Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 45(7): 4142-4157, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27998933

ABSTRACT

miRNAs play important roles during mammalian spermatogenesis. However, the function of most miRNAs in spermatogenesis and the underlying mechanisms remain unknown. Here, we report that miR-202 is highly expressed in mouse spermatogonial stem cells (SSCs), and is oppositely regulated by Glial cell-Derived Neurotrophic Factor (GDNF) and retinoic acid (RA), two key factors for SSC self-renewal and differentiation. We used inducible CRISPR-Cas9 to knockout miR-202 in cultured SSCs, and found that the knockout SSCs initiated premature differentiation accompanied by reduced stem cell activity and increased mitosis and apoptosis. Target genes were identified with iTRAQ-based proteomic analysis and RNA sequencing, and are enriched with cell cycle regulators and RNA-binding proteins. Rbfox2 and Cpeb1 were found to be direct targets of miR-202 and Rbfox2 but not Cpeb1, is essential for the differentiation of SSCs into meiotic cells. Accordingly, an SSC fate-regulatory network composed of signaling molecules of GDNF and RA, miR-202 and diverse downstream effectors has been identified.


Subject(s)
Adult Germline Stem Cells/metabolism , Cell Cycle/genetics , MicroRNAs/metabolism , RNA Splicing Factors/biosynthesis , Adult Germline Stem Cells/cytology , Animals , Gene Knockout Techniques , Male , Meiosis/genetics , Mice, Inbred C57BL , Mice, Inbred DBA , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Proteomics , Sequence Analysis, RNA , Spermatogenesis/genetics , Transcription Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/biosynthesis
2.
Dev Biol ; 426(1): 43-55, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28434803

ABSTRACT

Cytoplasmic polyadenylation is a mechanism of mRNA regulation prevalent in metazoan germ cells; it is largely dependent on Cytoplasmic Polyadenylation Element Binding proteins (CPEBs). Two CPEB homologs were identified in the planarian Schmidtea mediterranea. Smed-CPEB1 is expressed in ovaries and yolk glands of sexually mature planarians, and required for oocyte and yolk gland development. In contrast, Smed-CPEB2 is expressed in the testes and the central nervous system; its function is required for spermatogenesis as well as non-autonomously for development of ovaries and accessory reproductive organs. Transcriptome analysis of CPEB knockdown animals uncovered a comprehensive collection of molecular markers for reproductive structures in S. mediterranea, including ovaries, testes, yolk glands, and the copulatory apparatus. Analysis by RNA interference revealed contributions for a dozen of these genes during oogenesis, spermatogenesis, or capsule formation. We also present evidence suggesting that Smed-CPEB2 promotes translation of Neuropeptide Y-8, a prohormone required for planarian sexual maturation. These findings provide mechanistic insight into potentially conserved processes of germ cell development, as well as events involved in capsule deposition by flatworms.


Subject(s)
Germ Cells/cytology , Oogenesis/physiology , Ovary/growth & development , Planarians/anatomy & histology , Planarians/growth & development , Spermatogenesis/physiology , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Cell Differentiation/genetics , Female , Gene Expression Profiling , Ovary/metabolism , Polyadenylation , RNA Interference , RNA, Small Interfering/genetics , Receptors, Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/genetics , Sexual Maturation/genetics , Sexual Maturation/physiology , mRNA Cleavage and Polyadenylation Factors/biosynthesis
3.
Mol Reprod Dev ; 81(4): 376-87, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24474627

ABSTRACT

In many species, there is little transcription in the mature oocyte, and zygotic transcription does not begin immediately after fertilization. In zebrafish, zygotic transcription is not initiated until the mid-blastula transition, thus the production of new proteins during oogenesis and early embryogenesis is dependent on the translation of maternal mRNAs. In a growing number of species, the translation of key maternal transcripts is coupled to their cytoplasmic polyadenylation. One family of RNA-binding proteins implicated in this process is the cytoplasmic polyadenylation element (CPE)-binding proteins (CPEBs), which bind to a sequence in the 3'-untranslated regions of regulated transcripts and mediate their storage/repression or translation. In several species, there is evidence for two classes of CPEBs, a larger oocyte-type and a smaller CPEB that functions during embryogenesis. This appears to be the case in zebrafish as well, and we now provide evidence suggesting that the oocyte-type CPEB (zorba) regulates the translation of the embryonic-type (ElrA) by keeping the ElrA transcript in storage until fertilization. When zorba levels fall, ElrA protein is then produced and available to regulate the translation of additional mRNAs during embryogenesis. We have also identified a potential target of ElrA, the maternal mRNA for hnRNPab, which is a potential homolog of the Drosophila gene squid, whose product plays a role in patterning the Drosophila oocyte and embryo. These data suggest that during zebrafish embryogenesis, cytoplasmic polyadenylation mediates a cascade of translational control whose final targets play central patterning roles during embryogenesis.


Subject(s)
ELAV Proteins/biosynthesis , Gene Expression Regulation, Developmental , Oogenesis/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/biosynthesis , RNA-Binding Proteins/physiology , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/physiology , Zebrafish/genetics , mRNA Cleavage and Polyadenylation Factors/biosynthesis , 3' Untranslated Regions , Animals , Blastula/metabolism , Body Patterning/genetics , Cytoplasm/metabolism , ELAV Proteins/genetics , Embryo, Nonmammalian/metabolism , Female , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Oocytes/metabolism , Polyadenylation , Protein Biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Zebrafish/embryology , Zebrafish/physiology , Zebrafish Proteins/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/physiology
4.
Gut ; 61(8): 1115-23, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22052064

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a highly prevalent disease, being the fourth most common cancer and the second leading cause of cancer-associated deaths worldwide. Although many genes have been implicated in its development, many cases remain genetically unexplained. Hence, there is an urgent need to find new disease-related genes. METHODS: A transgenic Drosophila model was used to screen for novel genes putatively involved in GC. The authors evaluated the expression of the most interesting candidates in GC cell lines and primary tumours by semi-quantitative reverse transcription PCR, dissected the molecular mechanisms responsible for the deregulation of the most relevant one, and analysed its functional role in vitro and in a chicken embryo model. RESULTS: Six candidate genes were identified, of which cytoplasmic polyadenylation element binding protein 1 (CPEB1) was downregulated in all GC cell lines and in 11 of 12 primary GC tumours. The pivotal CPEB1 promoter CpG site was determined, and it was found that methylation at this 79th CpG site was associated with CPEB1 silencing in GC cell lines and primary tumours. It was also discovered that methylation of this site was significantly more prevalent in diffuse type GC (p=0.007) and in cases with lymph node metastases (p=0.042). In vitro, CPEB1 impaired invasion. Its antiangiogenic role was also discovered, which was associated with downregulation of MMP14 and VEGFA. CONCLUSIONS: The first evidence of CPEB1 involvement in GC is presented, along with the molecular mechanism underlying the regulation of its expression and its potential role in invasion and angiogenesis.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms, Experimental/genetics , RNA, Neoplasm/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Blotting, Western , Cell Line, Tumor , DNA Methylation , Drosophila Proteins/biosynthesis , Gene Silencing , Humans , Immunohistochemistry , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcription Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/biosynthesis
5.
J Biol Chem ; 286(4): 2853-63, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21098481

ABSTRACT

Precise control of the timing of translational activation of dormant mRNAs stored in oocytes is required for normal progression of oocyte maturation. We previously showed that Pumilio1 (Pum1) is specifically involved in the translational control of cyclin B1 mRNA during Xenopus oocyte maturation, in cooperation with cytoplasmic polyadenylation element-binding protein (CPEB). It was reported that another Pumilio, Pumilio2 (Pum2), exists in Xenopus oocytes and that this protein regulates the translation of RINGO mRNA, together with Deleted in Azoospermia-like protein (DAZL). In this study, we characterized Pum1 and Pum2 biochemically by using newly produced antibodies that discriminate between them. Pum1 and Pum2 are bound to several key proteins involved in translational control of dormant mRNAs, including CPEB and DAZL, in immature oocytes. However, Pum1 and Pum2 themselves have no physical interaction. Injection of anti-Pum1 or anti-Pum2 antibody accelerated CPEB phosphorylation, cyclin B1 translation, and oocyte maturation. Pum1 phosphorylation coincides with the dissociation of CPEB from Pum1 and the translational activation of cyclin B1 mRNA, a target of Pum1, whereas Pum2 phosphorylation occurred at timing earlier than that for Pum1. Some, but not all, of cyclin B1 mRNAs release the deadenylase PARN during oocyte maturation, whereas Pum1 remains associated with the mRNA. On the basis of these findings, we discuss the functions of Pum1 and Pum2 in translational control of mRNAs during oocyte maturation.


Subject(s)
Oocytes/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Antibodies/chemistry , Base Sequence , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cyclin B1/biosynthesis , Cyclin B1/genetics , Molecular Sequence Data , Phosphorylation/physiology , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Time Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Xenopus Proteins/biosynthesis , Xenopus Proteins/genetics , Xenopus laevis , mRNA Cleavage and Polyadenylation Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/genetics
6.
Mol Reprod Dev ; 79(6): 380-91, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22467188

ABSTRACT

During early embryo development, chromatin packaging is sustained by histones of maternal origin. Most histone messenger RNAs are not polyadenylated, but rather end in an evolutionarily conserved stem-loop that controls RNA processing, nucleocytoplasmic transport, stability, and translation via interactions with a specific protein named stem-loop-binding protein (SLBP). In mouse oocytes, mSLBP is synthesized abundantly during maturation and activates histone translation. In Xenopus, xSLBP is present in stage-VI oocytes, but histone mRNA is protected from premature translation by the oocyte-specific Xenopus SLBP2 (xSLBP2) protein; during maturation xSLBP2 replacement by xSLBP results in histone synthesis. Here, we report the first experimental evidence and characterization of a mammalian SLBP2 ortholog. Bovine bSLBP and bSLBP2 display distinct expression patterns throughout oocyte maturation and pre-implantation embryo development. From the immature oocyte to the morula, bSLBP2 is concentrated in the nucleus, while it is homogeneously distributed throughout the cytoplasm in mature oocytes. A putative SLBP2 gene is conserved in the genome of several mammalian species, and the corresponding transcripts were detected in rat, dog, horse, and pig oocytes. By contrast, a pseudogene is found in mouse, human, and rabbit. Altogether, our data suggest that the availability of histones in oocytes is regulated by an alternative mechanism in bovine and other species as compared to mouse and frog.


Subject(s)
Nuclear Proteins/biosynthesis , Oogenesis , RNA, Messenger/genetics , mRNA Cleavage and Polyadenylation Factors/biosynthesis , Animals , Base Sequence , Binding Sites/genetics , Cattle , Dogs , Embryonic Development , Histones/genetics , Horses , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/cytology , RNA-Binding Proteins , Rabbits , Rats , Sequence Alignment , Swine , Xenopus laevis , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
7.
Haematologica ; 94(9): 1236-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19734416

ABSTRACT

BACKGROUND: A T-cell clone, thought to be the source of eosinophilopoietic cytokines, identified by clonal rearrangement of the T-cell receptor and by the presence of aberrant T-cell immunophenotype in peripheral blood defines lymphocytic variant of hypereosinophilic syndrome (L-HES). DESIGN AND METHODS: Peripheral blood samples from 42 patients who satisfied the diagnostic criteria for HES were studied for T-cell receptor clonal rearrangement by polymerase chain reaction according to BIOMED-2. The T-cell immunophenotype population was assessed in peripheral blood by flow cytometry. The FIP1L1-PDGFRA fusion gene was detected by nested polymerase chain reaction. RESULTS: Forty-two HES patients (18 males and 24 females) with a median age at diagnosis of 56 years (range 17-84) were examined in this study. Their median white blood cell count was 12.9 x 10(9)/L (range 5.3-121), with an absolute eosinophil count of 4.5 x 10(9)/L (range 1.5-99) and a median eosinophilic bone marrow infiltration of 30% (range 11-64). Among the 42 patients, clonal T-cell receptor rearrangements were detected in 18 patients (42.8%). Patients with T-cell receptor clonality included: T-cell receptor beta in 15 patients (35%), T-cell receptor gamma in 9 (21%) and T-cell receptor delta in 9 (21%) patients, respectively. Clonality was detected in all three T-cell receptor loci in 4 cases, in two loci in 7 patients and in one T-cell receptor locus in the remaining 7 patients. The FIP1L1-PDGFRA fusion transcript was absent in all but 2 patients with T-cell receptor clonality. Three patients out of 42 revealed an aberrant T-cell immunophenotype. In some patients, an abnormal CD4:CD8 ratio was demonstrated. CONCLUSIONS: T-cell abnormalities are present at high frequencies in patients with HES.


Subject(s)
Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor/immunology , Hypereosinophilic Syndrome/immunology , Oncogene Proteins, Fusion/immunology , Receptor, Platelet-Derived Growth Factor alpha/immunology , T-Lymphocytes/immunology , mRNA Cleavage and Polyadenylation Factors/immunology , Adolescent , Adult , Aged , Aged, 80 and over , CD4-CD8 Ratio , Female , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor/genetics , Humans , Hypereosinophilic Syndrome/blood , Hypereosinophilic Syndrome/genetics , Male , Middle Aged , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Polymerase Chain Reaction , Prospective Studies , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , mRNA Cleavage and Polyadenylation Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/genetics
8.
Cancer Res ; 67(8): 3759-66, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440089

ABSTRACT

An interstitial deletion on chromosome 4q12 resulting in the formation of the FIP1L1-PDGFRA fusion protein is involved in the pathogenesis of imatinib-sensitive chronic eosinophilic leukemia. The molecular mechanisms underlying the development of disease are largely undefined. Human CD34(+) hematopoietic progenitor cells were used to investigate the role of FIP1L1-PDGFRA in modulating lineage development. FIP1L1-PDGFRA induced both proliferation and differentiation of eosinophils, neutrophils, and erythrocytes in the absence of cytokines, which could be inhibited by imatinib. Whereas expression of FIP1L1-PDGFRA in hematopoietic stem cells and common myeloid progenitors induced the formation of multiple myeloid lineages, expression in granulocyte-macrophage progenitors induced only the development of eosinophils, neutrophils, and myeloblasts. Deletion of amino acids 30 to 233 in the FIP1L1 gene [FIP1L1(1-29)-PDGFRA] gave rise to an intermediate phenotype, exhibiting a dramatic reduction in the number of erythrocytes. FIP1L1-PDGFRA and FIP1L1(1-29)-PDGFRA both induced the activation of p38 and extracellular signal-regulated kinase 1/2 (ERK1/2) in myeloid progenitors, whereas signal transducers and activators of transcription 5 (STAT5) and protein kinase B/c-akt were only activated by FIP1L1-PDGFRA. Dominant-negative STAT5 partially inhibited FIP1L1-PDGFRA-induced colony formation, whereas combined inhibition of phosphatidylinositol-3-kinase and ERK1/2 significantly reversed FIP1L1-PDGFRA-induced colony formation. Taken together, these results suggest that expression of FIP1L1-PDFGRA in human hematopoietic progenitors induce a myeloproliferative phenotype via activation of multiple signaling molecules including phosphatidylinositol-3-kinase, ERK1/2, and STAT5.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hypereosinophilic Syndrome/metabolism , Oncogene Proteins, Fusion/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , mRNA Cleavage and Polyadenylation Factors/biosynthesis , Benzamides , Cell Differentiation/physiology , Cell Growth Processes/physiology , Chronic Disease , Enzyme Activation , Hematopoietic Stem Cells/pathology , Humans , Hypereosinophilic Syndrome/enzymology , Hypereosinophilic Syndrome/pathology , Imatinib Mesylate , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , mRNA Cleavage and Polyadenylation Factors/antagonists & inhibitors , mRNA Cleavage and Polyadenylation Factors/metabolism
9.
Cancer Res ; 66(12): 6336-44, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778211

ABSTRACT

Hypereosinophilic syndrome (HES) has recently been recognized as a clonal leukemic lesion, which is due to a specific oncogenic event that generates hyperactive platelet-derived growth factor receptor-alpha-derived tyrosine kinase fusion proteins. In the present work, the effect of retinoids on the leukemic hypereosinophilia-derived EoL-1 cell line and on primary HES-derived cells has been investigated. We show that all-trans-retinoic acid (ATRA) inhibits eosinophil colony formation of HES-derived bone marrow cells and is a powerful inducer of apoptosis of the EoL-1 cell line. Apoptosis was shown in the nanomolar concentration range by phosphatidylserine externalization, proapoptotic shift of the Bcl-2/Bak ratio, drop in mitochondrial membrane potential, activation of caspases, and cellular morphology. Unlike in other ATRA-sensitive myeloid leukemia models, apoptosis was rapid and was not preceded by terminal cell differentiation. Use of isoform-selective synthetic retinoids indicated that retinoic acid receptor-alpha-dependent signaling is sufficient to induce apoptosis of EoL-1 cells. Our work shows that the scope of ATRA-induced apoptosis of malignancies may be wider within the myeloid lineage than thought previously, that the EoL-1 cell line constitutes a new and unique model for the study of ATRA-induced cell death, and that ATRA may have potential for the management of clonal HES.


Subject(s)
Apoptosis/drug effects , Drug Hypersensitivity/pathology , Eosinophils/drug effects , Hypereosinophilic Syndrome/pathology , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Cell Line, Tumor , Drug Hypersensitivity/etiology , Drug Hypersensitivity/metabolism , Eosinophils/metabolism , Eosinophils/pathology , HL-60 Cells , Humans , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Stem Cells/drug effects , Stem Cells/pathology , mRNA Cleavage and Polyadenylation Factors/biosynthesis
10.
Int Arch Allergy Immunol ; 143 Suppl 1: 28-32, 2007.
Article in English | MEDLINE | ID: mdl-17541273

ABSTRACT

BACKGROUND: EoL-1 cells have a FIP1L1-PDGFRA fusion gene which causes the transformation of eosinophilic precursor cells into leukemia cells. Recently, we suggested that the induction of differentiation of EoL-1 cells into eosinophils by the HDAC inhibitors apicidin and n-butyrate is due to the continuous inhibition of HDACs. However, neither apicidin nor n-butyrate inhibited the expression of FIP1L1-PDGFRA mRNA, although both these inhibitors suppressed cell proliferation. Therefore, in this study, we analyzed whether the levels of FIP1L1-PDGFRalpha protein and phosphorylated-Stat5 involved in the signaling for the proliferation of EoL-1 cells are attenuated by HDAC inhibitors. METHODS: EoL-1 cells were incubated in the presence of apicidin, TSA or n-butyrate. FIP1L1-PDGFRalpha and phosphorylated-Stat5 were detected by Western blotting. RESULTS: Treatment of EoL-1 cells with apicidin at 100 nM or n-butyrate at 500 microM decreased the levels of FIP1L1-PDGFRalpha protein and phosphorylated-Stat5, while that with trichostatin A at 30 nM did not. CONCLUSIONS: The decrease in the level of FIP1L1-PDGFRalpha protein caused by apicidin and n-butyrate might be one of the mechanisms by which EoL-1 cells are induced to differentiate into eosinophils by these HDAC inhibitors.


Subject(s)
Butyrates/pharmacology , Eosinophils/cytology , Histone Deacetylase Inhibitors , Hypereosinophilic Syndrome/pathology , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , Peptides, Cyclic/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/physiology , mRNA Cleavage and Polyadenylation Factors/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Hydroxamic Acids/pharmacology , Hypereosinophilic Syndrome/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/genetics , STAT5 Transcription Factor/metabolism , mRNA Cleavage and Polyadenylation Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/genetics
11.
Int J Hematol ; 83(5): 433-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16787876

ABSTRACT

A 38-year-old Japanese man was referred to our hospital in June 2003 for treatment of acute respiratory failure with severe eosinophilia. Idiopathic hypereosinophilic syndrome had been diagnosed in 1994. However, karyotypic examination of bone marrow cells revealed that chromosomal translocation with t(4;10)(q12;p11) had occurred in 2000, and chronic eosinophilic leukemia was diagnosed. At admission, the patient's respiratory condition was extremely serious, and mechanical support was necessary. Despite treatment with steroid pulse therapy and cytarabine, the blood eosinophil count did not decrease, and the patient's respiratory condition worsened. After obtaining informed consent, we administered imatinib mesylate at a dose of 200 mg/day for 2 days and 100 mg/day for 3 days. The blood eosinophil count decreased dramatically over 5 days, and the patient's condition rapidly improved, such that the patient could be discharged. In this case, we performed molecular analysis using peripheral blood. The FIP1-like 1 (FIP1L1)-platelet-derived growth factor receptor alpha (PDGFRalpha) fusion transcript was found with the reverse transcriptase polymerase chain reaction analysis. In this case, eosinophilia was possibly caused by constitutive activation of tyrosine kinase produced by the FIP1L1-PDGFRalpha fusion transcript.


Subject(s)
Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/enzymology , Oncogene Proteins, Fusion/antagonists & inhibitors , Piperazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , mRNA Cleavage and Polyadenylation Factors/antagonists & inhibitors , Adult , Asian People , Benzamides , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 4/genetics , Chronic Disease , Enzyme Activation/drug effects , Eosinophilia/drug therapy , Eosinophilia/enzymology , Eosinophilia/genetics , Follow-Up Studies , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Hypereosinophilic Syndrome/genetics , Imatinib Mesylate , Leukocyte Count , Male , Oncogene Proteins, Fusion/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Translocation, Genetic/genetics , mRNA Cleavage and Polyadenylation Factors/biosynthesis
12.
Mol Med Rep ; 14(5): 4271-4278, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27633853

ABSTRACT

MicroRNA-181a (miR-181a) is upregulated in osteosarcoma, and its overexpression promotes the proliferation and inhibits the apoptosis of osteosarcoma cells. However, the mechanism of miRĀ­181a as an oncogene remains to be fully elucidated in osteosarcoma. Cleavage factor (CF)Ā Im25 links alternative polyadenylation to glioblastoma tumor suppression, however, its role in osteosarcoma has not been reported. In the present study, it was confirmed that the expression of miRĀ­181a was upregulated in osteosarcoma, and that silencing miRĀ­181a inhibited the proliferation and promoted the apoptosis of osteosarcoma cells. miRNAs are short nonĀ­coding RNAs, which regulate target mRNAs by binding predominantly to the 3'untranslated region (3'UTR), inducing either translational repression or degradation of the target. In the present study, target genes of miRĀ­181a were screened using miRanda, which is a commonly used prediction algorithm. It was found that miRĀ­181a targeted the 3'UTR of CFIm25 mRNA. Subsequent experiments confirmed that miRĀ­181a downregulated the expression of CFIm25 in osteosarcoma cells. Finally, it was demonstrated that the CFIm25 protein was also downregulated in osteosarcoma tissues, and inhibited the proliferation and promoted the apoptosis of the cells. Elucidating the roles of miRĀ­181a and CFIm25 in osteosarcoma not only assists in further understanding the pathogenesis and progression of this disease, but also offers novel targets for effective therapies.


Subject(s)
Cell Proliferation/genetics , MicroRNAs/genetics , Osteosarcoma/genetics , mRNA Cleavage and Polyadenylation Factors/biosynthesis , Apoptosis/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Osteosarcoma/pathology , RNA, Messenger/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
13.
Methods Mol Biol ; 1255: 91-115, 2015.
Article in English | MEDLINE | ID: mdl-25487207

ABSTRACT

In the post-genomic era where gene sequences are available for many organisms, attention has shifted from DNA to the workhorses of the cell-RNA and protein. A number of proteins, as recent studies indicate, seem to possess RNA-binding and RNA cleavage activities. In order to understand the events that comprise RNA processing such as splicing, 3' end processing, and even RNA turnover, well established methods are necessary. Bacterial recombinant proteins afford an invaluable opportunity to produce proteins in an economical and reproducible fashion in order to study these activities. This chapter describes various experimental protocols to begin the elucidation of the many events that surround RNA processing at the 3' end of a transcript.


Subject(s)
Plant Proteins/isolation & purification , Plants/metabolism , Polyadenylation , RNA, Messenger/metabolism , Recombinant Proteins/isolation & purification , mRNA Cleavage and Polyadenylation Factors/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Plant Proteins/biosynthesis , Plant Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Ribonucleases/metabolism , mRNA Cleavage and Polyadenylation Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/genetics
15.
Zhonghua Yi Xue Za Zhi ; 84(18): 1541-4, 2004 Sep 17.
Article in Zh | MEDLINE | ID: mdl-15500716

ABSTRACT

OBJECTIVE: To determine whether FIP1L1-PDGFRA fusion exists in hypereosinophilic syndrome (HES) patients, explore the relationship between FIP1L1-PDGFRA fusion and clinical phenotypes, and observe and reveal the expression of signal transducer and activator of transcription 5 (STAT(5)) in granulocytes of HES and the biological significance thereof. METHODS: Specimens of peripheral blood were collected from 4 HES patients diagnosed based on the criteria of Chusid et al. Total RNA was extracted from granulocytes and cDNA was synthesized by reverse transcription. Nested-PCR was used to amplify the target fusion gene and the positive PCR fragments were sequenced directly. Total protein of the peripheral granulocytes was extracted. Western blotting was used to detect the expression of STAT(5) protein in the granulocyte lysates. RESULTS: FIP1L1-PDGFRA fusion genes were found in 3 of the 4 HES patients. The break points in PDGFRA were all located at exon 12, while in FIP1L1 the break points were highly variable, located at exon 8a, intron 8a, and exon 8 respectively. The patients with FIP1L1-PDGFRA fusion were susceptible to cardiac involvement. The expression of STAT(5) protein was upregulated in FIP1L1-PDGFRA positive HES patients, while STAT(5) protein expression was negative in HES patients without FIP1L1-PDGFRA fusion. CONCLUSION: FIP1L1-PDGFRA fusion has a universal significance for HES. The identification of FIP1L1-PDGFRA rearrangement is a useful molecular mark for HES diagnosis and works as the therapeutic target of imatinib. Furthermore, the activation of STAT(5), a downstream signal of the FIP1L1-PDGFRA fusion, indicates that HES is a malignant clonal disease of the hematopoietic tissue.


Subject(s)
DNA-Binding Proteins/genetics , Hypereosinophilic Syndrome/genetics , Milk Proteins/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Trans-Activators/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , DNA-Binding Proteins/biosynthesis , Gene Rearrangement , Humans , Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/drug therapy , Male , Milk Proteins/biosynthesis , Molecular Sequence Data , Oncogene Proteins, Fusion , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , STAT5 Transcription Factor , Trans-Activators/biosynthesis , mRNA Cleavage and Polyadenylation Factors/biosynthesis
16.
Nucleus ; 5(6): 613-25, 2014.
Article in English | MEDLINE | ID: mdl-25493544

ABSTRACT

The scaffolding protein Symplekin is part of multiple complexes involved in generating and modifying the 3' end of mRNAs, including cleavage-polyadenylation, histone pre-mRNA processing and cytoplasmic polyadenylation. To study these functions in vivo, we examined the localization of Symplekin during development and generated mutations of the Drosophila Symplekin gene. Mutations in Symplekin that reduce Symplekin protein levels alter the efficiency of both poly A(+) and histone mRNA 3' end formation resulting in lethality or sterility. Histone mRNA synthesis takes place at the histone locus body (HLB) and requires a complex composed of Symplekin and several polyadenylation factors that associates with the U7 snRNP. Symplekin is present in the HLB in the early embryo when Cyclin E/Cdk2 is active and histone genes are expressed and is absent from the HLB in cells that have exited the cell cycle. During oogenesis, Symplekin is preferentially localized to HLBs during S-phase in endoreduplicating follicle cells when histone mRNA is synthesized. After the completion of endoreplication, Symplekin accumulates in the cytoplasm, in addition to the nucleoplasm, and localizes to tricellular junctions of the follicle cell epithelium. This localization depends on the RNA binding protein ypsilon schachtel. CPSF-73 and a number of mRNAs are localized at this same site, suggesting that Symplekin participates in cytoplasmic polyadenylation at tricellular junctions.


Subject(s)
Drosophila Proteins/biosynthesis , Embryonic Development , Histones/genetics , Polyadenylation/genetics , mRNA Cleavage and Polyadenylation Factors/biosynthesis , Animals , Cytoplasm/genetics , Drosophila , Drosophila Proteins/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Histones/metabolism , Intranuclear Inclusion Bodies/genetics , Intranuclear Inclusion Bodies/metabolism , Mutation , Nucleoplasmins/genetics , Ribonucleoprotein, U7 Small Nuclear/genetics , S Phase/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
17.
Oncotarget ; 5(16): 6756-69, 2014 Aug 30.
Article in English | MEDLINE | ID: mdl-25216517

ABSTRACT

Glioma stemness has been recognized as the most important reason for glioma relapse and drug resistance. Differentiation of glioma stem cells (GSCs) has been implicated as a novel approach to target recurrent glioma. However, the detailed molecular mechanism involved in the differentiation of GSCs has not yet been elucidated. This study identified CPEB1 as the key modulator that induces the differentiation of GSCs at the post-transcriptional level. Gain and loss of function experiments showed that CPEB1 expression reduced sphere formation ability and the expression of stemness markers such as Nestin and Notch. To elucidate the detailed molecular mechanism underlying the action of CPEB1, we investigated the interacting ribonome of the CPEB1 complex using a Ribonomics approach. CPEB1 specifically suppressed the translation of HES1 and SIRT1 by interacting with a cytoplasmic polyadenylation element. The expression profile of CPEB1 negatively correlated with overall survival in glioma patients. Overexpression of CPEB1 decreased the number of GSCs in an orthotopically implanted glioma animal model. These results suggest that CPEB1-mediated translational control is essential for the differentiation of GSCs and provides novel therapeutic concepts for differentiation therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Glioma/pathology , Homeodomain Proteins/biosynthesis , Neoplastic Stem Cells/pathology , Sirtuin 1/biosynthesis , Transcription Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Differentiation/physiology , Down-Regulation , Female , Glioma/genetics , Glioma/metabolism , HEK293 Cells , Heterografts , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Sirtuin 1/genetics , Transcription Factor HES-1 , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transfection , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
19.
Mol Cell Biol ; 32(21): 4306-22, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22907757

ABSTRACT

Histone mRNAs are rapidly degraded at the end of S phase, and a 26-nucleotide stem-loop in the 3' untranslated region is a key determinant of histone mRNA stability. This sequence is the binding site for stem-loop binding protein (SLBP), which helps to recruit components of the RNA degradation machinery to the histone mRNA 3' end. SLBP is the only protein whose expression is cell cycle regulated during S phase and whose degradation is temporally correlated with histone mRNA degradation. Here we report that chemical inhibition of the prolyl isomerase Pin1 or downregulation of Pin1 by small interfering RNA (siRNA) increases the mRNA stability of all five core histone mRNAs and the stability of SLBP. Pin1 regulates SLBP polyubiquitination via the Ser20/Ser23 phosphodegron in the N terminus. siRNA knockdown of Pin1 results in accumulation of SLBP in the nucleus. We show that Pin1 can act along with protein phosphatase 2A (PP2A) in vitro to dephosphorylate a phosphothreonine in a conserved TPNK sequence in the SLBP RNA binding domain, thereby dissociating SLBP from the histone mRNA hairpin. Our data suggest that Pin1 and PP2A act to coordinate the degradation of SLBP by the ubiquitin proteasome system and the exosome-mediated degradation of the histone mRNA by regulating complex dissociation.


Subject(s)
Nuclear Proteins/metabolism , Peptidylprolyl Isomerase/metabolism , Protein Phosphatase 2/metabolism , RNA Stability , RNA, Messenger/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Down-Regulation , HEK293 Cells , HeLa Cells , Histones , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Nuclear Proteins/biosynthesis , Peptidylprolyl Isomerase/genetics , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/metabolism , Ubiquitination , mRNA Cleavage and Polyadenylation Factors/biosynthesis , mRNA Cleavage and Polyadenylation Factors/genetics
20.
SELECTION OF CITATIONS
SEARCH DETAIL