Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Trends Genet ; 39(11): 858-872, 2023 11.
Article in English | MEDLINE | ID: mdl-37481442

ABSTRACT

Transcription elongation requires elaborate coordination between the transcriptional machinery and chromatin regulatory factors to successfully produce RNA while preserving the epigenetic landscape. Recent structural and genomic studies have highlighted that suppressor of Ty 6 (Spt6), a conserved histone chaperone and transcription elongation factor, sits at the crux of the transcription elongation process. Other recent studies have revealed that Spt6 also promotes DNA replication and genome integrity. Here, we review recent studies of Spt6 that have provided new insights into the mechanisms by which Spt6 controls transcription and have revealed the breadth of Spt6 functions in eukaryotic cells.


Subject(s)
Histones , Humans , DNA Replication/genetics , Genomic Instability/genetics , Histone Chaperones/genetics , Histone Chaperones/chemistry , Histone Chaperones/metabolism , Histones/genetics , Histones/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/chemistry , Transcriptional Elongation Factors/metabolism , Animals
2.
J Clin Invest ; 129(11): 4657-4670, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31545299

ABSTRACT

Membrane repair is essential to cell survival. In skeletal muscle, injury often associates with plasma membrane disruption. Additionally, muscular dystrophy is linked to mutations in genes that produce fragile membranes or reduce membrane repair. Methods to enhance repair and reduce susceptibility to injury could benefit muscle in both acute and chronic injury settings. Annexins are a family of membrane-associated Ca2+-binding proteins implicated in repair, and annexin A6 was previously identified as a genetic modifier of muscle injury and disease. Annexin A6 forms the repair cap over the site of membrane disruption. To elucidate how annexins facilitate repair, we visualized annexin cap formation during injury. We found that annexin cap size positively correlated with increasing Ca2+ concentrations. We also found that annexin overexpression promoted external blebs enriched in Ca2+ and correlated with a reduction of intracellular Ca2+ at the injury site. Annexin A6 overexpression reduced membrane injury, consistent with enhanced repair. Treatment with recombinant annexin A6 protected against acute muscle injury in vitro and in vivo. Moreover, administration of recombinant annexin A6 in a model of muscular dystrophy reduced serum creatinine kinase, a biomarker of disease. These data identify annexins as mediators of membrane-associated Ca2+ release during membrane repair and annexin A6 as a therapeutic target to enhance membrane repair capacity.


Subject(s)
Annexin A6/pharmacology , Calcium/metabolism , Cell Membrane/metabolism , Muscle, Skeletal/injuries , Muscular Dystrophy, Animal/prevention & control , Animals , Annexin A6/genetics , Cell Membrane/pathology , Female , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
3.
JACC Basic Transl Sci ; 4(2): 251-268, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31061927

ABSTRACT

The adult myocardium relies on oxidative metabolism. In ischemic myocardium, such as the embryonic heart, glycolysis contributes more prominently as a fuel source. The sulfonylurea receptor 2 (SUR2) was previously implicated in the normal myocardial transition from glycolytic to oxidative metabolism that occurs during adaptation to postnatal life. This receptor was now selectively deleted in adult mouse myocardium resulting in protection from ischemia reperfusion injury. SUR2-deleted cardiomyocytes had enhanced glucose uptake, and SUR2 forms a complex with the major glucose transporter. These data identify the SUR2 receptor as a target to shift cardiac metabolism to protect against myocardial injury.

4.
PLoS Genet ; 13(10): e1007070, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29065150

ABSTRACT

Genetic disruption of the dystrophin complex produces muscular dystrophy characterized by a fragile muscle plasma membrane leading to excessive muscle degeneration. Two genetic modifiers of Duchenne Muscular Dystrophy implicate the transforming growth factor ß (TGFß) pathway, osteopontin encoded by the SPP1 gene and latent TGFß binding protein 4 (LTBP4). We now evaluated the functional effect of these modifiers in the context of muscle injury and repair to elucidate their mechanisms of action. We found that excess osteopontin exacerbated sarcolemmal injury, and correspondingly, that loss of osteopontin reduced injury extent both in isolated myofibers and in muscle in vivo. We found that ablation of osteopontin was associated with reduced expression of TGFß and TGFß-associated pathways. We identified that increased TGFß resulted in reduced expression of Anxa1 and Anxa6, genes encoding key components of the muscle sarcolemma resealing process. Genetic manipulation of Ltbp4 in dystrophic muscle also directly modulated sarcolemmal resealing, and Ltbp4 alleles acted in concert with Anxa6, a distinct modifier of muscular dystrophy. These data provide a model in which a feed forward loop of TGFß and osteopontin directly impacts the capacity of muscle to recover from injury, and identifies an intersection of genetic modifiers on muscular dystrophy.


Subject(s)
Genes, Modifier , Latent TGF-beta Binding Proteins/physiology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/genetics , Osteopontin/metabolism , Animals , Annexin A1/genetics , Annexin A1/metabolism , Annexin A6/genetics , Annexin A6/metabolism , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Muscle, Skeletal/injuries , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Osteopontin/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Recovery of Function , Sarcolemma/physiology
5.
Am J Pathol ; 187(11): 2520-2535, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28823869

ABSTRACT

The muscular dystrophies are genetically diverse. Shared pathological features among muscular dystrophies include breakdown, or loss of muscle, and accompanying fibrotic replacement. Novel strategies are needed to enhance muscle repair and function and to slow this pathological remodeling. Glucocorticoid steroids, like prednisone, are known to delay loss of ambulation in patients with Duchenne muscular dystrophy but are accompanied by prominent adverse effects. However, less is known about the effects of steroid administration in other types of muscular dystrophies, including limb-girdle muscular dystrophies (LGMDs). LGMD 2B is caused by loss of dysferlin, a membrane repair protein, and LGMD 2C is caused by loss of the dystrophin-associated protein, γ-sarcoglycan. Herein, we assessed the efficacy of steroid dosing on sarcolemmal repair, muscle function, histopathology, and the regenerative capacity of primary muscle cells. We found that in murine models of LGMD 2B and 2C, daily prednisone dosing reduced muscle damage and fibroinflammatory infiltration. However, daily prednisone dosing also correlated with increased muscle adipogenesis and atrophic remodeling. Conversely, intermittent dosing of prednisone, provided once weekly, enhanced muscle repair and did not induce atrophy or adipogenesis, and was associated with improved muscle function. These data indicate that dosing frequency of glucocorticoid steroids affects muscle remodeling in non-Duchenne muscular dystrophies, suggesting a positive outcome associated with intermittent steroid dosing in LGMD 2B and 2C muscle.


Subject(s)
Glucocorticoids/pharmacology , Muscle, Skeletal/drug effects , Muscular Dystrophies, Limb-Girdle/drug therapy , Animals , Dystrophin/drug effects , Dystrophin/metabolism , Glucocorticoids/administration & dosage , Membrane Proteins/metabolism , Mice , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Prednisone/administration & dosage , Prednisone/pharmacology , Sarcoglycans/drug effects , Sarcoglycans/metabolism
6.
J Clin Invest ; 127(6): 2418-2432, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28481224

ABSTRACT

Glucocorticoid steroids such as prednisone are prescribed for chronic muscle conditions such as Duchenne muscular dystrophy, where their use is associated with prolonged ambulation. The positive effects of chronic steroid treatment in muscular dystrophy are paradoxical because these steroids are also known to trigger muscle atrophy. Chronic steroid use usually involves once-daily dosing, although weekly dosing in children has been suggested for its reduced side effects on behavior. In this work, we tested steroid dosing in mice and found that a single pulse of glucocorticoid steroids improved sarcolemmal repair through increased expression of annexins A1 and A6, which mediate myofiber repair. This increased expression was dependent on glucocorticoid response elements upstream of annexins and was reinforced by the expression of forkhead box O1 (FOXO1). We compared weekly versus daily steroid treatment in mouse models of acute muscle injury and in muscular dystrophy and determined that both regimens provided comparable benefits in terms of annexin gene expression and muscle repair. However, daily dosing activated atrophic pathways, including F-box protein 32 (Fbxo32), which encodes atrogin-1. Conversely, weekly steroid treatment in mdx mice improved muscle function and histopathology and concomitantly induced the ergogenic transcription factor Krüppel-like factor 15 (Klf15) while decreasing Fbxo32. These findings suggest that intermittent, rather than daily, glucocorticoid steroid regimen promotes sarcolemmal repair and muscle recovery from injury while limiting atrophic remodeling.


Subject(s)
Glucocorticoids/administration & dosage , Muscle, Skeletal/drug effects , Prednisone/administration & dosage , Animals , Annexin A6/genetics , Annexin A6/metabolism , Cells, Cultured , Drug Administration Schedule , Drug Evaluation, Preclinical , Gene Expression , Glucocorticoids/adverse effects , Male , Mice, 129 Strain , Mice, Inbred DBA , Mice, Inbred mdx , Muscle, Skeletal/physiopathology , Muscular Atrophy/chemically induced , Muscular Dystrophy, Duchenne/drug therapy , Prednisone/adverse effects , Protein Binding , Receptors, Glucocorticoid/metabolism , Regeneration , Sarcolemma/drug effects , Sarcolemma/physiology , Transcriptional Activation/drug effects
7.
Am J Pathol ; 186(6): 1610-22, 2016 06.
Article in English | MEDLINE | ID: mdl-27070822

ABSTRACT

Dysferlin is a membrane-associated protein implicated in membrane resealing; loss of dysferlin leads to muscular dystrophy. We examined the same loss-of-function Dysf mutation in two different mouse strains, 129T2/SvEmsJ (Dysf(129)) and C57BL/6J (Dysf(B6)). Although there are many genetic differences between these two strains, we focused on polymorphisms in Anxa6 because these variants were previously associated with modifying a pathologically distinct form of muscular dystrophy and increased the production of a truncated annexin A6 protein. Dysferlin deficiency in the C57BL/6J background was associated with increased Evan's Blue dye uptake into muscle and increased serum creatine kinase compared to the 129T2/SvEmsJ background. In the C57BL/6J background, dysferlin loss was associated with enhanced pathologic severity, characterized by decreased mean fiber cross-sectional area, increased internalized nuclei, and increased fibrosis, compared to that in Dysf(129) mice. Macrophage infiltrate was also increased in Dysf(B6) muscle. High-resolution imaging of live myofibers demonstrated that fibers from Dysf(B6) mice displayed reduced translocation of full-length annexin A6 to the site of laser-induced sarcolemmal disruption compared to Dysf(129) myofibers, and impaired translocation of annexin A6 associated with impaired resealing of the sarcolemma. These results provide one mechanism by which the C57BL/6J background intensifies dysferlinopathy, giving rise to a more severe form of muscular dystrophy in the Dysf(B6) mouse model through increased membrane leak and inflammation.


Subject(s)
Annexin A6/metabolism , Membrane Proteins/deficiency , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Animals , Annexin A6/genetics , Dysferlin , Immunoblotting , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Muscular Dystrophy, Animal/metabolism , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Protein Transport , Sarcolemma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL