Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Gene Ther ; 31(5-6): 234-241, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38135787

RESUMEN

EPM1 is the most common form of Progressive Myoclonus Epilepsy characterized by late-childhood onset, ever-worsening and disabling myoclonus, seizures, ataxia, psychiatric disease, and shortened lifespan. EPM1 is caused by expansions of a dodecamer repeat sequence in the promoter of CSTB (cystatin B), which dramatically reduces, but does not eliminate, gene expression. The relatively late onset and consistent presence of a minimal amount of protein product makes EPM1 a favorable target for gene replacement therapy. If treated early, these children's normally developed brains could be rescued from the neurodegeneration that otherwise follows, and their cross-reactive immunological material (CRIM) positive status greatly reduces transgene related toxicity. We performed a proof-of-concept CSTB gene replacement study in Cstb knockout mice by introducing full-length human CSTB driven by the CBh promoter packaged in AAV9 and administered at postnatal days 21 and 60. Mice were sacrificed at 2 or 9 months of age, respectively. We observed significant improvements in expression levels of neuroinflammatory pathway genes and cerebellar granule cell layer apoptosis, as well as amelioration of motor impairment. The data suggest that gene replacement is a promising therapeutic modality for EPM1 and could spare affected children and families the ravages of this otherwise severe neurodegenerative disease.


Asunto(s)
Cistatina B , Terapia Genética , Ratones Noqueados , Enfermedades Neuroinflamatorias , Animales , Ratones , Terapia Genética/métodos , Cistatina B/genética , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/genética , Humanos , Ataxia/genética , Ataxia/terapia , Epilepsias Mioclónicas Progresivas/genética , Epilepsias Mioclónicas Progresivas/terapia , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación
2.
Acta Neuropathol ; 147(1): 46, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-38411740

RESUMEN

At least five enzymes including three E3 ubiquitin ligases are dedicated to glycogen's spherical structure. Absence of any reverts glycogen to a structure resembling amylopectin of the plant kingdom. This amylopectinosis (polyglucosan body formation) causes fatal neurological diseases including adult polyglucosan body disease (APBD) due to glycogen branching enzyme deficiency, Lafora disease (LD) due to deficiencies of the laforin glycogen phosphatase or the malin E3 ubiquitin ligase and type 1 polyglucosan body myopathy (PGBM1) due to RBCK1 E3 ubiquitin ligase deficiency. Little is known about these enzymes' functions in glycogen structuring. Toward understanding these functions, we undertake a comparative murine study of the amylopectinoses of APBD, LD and PGBM1. We discover that in skeletal muscle, polyglucosan bodies form as two main types, small and multitudinous ('pebbles') or giant and single ('boulders'), and that this is primarily determined by the myofiber types in which they form, 'pebbles' in glycolytic and 'boulders' in oxidative fibers. This pattern recapitulates what is known in the brain in LD, innumerable dust-like in astrocytes and single giant sized in neurons. We also show that oxidative myofibers are relatively protected against amylopectinosis, in part through highly increased glycogen branching enzyme expression. We present evidence of polyglucosan body size-dependent cell necrosis. We show that sex influences amylopectinosis in genotype, brain region and myofiber-type-specific fashion. RBCK1 is a component of the linear ubiquitin chain assembly complex (LUBAC), the only known cellular machinery for head-to-tail linear ubiquitination critical to numerous cellular pathways. We show that the amylopectinosis of RBCK1 deficiency is not due to loss of linear ubiquitination, and that another function of RBCK1 or LUBAC must exist and operate in the shaping of glycogen. This work opens multiple new avenues toward understanding the structural determinants of the mammalian carbohydrate reservoir critical to neurologic and neuromuscular function and disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo IV , Enfermedad del Almacenamiento de Glucógeno , Enfermedades del Sistema Nervioso , Animales , Ratones , Glucógeno , Ubiquitina-Proteína Ligasas , Ubiquitinas , Mamíferos
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34518374

