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.
Leukemia ; 30(7): 1552-61, 2016 07.
Article in English | MEDLINE | ID: mdl-27055868

ABSTRACT

Aberrant ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) expression drives leukemic transformation in mice and high expression is associated with poor patient outcomes in acute myeloid leukemia (AML) and T-acute lymphoblastic leukemia (T-ALL). Protein phosphorylation regulates the activity of many ETS factors but little is known about ERG in leukemic cells. To characterize ERG phosphorylation in leukemic cells, we applied liquid chromatography coupled tandem mass spectrometry and identified five phosphorylated serines on endogenous ERG in T-ALL and AML cells. S283 was distinct as it was abundantly phosphorylated in leukemic cells but not in healthy hematopoietic stem and progenitor cells (HSPCs). Overexpression of a phosphoactive mutant (S283D) increased expansion and clonogenicity of primary HSPCs over and above wild-type ERG. Using a custom antibody, we screened a panel of primary leukemic xenografts and showed that ERG S283 phosphorylation was mediated by mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling and in turn regulated expression of components of this pathway. S283 phosphorylation facilitates ERG enrichment and transactivation at the ERG +85 HSPC enhancer that is active in AML and T-ALL with poor prognosis. Taken together, we have identified a specific post-translational modification in leukemic cells that promotes progenitor proliferation and is a potential target to modulate ERG-driven transcriptional programs in leukemia.


Subject(s)
Leukemia/pathology , MAP Kinase Signaling System/physiology , Binding Sites , Cell Line, Tumor , Cell Proliferation , Hematopoietic Stem Cells , Humans , Leukemia, Myeloid, Acute/pathology , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Processing, Post-Translational , Serine/metabolism , Transcriptional Regulator ERG/metabolism , Transcriptome
2.
Biol Reprod ; 81(1): 234-42, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630168

ABSTRACT

A universal response to cellular stress is the expression of transformation-related protein 53 (TRP53). This transcription factor reduces cell proliferation and/or survival and is classed as a tumour suppressor protein. Several stresses (including culture) cause increased TRP53 expression in blastocysts and their reduced long-term developmental potential. This study shows that culture from the zygote stage (but not the 2-cell stage) reduced the development of C57BL6 inbred (but not hybrid) strain mouse embryos. Reduced viability was TRP53 dependent, being partially reversed by a TRP53 inhibitor (Pifithrin-alpha). However, the presence of culture did not cause an increase in Trp53 mRNA levels (levels were reduced following culture, P < 0.001). Transformed mouse 3T3 cell double minute 2 (MDM2) causes the ubiquitination and degradation of TRP53. MDM2 activation is accompanied by phosphorylation of Ser-166, and this is commonly catalyzed by the phosphatidylinositol-3 kinase and RAC-alpha serine/threonine-protein kinase (AKT) signaling pathway. Paf is an autocrine embryotrophin that activates the phosphatidylinositol-3 kinase/AKT pathway. High levels of TRP53 expression occurred following the culture of zygotes lacking the Paf receptor (Ptafr(-/-)) and following inhibition of phosphatidylinositol-3 kinase or AKT. Inhibition of MDM2 caused a Trp53-dependent reduction in zygote development. Inbred strain embryos cultured from the zygote stage expressed less phosphorylated MDM2 than similar embryos collected from the uterus. The addition of Paf to the media caused increased phosphorylation of MDM2, and this was blocked by inhibitors of phosphatidylinositol-3 kinase and AKT. The study identifies trophic ligand signaling via the activation of phosphatidylinositol-3 kinase and AKT as a mechanism resulting in the activation of MDM2.


Subject(s)
Embryonic Development/genetics , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/genetics , Animals , Enzyme Activation , Female , Gene Expression Regulation, Developmental , Ligands , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Models, Biological , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/physiology , Time Factors , Tumor Suppressor Protein p53/metabolism
3.
Biol Reprod ; 80(2): 286-94, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18923161

ABSTRACT

A universal response to cellular stress is the expression of transformation-related protein 53 (TRP53). This transcription factor reduces cell proliferation and/or survival and is classed as a tumour suppressor protein. Several stresses (including culture) cause increased TRP53 expression in blastocysts and their reduced long-term developmental potential. This study shows that culture from the zygote stage (but not the 2-cell stage) reduced the development of C57BL6 inbred (but not hybrid) strain mouse embryos. Reduced viability was TRP53 dependent, being partially reversed by a TRP53 inhibitor (Pifithrin-alpha). However, the presence of culture did not cause an increase in Trp53 mRNA levels (levels were reduced following culture, P < 0.001). Transformed mouse 3T3 cell double minute 2 (MDM2) causes the ubiquitination and degradation of TRP53. MDM2 activation is accompanied by phosphorylation of Ser-166, and this is commonly catalyzed by the phosphatidylinositol-3 kinase and RAC-alpha serine/threonine-protein kinase (AKT) signaling pathway. Paf is an autocrine embryotrophin that activates the phosphatidylinositol-3 kinase/AKT pathway. High levels of TRP53 expression occurred following the culture of zygotes lacking the Paf receptor (Ptafr(-/-)) and following inhibition of phosphatidylinositol-3 kinase or AKT. Inhibition of MDM2 caused a Trp53-dependent reduction in zygote development. Inbred strain embryos cultured from the zygote stage expressed less phosphorylated MDM2 than similar embryos collected from the uterus. The addition of Paf to the media caused increased phosphorylation of MDM2, and this was blocked by inhibitors of phosphatidylinositol-3 kinase and AKT. The study identifies trophic ligand signaling via the activation of phosphatidylinositol-3 kinase and AKT as a mechanism resulting in the activation of MDM2.


Subject(s)
Embryonic Development/genetics , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/genetics , Animals , Cells, Cultured , Enzyme Activation , Female , Gene Expression Regulation, Developmental , Ligands , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Models, Biological , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/physiology , Up-Regulation
4.
Biol Reprod ; 76(3): 362-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17093197

ABSTRACT

The expression of TRP53 in blastocysts that had been cultured from the zygote stage in vitro for 90 h was compared with that in blastocysts collected from the uterus in C57BL6 (B6) and in F1 hybrid (B6CBF1) strain mice. In both strains, there was little TRP53 detected in blastocysts collected from the uterus. There was some increased expression in cultured embryos from B6CBF1 mice and marked increased expression in cultured B6 blastocysts. In cultured B6 embryos, there was obvious accumulation of TRP53 within the nuclear region of embryonic cells. Cultured B6 zygotes had significantly poorer rates of blastocyst formation and of capacity to undergo implantation or form viable fetuses than cultured zygotes from B6CBF1 mice or B6 blastocysts collected from the uterus. Trp53-/- zygotes (B6 background) were significantly more likely to form blastocysts than sibling wild-type embryos, with Trp53+/- embryos having an intermediate level of viability (P<0.01). On transfer of blastocysts to recipient females, Trp53-/- blastocysts were more likely to form viable fetuses than wild-type or heterozygous sibling blastocysts when the embryos resulted from culture of zygotes (P<0.001). This shift in viability did not occur when embryos were only subjected to 24 h of culture from the compacted embryo stage. Culture in vitro in the B6 strain caused a marked increase in the expression and nuclear accumulation of TRP53. This expression was a significant cause of the loss of viability that occurs on culture of zygotes from this strain in vitro.


Subject(s)
Embryo Culture Techniques , Embryo, Mammalian/physiology , Tumor Suppressor Protein p53/metabolism , Zygote , Animals , Blastocyst/cytology , Embryo Loss , Embryo Transfer , Embryo, Mammalian/cytology , Female , Gene Dosage , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pregnancy , Tumor Suppressor Protein p53/genetics
5.
J Cell Sci ; 117(Pt 8): 1567-76, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15020683

ABSTRACT

The growth and survival of the preimplantation mammalian embryo may be regulated by several autocrine trophic factors that have redundant or overlapping actions. One of the earliest trophic factors to be produced is embryo-derived platelet-activating factor (1-O-alky-2-acetyl-sn-glyceryl-3-phosphocholine). The addition of platelet-activating factor to embryo culture media exerted a trophic effect, but structurally related lipids (3-O-alky-2-acetyl-sn-glyceryl-1-phosphocholine, 1-O-alky-sn-glyceryl-3-phosphocholine, octadecyl-phosphocholine) had no effect. Platelet-activating factor induced a pertussis toxin-sensitive [Ca(2+)](i) transient in two-cell embryos that did not occur in platelet-activating factor-receptor null (Pafr-/-) genotype embryos. Fewer Pafr-/- mouse zygotes developed to the blastocyst stage in vitro compared with Pafr+/+ zygotes (P<0.02), those that developed to blastocysts had fewer cells (P<0.001) and more cells with fragmented nuclei (P<0.001). The inhibition of 1-O-phosphatidylinositol 3-kinase (LY294002 (3 microM and 15 microM) and wortmannin (10 nM and 50 nM)) caused a dose-dependent inhibition of platelet-activating factor-induced [Ca(2+)](i) transients (P<0.001). The two-cell embryo expressed 1-O-phosphatidylinositol 3-kinase catalytic subunits p110 alpha, beta, gamma and delta, and regulatory subunits p85 alpha and beta. LY294002 and wortmannin each caused a significant reduction in the proportion of embryos developing to the morula and blastocyst stages in vitro, reduced the number of cells within each blastocyst, and significantly increased the proportion of cells in blastocysts with fragmented nuclei. The results indicate that embryo-derived platelet-activating factor (and other embryotrophic factors) act through its membrane receptor to enhance embryo survival through a 1-O-phosphatidylinositol 3-kinase-dependent survival pathway.


Subject(s)
Blastocyst , Embryonic Development , Phosphatidylinositol 3-Kinases/metabolism , Androstadienes/pharmacology , Animals , Blastocyst/cytology , Blastocyst/drug effects , Calcium Signaling/drug effects , Chromones/pharmacology , Culture Media/pharmacology , Culture Techniques , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Mice , Mice, Knockout , Morpholines/pharmacology , Morula/cytology , Platelet Activating Factor/antagonists & inhibitors , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/metabolism , Pregnancy , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Wortmannin , Zygote
SELECTION OF CITATIONS
SEARCH DETAIL
...