Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Gene Ther ; 29(9): 520-535, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35105949

ABSTRACT

Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the gene encoding dystrophin. Gene therapy using micro-dystrophin (MD) transgenes and recombinant adeno-associated virus (rAAV) vectors hold great promise. To overcome the limited packaging capacity of rAAV vectors, most MD do not include dystrophin carboxy-terminal (CT) domain. Yet, the CT domain is known to recruit α1- and ß1-syntrophins and α-dystrobrevin, a part of the dystrophin-associated protein complex (DAPC), which is a signaling and structural mediator of muscle cells. In this study, we explored the impact of inclusion of the dystrophin CT domain on ΔR4-23/ΔCT MD (MD1), in DMDmdx rats, which allows for relevant evaluations at muscular and cardiac levels. We showed by LC-MS/MS that MD1 expression is sufficient to restore the interactions at a physiological level of most DAPC partners in skeletal and cardiac muscles, and that inclusion of the CT domain increases the recruitment of some DAPC partners at supra-physiological levels. In parallel, we demonstrated that inclusion of the CT domain does not improve MD1 therapeutic efficacy on DMD muscle and cardiac pathologies. Our work highlights new evidences of the therapeutic potential of MD1 and strengthens the relevance of this candidate for gene therapy of DMD.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Animals , Chromatography, Liquid , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin-Associated Protein Complex/metabolism , Genetic Therapy , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Rats , Tandem Mass Spectrometry
2.
Biochim Biophys Acta ; 1839(1): 50-61, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24321385

ABSTRACT

Runx1 is a transcription factor essential for definitive hematopoiesis. In all vertebrates, the Runx1 gene is transcribed from two promoters: a proximal promoter (P2), and a distal promoter (P1). We previously found that runx1 expression in a specific hematopoietic cell population in zebrafish embryos depends on cohesin. Here we show that zebrafish runx1 is directly bound by cohesin and CCCTC binding factor (CTCF) at the P1 and P2 promoters, and within the intron between P1 and P2. Cohesin initiates expression of runx1 in the posterior lateral mesoderm and influences promoter use, while CTCF represses its expression in the newly emerging cells of the tail bud. The intronic binding sites for cohesin and CTCF coincide with histone modifications that confer enhancer-like properties, and two of the cohesin/CTCF sites behaved as insulators in an in vivo assay. The identified cohesin and CTCF binding sites are likely to be cis-regulatory elements (CREs) for runx1 since they also recruit RNA polymerase II (RNAPII). CTCF depletion excluded RNAPII from two intronic CREs but not the promoters of runx1. We propose that cohesin and CTCF have distinct functions in the regulation of runx1 during zebrafish embryogenesis, and that these regulatory functions are likely to involve runx1 intronic CREs. Cohesin (but not CTCF) depletion enhanced RUNX1 expression in a human leukemia cell line, suggesting conservation of RUNX1 regulation through evolution.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Repressor Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , Animals , CCCTC-Binding Factor , Cell Line, Tumor , Embryo, Nonmammalian , Embryonic Development , Gene Expression Regulation, Developmental , Humans , Mesoderm , Promoter Regions, Genetic , Zebrafish/genetics , Cohesins
3.
FASEB J ; 28(4): 1610-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24371119

ABSTRACT

The clinical symptoms of hemoglobin disorders such as ß-thalassemia and sickle cell anemia are significantly ameliorated by the persistent expression of γ-globin after birth. This knowledge has driven the discovery of important regulators that silence γ-globin postnatally. Improved understanding of the γ- to ß-globin switching mechanism holds the key to devising targeted therapies for ß-hemoglobinopathies. To further investigate this mechanism, we used the murine erythroleukemic (MEL) cell line containing an intact 183-kb human ß-globin locus, in which the (G)γ- and ß-globin genes are replaced by DsRed and eGFP fluorescent reporters, respectively. Following RNA interference (RNAi)-mediated knockdown of two key transcriptional regulators, Myb and BCL11A, we observed a derepression of γ-globin, measured by DsRed fluorescence and qRT-PCR (P<0.001). Interestingly, double knockdown of Myb and DNA methyltransferase 1 (DNMT1) resulted in a robust induction of ε-globin, (up to 20% of total ß-like globin species) compared to single knockdowns (P<0.001). Conversely, double knockdowns of BCL11A and DNMT1 enhanced γ-globin expression (up to 90% of total ß-like globin species) compared to single knockdowns (P<0.001). Moreover, following RNAi treatment, expression of human ß-like globin genes mirrored the expression levels of their endogenous murine counterparts. These results demonstrate that Myb and BCL11A cooperate with DNMT1 to achieve developmental repression of embryonic and fetal ß-like globin genes in the adult erythroid environment.


Subject(s)
Carrier Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Fetal Hemoglobin/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myb/genetics , RNA Interference , Animals , Blotting, Western , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins , Erythropoiesis/genetics , Fetal Hemoglobin/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transgenes/genetics , beta-Globins/genetics , beta-Globins/metabolism , epsilon-Globins/genetics , epsilon-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
4.
FASEB J ; 28(5): 2306-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24443374

ABSTRACT

Expression of fetal γ-globin in adulthood ameliorates symptoms of ß-hemoglobinopathies by compensating for the mutant ß-globin. Reactivation of the silenced γ-globin gene is therefore of substantial clinical interest. To study the regulation of γ-globin expression, we created the GG mice, which carry an intact 183-kb human ß-globin locus modified to express enhanced green fluorescent protein (eGFP) from the Gγ-globin promoter. GG embryos express eGFP first in the yolk sac blood islands and then in the aorta-gonad mesonephros and the fetal liver, the sites of normal embryonic hematopoiesis. eGFP expression in erythroid cells peaks at E9.5 and then is rapidly silenced (>95%) and maintained at low levels into adulthood, demonstrating appropriate developmental regulation of the human ß-globin locus. In vitro knockdown of the epigenetic regulator DNA methyltransferase-1 in GG primary erythroid cells increases the proportion of eGFP(+) cells in culture from 41.9 to 74.1%. Furthermore, eGFP fluorescence is induced >3-fold after treatment of erythroid precursors with epigenetic drugs known to induce γ-globin expression, demonstrating the suitability of the Gγ-globin eGFP reporter for evaluation of γ-globin inducers. The GG mouse model is therefore a valuable model system for genetic and pharmacologic studies of the regulation of the ß-globin locus and for discovery of novel therapies for the ß-hemoglobinopathies.


Subject(s)
Erythropoiesis/physiology , Gene Expression Regulation , gamma-Globins/metabolism , Animals , DNA Modification Methylases/metabolism , Disease Models, Animal , Epigenesis, Genetic , Erythroid Cells/metabolism , Female , Fetal Hemoglobin/metabolism , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Promoter Regions, Genetic , RNA Interference , Time Factors , Transgenes , beta-Globins/metabolism
5.
Nucleic Acid Ther ; 31(6): 457-464, 2021 12.
Article in English | MEDLINE | ID: mdl-34081545

ABSTRACT

Ribosomal protein L3-like (RPL3L) is a poorly characterized ribosomal protein that is exclusively expressed in skeletal and cardiac muscle. RPL3L is also downregulated in Duchenne muscular dystrophy (DMD), suggesting that it may play an important role in muscle biology. In this study, we investigated the role of RPL3L in skeletal muscle of healthy C57 and dystrophic mdx mice. We show that RPL3L is developmentally regulated and that intramuscular adeno-associated virus (AAV)-mediated RPL3L knockdown in the tibialis anterior of C57 and mdx mice results in increased specific force with improved resistance to eccentric contraction induced muscle damage in dystrophic muscles. The mechanism by which RPL3L knockdown improves muscle function remains unclear. Histological observations showed a significant increase in muscle length and decrease in muscle cross-sectional area after RPL3L inhibition suggesting that this ribosomal protein may play a role in myofiber morphology. The endogenous downregulation of RPL3L in DMD may be a protective mechanism that attempts to improve skeletal muscle function and counteract the dystrophic phenotype.


Subject(s)
Muscular Dystrophy, Duchenne , Ribosomal Protein L3 , Animals , Disease Models, Animal , Dystrophin , Mice , Mice, Inbred mdx , Muscle Contraction , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy
6.
Methods Mol Biol ; 1227: 323-43, 2015.
Article in English | MEDLINE | ID: mdl-25239756

ABSTRACT

Bacterial artificial chromosome (BAC) reporter cell lines are generated through stable transfection of a BAC reporter construct wherein the gene of interest is tagged with a reporter gene such as eGFP. The large capacity of BACs (up to 350 kb of genomic sequence) enables the inclusion of all regulatory elements that ensure appropriate regulation of the gene of interest. Furthermore, the reporter gene allows the expression of the gene of interest to be readily detected by flow cytometry. Cell lines can also be easily cultured for extended periods with minimal cost. These features of BAC reporter cell lines make them highly amenable for use in high-throughput screening of large drug libraries for compounds that induce the expression of the gene of interest. This chapter describes a method for generation of BAC reporter cell lines that are suitable as cellular assay systems in high-throughput screening. Briefly, this method involves (A) generation of cell clones stably transfected with a BAC reporter construct, (B) selection of "candidate" cell clones based on the responsiveness to known inducers, (C) confirmation of the integrity of the BAC reporter construct integrated within the candidate clones, and (D) assessment of the developmental regulation of the BAC reporter construct. As an example, we describe the generation of a BAC reporter cell line containing the human ß-globin locus modified to express γ-globin as eGFP for use as a cellular reporter assay for screening of drugs that can reactivate expression of developmentally silenced γ-globin for the treatment of ß-hemoglobin disorders.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Green Fluorescent Proteins/genetics , High-Throughput Screening Assays , Small Molecule Libraries/pharmacology , beta-Globins/genetics , gamma-Globins/genetics , Animals , Chromosomes, Artificial, Bacterial/metabolism , Drug Discovery , Electroporation , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , K562 Cells , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , beta-Globins/metabolism , gamma-Globins/metabolism
7.
J Appl Behav Anal ; 44(4): 909-13, 2011.
Article in English | MEDLINE | ID: mdl-22219539

ABSTRACT

The current study evaluated the effects of conditional discrimination (listener) training with coins on the emergence of novel stimulus relations, textual behavior, tacts, and intraverbals. Two preschoolers with autism were taught 3 relations among coins, their names, and values. After initial training, 4 relations emerged for the first participant and 7 for the second participant, suggesting that this technology can be incorporated into educational curricula for teaching prerequisite money skills to children with autism.


Subject(s)
Autistic Disorder/psychology , Discrimination Learning , Pattern Recognition, Visual , Attention , Child , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL