Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Anticancer Res ; 43(12): 5437-5446, 2023 Dec.
Article En | MEDLINE | ID: mdl-38030167

BACKGROUND/AIM: PHD and RING finger domain-containing protein 1 (PHRF1) ubiquitinates TGIP (TG-interacting protein) and redistributes cPML (cytoplasmic variant of PML) to the cytoplasm to enhance TGF-ß signaling by. It is unclear whether PHRF1 affects invasion and survival when both mutations of the activated oncogene Kras and inactivation of the tumor suppressor p53 are present. MATERIALS AND METHODS: We knockout PHRF1 expression using Crispr-Cas9 editing in HCT116-p53-/- (KrasG13D/p53-/-) cells and analyzed the expression profile in HCT116-p53-/-PHRF1-/- cells. RESULTS: In contrast to lung cancer A549 (KrasG12S/p53wt) cells, the expression of Zeb1, a transcription factor for epidermal-mesenchymal transition (EMT), was not affected in PHRF1-knockout HCT116 p53-/- cells. Instead, SOX4 displayed a significant contribution to the impaired invasion in HCT116-p53-/-PHRF1-/- cells. Mechanistically, the C-terminal SRI domain of PHRF1 was required for both transwell invasion and SOX4 expression. The reintroduction of SOX4 into HCT116-p53-/- PHRF1-/- cells partially restored their invasive capability. CONCLUSION: This study sheds light on the role of PHRF1 in the invasion of colorectal cancer HCT116-p53-/- cells, which harbor the oncogenic KrasG13D mutation and lack p53. These findings provide novel insights regarding the role of PHRF1 in invasion by modulating SOX4 expression in colorectal cancer HCT116-p53-/- cells.


Colorectal Neoplasms , Membrane Proteins , SOXC Transcription Factors , Humans , Cell Line, Tumor , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism
2.
PLoS One ; 18(8): e0285159, 2023.
Article En | MEDLINE | ID: mdl-37540725

PHRF1 is an E3 ligase that promotes TGF-ß signaling by ubiquitinating a homeodomain repressor TG-interacting factor (TGIF). The suppression of PHRF1 activity by PML-RARα facilitates the progression of acute promyelocytic leukemia (APL). PHRF1 also contributes to non-homologous end-joining in response to DNA damage by linking H3K36me3 and NBS1 with DNA repair machinery. However, its role in class switch recombination (CSR) is not well understood. In this study, we report the importance of PHRF1 in IgA switching in CH12F3-2A cells and CD19-Cre mice. Our studies revealed that Crispr-Cas9 mediated PHRF1 knockout and shRNA-silenced CH12F3-2A cells reduced IgA production, as well as decreased the amounts of PARP1, NELF-A, and NELF-D. The introduction of PARP1 could partially restore IgA production in PHRF1 knockout cells. Intriguingly, IgA, as well as IgG1, IgG2a, and IgG3, switchings were not significantly decreased in PHRF1 deficient splenic B lymphocytes isolated from CD19-Cre mice. The levels of PARP1 and NELF-D were not decreased in PHRF1-depleted primary splenic B cells. Overall, our findings suggest that PHRF1 may modulate IgA switching in CH12F3-2A cells.


DNA-Binding Proteins , Immunoglobulin Class Switching , Mice , Animals , DNA-Binding Proteins/genetics , Immunoglobulin Class Switching/genetics , DNA Repair , DNA End-Joining Repair , Immunoglobulin A/genetics
3.
PLoS One ; 16(8): e0256282, 2021.
Article En | MEDLINE | ID: mdl-34407138

Human PUF-A/PUM3 is a RNA and DNA binding protein participating in the nucleolar processing of 7S to 5.8S rRNA. The nucleolar localization of PUF-A redistributes to the nucleoplasm upon the exposure to genotoxic agents in cells. However, little is known regarding the roles of PUF-A in tumor progression. Phosphoprotein database analysis revealed that Y259 phosphorylation of PUF-A is the most prevalent residue modified. Here, we reported the importance of PUF-A's phosphorylation on Y259 in tumorigenesis. PUF-A gene was knocked out by the Crispr/Cas9 method in human cervix epithelial HeLa cells. Loss of PUF-A in HeLa cells resulted in reduced clonogenic and lower transwell invasion capacity. Introduction of PUF-AY259F to PUF-A deficient HeLa cells was unable to restore colony formation. In addition, the unphosphorylated mutant of PUF-A, PUF-AY259F, attenuated PUF-A protein stability. Our results suggest the important role of Y259 phosphorylation of PUF-A in cell proliferation.


Carcinogenesis/metabolism , Minor Histocompatibility Antigens/metabolism , Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Processing, Post-Translational , Tyrosine/metabolism , Atlases as Topic , CRISPR-Cas Systems , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Movement , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Cell Proliferation , Databases, Genetic , Female , Gene Deletion , HeLa Cells , Humans , Minor Histocompatibility Antigens/genetics , Mutation , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Phosphoproteins/genetics , Phosphorylation , Protein Stability , Survival Analysis
4.
PLoS One ; 15(7): e0236876, 2020.
Article En | MEDLINE | ID: mdl-32730336

PHRF1 (PHD and RING finger domain-containing protein 1) suppresses acute promyelocytic leukemia (APL) by promoting TGIF (TG-interacting factor) ubiquitination, while the PML-RARα protein interferes with PHRF1-mediated TGIF breakdown to facilitate APL. Beyond its role in APL tumorigenesis, PHRF1 contributes to non-homologous end-joining by linking H3K36 methylation and Nbs1 upon DNA damage insults. However, little is known regarding its function in tumor invasion. Here we highlight the unreported details of PHRF1 in the invasion of lung cancer cells by modulating the transcriptional level of ZEB1, a prominent regulator involved in epithelial-mesenchymal transition. PHRF1 associated with the phosphorylated C-terminal repeat domain of Rpb1, the large subunit of RNA polymerase II, through its C-terminal Set2 Rpb1 Interacting (SRI) domain. Chromatin immunoprecipitation revealed that PHRF1 bound to the proximal region adjacent to the transcription start site of ZEB1. SRI-deleted PHRF1 neither associated with Rpb1 nor increased ZEB1's expression. Collectively, PHRF1 might take the stage at migration and invasion by modulating the expression of ZEB1.


Cell Movement , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Mice , Neoplasm Invasiveness , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1/genetics
5.
Sci Rep ; 10(1): 10857, 2020 07 02.
Article En | MEDLINE | ID: mdl-32616804

PHRF1 is involved in transforming growth factor ß (TGF-ß) signaling to constrain the formation of acute promyelocytic leukemia (APL) in mouse APL models. PHRF1 also participates in modulating non-homologous end-joining. However, the role of PHRF1 in mammalian dendrite architecture and synaptic plasticity is unclear. Here, we investigated the role of PHRF1 in dendritic formation in the murine hippocampus using Camk2a promoter driven-iCre recombinase to conduct a PHRF1 conditional knockout, namely PHRF1Δ/Δ, in the forebrain region. PHRF1Δ/Δ mice developed normally, but exhibited anxiety-like behaviors and displayed defective spatial memory. Alterations of dendritic complexity in apical and basal dendrites of pyramidal neurons were noticed in PHRF1Δ/Δ mutants. Furthermore, electrical stimulation in the hippocampal CA1 region after the TGF-ß1 treatment showed a reduced synaptic plasticity in PHRF1Δ/Δ mice. Immunoblotting analysis indicated that PHRF1 ablation affected the TGF-ß signaling. Collectively, our results demonstrate that PHRF1 is important for the dendritic architecture and required for spatial memory formation in the hippocampus.


Dendrites/chemistry , Hippocampus/metabolism , Membrane Proteins/physiology , Pyramidal Cells/metabolism , RING Finger Domains/physiology , Spatial Memory/physiology , Transforming Growth Factor beta/metabolism , Animals , Dendrites/physiology , Female , Male , Mice , Mice, Knockout , Neuronal Plasticity , Pyramidal Cells/cytology , Signal Transduction , Transforming Growth Factor beta/genetics
6.
Antioxidants (Basel) ; 8(11)2019 Nov 15.
Article En | MEDLINE | ID: mdl-31731716

Flavonoids are well-known antioxidants and have shown the ability to prevent tumor formation and recurrence. Especially in dietary flavonoids, they have provided convenience and consistence of intake for long-term prevention of tumor formation. Previous reports suggested that S100 calcium-binding protein A7 (S100A7) might activate epithelial-mesenchymal transition (EMT) signaling and promote the metastasis of tumor cells; however, the regulatory signaling was unclear. In this study, we found that S100A7 was highly expressed in cancer cells and could be reduced by luteolin (Lu) and quercetin (Qu) through Src/Stat3 signaling. We found that the protein levels of S100A7, phosphorylated Src (p-Src), and p-Stat3 were increased in A431-III cells. Flavonoids Lu and Qu reduce protein levels of p-Src, p-Stat3 and S100A7 in A431-III cells. Treatment of A431-III cells with Src inhibitor SU6656 and Stat3 inhibitor S3I-201 also reduced the protein levels of S100A7. Transactivation activity of 5'-upstream regions of S100A7 was activated by Stat3 but was reduced by treatment with Lu, Qu, SU6656 and S3I-201. The treatment also reduced the migratory and invasive abilities of A431-III cells. In a further analysis of EMT markers, the protein level of E-cad increased and that of Twist decreased after treatment with the inhibitors and flavonoids. Overexpression of S100A7 decreased the protein level of E-cad and increased the Twist level, whereas knockdown of S100A7 had the opposite effects. Treatment with S3I-201, Lu and Qu, compared to the control, were found to decrease metastasis of tumor cells in zebrafish larvae. These results suggest that Lu and Qu may inhibit Src/Stat3/S100A7 signaling to reduce tumorigenesis of cancer cells.

7.
Sci Rep ; 7: 45646, 2017 04 03.
Article En | MEDLINE | ID: mdl-28368047

Vilse/Arhgap39 is a Rho GTPase activating protein (RhoGAP) and utilizes its WW domain to regulate Rac/Cdc42-dependent morphogenesis in Drosophila and murine hippocampal neurons. However, the function of Vilse in mammalian dendrite architecture and synaptic plasticity remained unclear. In the present study, we aimed to explore the possible role of Vilse in dendritic structure and synaptic function in the brain. Homozygous knockout of Vilse resulted in premature embryonic lethality in mice. Changes in dendritic complexity and spine density were noticed in hippocampal neurons of Camk2a-Cre mediated forebrain-specific Vilse knockout (VilseΔ/Δ) mice. VilseΔ/Δ mice displayed impaired spatial memory in water maze and Y-maze tests. Electrical stimulation in hippocampal CA1 region revealed that the synaptic transmission and plasticity were defected in VilseΔ/Δ mice. Collectively, our results demonstrate that Vilse is essential for embryonic development and required for spatial memory.


CA1 Region, Hippocampal/physiology , Dendrites/physiology , GTPase-Activating Proteins/metabolism , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Dendrites/metabolism , Female , GTPase-Activating Proteins/genetics , Genes, Lethal/genetics , Male , Maze Learning/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/genetics , Pyramidal Cells/metabolism , Spatial Memory/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
8.
Genetics ; 202(1): 77-92, 2016 Jan.
Article En | MEDLINE | ID: mdl-26564157

DNA double-strand breaks (DSBs) represent one of the most threatening lesions to the integrity of genomes. In yeast Saccharomyces cerevisiae, NuA4, a histone acetylation complex, is recruited to DSBs, wherein it acetylates histones H2A and H4, presumably relaxing the chromatin and allowing access to repair proteins. Two subunits of NuA4, Yng2 and Eaf3, can interact in vitro with methylated H3K4 and H3K36 via their plant homeodomain (PHD) and chromodomain. However, the roles of the two domains and how they interact in a combinatorial fashion are still poorly characterized. In this study, we generated mutations in the PHD and chromodomain that disrupt their interaction with methylated H3K4 and H3K36. We demonstrate that the combined mutations in both the PHD and chromodomain impair the NuA4 recruitment, reduce H4K12 acetylation at the DSB site, and confer sensitivity to bleomycin that induces DSBs. In addition, the double mutant cells are defective in DSB repair as judged by Southern blot and exhibit prolonged activation of phospho-S129 of H2A. Cells harboring the H3K4R, H3K4R, K36R, or set1Δ set2Δ mutant that disrupts H3K4 and H3K36 methylation also show very similar phenotypes to the PHD and chromodomain double mutant. Our results suggest that multivalent interactions between the PHD, chromodomain, and methylated H3K4 and H3K36 act in a combinatorial manner to recruit NuA4 and regulate the NuA4 activity at the DSB site.


DNA, Fungal/metabolism , Histone Acetyltransferases/metabolism , Homeodomain Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Acetylation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Binding Sites , Bleomycin/pharmacology , Chromatin/metabolism , DNA Breaks, Double-Stranded , Drug Resistance, Fungal/genetics , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Histones/metabolism , Methylation , Mutation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
9.
Fish Shellfish Immunol ; 38(1): 230-43, 2014 May.
Article En | MEDLINE | ID: mdl-24680831

Induction of interferons (IFNs) produces an innate immune response through activation of the JAK-STAT signaling pathway. Type I IFN signaling activates downstream gene expression through the IFN-stimulated gene factor 3 (ISGF3) complex, while type II IFN (IFN-γ) signaling is mediated through active STAT1 protein. The IFN target gene Mx is involved in the defense against viral infection. However, the mechanism by which Tetraodon (pufferfish) Mx is regulated by IFN signaling has not been identified. In this study, we describe the cloning and expression of Tetraodon STAT1, STAT2, and IFN regulatory factor 9 (IRF9). By combining constitutively-active STAT1 (STAT1-JH1) and STAT2 (STA2-JH1) fusion proteins with IRF9, we demonstrate that a constitutively-active ISGF3 complex increases the transcriptional activity of the Tetraodon Mx promoter via direct binding to two IFN-stimulated response element (ISRE) sites. In addition, a constitutively-active TnIRF9-S2C containing a fusion of the C-terminal region of STAT2 and IRF9 also activated the Mx promoter through binding to the ISRE sites. Furthermore, constitutively-active STAT1-JH1 elevates Mx promoter activity through two IFN gamma-activated sequence (GAS) elements. The Mx promoter is also activated by constitutively-active TnIRF9-S2C and STAT1-JH1 protein, as determined using an in vivo luciferase assay. We conclude that the Tetraodon Mx gene is activated via Type I (IFN-1) and Type II (IFN-γ) signaling. These results provide mechanistic insights into the role of IFN signaling in teleosts, and the in vivo luciferase assay may be suitable as a tool for studying induction and regulation by IFNs in teleost fish.


Gene Expression Regulation/physiology , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Myxovirus Resistance Proteins/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Tetraodontiformes/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Molecular Sequence Data , Myxovirus Resistance Proteins/genetics , Phylogeny , Promoter Regions, Genetic , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , Signal Transduction
10.
Exp Cell Res ; 322(2): 227-35, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24424245

We earlier identified OSBP-related protein 8 (ORP8) as an endoplasmic reticulum/nuclear envelope oxysterol-binding protein implicated in cellular lipid homeostasis, migration, and organization of the microtubule cytoskeleton. Here, a yeast two-hybrid screen identified Homo sapiens sperm associated antigen 5 (SPAG5)/Astrin as interaction partner of ORP8. The putative interaction was further confirmed by pull-down and co-immunoprecipitation assays. ORP8 did not colocalize with kinetochore-associated SPAG5 in mitotic HepG2 or HuH7 cells, but overexpressed ORP8 was capable of recruiting SPAG5 onto endoplasmic reticulum membranes in interphase cells. In our experiments, 25-hydroxycholesterol (25OHC) retarded the HepG2 cell cycle, causing accumulation in G2/M phase; ORP8 overexpression resulted in the same phenotype. Importantly, ORP8 knock-down dramatically inhibited the oxysterol effect on HepG2 cell cycle, suggesting a mediating role of ORP8. Furthermore, knock-down of SPAG5 significantly reduced the effects of both ORP8 overexpression and 25OHC on the cell cycle, placing SPAG5 downstream of the two cell-cycle interfering factors. Taken together, the present results suggest that ORP8 may via SPAG5 mediate oxysterol interference of the HepG2 cell cycle.


Cell Cycle Proteins/metabolism , Cell Cycle/drug effects , Hydroxycholesterols/pharmacology , Receptors, Steroid/metabolism , Blotting, Western , Cell Cycle/physiology , Fluorescent Antibody Technique , Hep G2 Cells , Humans , Immunoprecipitation , Microscopy, Fluorescence , Subcellular Fractions , Two-Hybrid System Techniques
11.
Tumour Biol ; 34(5): 2557-64, 2013 Oct.
Article En | MEDLINE | ID: mdl-23625657

hPuf-A is a member of RNA-binding PUF family that regulates mRNA translation. Redistribution of hPuf-A from the nucleolus to the nucleoplasm upon genotoxic stress modulates the poly(ADP-ribosyl)ation activity of PARP-1. Here, we report a novel function of hPuf-A involved in promoting breast cancer progression. Immunohistochemical studies showed higher expression levels of hPuf-A in stage I, II, III, and IV breast cancer specimens in contrast with those of hPuf-A in ductal carcinoma in situ. The presence of hPuf-A is highly associated with colony formation capacities in breast cancer T47D and MDA-MB-231 cells. Xenograft growth of hPuf-A-silenced and hPuf-A overexpressing MDA-MB-231 cells in nude mice was substantially in concert with colony formation capacities. This promoting effect of hPuf-A in tumorigenesis might be correlated with the regulation of its associated mRNAs, such as RbAp48 and DDX3. Collectively, hPuf-A may have diagnostic values in breast cancer progression.


Breast Neoplasms/metabolism , Carcinogenesis/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Lung Neoplasms/metabolism , RNA-Binding Proteins/genetics , Animals , Breast Neoplasms/pathology , Carcinogenesis/pathology , Carcinoma, Intraductal, Noninfiltrating/secondary , Cell Line, Tumor , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Minor Histocompatibility Antigens , Neoplasm Staging , Neoplasm Transplantation , RNA-Binding Proteins/metabolism , Tumor Burden , Up-Regulation
12.
Mol Biol Rep ; 40(4): 2867-77, 2013 Apr.
Article En | MEDLINE | ID: mdl-23212617

Lipopolysaccharide (LPS) treatment causes the marked changes of gene expression in macrophages. Tristetraprolin (TTP), which is an mRNA-destabilizing protein that negatively regulates the expression of pro-inflammatory mediators, is induced by LPS. To delineate the molecular mechanism of LPS-stimulated TTP expression, several inhibitors blocking different signaling pathways were used initially. We observed that inhibitors of the NF-κB signaling pathway could down-regulate the TTP expression during LPS-induction. Consistently, TTP expression was increased upon recombinant TNFα stimulation which activates NF-κB signaling. The 5' regulatory region of zfp36 gene spanning from -2 k to +50 was isolated, which contained a putative NF-κB-binding site located in -1859 to -1850. Analysis of luciferase reporter activity driven by a serial 5'-deletion of TTP promoter showed that NF-κB inhibitor-mediated suppression of LPS or TNFα induced activity was through the predicted κB-binding sites. When the NF-κB-binding site was mutated, the TTP promoter decreased its response to the ectopic expression of NF-κB. Physical interaction analysis including oligonucleotides competition, gel shift and chromatin immunoprecipitation assays demonstrated that NF-κB activated by LPS or TNFα bound to the TTP promoter specifically. These results suggested that during LPS stimulation, NF-κB signaling were activated to regulate the transcription of TTP mRNA.


Macrophages/metabolism , NF-kappa B/genetics , Transcription, Genetic , Tristetraprolin/genetics , Animals , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/toxicity , Macrophages/drug effects , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , RNA, Messenger/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics
13.
Int J Biol Sci ; 8(5): 606-19, 2012.
Article En | MEDLINE | ID: mdl-22553461

The Tristetraprolin (TTP) protein family includes four mammalian members (TTP, TIS11b, TIS11d, and ZFP36L3), but only one in Drosophila melanogaster (DTIS11). These proteins bind target mRNAs with AU-rich elements (AREs) via two C3H zinc finger domains and destabilize the mRNAs. We found that overexpression of mouse TIS11b or DTIS11 in the Drosophila retina dramatically reduced eye size, similar to the phenotype of eyes absent (eya) mutants. The eya transcript is one of many ARE-containing mRNAs in Drosophila. We showed that TIS11b reduced levels of eya mRNA in vivo. In addition, overexpression of Eya rescued the TIS11b overexpression phenotype. RNA pull-down and luciferase reporter analyses demonstrated that the DTIS11 RNA-binding domain is required for DTIS11 to bind the eya 3' UTR and reduce levels of eya mRNA. Moreover, ectopic expression of DTIS11 in Drosophila S2 cells decreased levels of eya mRNA and reduced cell viability. Consistent with these results, TTP proteins overexpressed in MCF7 human breast cancer cells were associated with eya homologue 2 (EYA2) mRNA, and caused a decrease in EYA2 mRNA stability and cell viability. Our results suggest that eya mRNA is a target of TTP proteins, and that downregulation of EYA by TTP may lead to reduced cell viability in Drosophila and human cells.


Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye Proteins/genetics , RNA, Messenger/metabolism , Tristetraprolin/metabolism , 3' Untranslated Regions/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Cell Survival , Down-Regulation , Drosophila melanogaster , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Nuclear Proteins/genetics , Protein Tyrosine Phosphatases/genetics , RNA Stability/genetics , RNA-Binding Proteins , Sequence Alignment , Tristetraprolin/genetics
14.
PLoS One ; 7(3): e33538, 2012.
Article En | MEDLINE | ID: mdl-22448250

Human p29 is a putative component of spliceosomes, but its role in pre-mRNA is elusive. By siRNA knockdown and stable overexpression, we demonstrated that human p29 is involved in DNA damage response and Fanconi anemia pathway in cultured cells. In this study, we generated p29 knockout mice (mp29(GT/GT)) using the mp29 gene trap embryonic stem cells to study the role of mp29 in DNA damage response in vivo. Interruption of mp29 at both alleles resulted in embryonic lethality. Embryonic abnormality occurred as early as E6.5 in mp29(GT/GT) mice accompanied with decreased mRNA levels of α-tubulin and Chk1. The reduction of α-tubulin and Chk1 mRNAs is likely due to an impaired post-transcriptional event. An aberrant G2/M checkpoint was found in mp29 gene trap embryos when exposed to aphidicolin and UV light. This embryonic lethality was rescued by crossing with mp29 transgenic mice. Additionally, the knockdown of zfp29 in zebrafish resulted in embryonic death at 72 hours of development postfertilization (hpf). A lower level of acetylated α-tubulin was also observed in zfp29 morphants. Together, these results illustrate an indispensable role of mp29 in DNA checkpoint response during embryonic development.


