle necrosis. METHODS: To assess the pathogenicity of GAS and S. aureus for muscles in comparison to keratinocytes, adhesion and invasion of NSTI-GAS and NSTI-SA isolates were assessed in these cells. Bloodstream infections (BSI-SA) and noninvasive coagulase-negative staphylococci (CNS) isolates were used as controls. RESULTS: NSTI-SA and BSI-SA exhibited stronger internalization into human keratinocytes and myoblasts than NSTI-GAS or CNS. S. aureus internalization reached over 30% in human myoblasts due to a higher percentage of infected myoblasts (>11%) as compared to keratinocytes (<3%). Higher cytotoxicity for myoblasts of NSTI-SA as compared to BSI-SA was attributed to higher levels of psmα and RNAIII transcripts in NSTI-SA. However, the 2 groups were not discriminated at the genomic level. The cellular basis of high internalization rate in myoblasts was attributed to higher expression of α5ß1 integrin in myoblasts. Major contribution of FnbpAB-integrin α5ß1 pathway to internalization was confirmed by isogenic mutants. CONCLUSIONS: Our findings suggest a factor in NSTI-SA severity is the strong invasiveness of S. aureus in muscle cells, a property not shared by NSTI-GAS isolates.


Asunto(s)
Fascitis Necrotizante/microbiología , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Infecciones Estreptocócicas/microbiología , Anciano , Femenino , Humanos , Queratinocitos/microbiología , Masculino , Células Musculares/microbiología , Mioblastos/microbiología , Staphylococcus aureus/genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Adulto Joven
5.
Nucleic Acids Res ; 43(W1): W571-5, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25883154

RESUMEN

Given a query list of genes or proteins, CellWhere produces an interactive graphical display that mimics the structure of a cell, showing the local interaction network organized into subcellular locations. This user-friendly tool helps in the formulation of mechanistic hypotheses by enabling the experimental biologist to explore simultaneously two elements of functional context: (i) protein subcellular localization and (ii) protein-protein interactions or gene functional associations. Subcellular localization terms are obtained from public sources (the Gene Ontology and UniProt-together containing several thousand such terms) then mapped onto a smaller number of CellWhere localizations. These localizations include all major cell compartments, but the user may modify the mapping as desired. Protein-protein interaction listings, and their associated evidence strength scores, are obtained from the Mentha interactome server, or power-users may upload a pre-made network produced using some other interactomics tool. The Cytoscape.js JavaScript library is used in producing the graphical display. Importantly, for a protein that has been observed at multiple subcellular locations, users may prioritize the visual display of locations that are of special relevance to their research domain. CellWhere is at http://cellwhere-myology.rhcloud.com.


Asunto(s)
Mapeo de Interacción de Proteínas , Proteínas/análisis , Programas Informáticos , Gráficos por Computador , Genes , Internet , Espacio Intracelular/química
6.
Hum Mol Genet ; 23(18): 4745-57, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24760765

RESUMEN

While spinal muscular atrophy (SMA) is characterized by motor neuron degeneration, it is unclear whether and how much survival motor neuron (SMN) protein deficiency in muscle contributes to the pathophysiology of the disease. There is increasing evidence from patients and SMA model organisms that SMN deficiency causes intrinsic muscle defects. Here we investigated the role of SMN in muscle development using muscle cell lines and primary myoblasts. Formation of multinucleate myotubes by SMN-deficient muscle cells is inhibited at a stage preceding plasma membrane fusion. We found increased expression and reduced induction of key muscle development factors, such as MyoD and myogenin, with differentiation of SMN-deficient cells. In addition, SMN-deficient muscle cells had impaired cell migration and altered organization of focal adhesions and the actin cytoskeleton. Partially restoring SMN inhibited the premature expression of muscle differentiation markers, corrected the cytoskeletal abnormalities and improved myoblast fusion. These findings are consistent with a role for SMN in myotube formation through effects on muscle differentiation and cell motility.


Asunto(s)
Adhesiones Focales/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Miogenina/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Diferenciación Celular , Línea Celular , Movimiento Celular , Regulación de la Expresión Génica , Humanos , Ratones , Fibras Musculares Esqueléticas/citología , Proteína MioD/genética , Proteína MioD/metabolismo , Miogenina/metabolismo , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética
7.
Ann Neurol ; 78(3): 387-400, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26018399

RESUMEN

OBJECTIVE: Facioscapulohumeral muscular dystrophy (FSHD) is linked to either contraction of D4Z4 repeats on chromosome 4 or to mutations in the SMCHD1 gene, both of which result in the aberrant expression of the transcription factor DUX4. However, it is still difficult to correlate these genotypes with the phenotypes observed in patients. Because we have recently shown that mice with disrupted Fat1 functions exhibit FSHD-like phenotypes, we have investigated the expression of the human FAT1 gene in FSHD. METHODS: We first analyzed FAT1 expression in FSHD adult muscles and determined whether FAT1 expression was driven by DUX4. We next determined FAT1 expression levels in 64 muscles isolated from 16 control fetuses. These data were further complemented with analysis of Fat1 expression in developing mouse embryos. RESULTS: We demonstrated that FAT1 expression is independent of DUX4. Moreover, we observed that (1) in control fetal human biopsies or in developing mouse embryos, FAT1 is expressed at lower levels in muscles that are affected at early stages of FSHD progression than in muscles that are affected later or are nonaffected; and (2) in adult muscle biopsies, FAT1 expression is lower in FSHD muscles compared to control muscles. INTERPRETATION: We propose a revised model for FSHD in which FAT1 levels might play a role in determining which muscles will exhibit early and late disease onset, whereas DUX4 may worsen the muscle phenotype.


Asunto(s)
Cadherinas/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Distrofia Muscular Facioescapulohumeral/diagnóstico , Distrofia Muscular Facioescapulohumeral/metabolismo , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , Adulto , Animales , Células Cultivadas , Femenino , Feto , Humanos , Masculino , Ratones , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Cuádriceps/embriología
8.
Proc Natl Acad Sci U S A ; 109(34): 13763-8, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869723

RESUMEN

Duchenne muscular dystrophy (DMD), the commonest form of muscular dystrophy, is caused by lack of dystrophin. One of the most promising therapeutic approaches is antisense-mediated elimination of frame-disrupting mutations by exon skipping. However, this approach faces two major hurdles: limited applicability of each individual target exon and uncertain function and stability of each resulting truncated dystrophin. Skipping of exons 45-55 at the mutation hotspot of the DMD gene would address both issues. Theoretically it could rescue more than 60% of patients with deletion mutations. Moreover, spontaneous deletions of this specific region are associated with asymptomatic or exceptionally mild phenotypes. However, such multiple exon skipping of exons 45-55 has proved technically challenging. We have therefore designed antisense oligo (AO) morpholino mixtures to minimize self- or heteroduplex formation. These were tested as conjugates with cell-penetrating moieties (vivo-morpholinos). We have tested the feasibility of skipping exons 45-55 in H2K-mdx52 myotubes and in mdx52 mice, which lack exon 52. Encouragingly, with mixtures of 10 AOs, we demonstrated skipping of all 10 exons in vitro, in H2K-mdx52 myotubes and on intramuscular injection into mdx52 mice. Moreover, in mdx52 mice in vivo, systemic injections of 10 AOs induced extensive dystrophin expression at the subsarcolemma in skeletal muscles throughout the body, producing up to 15% of wild-type dystrophin protein levels, accompanied by improved muscle strength and histopathology without any detectable toxicity. This is a unique successful demonstration of effective rescue by exon 45-55 skipping in a dystrophin-deficient animal model.


Asunto(s)
Distrofina/genética , Exones , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Oligonucleótidos Antisentido/genética , Animales , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Fibras Musculares Esqueléticas , Medicina de Precisión
9.
Mol Ther ; 21(10): 1950-7, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23975040

RESUMEN

The development of innovative therapeutic strategies for muscular dystrophies, particularly cell-based approaches, is still a developing field. Although positive results have been obtained in animal models, they have rarely been confirmed in patients and resulted in very limited clinical improvements, suggesting some specificity in humans. These findings emphasized the need for an appropriate animal model (i.e., immunodeficient and dystrophic) to investigate in vivo the behavior of transplanted human myogenic stem cells. We report a new model, the Rag2(-)Il2rb(-)Dmd(-) mouse, which lacks T, B, and NK cells, and also carries a mutant Dmd allele that prevents the production of any dystrophin isoform. The dystrophic features of this new model are comparable with those of the classically used mdx mouse, but with the total absence of any revertant dystrophin positive fiber. We show that Rag2(-)Il2rb(-)Dmd(-) mice allow long-term xenografts of human myogenic cells. Altogether, our findings indicate that the Rag2(-)Il2rb(-)Dmd(-) mouse represents an ideal model to gain further insights into the behavior of human myogenic stem cells in a dystrophic context, and can be used to assess innovative therapeutic strategies for muscular dystrophies.


Asunto(s)
Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Distrofina/genética , Subunidad beta del Receptor de Interleucina-2/genética , Ratones Endogámicos mdx/genética , Distrofias Musculares/patología , Distrofia Muscular Animal/patología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Técnicas de Inactivación de Genes , Humanos , Recién Nacido , Masculino , Ratones , Ratones Endogámicos C57BL , Distrofias Musculares/terapia , Distrofia Muscular Animal/terapia , Mioblastos/trasplante , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell Mol Life Sci ; 70(12): 2159-74, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23344255

RESUMEN

Duchenne muscular dystrophy results from loss of the protein dystrophin, which links the intracellular cytoskeletal network with the extracellular matrix, but deficiency in this function does not fully explain the onset or progression of the disease. While some intracellular events involved in the degeneration of dystrophin-deficient muscle fibers have been well characterized, changes in their secretory profile are undescribed. To analyze the secretome profile of mdx myotubes independently of myonecrosis, we labeled the proteins of mdx and wild-type myotubes with stable isotope-labeled amino acids (SILAC), finding marked enrichment of vesicular markers in the mdx secretome. These included the lysosomal-associated membrane protein, LAMP1, that co-localized in vesicles with an over-secreted cytoskeletal protein, myosin light chain 1. These LAMP1/MLC1-3-positive vesicles accumulated in the cytosol of mdx myotubes and were secreted into the culture medium in a range of abnormal densities. Restitution of dystrophin expression, by exon skipping, to some 30 % of the control value, partially normalized the secretome profile and the excess LAMP1 accumulation. Together, our results suggest that a lack of dystrophin leads to a general dysregulation of vesicle trafficking. We hypothesize that disturbance of the export of proteins through vesicles occurs before, and then concurrently with, the myonecrotic cascade and contributes chronically to the pathophysiology of DMD, thereby presenting us with a range of new potential therapeutic targets.


Asunto(s)
Distrofina/deficiencia , Proteínas de Membrana de los Lisosomas/metabolismo , Fibras Musculares Esqueléticas/patología , Distrofia Muscular de Duchenne/metabolismo , Vesículas Secretoras/metabolismo , Actinas/análisis , Aminoácidos/metabolismo , Animales , Western Blotting , Línea Celular , Cromatografía Liquida , Biología Computacional , Immunoblotting , Marcaje Isotópico , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica de Transmisión , Fibras Musculares Esqueléticas/metabolismo , Vesículas Secretoras/ultraestructura , Estadísticas no Paramétricas , Espectrometría de Masas en Tándem
11.
Biogerontology ; 14(3): 339-52, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624703

RESUMEN

Skeletal muscle ageing is characterized by a progressive and dramatic loss of muscle mass and strength leading to decreased muscular function resulting in muscle weakness which is often referred to as sarcopenia. Following the standardisation of "omics" approaches to study the genome (genomics) and the transcriptome (transcriptomics), the study of the proteins encoded by the genome, referred to as proteomics, is a tremendous challenge. Unlike the genome, the proteome varies in response to many physiological or pathological factors. In addition, the proteome is orders of magnitude more complex than the transcriptome due to post-translational modifications, protein oxidation and limited protein degradation. Proteomic studies, including the analysis of protein abundance as well as post-translational modified proteins have been shown to provide valuable information to unravel the key molecular pathways implicated in complex biological processes, such as tissue and organ ageing. In this article, we will describe proteomic approaches for the analysis of protein abundance as well as the specific protein targets for oxidative damage upon oxidative stress and/or during skeletal muscle ageing.


Asunto(s)
Envejecimiento/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas Musculares/fisiología , Músculo Esquelético/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Proteómica/métodos , Envejecimiento/genética , Animales , Citoesqueleto/fisiología , Metabolismo Energético/fisiología , Europa (Continente) , Humanos , Modelos Animales , Proteínas Musculares/genética , Estrés Oxidativo/fisiología , Sarcopenia/fisiopatología
12.
Biogerontology ; 14(3): 273-92, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23666344

RESUMEN

Although adult skeletal muscle is composed of fully differentiated fibers, it retains the capacity to regenerate in response to injury and to modify its contractile and metabolic properties in response to changing demands. The major role in the growth, remodeling and regeneration is played by satellite cells, a quiescent population of myogenic precursor cells that reside between the basal lamina and plasmalemma and that are rapidly activated in response to appropriate stimuli. However, in pathologic conditions or during aging, the complete regenerative program can be precluded by fibrotic tissue formation and resulting in functional impairment of the skeletal muscle. Our study, along with other studies, demonstrated that although the regenerative program can also be impaired by the limited proliferative capacity of satellite cells, this limit is not reached during normal aging, and it is more likely that the restricted muscle repair program in aging is presumably due to missing signals that usually render the damaged muscle a permissive environment for regenerative activity.


Asunto(s)
Envejecimiento/fisiología , Proliferación Celular , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Proteínas de Neoplasias/metabolismo , Estrés Oxidativo/fisiología , Sarcopenia/metabolismo , Sarcopenia/patología , Sarcopenia/fisiopatología , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Telómero/ultraestructura , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
13.
Life (Basel) ; 13(6)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37374084

RESUMEN

Neurofilament levels are elevated in many neurodegenerative diseases and have shown promise as diagnostic and prognostic biomarkers in Amyotrophic Lateral Sclerosis (ALS), the most common form of Motor Neuron Disease (MND). This study assesses serum neurofilament light (NFL) and neurofilament heavy (NFH) chain concentrations in patients with ALS, other variants of motor neuron disease such as Progressive Muscular Atrophy (PMA) and Primary Lateral Sclerosis (PLS), and a range of other neurological diseases. It aims to evaluate the use of NFL and NFH to differentiate these conditions and for the prognosis of MND disease progression. NFL and NFH levels were quantified using electrochemiluminescence immunoassays (ECLIA). Both were elevated in 47 patients with MND compared to 34 patients with other neurological diseases and 33 healthy controls. NFL was able to differentiate patients with MND from the other groups with a Receiver Operating Characteristic (ROC) curve area under the curve (AUC) of 0.90 (p < 0.001). NFL correlated with the rate of disease progression in MND (rho 0.758, p < 0.001) and with the ALS Functional Rating Scale (rho -0.335, p = 0.021). NFL levels were higher in patients with ALS compared to both PMA (p = 0.032) and PLS (p = 0.012) and were able to distinguish ALS from both PMA and PLS with a ROC curve AUC of 0.767 (p = 0.005). These findings support the use of serum NFL to help diagnose and differentiate types of MND, in addition to providing prognostic information to patients and their families.

14.
Cells ; 12(3)2023 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-36766730

RESUMEN

Genes with similar expression patterns in a set of diverse samples may be considered coexpressed. Human Gene Coexpression Analysis 2.0 (HGCA2.0) is a webtool which studies the global coexpression landscape of human genes. The website is based on the hierarchical clustering of 55,431 Homo sapiens genes based on a large-scale coexpression analysis of 3500 GTEx bulk RNA-Seq samples of healthy individuals, which were selected as the best representative samples of each tissue type. HGCA2.0 presents subclades of coexpressed genes to a gene of interest, and performs various built-in gene term enrichment analyses on the coexpressed genes, including gene ontologies, biological pathways, protein families, and diseases, while also being unique in revealing enriched transcription factors driving coexpression. HGCA2.0 has been successful in identifying not only genes with ubiquitous expression patterns, but also tissue-specific genes. Benchmarking showed that HGCA2.0 belongs to the top performing coexpression webtools, as shown by STRING analysis. HGCA2.0 creates working hypotheses for the discovery of gene partners or common biological processes that can be experimentally validated. It offers a simple and intuitive website design and user interface, as well as an API endpoint.


Asunto(s)
Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Genes , Humanos , RNA-Seq , Factores de Transcripción , Genes/genética , Genes/fisiología
15.
J Pharmacol Exp Ther ; 343(1): 225-32, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22743576

RESUMEN

Glucocorticoids are standard of care for many inflammatory conditions, but chronic use is associated with a broad array of side effects. This has led to a search for dissociative glucocorticoids--drugs able to retain or improve efficacy associated with transrepression [nuclear factor-κB (NF-κB) inhibition] but with the loss of side effects associated with transactivation (receptor-mediated transcriptional activation through glucocorticoid response element gene promoter elements). We investigated a glucocorticoid derivative with a Δ-9,11 modification as a dissociative steroid. The Δ-9,11 analog showed potent inhibition of tumor necrosis factor-α-induced NF-κB signaling in cell reporter assays, and this transrepression activity was blocked by 17ß-hydroxy-11ß-[4-dimethylamino phenyl]-17α-[1-propynyl]estra-4,9-dien-3-one (RU-486), showing the requirement for the glucocorticoid receptor (GR). The Δ-9,11 analog induced the nuclear translocation of GR but showed the loss of transactivation as assayed by GR-luciferase constructs as well as mRNA profiles of treated cells. The Δ-9,11 analog was tested for efficacy and side effects in two mouse models of muscular dystrophy: mdx (dystrophin deficiency), and SJL (dysferlin deficiency). Daily oral delivery of the Δ-9,11 analog showed a reduction of muscle inflammation and improvements in multiple muscle function assays yet no reductions in body weight or spleen size, suggesting the loss of key side effects. Our data demonstrate that a Δ-9,11 analog dissociates the GR-mediated transcriptional activities from anti-inflammatory activities. Accordingly, Δ-9,11 analogs may hold promise as a source of safer therapeutic agents for chronic inflammatory disorders.


Asunto(s)
Dronabinol/análogos & derivados , Glucocorticoides/efectos adversos , Glucocorticoides/farmacología , FN-kappa B/antagonistas & inhibidores , Elementos de Respuesta/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Dronabinol/química , Dronabinol/farmacología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , FN-kappa B/metabolismo , Elementos de Respuesta/fisiología , Bazo/efectos de los fármacos , Bazo/metabolismo , Resultado del Tratamiento
16.
Expert Rev Proteomics ; 9(3): 337-45, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22809211

RESUMEN

Studies of the cell secretome have greatly increased in recent years owing to improvements in proteomic platforms, mass spectrometry instrumentation and to the increased interaction between analytical chemists, biologists and clinicians. Several secretome studies have been implemented in different areas of research, leading to the generation of a valuable secretome catalogs. Secreted proteins continue to be an important source of biomarkers and therapeutic target discovery and are equally valuable in the field of microbiology. Several discoveries have been achieved in vitro using cell culture systems, ex vivo using human tissue specimens and in vivo using animal models. In this review, some of the most recent advances in secretome studies and the fields that have benefited the most from this evolving technology are highlighted.


Asunto(s)
Proteínas/análisis , Proteínas/metabolismo , Proteómica/métodos , Líquido Amniótico/química , Biomarcadores de Tumor/análisis , Secreciones Corporales/química , Líquidos Corporales/química , Exosomas/química , Humanos , Espectrometría de Masas/métodos , Neoplasias/metabolismo
17.
Exp Cell Res ; 317(14): 1979-93, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21635888

RESUMEN

Research into muscle atrophy and hypertrophy is hampered by limitations of the available experimental models. Interpretation of in vivo experiments is confounded by the complexity of the environment while in vitro models are subject to the marked disparities between cultured myotubes and the mature myofibres of living tissues. Here we develop a method (PhAct) based on ex vivo maintenance of the isolated myofibre as a model of disuse atrophy, using standard microscopy equipment and widely available analysis software, to measure f-actin content per myofibre and per nucleus over two weeks of ex vivo maintenance. We characterize the 35% per week atrophy of the isolated myofibre in terms of early changes in gene expression and investigate the effects on loss of muscle mass of modulatory agents, including Myostatin and Follistatin. By tracing the incorporation of a nucleotide analogue we show that the observed atrophy is not associated with loss or replacement of myonuclei. Such a completely controlled investigation can be conducted with the myofibres of a single muscle. With this novel method we can distinguish those features and mechanisms of atrophy and hypertrophy that are intrinsic to the muscle fibre from those that include activities of other tissues and systemic agents.


Asunto(s)
Actinas/análisis , Fibras Musculares Esqueléticas/patología , Atrofia Muscular/patología , Programas Informáticos , Actinas/genética , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Pers Med ; 12(11)2022 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-36422108

RESUMEN

The rapid increase in the number of genetic variants identified to be associated with Amyotrophic Lateral Sclerosis (ALS) through genome-wide association studies (GWAS) has created an emerging need to understand the functional pathways that are implicated in the pathology of ALS. Gene-set analysis (GSA) is a powerful method that can provide insight into the associated biological pathways, determining the joint effect of multiple genetic markers. The main contribution of this review is the collection of ALS GSA studies that employ GWAS or individual-based genotype data, investigating their methodology and results related to ALS-associated molecular pathways. Furthermore, the limitations in standard single-gene analyses are summarized, highlighting the power of gene-set analysis, and a brief overview of the statistical properties of gene-set analysis and related concepts is provided. The main aims of this review are to investigate the reproducibility of the collected studies and identify their strengths and limitations, in order to enhance the experimental design and therefore the quality of the results of future studies, deepening our understanding of this devastating disease.

19.
J Pers Med ; 12(11)2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36579600

RESUMEN

Amyotrophic lateral sclerosis is a disease affecting upper and lower motor neurons. Although motor neuron death is the core event of ALS pathology, it is increasingly recognized that other tissues and cell types are affected in the disease, making potentially major contributions to the occurrence and progression of pathology. We review here the known cellular and molecular characteristics of muscle tissue affected by ALS. Evidence of toxicity in skeletal muscle tissue is considered, including metabolic dysfunctions, impaired proteostasis, and deficits in muscle regeneration and RNA metabolism. The role of muscle as a secretory organ, and effects on the skeletal muscle secretome are also covered, including the increase in secretion of toxic factors or decrease in essential factors that have consequences for neuronal function and survival.

20.
Life (Basel) ; 13(1)2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36676070

RESUMEN

Amyotrophic Lateral Sclerosis is a progressive neurodegenerative disease and is the most common adult motor neuron disease. The disease pathogenesis is complex with the perturbation of multiple pathways proposed, including mitochondrial dysfunction, RNA processing, glutamate excitotoxicity, endoplasmic reticulum stress, protein homeostasis and endosomal transport/extracellular vesicle (EV) secretion. EVs are nanoscopic membrane-bound particles that are released from cells, involved in the intercellular communication of proteins, lipids and genetic material, and there is increasing evidence of their role in ALS. After discussing the biogenesis of EVs, we review their roles in the propagation of pathological proteins in ALS, such as TDP-43, SOD1 and FUS, and their contribution to disease pathology. We also discuss the ALS related genes which are involved in EV formation and vesicular trafficking, before considering the EV protein and RNA dysregulation found in ALS and how these have been investigated as potential biomarkers. Finally, we highlight the potential use of EVs as therapeutic agents in ALS, in particular EVs derived from mesenchymal stem cells and EVs as drug delivery vectors for potential treatment strategies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA