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.
Dev Neurorehabil ; 18(2): 75-81, 2015 Apr.
Article in English | MEDLINE | ID: mdl-23815083

ABSTRACT

OBJECTIVE: This study investigated the effectiveness of iPad-presented social stories in increasing the on-task behaviour of three young children with autism. METHOD: A single-subject with multiple baseline across participants design was employed with three 4-year-old children to assess intervention effectiveness during structured table top activities. Observational data were digitally recorded, scored, graphed, and interpreted using 10-second interval measures over 5-min periods across baseline, intervention, and withdrawal phases. RESULTS: The combination of the social story together with the iPad proved to be an effective intervention for one of the three child participants. These findings confirm that the intervention may be effective with some children, but not others. CONCLUSION: Overall, this study builds on existing research that supports social stories as a promising practice. Further research into the use of iPad-presented social stories, particularly for children of varying ages, abilities, and learning styles is recommended.


Subject(s)
Autistic Disorder/rehabilitation , Behavior Therapy/methods , Social Behavior , Autistic Disorder/psychology , Child, Preschool , Female , Humans , Male , Treatment Outcome
2.
PLoS One ; 9(4): e94032, 2014.
Article in English | MEDLINE | ID: mdl-24710205

ABSTRACT

Numerous human diseases can lead to atrophy of skeletal muscle, and loss of this tissue has been correlated with increased mortality and morbidity rates. Clinically addressing muscle atrophy remains an unmet medical need, and the development of preclinical tools to assist drug discovery and basic research in this effort is important for advancing this goal. In this report, we describe the development of a bioluminescent gene reporter rat, based on the zinc finger nuclease-targeted insertion of a bicistronic luciferase reporter into the 3' untranslated region of a muscle specific E3 ubiquitin ligase gene, MuRF1 (Trim63). In longitudinal studies, we noninvasively assess atrophy-related expression of this reporter in three distinct models of muscle loss (sciatic denervation, hindlimb unloading and dexamethasone-treatment) and show that these animals are capable of generating refined detail on in vivo MuRF1 expression with high temporal and anatomical resolution.


Subject(s)
Luminescent Measurements/methods , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Genes, Reporter , Hindlimb Suspension , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Rats , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
3.
FASEB J ; 28(7): 2790-803, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24671708

ABSTRACT

Controlled mechanical ventilation (CMV) is associated with the development of diaphragm atrophy and contractile dysfunction, and respiratory muscle weakness is thought to contribute significantly to delayed weaning of patients. Therefore, therapeutic strategies for preventing these processes may have clinical benefit. The aim of the current study was to investigate the role of the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in CMV-mediated diaphragm wasting and weakness in rats. CMV-induced diaphragm atrophy and contractile dysfunction coincided with marked increases in STAT3 phosphorylation on both tyrosine 705 (Tyr705) and serine 727 (Ser727). STAT3 activation was accompanied by its translocation into mitochondria within diaphragm muscle and mitochondrial dysfunction. Inhibition of JAK signaling during CMV prevented phosphorylation of both target sites on STAT3, eliminated the accumulation of phosphorylated STAT3 within the mitochondria, and reversed the pathologic alterations in mitochondrial function, reduced oxidative stress in the diaphragm, and maintained normal diaphragm contractility. In addition, JAK inhibition during CMV blunted the activation of key proteolytic pathways in the diaphragm, as well as diaphragm atrophy. These findings implicate JAK/STAT3 signaling in the development of diaphragm muscle atrophy and dysfunction during CMV and suggest that the delayed extubation times associated with CMV can be prevented by inhibition of Janus kinase signaling.-Smith, I. J., Godinez, G. L., Singh, B. K., McCaughey, K. M., Alcantara, R. R., Gururaja, T., Ho, M. S., Nguyen, H. N., Friera, A. M., White, K. A., McLaughlin, J. R., Hansen, D., Romero, J. M., Baltgalvis, K. A., Claypool, M. D., Li, W., Lang, W., Yam, G. C., Gelman, M. S., Ding, R., Yung, S. L., Creger, D. P., Chen, Y., Singh, R., Smuder, A. J., Wiggs, M. P., Kwon, O.-S., Sollanek, K. J., Powers, S. K., Masuda, E. S., Taylor, V. C., Payan, D. G., Kinoshita, T., Kinsella, T. M. Inhibition of Janus kinase signaling during controlled mechanical ventilation prevents ventilation-induced diaphragm dysfunction.


Subject(s)
Diaphragm/metabolism , Janus Kinases/metabolism , Respiration, Artificial/adverse effects , Signal Transduction/physiology , Animals , Interleukin-6/metabolism , Male , Mitochondria/metabolism , Muscle Weakness/metabolism , Muscular Atrophy/metabolism , Oxidative Stress/physiology , Phosphorylation/physiology , Proteolysis , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Serine/metabolism , Tyrosine/metabolism
4.
Am J Physiol Heart Circ Physiol ; 306(8): H1128-45, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24561866

ABSTRACT

Intermittent claudication is a form of exercise intolerance characterized by muscle pain during walking in patients with peripheral artery disease (PAD). Endothelial cell and muscle dysfunction are thought to be important contributors to the etiology of this disease, but a lack of preclinical models that incorporate these elements and measure exercise performance as a primary end point has slowed progress in finding new treatment options for these patients. We sought to develop an animal model of peripheral vascular insufficiency in which microvascular dysfunction and exercise intolerance were defining features. We further set out to determine if pharmacological activation of 5'-AMP-activated protein kinase (AMPK) might counteract any of these functional deficits. Mice aged on a high-fat diet demonstrate many functional and molecular characteristics of PAD, including the sequential development of peripheral vascular insufficiency, increased muscle fatigability, and progressive exercise intolerance. These changes occur gradually and are associated with alterations in nitric oxide bioavailability. Treatment of animals with an AMPK activator, R118, increased voluntary wheel running activity, decreased muscle fatigability, and prevented the progressive decrease in treadmill exercise capacity. These functional performance benefits were accompanied by improved mitochondrial function, the normalization of perfusion in exercising muscle, increased nitric oxide bioavailability, and decreased circulating levels of the endogenous endothelial nitric oxide synthase inhibitor asymmetric dimethylarginine. These data suggest that aged, obese mice represent a novel model for studying exercise intolerance associated with peripheral vascular insufficiency, and pharmacological activation of AMPK may be a suitable treatment for intermittent claudication associated with PAD.


Subject(s)
AMP-Activated Protein Kinases/physiology , Diet, High-Fat , Enzyme Activators/administration & dosage , Obesity/complications , Peripheral Vascular Diseases/physiopathology , Physical Exertion/physiology , Aging , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Arginine/analogs & derivatives , Arginine/blood , Cilostazol , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Intermittent Claudication/complications , Intermittent Claudication/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fatigue/drug effects , Muscle, Skeletal/blood supply , Nitric Oxide Synthase Type III/metabolism , Peripheral Vascular Diseases/etiology , Phosphodiesterase 3 Inhibitors/administration & dosage , Tetrazoles/administration & dosage , Vasodilator Agents
5.
Blood ; 117(25): 6866-75, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21531978

ABSTRACT

The activating mutations in JAK2 (including JAK2V617F) that have been described in patients with myeloproliferative neoplasms (MPNs) are linked directly to MPN pathogenesis. We developed R723, an orally bioavailable small molecule that inhibits JAK2 activity in vitro by 50% at a concentration of 2nM, while having minimal effects on JAK3, TYK2, and JAK1 activity. R723 inhibited cytokine-independent CFU-E growth and constitutive activation of STAT5 in primary hematopoietic cells expressing JAK2V617F. In an anemia mouse model induced by phenylhydrazine, R723 inhibited erythropoiesis. In a leukemia mouse model using Ba/F3 cells expressing JAK2V617F, R723 treatment prolonged survival and decreased tumor burden. In V617F-transgenic mice that closely mimic human primary myelofibrosis, R723 treatment improved survival, hepatosplenomegaly, leukocytosis, and thrombocytosis. R723 preferentially targeted the JAK2-dependent pathway rather than the JAK1- and JAK3-dependent pathways in vivo, and its effects on T and B lymphocytes were mild compared with its effects on myeloid cells. Our preclinical data indicate that R723 has a favorable safety profile and the potential to become an efficacious treatment for patients with JAK2V617F-positive MPNs.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Janus Kinase 2/antagonists & inhibitors , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Anemia, Hemolytic/chemically induced , Animals , Cell Line , Cells, Cultured , Erythropoiesis/drug effects , Female , Humans , Janus Kinase 2/genetics , Leukemia/drug therapy , Leukemia/genetics , Leukocytosis/drug therapy , Mice , Mice, Inbred BALB C , Mice, SCID , Mutation/drug effects
6.
Mol Biol Cell ; 16(12): 5621-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16195352

ABSTRACT

Early cellular events associated with tumorigenesis often include loss of cell cycle checkpoints or alteration in growth signaling pathways. Identification of novel genes involved in cellular proliferation may lead to new classes of cancer therapeutics. By screening a tetracycline-inducible cDNA library in A549 cells for genes that interfere with proliferation, we have identified a fragment of UHRF1 (ubiquitin-like protein containing PHD and RING domains 1), a nuclear RING finger protein, that acts as a dominant negative effector of cell growth. Reduction of UHRF1 levels using an UHRF1-specific shRNA decreased growth rates in several tumor cell lines. In addition, treatment of A549 cells with agents that activated different cell cycle checkpoints resulted in down-regulation of UHRF1. The primary sequence of UHRF1 contains a PHD and a RING motif, both of which are structural hallmarks of ubiquitin E3 ligases. We have confirmed using an in vitro autoubiquitination assay that UHRF1 displays RING-dependent E3 ligase activity. Overexpression of a GFP-fused UHRF1 RING mutant that lacks ligase activity sensitizes cells to treatment with various chemotherapeutics. Taken together, our results suggest a general requirement for UHRF1 in tumor cell proliferation and implicate the RING domain of UHRF1 as a functional determinant of growth regulation.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Division/physiology , Neoplasms/enzymology , Binding Sites , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor , Cloning, Molecular , HeLa Cells , Humans , Kinetics , Oligonucleotides, Antisense , Recombinant Proteins/metabolism , Retroviridae/genetics , Transcription, Genetic , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...