Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Epigenetics Chromatin ; 12(1): 3, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30606231

ABSTRACT

BACKGROUND: The 5 hydroxymethylation (5hmC) mark and TET DNA dioxygenases play a pivotal role in embryonic stem cell differentiation and animal development. However, very little is known about TET enzymes in lineage determination of human bone marrow-derived mesenchymal stem/stromal cells (BMSC). We examined the function of all three TET DNA dioxygenases, responsible for DNA hydroxymethylation, in human BMSC cell osteogenic and adipogenic differentiation. RESULTS: We used siRNA knockdown and retroviral mediated enforced expression of TET molecules and discovered TET1 to be a repressor of both osteogenesis and adipogenesis. TET1 was found to recruit the co-repressor proteins, SIN3A and the histone lysine methyltransferase, EZH2 to osteogenic genes. Conversely, TET2 was found to be a promoter of both osteogenesis and adipogenesis. The data showed that TET2 was directly responsible for 5hmC levels on osteogenic and adipogenic lineage-associated genes, whereas TET1 also played a role in this process. Interestingly, TET3 showed no functional effect in BMSC osteo-/adipogenic differentiation. Finally, in a mouse model of ovariectomy-induced osteoporosis, the numbers of clonogenic BMSC were dramatically diminished corresponding to lower trabecular bone volume and reduced levels of TET1, TET2 and 5hmC. CONCLUSION: The present study has discovered an epigenetic mechanism mediated through changes in DNA hydroxymethylation status regulating the activation of key genes involved in the lineage determination of skeletal stem cells, which may have implications in BMSC function during normal bone regulation. Targeting TET molecules or their downstream targets may offer new therapeutic strategies to help prevent bone loss and repair following trauma or disease.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Adipogenesis , Animals , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mixed Function Oxygenases/genetics , Osteogenesis , Protein Binding , Proto-Oncogene Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
2.
Skelet Muscle ; 7(1): 22, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29065908

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a lethal X-linked muscle wasting disorder caused by the absence of dystrophin, a large cytoskeletal muscle protein. Increasing the levels of the dystrophin-related-protein utrophin is a highly promising therapy for DMD and has been shown to improve pathology in dystrophin-deficient mice. One contributing factor to muscle wasting in DMD is mitochondrial pathology that contributes to oxidative stress and propagates muscle damage. The purpose of this study was to assess whether utrophin could attenuate mitochondria pathology and oxidative stress. METHODS: Skeletal muscles from wildtype C57BL/10, dystrophin-deficient mdx, dystrophin/utrophin double knockout (dko) and dystrophin-deficient mdx/utrophin over-expressing mdx-Fiona transgenic mice were assessed for markers of mitochondrial damage. RESULTS: Using transmission electron microscopy, we show that high levels of utrophin ameliorate the aberrant structure and localisation of mitochondria in mdx mice whereas absence of utrophin worsened these features in dko mice. Elevated utrophin also reverts markers of protein oxidation and oxidative stress, elevated in mdx and dko mice, to wildtype levels. These changes were observed independently of a shift in oxidative phenotype. CONCLUSION: These findings show that utrophin levels influence mitochondrial pathology and oxidative stress. While utrophin deficiency worsens the pathology, utrophin over-expression in dystrophic muscle benefits mitochondria and attenuates the downstream pathology associated with aberrant mitochondrial function.


Subject(s)
Mitochondria, Muscle/metabolism , Muscular Dystrophy, Duchenne/metabolism , Oxidative Stress , Utrophin/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Mitochondria, Muscle/ultrastructure , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Utrophin/metabolism
3.
FASEB J ; 31(3): 1011-1027, 2017 03.
Article in English | MEDLINE | ID: mdl-27934660

ABSTRACT

In this study, we examined the functional importance of EZH2 during skeletal development and homeostasis using the conditional deletion of Ezh2 (Ezh2fl/fl ) in early mesenchyme with the use of a Prrx-1-cre driver mouse (Ezh2+/+). Heterozygous (Ezh2+/-) newborn and 4-wk-old mice exhibited increased skeletal size, growth plate size, and weight when compared to the wild-type control (Ezh2+/+), whereas homozygous deletion of Ezh2 (Ezh2-/-) resulted in skeletal deformities and reduced skeletal size, growth plate size, and weight in newborn and 4-wk-old mice. Ezh2-/- mice exhibited enhanced trabecular patterning. Osteogenic cortical and trabecular bone formation was enhanced in Ezh2+/- and Ezh2-/- animals. Ezh2+/- and Ezh2-/- mice displayed thinner cortical bone and decreased mechanical strength compared to the wild-type control. Differences in cortical bone thickness were attributed to an increased number of osteoclasts, corresponding with elevated levels of the bone turnover markers cross-linked C-telopeptide-1 and tartrate-resistant acid phosphatase, detected within serum. Moreover, Ezh2+/- mice displayed increased osteoclastogenic potential coinciding with an upregulation of Rankl and M-csf expression by mesenchymal stem cells (MSCs). MSCs isolated from Ezh2+/- mice also exhibited increased trilineage potential compared with wild-type bone marrow stromal/stem cells (BMSCs). Gene expression studies confirmed the upregulation of known Ezh2 target genes in Ezh2-/- bone tissue, many of which are involved in Wnt/BMP signaling as promoters of osteogenesis and inhibitors of adipogenesis. In summary, EZH2 appears to be an important orchestrator of skeletal development, postnatal bone remodelling and BMSC fate determination in vitro and in vivo-Hemming, S., Cakouros, D., Codrington, J., Vandyke, K., Arthur, A., Zannettino, A., Gronthos, S. EZH2 deletion in early mesenchyme compromises postnatal bone microarchitecture and structural integrity and accelerates remodeling.


Subject(s)
Bone Remodeling , Cancellous Bone/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Mesoderm/metabolism , Osteogenesis , Animals , Cancellous Bone/cytology , Cancellous Bone/growth & development , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Deletion , Heterozygote , Homozygote , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Wnt Signaling Pathway
4.
Bone ; 93: 12-21, 2016 12.
Article in English | MEDLINE | ID: mdl-27622886

ABSTRACT

The EphB receptor tyrosine kinase family and their ephrinB ligands have been implicated as mediators of skeletal development and bone homeostasis in humans, where mutations in ephrinB1 contribute to frontonasal dysplasia and coronal craniosynostosis. In mouse models, ephrinB1 has been shown to be a critical factor mediating osteoblast function. The present study examined the functional importance of ephrinB1 during endochondral ossification using the Cre recombination system with targeted deletion of ephrinB1 (EfnB1fl/fl) in osteogenic progenitor cells, under the control of the osterix (Osx:Cre) promoter. The Osx:EfnB1-/- mice displayed aberrant bone growth during embryonic and postnatal skeletal development up to 4weeks of age, when compared to the Osx:Cre controls. Furthermore, compared to the Osx:Cre control mice, the Osx:EfnB1-/- mice exhibited significantly weaker and less rigid bones, with a reduction in trabecular/ cortical bone formation, reduced trabecular architecture and a reduction in the size of the growth plates at the distal end of the femora from newborn through to 4weeks of age. The aberrant bone formation correlated with increased numbers of tartrate resistant acid phosphatase positive osteoclasts and decreased numbers of bone lining osteoblasts in 4week old Osx:EfnB1-/- mice, compared to Osx:Cre control mice. Taken together, these observations demonstrate the importance of ephrinB1 signalling between cells of the skeleton required for endochondral ossification.


Subject(s)
Bone and Bones/physiology , Chondrogenesis , Ephrin-B1/deficiency , Osteogenesis , Stem Cells/metabolism , Animals , Bone and Bones/embryology , Cancellous Bone/growth & development , Cortical Bone/growth & development , Embryonic Development , Ephrin-B1/metabolism , Female , Growth Plate/growth & development , Male , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoclasts/metabolism , Promoter Regions, Genetic , Sp7 Transcription Factor/genetics , Sp7 Transcription Factor/metabolism , Survival Analysis
5.
Stem Cells Dev ; 25(12): 909-21, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27168161

ABSTRACT

Histone three lysine 27 (H3K27) methyltransferase enhancer of zeste homolog 2 (EZH2) is a critical epigenetic modifier, which regulates gene transcription through the trimethylation of the H3K27 residue leading to chromatin compaction and gene repression. EZH2 has previously been identified to regulate human bone marrow-derived mesenchymal stem cells (MSC) lineage specification. MSC lineage specification is regulated by the presence of EZH2 and its H3K27me3 modification or the removal of the H3K27 modification by lysine demethylases 6A and 6B (KDM6A and KDM6B). This study used a bioinformatics approach to identify novel genes regulated by EZH2 during MSC osteogenic differentiation. In this study, we identified the EZH2 targets, ZBTB16, MX1, and FHL1, which were expressed at low levels in MSC. EZH2 and H3K27me3 were found to be present along the transcription start site of their respective promoters. During osteogenesis, these genes become actively expressed coinciding with the disappearance of EZH2 and H3K27me3 on the transcription start site of these genes and the enrichment of the active H3K4me3 modification. Overexpression of EZH2 downregulated the transcript levels of ZBTB16, MX1, and FHL1 during osteogenesis. Small interfering RNA targeting of MX1 and FHL1 was associated with a downregulation of the key osteogenic transcription factor, RUNX2, and its downstream targets osteopontin and osteocalcin. These findings highlight that EZH2 not only acts through the direct regulation of signaling modules and lineage-specific transcription factors but also targets many novel genes important for mediating MSC osteogenic differentiation.


Subject(s)
Cell Differentiation , Enhancer of Zeste Homolog 2 Protein/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis , Adult , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Gene Knockdown Techniques , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Lysine/metabolism , Methylation , Muscle Proteins/metabolism , Myxovirus Resistance Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism , Up-Regulation/genetics
6.
Stem Cells Dev ; 24(11): 1297-308, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25579220

ABSTRACT

Basic helix-loop-helix (bHLH) transcription factors are pivotal regulators of cellular differentiation and development. The bHLH factor, Twist-1 has previously been found to control bone marrow stromal/stem cells (BMSC) self-renewal, life span, and differentiation, however not much is known about its mechanism of action. In this study, we have discovered a novel Twist-1 regulated bHLH gene, Hes4, expressed in humans, but not in mice. Its closest homologue in both humans and mice is Hes1. Overexpression and knockdown studies demonstrated that Hes4 promotes osteogenesis resulting in an increase in Runx2, osteocalcin, osteopontin, and bone sialoprotein expression. Conversely, Hes4 was found to inhibit adipogenesis accompanied by a decrease in PPARγ2, adiponectin, and adipsin expression. In vitro studies indicate that Hes4 employs a mechanism to counteract the negative function of Twist-1 on osteogenesis by binding to Twist-1 and inhibiting the ability of Twist-1 to bind and inhibit Runx2. In vivo chromatin immunoprecipitation and in vitro reporter assays illustrated that Runx2 recruitment to the osterix promoter, was found to be enhanced in the presence of Hes4 and inhibited in the presence of Twist-1. Therefore, Hes4 antagonizes the function of Twist-1 to regulate lineage commitment of BMSC. These studies highlight the potential differences in molecular mechanisms that regulate BMSC osteogenic differentiation between human and mouse.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Lineage , Mesenchymal Stem Cells/metabolism , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Twist-Related Protein 1/metabolism , Adipogenesis , Adiponectin/genetics , Adiponectin/metabolism , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cells, Cultured , Complement Factor D/genetics , Complement Factor D/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Mesenchymal Stem Cells/cytology , Molecular Sequence Data , Nuclear Proteins/genetics , Osteoblasts/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteopontin/genetics , Osteopontin/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Species Specificity , Twist-Related Protein 1/genetics
8.
Stem Cells ; 32(3): 802-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24123378

ABSTRACT

The methyltransferase, Enhancer of Zeste homology 2 (EZH2), trimethylates histone 3 lysine 27 (H3K27me3) on chromatin and this repressive mark is removed by lysine demethylase 6A (KDM6A). Loss of these epigenetic modifiers results in developmental defects. We demonstrate that Ezh2 and Kdm6a transcript levels change during differentiation of multipotential human bone marrow-derived mesenchymal stem cells (MSC). Enforced expression of Ezh2 in MSC promoted adipogenic in vitro and inhibited osteogenic differentiation potential in vitro and in vivo, whereas Kdm6a inhibited adipogenesis in vitro and promoted osteogenic differentiation in vitro and in vivo. Inhibition of EZH2 activity and knockdown of Ezh2 gene expression in human MSC resulted in decreased adipogenesis and increased osteogenesis. Conversely, knockdown of Kdm6a gene expression in MSC leads to increased adipogenesis and decreased osteogenesis. Both Ezh2 and Kdm6a were shown to affect expression of master regulatory genes involved in adipogenesis and osteogenesis and H3K27me3 on the promoters of master regulatory genes. These findings demonstrate an important epigenetic switch centered on H3K27me3 which dictates MSC lineage determination.


Subject(s)
Cell Lineage/genetics , Epigenesis, Genetic , Histone Demethylases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Adolescent , Adult , Animals , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation , Gene Knockdown Techniques , Histone Demethylases/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mice , Mice, SCID , Nuclear Proteins/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...