RESUMEN

Most endangered species exist today in small populations, many of which are isolated. Evolution in such populations is largely governed by genetic drift. Empirical evidence for drift affecting striking phenotypes based on substantial genetic data are rare. Approximately 37% of tigers (Panthera tigris) in the Similipal Tiger Reserve (in eastern India) are pseudomelanistic, characterized by wide, merged stripes. Camera trap data across the tiger range revealed the presence of pseudomelanistic tigers only in Similipal. We investigated the genetic basis for pseudomelanism and examined the role of drift in driving this phenotype's frequency. Whole-genome data and pedigree-based association analyses from captive tigers revealed that pseudomelanism cosegregates with a conserved and functionally important coding alteration in Transmembrane Aminopeptidase Q (Taqpep), a gene responsible for similar traits in other felid species. Noninvasive sampling of tigers revealed a high frequency of the Taqpep p.H454Y mutation in Similipal (12 individuals, allele frequency = 0.58) and absence from all other tiger populations (395 individuals). Population genetic analyses confirmed few (minimal number) tigers in Similipal, and its genetic isolation, with poor geneflow. Pairwise FST (0.33) at the mutation site was high but not an outlier. Similipal tigers had low diversity at 81 single nucleotide polymorphisms (mean heterozygosity = 0.28, SD = 0.27). Simulations were consistent with founding events and drift as possible drivers for the observed stark difference of allele frequency. Our results highlight the role of stochastic processes in the evolution of rare phenotypes. We highlight an unusual evolutionary trajectory in a small and isolated population of an endangered species.


Asunto(s)
Evolución Biológica , Flujo Genético , Variación Genética , Genética de Población , Melanosis/genética , Fenotipo , Tigres/fisiología , Secuencia de Aminoácidos , Animales , Conservación de los Recursos Naturales , Especies en Peligro de Extinción , Genoma , Genotipo , India , Repeticiones de Microsatélite , Homología de Secuencia , Tigres/genética
4.
PLoS Genet ; 15(12): e1008468, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31877123

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease in which the dystrophin coding for a membrane stabilizing protein is mutated. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice. Recent DMD transcriptomics revealed the defects were correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with constitutive (CAGCreERTM:Flt1LoxP/LoxP) and endothelial cell-specific conditional gene knockout mice (Cdh5CreERT2:Flt1LoxP/LoxP) for Flt1 (VEGFR1) which is a decoy receptor for VEGF. Here, we showed that while constitutive deletion of Flt1 is detrimental to the skeletal muscle function, endothelial cell-specific Flt1 deletion resulted in increased vascular density, increased satellite cell number and improvement in the DMD-associated phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Distrofia Muscular de Duchenne/tratamiento farmacológico , Péptidos/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Especificidad de Órganos , Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
5.
Circulation ; 137(23): 2478-2493, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29386201

RESUMEN

BACKGROUND: Valvular heart disease is common and affects the mitral valve (MV) most frequently. Despite the prevalence of MV disease (MVD), the cellular and molecular pathways that initiate and perpetuate it are not well understood. METHODS: K/B.g7 T-cell receptor transgenic mice spontaneously develop systemic autoantibody-associated autoimmunity, leading to fully penetrant fibroinflammatory MVD and arthritis. We used multiparameter flow cytometry, intracellular cytokine staining, and immunofluorescent staining to characterize the cells in inflamed K/B.g7 MVs. We used genetic approaches to study the contribution of mononuclear phagocytes (MNPs) to MVD in this model. Specifically, we generated K/B.g7 mice in which either CX3CR1 or CD301b/macrophage galactose N-acetylgalactosamine-specific lectin 2 (MGL2)-expressing MNPs were ablated. Using K/B.g7 mice expressing Cx3Cr1-Cre, we conditionally deleted critical inflammatory molecules from MNPs, including the Fc-receptor signal-transducing tyrosine kinase Syk and the cell adhesion molecule very late antigen-4. We performed complementary studies using monoclonal antibodies to block key inflammatory molecules. We generated bone marrow chimeric mice to define the origin of the inflammatory cells present in the MV and to determine which valve cells respond to the proinflammatory cytokine tumor necrosis factor (TNF). Finally, we examined specimens from patients with rheumatic heart disease to correlate our findings to human pathology. RESULTS: MNPs comprised the vast majority of MV-infiltrating cells; these MNPs expressed CX3CR1 and CD301b/MGL2. Analogous cells were present in human rheumatic heart disease valves. K/B.g7 mice lacking CX3CR1 or in which CD301b/MGL2-expressing MNPs were ablated were protected from MVD. The valve-infiltrating CD301b/MGL2+ MNPs expressed tissue-reparative molecules including arginase-1 and resistin-like molecule α. These MNPs also expressed the proinflammatory cytokines TNF and interleukin-6, and antibody blockade of these cytokines prevented MVD. Deleting Syk from CX3CR1-expressing MNPs reduced their TNF and interleukin-6 production and also prevented MVD. TNF acted through TNF receptor-1 expressed on valve-resident cells to increase the expression of vascular cell adhesion molecule-1. Conditionally deleting the vascular cell adhesion molecule-1 ligand very late antigen-4 from CX3CR1-expressing MNPs prevented MVD. CONCLUSIONS: CD301b/MGL2+ MNPs are key drivers of autoimmune MVD in K/B.g7 mice and are also present in human rheumatic heart disease. We define key inflammatory molecules that drive MVD in this model, including Syk, TNF, interleukin-6, very late antigen-4, and vascular cell adhesion molecule-1.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades de las Válvulas Cardíacas/inmunología , Lectinas Tipo C/inmunología , Fagocitos/inmunología , Células Alogénicas , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Trasplante de Médula Ósea , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Fibrosis , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Inflamación , Interleucina-6/genética , Interleucina-6/inmunología , Lectinas Tipo C/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Fagocitos/patología , Cardiopatía Reumática/patología , Quimera por Trasplante/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
6.
Muscle Nerve ; 59(5): 594-602, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30698289

RESUMEN

INTRODUCTION: The vasculature and blood flow in muscle are perturbed in Duchenne muscular dystrophy (DMD) and its mdx mouse model. MicroRNA-92a (miR-92a) is enriched in endothelial cells, especially during ischemic injury. METHODS: Because antagonizing miR-92a was shown to result in increased proliferation and migration of endothelial cells and recovery from ischemia, we assessed the effects of Antagomir-92a in vitro in muscle stem cell culture and in vivo in mdx mice. RESULTS: miR-92a was found to be highly expressed in muscle endothelial cells and satellite cells. Treatment with Antagomir-92a increased capillary density and tissue perfusion, which was accompanied by an increase in satellite cells. However, Antagomir-92a-treated mdx mice showed no histological improvement and had worse muscle function. Antagomir-92a suppressed myogenic differentiation in satellite cell culture. DISCUSSION: AntagomiR-92a improves the vasculature but not the muscle in mdx mice, possibly due to its side effects on satellite cell differentiation. Muscle Nerve 59:594-594, 2019.


Asunto(s)
Antagomirs/farmacología , MicroARNs/antagonistas & inhibidores , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular de Duchenne , Células Satélite del Músculo Esquelético/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Fuerza de la Mano , Flujometría por Láser-Doppler , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Satélite del Músculo Esquelético/citología
7.
Dev Cell ; 59(11): 1457-1474.e5, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38569550

RESUMEN

The function of many organs, including skeletal muscle, depends on their three-dimensional structure. Muscle regeneration therefore requires not only reestablishment of myofibers but also restoration of tissue architecture. Resident muscle stem cells (SCs) are essential for regeneration, but how SCs regenerate muscle architecture is largely unknown. We address this problem using genetic labeling of mouse SCs and whole-mount imaging to reconstruct, in three dimensions, muscle regeneration. Unexpectedly, we found that myofibers form via two distinct phases of fusion and the residual basement membrane of necrotic myofibers is critical for promoting fusion and orienting regenerated myofibers. Furthermore, the centralized myonuclei characteristic of regenerated myofibers are associated with myofibrillogenesis and endure months post injury. Finally, we elucidate two cellular mechanisms for the formation of branched myofibers, a pathology characteristic of diseased muscle. We provide a synthesis of the cellular events of regeneration and show that these differ from those used during development.


Asunto(s)
Imagenología Tridimensional , Músculo Esquelético , Regeneración , Animales , Regeneración/fisiología , Ratones , Músculo Esquelético/fisiología , Imagenología Tridimensional/métodos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/citología , Desarrollo de Músculos/fisiología , Células Madre/citología , Células Madre/metabolismo , Membrana Basal/metabolismo
8.
Elife ; 132024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38842166

RESUMEN

Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.


Asunto(s)
Supervivencia Celular , Células Endoteliales , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Animales , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Ratones Endogámicos C57BL , Masculino
9.
J Physiol ; 591(15): 3765-76, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23753524

RESUMEN

A primary feature of skeletal muscle lacking the protein dystrophin, as occurring in Duchenne muscular dystrophy, is a hypersensitivity to contraction-induced strength loss. We tested the hypothesis that the extensive strength loss results from an impairment in the electrophysiological function of the plasmalemma specifically impaired action potential development. Anterior crural muscles from mdx and wildtype mice performed a single bout of 100 electrically stimulated eccentric contractions in vivo. Electromyography, specifically the M-wave, was analysed during muscle contraction to assess the ability of the tibialis anterior muscle plasmalemma to generate and conduct action potentials. During eccentric contractions, wildtype mice exhibited a 36% loss in torque about the ankle but mdx mice exhibited a greater torque loss of 73% (P < 0.001). Despite the loss of torque, there was no reduction in M-wave root mean square (RMS) for wildtype mice, which was in stark contrast to mdx mice that had a 55% reduction in M-wave RMS (P < 0.001). This impairment resolved within 24 h and coincided with a significant improvement in strength and membrane integrity. Intracellular measurements of resting membrane potential (RMP) in uninjured and injured extensor digitorum longus muscles were made to determine if a chronic depolarization had occurred, which could lead to impaired fibre excitability and/or altered action potential conduction properties. The distributions of RMP were not different between wildtype uninjured and injured muscle cells (median: -73.2 mV vs. -72.7 mV, P = 0.46) whereas there was a significant difference between mdx uninjured and injured cells (median: -71.5 mV vs. -56.6 mV, P < 0.001). These data show that mdx muscle fibres are depolarized after an injurious bout of eccentric contractions. These findings (i) suggest a major plasmalemma-based mechanism of strength loss underlying contraction-induced injury in Duchenne muscular dystrophy distinctly different from that for healthy muscle, and (ii) demonstrate dystrophin is critical for maintaining action potential generation and conduction after eccentric contractions.


Asunto(s)
Potenciales de Acción/fisiología , Contracción Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Animales , Modelos Animales de Enfermedad , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/lesiones
10.
Methods Mol Biol ; 2640: 453-462, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995613

RESUMEN

Skeletal muscle is a highly ordered tissue composed of a complex network of a diverse variety of cells. The dynamic spatial and temporal interaction between these cells during homeostasis and during times of injury gives the skeletal muscle its regenerative capacity. To properly understand the process of regeneration, a three-dimensional (3-D) imaging process must be conducted. With the advancement of imaging and computing technology, it has become powerful to analyze spatial data from confocal microscope images. In order to prepare whole tissue skeletal muscle samples for confocal imaging, the muscle must be subjected to tissue clearing. With the use of an ideal optical clearing protocol - one that minimizes light scattering via refractive index mismatching - a more accurate 3-D image of the muscle can be produced as it does not involve the physical sectioning of the muscle. While there have been several protocols relating to the study of 3-D biology in whole tissue, these protocols have primarily been focused on the nervous system. In this chapter, we present a new method for skeletal muscle tissue clearing. In addition, this protocol aims to outline the specific parameters required for taking 3-D images of immunofluorescence-stained skeletal muscle samples using a confocal microscope.


Asunto(s)
Imagenología Tridimensional , Músculo Esquelético , Microscopía Confocal , Técnica del Anticuerpo Fluorescente , Imagenología Tridimensional/métodos
11.
Methods Mol Biol ; 2640: 463-477, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995614

RESUMEN

Skeletal muscle is a highly ordered tissue composed of a complex network of a diverse variety of cells. The dynamic spatial and temporal interaction between these cells during homeostasis and during times of injury gives the skeletal muscle its regenerative capacity. In order to properly understand the process of regeneration, a three-dimensional (3-D) imaging process must be conducted. While there have been several protocols studying 3-D imaging, it has primarily been focused on the nervous system. This protocol aims to outline the workflow for rendering a 3-D image of the skeletal muscle using spatial data from confocal microscope images. This protocol uses the ImageJ, Ilastik, and Imaris software for 3-D rendering and computational image analysis as both are relatively easy to use and have powerful segmentation capabilities.


Asunto(s)
Imagenología Tridimensional , Células Satélite del Músculo Esquelético , Células Satélite del Músculo Esquelético/fisiología , Músculo Esquelético/fisiología , Procesamiento de Imagen Asistido por Computador , Desarrollo de Músculos/fisiología , Diferenciación Celular
12.
Hum Mol Genet ; 19(21): 4145-59, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20705734

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease caused by mutations in the gene coding for the protein dystrophin. Recent work demonstrates that dystrophin is also found in the vasculature and its absence results in vascular deficiency and abnormal blood flow. This induces a state of ischemia further aggravating the muscular dystrophy pathogenesis. For an effective form of therapy of DMD, both the muscle and the vasculature need to be addressed. To reveal the developmental relationship between muscular dystrophy and vasculature, mdx mice, an animal model for DMD, were crossed with Flt-1 gene knockout mice to create a model with increased vasculature. Flt-1 is a decoy receptor for vascular endothelial growth factor, and therefore both homozygous (Flt-1(-/-)) and heterozygous (Flt-1(+/-)) Flt-1 gene knockout mice display increased endothelial cell proliferation and vascular density during embryogenesis. Here, we show that Flt-1(+/-) and mdx:Flt-1(+/-) adult mice also display a developmentally increased vascular density in skeletal muscle compared with the wild-type and mdx mice, respectively. The mdx:Flt-1(+/-) mice show improved muscle histology compared with the mdx mice with decreased fibrosis, calcification and membrane permeability. Functionally, the mdx:Flt-1(+/-) mice have an increase in muscle blood flow and force production, compared with the mdx mice. Consequently, the mdx:utrophin(-/-):Flt-1(+/-) mice display improved muscle histology and significantly higher survival rates compared with the mdx:utrophin(-/-) mice, which show more severe muscle phenotypes than the mdx mice. These data suggest that increasing the vasculature in DMD may ameliorate the histological and functional phenotypes associated with this disease.


Asunto(s)
Haploinsuficiencia , Distrofia Muscular Animal/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Proliferación Celular , Heterocigoto , Homocigoto , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Contracción Muscular , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Fenotipo
13.
Stem Cells ; 29(3): 505-16, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21425413

RESUMEN

Expression of the four transcription factors, that is, Oct4, Sox2, cMyc, and Klf4 has been shown to generate induced pluripotent stem cells (iPSCs) from many types of specialized differentiated somatic cells. It remains unclear, however, whether fully committed skeletal muscle progenitor cells (myoblasts) have the potency to undergo reprogramming to develop iPSCs in line with previously reported cases. To test this, we have isolated genetically marked myoblasts derived from satellite cell of adult mouse muscles using the Cre-loxP system (Pax7-CreER:R26R and Myf5-Cre:R26R). On infection with retroviral vectors expressing the four factors, these myoblasts gave rise to myogenic lineage tracer lacZ-positive embryonic stem cell (ESC)-like colonies. These cells expressed ESC-specific genes and were competent to differentiate into all three germ layers and germ cells, indicating the successful generation of myoblast-derived iPSCs. Continuous expression of the MyoD gene, a master transcription factor for skeletal muscle specification, inhibited this reprogramming process in myoblasts. In contrast, reprogramming myoblasts isolated from mice lacking the MyoD gene led to an increase in reprogramming efficiency. Our data also indicated that Oct4 acts as a transcriptional suppressor of MyoD gene expression through its interaction with the upstream enhancer region. Taken together, these results indicate that suppression of MyoD gene expression by Oct4 is required for the initial reprogramming step in the development of iPSCs from myoblasts. This data suggests that the skeletal muscle system provides a well-defined differentiation model to further elaborate on the effects of iPSC reprogramming in somatic cells.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/fisiología , Proteína MioD/genética , Mioblastos/fisiología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Animales , Células Cultivadas , Regulación hacia Abajo/genética , Femenino , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos ICR , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteína MioD/metabolismo , Proteína MioD/fisiología , Mioblastos/citología , Mioblastos/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Embarazo
14.
Int J STEM Educ ; 9(1): 49, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35915654

RESUMEN

Background: The University of California system has a novel tenure-track education-focused faculty position called Lecturer with Security of Employment (working titles: Teaching Professor or Professor of Teaching). We focus on the potential difference in implementation of active-learning strategies by faculty type, including tenure-track education-focused faculty, tenure-track research-focused faculty, and non-tenure-track lecturers. In addition, we consider other instructor characteristics (faculty rank, years of teaching, and gender) and classroom characteristics (campus, discipline, and class size). We use a robust clustering algorithm to determine the number of clusters, identify instructors using active learning, and to understand the instructor and classroom characteristics in relation to the adoption of active-learning strategies. Results: We observed 125 science, technology, engineering, and mathematics (STEM) undergraduate courses at three University of California campuses using the Classroom Observation Protocol for Undergraduate STEM to examine active-learning strategies implemented in the classroom. Tenure-track education-focused faculty are more likely to teach with active-learning strategies compared to tenure-track research-focused faculty. Instructor and classroom characteristics that are also related to active learning include campus, discipline, and class size. The campus with initiatives and programs to support undergraduate STEM education is more likely to have instructors who adopt active-learning strategies. There is no difference in instructors in the Biological Sciences, Engineering, or Information and Computer Sciences disciplines who teach actively. However, instructors in the Physical Sciences are less likely to teach actively. Smaller class sizes also tend to have instructors who teach more actively. Conclusions: The novel tenure-track education-focused faculty position within the University of California system represents a formal structure that results in higher adoption of active-learning strategies in undergraduate STEM education. Campus context and evolving expectations of the position (faculty rank) contribute to the symbols related to learning and teaching that correlate with differential implementation of active learning. Supplementary Information: The online version contains supplementary material available at 10.1186/s40594-022-00365-9.

15.
CBE Life Sci Educ ; 20(1): ar3, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33444101

RESUMEN

The Classroom Observation Protocol for Undergraduate STEM (COPUS) provides descriptive feedback to instructors by capturing student and instructor behaviors occurring in the classroom. Due to the increasing prevalence of COPUS data collection, it is important to recognize how researchers determine whether groups of courses or instructors have unique classroom characteristics. One approach uses cluster analysis, highlighted by a recently developed tool, the COPUS Analyzer, that enables the characterization of COPUS data into one of seven clusters representing three groups of instructional styles (didactic, interactive, and student centered). Here, we examine a novel 250 course data set and present evidence that a predictive cluster analysis tool may not be appropriate for analyzing COPUS data. We perform a de novo cluster analysis and compare results with the COPUS Analyzer output and identify several contrasting outcomes regarding course characterizations. Additionally, we present two ensemble clustering algorithms: 1) k-means and 2) partitioning around medoids. Both ensemble algorithms categorize our classroom observation data into one of two clusters: traditional lecture or active learning. Finally, we discuss implications of these findings for education research studies that leverage COPUS data.


Asunto(s)
Aprendizaje Basado en Problemas , Estudiantes , Análisis por Conglomerados , Recolección de Datos , Humanos
16.
Mol Ther Methods Clin Dev ; 21: 369-381, 2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-33898634

RESUMEN

Duchenne muscular dystrophy is characterized by structural degeneration of muscle, which is exacerbated by localized functional ischemia due to loss of nitric oxide synthase-induced vasodilation. Treatment strategies aimed at increasing vascular perfusion have been proposed. Toward this end, we have developed monoclonal antibodies (mAbs) that bind to the vascular endothelial growth factor (VEGF) receptor VEGFR-1 (Flt-1) and its soluble splice variant isoform (sFlt-1) leading to increased levels of free VEGF and proangiogenic signaling. The lead chimeric mAb, 21B3, had high affinity and specificity for both human and mouse sFlt-1 and inhibited VEGF binding to sFlt-1 in a competitive manner. Proof-of-concept studies in the mdx mouse model of Duchenne muscular dystrophy showed that intravenous administration of 21B3 led to elevated VEGF levels, increased vascularization and blood flow to muscles, and decreased fibrosis after 6-12 weeks of treatment. Greater muscle strength was also observed after 4 weeks of treatment. A humanized form of the mAb, 27H6, was engineered and demonstrated a comparable pharmacologic effect. Overall, administration of anti-Flt-1 mAbs in mdx mice inhibited the VEGF:Flt-1 interaction, promoted angiogenesis, and improved muscle function. These studies suggest a potential therapeutic benefit of Flt-1 inhibition for patients with Duchenne muscular dystrophy.

17.
Biochem Biophys Res Commun ; 391(1): 299-304, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19914205

RESUMEN

Nucleostemin (NS) is a nucleolar protein abundantly expressed in a variety of proliferating cells and undifferentiated cells. Its known functions include cell cycle regulation and the control of pre-rRNA processing. It also has been proposed that NS has an additional role in undifferentiated cells due to its downregulation during stem cell differentiation and its upregulation during tissue regeneration. Here, however, we demonstrate that skeletal muscle cell differentiation has a unique expression profile of NS in that it is continuously expressed during differentiation. NS was expressed at similar levels in non-proliferating muscle stem cells (satellite cells), rapidly proliferating precursor cells (myoblasts) and post-mitotic terminally differentiated cells (myotubes and myofibers). The sustained expression of NS during terminal differentiation is necessary to support increased protein synthesis during this process. Downregulation of NS inhibited differentiation of myoblasts to myotubes, accompanied by striking downregulation of key myogenic transcription factors, such as myogenin and MyoD. In contrast, upregulation of NS inhibited proliferation and promoted muscle differentiation in a p53-dependent manner. Our findings provide evidence that NS has an unexpected role in post-mitotic terminal differentiation. Importantly, these findings also indicate that, contrary to suggestions in the literature, the expression of NS cannot always be used as a reliable indicator for undifferentiated cells or proliferating cells.


Asunto(s)
Proteínas Portadoras/fisiología , Diferenciación Celular/genética , Desarrollo de Músculos/genética , Músculo Esquelético/citología , Proteínas Nucleares/fisiología , Células Satélite del Músculo Esquelético/citología , Animales , Proteínas Portadoras/genética , Proteínas de Unión al GTP , Técnicas de Silenciamiento del Gen , Ratones , Ratones Mutantes , Mitosis , Proteínas Nucleares/genética , Proteínas de Unión al ARN , Células Satélite del Músculo Esquelético/metabolismo , Proteína p53 Supresora de Tumor/genética
18.
J Clin Invest ; 130(5): 2465-2477, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32250341

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is caused by loss of repression of the DUX4 gene; however, the DUX4 protein is rare and difficult to detect in human muscle biopsies, and pathological mechanisms are obscure. FSHD is also a chronic disease that progresses slowly over decades. We used the sporadic, low-level, muscle-specific expression of DUX4 enabled by the iDUX4pA-HSA mouse to develop a chronic long-term muscle disease model. After 6 months of extremely low sporadic DUX4 expression, dystrophic muscle presented hallmarks of FSHD histopathology, including muscle degeneration, capillary loss, fibrosis, and atrophy. We investigated the transcriptional profile of whole muscle as well as endothelial cells and fibroadiopogenic progenitors (FAPs). Strikingly, differential gene expression profiles of both whole muscle and, to a lesser extent, FAPs, showed significant overlap with transcriptional profiles of MRI-guided human FSHD muscle biopsies. These results demonstrate a pathophysiological similarity between disease in muscles of iDUX4pA-HSA mice and humans with FSHD, solidifying the value of chronic rare DUX4 expression in mice for modeling pathological mechanisms in FSHD and highlighting the importance FAPs in this disease.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapulohumeral/metabolismo , Transcripción Genética , Animales , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/patología , Femenino , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Transgénicos , Músculo Esquelético/patología , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/patología
19.
J Appl Physiol (1985) ; 126(2): 363-375, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30571283

RESUMEN

Mutation to the dystrophin gene causes skeletal muscle weakness in patients with Duchenne muscular dystrophy (DMD) or Becker muscular dystrophy (BMD). Deliberation continues regarding implications of prescribing exercise for these patients. The purpose of this study was to determine whether isometric resistance exercise (~10 tetanic contractions/session) improves skeletal muscle strength and histopathology in the mdx mouse model of DMD. Three isometric training sessions increased in vivo isometric torque (22%) and contractility rates (54%) of anterior crural muscles of mdx mice. Mice expressing a BMD-causing missense mutated dystrophin on the mdx background showed comparable increases in torque (22%), while wild-type mice showed less change (11%). Increases in muscle function occurred within 1 h and peaked 3 days posttraining; however, the adaptation was lost after 7 days unless retrained. Six isometric training sessions over 4 wk caused increased isometric torque (28%) and contractility rates (22-28%), reduced fibrosis, as well as greater uniformity of fiber cross-sectional areas, fewer embryonic myosin heavy-chain-positive fibers, and more satellite cells in tibialis anterior muscle compared with the contralateral untrained muscle. Ex vivo functional analysis of isolated extensor digitorum longus (EDL) muscle from the trained hindlimb revealed greater absolute isometric force, lower passive stiffness, and a lower susceptibility to eccentric contraction-induced force loss compared with untrained EDL muscle. Overall, these data support the concept that exercise training in the form of isometric tetanic contractions can improve contractile function of dystrophin-deficient muscle, indicating a potential role for enhancing muscle strength in patients with DMD and BMD. NEW & NOTEWORTHY We focused on adaptive responses of dystrophin-deficient mouse skeletal muscle to isometric contraction training and report that in the absence of dystrophin (or in the presence of a mutated dystrophin), strength and muscle histopathology are improved. Results suggest that the strength gains are associated with fiber hypertrophy, reduced fibrosis, increased number of satellite cells, and blunted eccentric contraction-induced force loss in vitro. Importantly, there was no indication that the isometric exercise training was deleterious to dystrophin-deficient muscle.


Asunto(s)
Distrofina/deficiencia , Contracción Isométrica , Fuerza Muscular , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/terapia , Entrenamiento de Fuerza , Adaptación Fisiológica , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Fibrosis , Masculino , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Mutación Missense , Recuperación de la Función , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Factores de Tiempo
20.
Cell Stem Cell ; 23(4): 530-543.e9, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290177

RESUMEN

Skeletal muscle is a complex tissue containing tissue resident muscle stem cells (satellite cells) (MuSCs) important for postnatal muscle growth and regeneration. Quantitative analysis of the biological function of MuSCs and the molecular pathways responsible for a potential juxtavascular niche for MuSCs is currently lacking. We utilized fluorescent reporter mice and muscle tissue clearing to investigate the proximity of MuSCs to capillaries in 3 dimensions. We show that MuSCs express abundant VEGFA, which recruits endothelial cells (ECs) in vitro, whereas blocking VEGFA using both a vascular endothelial growth factor (VEGF) inhibitor and MuSC-specific VEGFA gene deletion reduces the proximity of MuSCs to capillaries. Importantly, this proximity to the blood vessels was associated with MuSC self-renewal in which the EC-derived Notch ligand Dll4 induces quiescence in MuSCs. We hypothesize that MuSCs recruit capillary ECs via VEGFA, and in return, ECs maintain MuSC quiescence though Dll4.


Asunto(s)
Receptores Notch/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Ratones , Células Satélite del Músculo Esquelético/citología , Nicho de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA