Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Pers Med ; 8(2)2018 Apr 30.
Article in English | MEDLINE | ID: mdl-29710874

ABSTRACT

As part of the Heart Healthy Lenoir Project, we developed a practice level intervention to improve blood pressure control. The goal of this study was: (i) to determine if single nucleotide polymorphisms (SNPs) that associate with blood pressure variation, identified in large studies, are applicable to blood pressure control in subjects from a rural population; (ii) to measure the association of these SNPs with subjects' responsiveness to the hypertension intervention; and (iii) to identify other SNPs that may help understand patient-specific responses to an intervention. We used a combination of candidate SNPs and genome-wide analyses to test associations with either baseline systolic blood pressure (SBP) or change in systolic blood pressure one year after the intervention in two genetically defined ancestral groups: African Americans (AA) and Caucasian Americans (CAU). Of the 48 candidate SNPs, 13 SNPs associated with baseline SBP in our study; however, one candidate SNP, rs592582, also associated with a change in SBP after one year. Using our study data, we identified 4 and 15 additional loci that associated with a change in SBP in the AA and CAU groups, respectively. Our analysis of gene-age interactions identified genotypes associated with SBP improvement within different age groups of our populations. Moreover, our integrative analysis identified AQP4-AS1 and PADI2 as genes whose expression levels may contribute to the pleiotropy of complex traits involved in cardiovascular health and blood pressure regulation in response to an intervention targeting hypertension. In conclusion, the identification of SNPs associated with the success of a hypertension treatment intervention suggests that genetic factors in combination with age may contribute to an individual's success in lowering SBP. If these findings prove to be applicable to other populations, the use of this genetic variation in making patient-specific interventions may help providers with making decisions to improve patient outcomes. Further investigation is required to determine the role of this genetic variance with respect to the management of hypertension such that more precise treatment recommendations may be made in the future as part of personalized medicine.

2.
Skelet Muscle ; 5: 27, 2015.
Article in English | MEDLINE | ID: mdl-26301073

ABSTRACT

BACKGROUND: The plasma membranes of striated muscle cells are particularly susceptible to rupture as they endure significant mechanical stress and strain during muscle contraction, and studies have shown that defects in membrane repair can contribute to the progression of muscular dystrophy. The synaptotagmin-related protein, dysferlin, has been implicated in mediating rapid membrane repair through its ability to direct intracellular vesicles to sites of membrane injury. However, further work is required to identify the precise molecular mechanisms that govern dysferlin targeting and membrane repair. We previously showed that the bin-amphiphysin-Rvs (BAR)-pleckstrin homology (PH) domain containing Rho-GAP GTPase regulator associated with focal adhesion kinase-1 (GRAF1) was dynamically recruited to the tips of fusing myoblasts wherein it promoted membrane merging by facilitating ferlin-dependent capturing of intracellular vesicles. Because acute membrane repair responses involve similar vesicle trafficking complexes/events and because our prior studies in GRAF1-deficient tadpoles revealed a putative role for GRAF1 in maintaining muscle membrane integrity, we postulated that GRAF1 might also play an important role in facilitating dysferlin-dependent plasma membrane repair. METHODS: We used an in vitro laser-injury model to test whether GRAF1 was necessary for efficient muscle membrane repair. We also generated dystrophin/GRAF1 doubledeficient mice by breeding mdx mice with GRAF1 hypomorphic mice. Evans blue dye uptake and extensive morphometric analyses were used to assess sarcolemmal integrity and related pathologies in cardiac and skeletal muscles isolated from these mice. RESULTS: Herein, we show that GRAF1 is dynamically recruited to damaged skeletal and cardiac muscle plasma membranes and that GRAF1-depleted muscle cells have reduced membrane healing abilities. Moreover, we show that dystrophin depletion exacerbated muscle damage in GRAF1-deficient mice and that mice with dystrophin/GRAF1 double deficiency phenocopied the severe muscle pathologies observed in dystrophin/dysferlin-double null mice. Consistent with a model that GRAF1 facilitates dysferlin-dependent membrane patching, we found that GRAF1 associates with and regulates plasma membrane deposition of dysferlin. CONCLUSIONS: Overall, our work indicates that GRAF1 facilitates dysferlin-dependent membrane repair following acute muscle injury. These findings indicate that GRAF1 might play a role in the phenotypic variation and pathological progression of cardiac and skeletal muscle degeneration in muscular dystrophy patients.

3.
Dev Biol ; 393(2): 298-311, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25019370

ABSTRACT

Myoblast fusion (a critical process by which muscles grow) occurs in a multi-step fashion that requires actin and membrane remodeling; but important questions remain regarding the spatial/temporal regulation of and interrelationship between these processes. We recently reported that the Rho-GAP, GRAF1, was particularly abundant in muscles undergoing fusion to form multinucleated fibers and that enforced expression of GRAF1 in cultured myoblasts induced robust fusion by a process that required GAP-dependent actin remodeling and BAR domain-dependent membrane sculpting. Herein we developed a novel line of GRAF1-deficient mice to explore a role for this protein in the formation/maturation of myotubes in vivo. Post-natal muscles from GRAF1-depleted mice exhibited a significant and persistent reduction in cross-sectional area, impaired regenerative capacity and a significant decrease in force production indicative of lack of efficient myoblast fusion. A significant fusion defect was recapitulated in isolated myoblasts depleted of GRAF1 or its closely related family member GRAF2. Mechanistically, we show that GRAF1 and 2 facilitate myoblast fusion, at least in part, by promoting vesicle-mediated translocation of fusogenic ferlin proteins to the plasma membrane.


Subject(s)
GTPase-Activating Proteins/genetics , Membrane Proteins/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Myoblasts/metabolism , Animals , Biological Transport , Brefeldin A/pharmacology , Cell Fusion , Cell Membrane/metabolism , Cells, Cultured , Golgi Apparatus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Synthesis Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering , Vesicular Transport Proteins
4.
Nat Commun ; 4: 2910, 2013.
Article in English | MEDLINE | ID: mdl-24335996

ABSTRACT

Although hypertension is a worldwide health issue, an incomplete understanding of its aetiology has hindered our ability to treat this complex disease. Here we identify arhgap42 (also known as GRAF3) as a Rho-specific GAP expressed specifically in smooth muscle cells (SMCs) in mice and humans. We show that GRAF3-deficient mice exhibit significant hypertension and increased pressor responses to angiotensin II and endothelin-1; these effects are prevented by treatment with the Rho-kinase inhibitor, Y27632. RhoA activity and myosin light chain phosphorylation are elevated in GRAF3-depleted SMCs in vitro and in vivo, and isolated vessel segments from GRAF3-deficient mice show increased contractility. Taken together, our data indicate that GRAF3-mediated inhibition of RhoA activity in vascular SMCs is necessary for maintaining normal blood pressure homoeostasis. Moreover, these findings provide a potential mechanism for a hypertensive locus recently identified within arhgap42 and provide a foundation for the future development of innovative hypertension therapies.


Subject(s)
GTPase-Activating Proteins/metabolism , Hypertension/physiopathology , Muscle, Smooth, Vascular/metabolism , Amino Acid Sequence , Angiotensin II/pharmacology , Animals , Aorta/metabolism , Blood Pressure/genetics , Blood Vessels/physiopathology , Cells, Cultured , Female , GTPase-Activating Proteins/genetics , Humans , Hypertension/drug therapy , Hypertension/etiology , Hypertension/genetics , In Vitro Techniques , Male , Mice , Mice, Mutant Strains , Molecular Sequence Data , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Myosin Light Chains/metabolism , Phosphorylation , Rats , rhoA GTP-Binding Protein/metabolism
5.
J Biol Chem ; 286(29): 25903-21, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21622574

ABSTRACT

Although RhoA activity is necessary for promoting myogenic mesenchymal stem cell fates, recent studies in cultured cells suggest that down-regulation of RhoA activity in specified myoblasts is required for subsequent differentiation and myotube formation. However, whether this phenomenon occurs in vivo and which Rho modifiers control these later events remain unclear. We found that expression of the Rho-GTPase-activating protein, GRAF1, was transiently up-regulated during myogenesis, and studies in C2C12 cells revealed that GRAF1 is necessary and sufficient for mediating RhoA down-regulation and inducing muscle differentiation. Moreover, forced expression of GRAF1 in pre-differentiated myoblasts drives robust muscle fusion by a process that requires GTPase-activating protein-dependent actin remodeling and BAR-dependent membrane binding or sculpting. Moreover, morpholino-based knockdown studies in Xenopus laevis determined that GRAF1 expression is critical for muscle development. GRAF1-depleted embryos exhibited elevated RhoA activity and defective myofibrillogenesis that resulted in progressive muscle degeneration, defective motility, and embryonic lethality. Our results are the first to identify a GTPase-activating protein that regulates muscle maturation and to highlight the functional importance of BAR domains in myotube formation.


Subject(s)
Cell Differentiation , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Actins/metabolism , Animals , Base Sequence , Cell Fusion , Cell Line , Cell Membrane/metabolism , Embryo, Nonmammalian/metabolism , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Humans , Larva/growth & development , Larva/metabolism , Lipid Metabolism , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/growth & development , Myoblasts/cytology , Myoblasts/metabolism , Rats , Swimming , Xenopus Proteins/chemistry , Xenopus Proteins/deficiency , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis , rho GTP-Binding Proteins/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...