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.
Stem Cells ; 35(3): 597-610, 2017 03.
Article in English | MEDLINE | ID: mdl-27734557

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by the loss of the protein dystrophin, leading to muscle fragility, progressive weakening, and susceptibility to mechanical stress. Although dystrophin-negative mdx mouse models have classically been used to study DMD, phenotypes appear mild compared to patients. As a result, characterization of muscle pathology, especially in the heart, has proven difficult. We report that injection of mdx embryonic stem cells (ESCs) into Wild Type blastocysts produces adult mouse chimeras with severe DMD phenotypes in the heart and skeletal muscle. Inflammation, regeneration and fibrosis are observed at the whole organ level, both in dystrophin-negative and dystrophin-positive portions of the chimeric tissues. Skeletal and cardiac muscle function are also decreased to mdx levels. In contrast to mdx heterozygous carriers, which show no significant phenotypes, these effects are even observed in chimeras with low levels of mdx ESC incorporation (10%-30%). Chimeric mice lack typical compensatory utrophin upregulation, and show pathological remodeling of Connexin-43. In addition, dystrophin-negative and dystrophin-positive isolated cardiomyocytes show augmented calcium response to mechanical stress, similar to mdx cells. These global effects highlight a novel role of mdx ESCs in triggering muscular dystrophy even when only low amounts are present. Stem Cells 2017;35:597-610.


Subject(s)
Aging/pathology , Chimera/metabolism , Embryonic Stem Cells/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Myocardium/pathology , Animals , Calcium/metabolism , Connexin 43/metabolism , Dystrophin/metabolism , Female , Heart Function Tests , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Myocytes, Cardiac/metabolism , Regeneration
2.
Biol Open ; 3(9): 821-31, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25150276

ABSTRACT

PDCD2 (programmed cell death domain 2) is a highly conserved, zinc finger MYND domain-containing protein essential for normal development in the fly, zebrafish and mouse. The molecular functions and cellular activities of PDCD2 remain unclear. In order to better understand the functions of PDCD2 in mammalian development, we have examined PDCD2 activity in mouse blastocyst embryos, as well as in mouse embryonic stem cells (ESCs) and embryonic fibroblasts (MEFs). We have studied mice bearing a targeted PDCD2 locus functioning as a null allele through a splicing gene trap, or as a conditional knockout, by deletion of exon2 containing the MYND domain. Tamoxifen-induced knockout of PDCD2 in MEFs, as well as in ESCs, leads to defects in progression from the G1 to the S phase of cell cycle, associated with increased levels of p53 protein and p53 target genes. G1 prolongation in ESCs was not associated with induction of differentiation. Loss of entry into S phase of the cell cycle and marked induction of nuclear p53 were also observed in PDCD2 knockout blastocysts. These results demonstrate a unique role for PDCD2 in regulating the cell cycle and p53 activation during early embryonic development of the mouse.

3.
Stem Cells Dev ; 22(1): 58-72, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22800338

ABSTRACT

Programmed cell death 2 (Pdcd2) is a highly conserved protein of undefined function, and is widely expressed in embryonic and adult tissues. The observations that knockout of Pdcd2 in the mouse is embryonic lethal at preimplantation stages, and that in Drosophila, Zfrp8, the ortholog of Pdcd2, is required for normal lymph gland development suggest that Pdcd2 is important for regulating hematopoietic development. Through genetic and functional studies, we investigated pdcd2 function during the zebrafish ontogeny. Knockdown of pdcd2 expression in zebrafish embryos resulted in defects in embryonic hematopoietic development. Loss of pdcd2 function caused increased expression of progenitor markers, and accumulation of erythroid progenitors during primitive hematopoiesis. Additionally, hematopoietic stem cells (HSCs) failed to appear in the aorta-gonad mesonephros, and were not able to terminally differentiate or reconstitute hematopoiesis. Pdcd2 effects on HSC emergence were cell autonomous and P53-independent, and loss of pdcd2 function was associated with mitotic defects and apoptosis. Restoration of runx1 function(s) and modulation of apoptosis through the inhibition of Jak/Stat signaling rescued the hematopoietic and erythroid defects resulting from pdcd2 knockdown. Our studies suggest that pdcd2 plays a critical role in regulating the transcriptional hierarchy controlling hematopoietic lineage determination. Furthermore, the effects of pdcd2 in regulating mitotic cell death may contribute to its role(s) in directing hematopoietic differentiation during development.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Differentiation , Embryonic Development , Hematopoietic Stem Cells/physiology , Zebrafish Proteins/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Erythroid Cells/metabolism , Erythropoiesis , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Mice , Mitosis , Morpholinos/genetics , Neovascularization, Physiologic , Organ Specificity , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
4.
Exp Hematol ; 40(12): 1028-1042.e3, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22922207

ABSTRACT

Programmed cell death-2 (PDCD2) protein is enriched in embryonic, hematopoietic, and neural stem cells, however, its function in stem/progenitor cell differentiation is unclear. We investigated the effects of PDCD2 knockdown on the development and differentiation of hematopoietic progenitor cells (HPC). CD34(+) cells derived from normal human bone marrow and K562 leukemic cells were effectively transduced with short-hairpin RNA to knockdown PDCD2. Colony-forming assays were used to investigate the effects of PDCD2 loss on HPC clonogenic potential and on 12-O-tetradecanoyl-phorbol-13-acetate-and arabinofuranosylcytosine-induced terminal differentiation. In CD34(+) clonogenic progenitors, PDCD2 knockdown decreased the total number of colony-forming units, increased the number of colony-forming units-granulocyte-erythroid-macrophage-megakaryocyte and burst-forming unit-erythroid primitive colonies, and decreased the number of burst-forming unit-erythroid mature colonies. Similar results were observed in K562 cells, suggesting that PDCD2 is important for HPC differentiation and/or survival, and for erythroid lineage commitment. Furthermore, 12-O-tetradecanoyl-phorbol-13-acetate-induced megakaryocytic differentiation and proliferation of K562 cells was not affected by PDCD2 knockdown. In contrast, arabinofuranosylcytosine-induced erythroid differentiation of K562 cells was significantly reduced with PDCD2 knockdown, with no effect on cell proliferation. The effects of PDCD2 knockdown were attributed to a cell cycle arrest at G(0)/G(1), along with increased messenger RNA expression of early progenitor factors c-MYB and GATA-2, and decreased expression of erythroid factors GATA-1, EpoR, and γ-globin. We conclude that PDCD2 loss of function(s) impedes erythroid differentiation by inducing cell cycle arrest and increasing expression of early hematopoietic progenitor factors. These findings suggest that PDCD2 has a novel regulatory role in human hematopoiesis and is essential for erythroid development.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Cell Differentiation/genetics , Erythroid Cells/cytology , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Antigens, CD34/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation , Cytarabine/pharmacology , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Gene Knockdown Techniques , Genetic Vectors/genetics , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Lentivirus/genetics , Leukemia/genetics , Megakaryocytes/drug effects , Megakaryocytes/metabolism , RNA Interference , Transduction, Genetic
5.
Dev Biol ; 349(2): 350-62, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21047506

ABSTRACT

Nodal, a secreted factor known for its conserved functions in cell-fate specification and the establishment of embryonic axes, is also required in mammals to maintain the pluripotency of the epiblast, the tissue that gives rise to all fetal lineages. Although Nodal is expressed as early as E3.5 in the mouse embryo, its regulation and functions at pre- and peri-implantation stages are currently unknown. Sensitive reporter transgenes for two Nodal cis-regulatory regions, the PEE and the ASE, exhibit specific expression profiles before implantation. Mutant and inhibitor studies find them respectively regulated by Wnt/ß-catenin signaling and Activin/Nodal signaling, and provide evidence for localized and heterogeneous activities of these pathways in the inner cell mass, the epiblast and the primitive endoderm. These studies also show that Nodal and its prime effector, FoxH1, are not essential to preimplantation Activin/Nodal signaling. Finally, a strong upregulation of the ASE reporter in implanting blastocysts correlates with a downregulation of the pluripotency factor Nanog in the maturing epiblast. This study uncovers conservation in the mouse blastocyst of Wnt/ß-catenin and Activin/Nodal-dependent activities known to govern Nodal expression and the establishment of polarity in the blastula of other deuterostomes. Our results indicate that these pathways act early on to initiate distinct cell-specification processes in the ICM derivatives. Our data also suggest that the activity of the Activin/Nodal pathway is dampened by interactions with the molecular machinery of pluripotency until just before implantation, possibly delaying cell-fate decisions in the mouse embryo.


Subject(s)
Embryo, Mammalian/embryology , Endoderm/physiology , Gene Expression Regulation, Developmental/physiology , Germ Layers/physiology , Nodal Protein/metabolism , Signal Transduction/physiology , Activins/metabolism , Animals , Binding Sites/genetics , Computational Biology , Conserved Sequence/genetics , DNA Primers/genetics , Embryo, Mammalian/metabolism , Endoderm/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Genotype , Germ Layers/metabolism , Homeodomain Proteins/metabolism , In Situ Hybridization , Likelihood Functions , Mice , Mice, Transgenic , Microscopy, Confocal , Models, Genetic , Nanog Homeobox Protein , Nodal Protein/genetics , Signal Transduction/genetics , beta-Galactosidase
SELECTION OF CITATIONS
SEARCH DETAIL
...