Cell Cycle Checkpoints , Embryo Loss , Embryo, Mammalian/pathology , Embryo, Nonmammalian/pathology , Nuclear Proteins/physiology , Animals , Blotting, Western , Cell Cycle Proteins , Checkpoint Kinase 1 , Embryonic Stem Cells/metabolism , Female , Fluorescent Antibody Technique , Genetic Complementation Test , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/antagonists & inhibitors , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Tubulin/genetics , Tubulin/metabolism , Ultraviolet Rays , Zebrafish/embryology , Zebrafish/genetics
15.
Cancer Res ; 71(3): 1126-34, 2011 Feb 01.
Article En | MEDLINE | ID: mdl-21266351

Human hPuf-A/KIAA0020 was first identified as a new minor histocompatibility antigen in 2001. Its zebrafish orthologue contains six Pumilio-homology RNA-binding domains and has been shown to participate in the development of eyes and primordial germ cells, but the cellular function of hPuf-A remains unclear. In this report, we showed that hPuf-A predominantly localized in the nucleoli with minor punctate signals in the nucleoplasm. The nucleolar localization of hPuf-A would redistribute to the nucleoplasm after the treatment of RNA polymerase inhibitors (actinomycin D and 5,6-dichlorobenzimidazole riboside) and topoisomerase inhibitors [camptothecin (CPT) and etoposide]. Interestingly, knockdown of hPuf-A sensitized cells to CPT and UV treatment and cells constitutively overexpressing hPuf-A became more resistant to genotoxic exposure. Affinity gel pull-down coupled with mass spectrometric analysis identified PARP-1 as one of the hPuf-A interacting proteins. hPuf-A specifically interacts with the catalytic domain of PARP-1 and inhibits poly(ADP-ribosyl)ation of PARP-1 in vitro. Depletion of hPuf-A increased the cleaved PARP-1 and overexpression of hPuf-A lessened PARP-1 cleavage when cells were exposed to CPT and UV light. Collectively, hPuf-A may regulate cellular response to genotoxic stress by inhibiting PARP-1 activity and thus preventing PARP-1 degradation by caspase-3.


DNA Damage , Poly(ADP-ribose) Polymerases/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Caspase 3/metabolism , Cell Line, Tumor , Cell Nucleolus/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Minor Histocompatibility Antigens , Molecular Sequence Data , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Sequence Alignment , Transfection
16.
Carcinogenesis ; 30(10): 1710-6, 2009 Oct.
Article En | MEDLINE | ID: mdl-19748926

Human p29 is a chromatin-associated protein and the silencing of p29 expression increases cell population in G(1) phase and decreases phosphorylation levels of Chk1 and Chk2 in response to UV treatment. To further characterize the function of p29, U2OS and Fanconi anemia complementation group G (FA-G) cells with constitutive p29 expression have been established. Analyses of these cells identified increased phosphorylation levels of Chk1 and Chk2, which were accompanied by elevated amounts of chromatin-associated Mre11-Rad50-Nbs1 complex and ATR-IP. Monoubiquitination of the FA ID complex was restored in p29 stably expressing FA-G cells. Moreover, lower tumor incidence was observed in mp29 transgenic mice after UV irradiation. These results suggest the involvement of p29 in the DNA damage responses and Fanconi anemia pathway.


Fanconi Anemia/physiopathology , Animals , Apoptosis , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Fanconi Anemia Complementation Group D2 Protein/metabolism , HeLa Cells , Humans , Mice , Mice, Transgenic , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Ubiquitin/metabolism , Ultraviolet Rays
17.
J Biol Chem ; 283(4): 2454-64, 2008 Jan 25.
Article En | MEDLINE | ID: mdl-18055457

Emerging evidence shows that glycogen synthase kinase 3beta (GSK3beta) is involved in mitotic division and that inhibiting of GSK3beta kinase activity causes defects in spindle microtubule length and chromosome alignment. However, the purpose of GSK3beta involvement in spindle microtubule assembly and accurate chromosome segregation remains obscure. Here, we report that GSK3beta interacts with the spindle-associated protein Astrin both in vitro and in vivo. Additionally, Astrin acts as a substrate for GSK3beta and is phosphorylated at Thr-111, Thr-937 ((S/T)P motif) and Ser-974/Thr-978 ((S/T)XXX(S/T)-p motif; p is a phosphorylatable residue). Inhibition of GSK3beta impairs spindle and kinetochore accumulation of Astrin and spindle formation at mitosis, suggesting that Astrin association with the spindle microtubule and kinetochore may be dependent on phosphorylation by GSK3beta. Conversely, depletion of Astrin by small interfering RNA has no detectable influence on the localization of GSK3beta. Interestingly, in vitro assays demonstrated that Astrin enhances GSK3beta-mediated phosphorylation of other substrates. Moreover, we showed that coexpression of Astrin and GSK3beta differentially increases GSK3beta-mediated Tau phosphorylation on an unprimed site. Collectively, these data indicate that GSK3beta interacts with and phosphorylates the spindle-associated protein Astrin, resulting in targeting Astrin to the spindle microtubules and kinetochores. In turn, the GSK3beta-Astrin complex may also facilitate further physiological and pathological phosphorylation.


Cell Cycle Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Kinetochores/metabolism , Mitosis/physiology , Spindle Apparatus/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Microtubules/genetics , Microtubules/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Spindle Apparatus/genetics , tau Proteins/genetics , tau Proteins/metabolism
18.
Blood ; 110(13): 4526-34, 2007 Dec 15.
Article En | MEDLINE | ID: mdl-17855628

The histo-blood group i and I antigens have been characterized as straight and branched repeats of N-acetyllactosamine, respectively, and the conversion of the straight-chain i to the branched-chain I structure on red cells is regulated to occur after birth. It has been demonstrated that the human I locus expresses 3 IGnT transcripts, IGnTA, IGnTB, and IGnTC, and that the last of these is responsible for the I branching formation on red cells. In the present investigation, the K-562 cell line was used as a model to show that the i-to-I transition in erythroid differentiation is determined by the transcription factor CCAAT/enhancer binding protein alpha (C/EBPalpha), which enhances transcription of the IGnTC gene, consequently leading to formation of the I antigen. Further investigation suggested that C/EBPalpha IGnTC-activation activity is modulated at a posttranslational level, and that the phosphorylation status of C/EBPalpha may have a crucial effect. Results from studies using adult and cord erythropoietic cells agreed with those derived using the K-562 cell model, with lentiviral expression of C/EBPalpha in CD34(+) hemopoietic cells demonstrating the determining role of C/EBPalpha in the induction of the IGnTC gene as well as in I antigen expression.


CCAAT-Enhancer-Binding Protein-alpha/physiology , Cell Differentiation , Erythrocytes/cytology , I Blood-Group System/metabolism , N-Acetylglucosaminyltransferases/genetics , Adult , Fetal Blood , Gene Expression Regulation, Enzymologic , Hematopoietic Stem Cells , Humans , K562 Cells , N-Acetylglucosaminyltransferases/metabolism , Phosphorylation
19.
Exp Cell Res ; 313(8): 1710-21, 2007 May 01.
Article En | MEDLINE | ID: mdl-17383637

Human Ninein (hNinein) is implicated in centrosomal microtubule nucleation and microtubule anchoring in interphase cells and may act as a scaffold protein, but its direct interaction partners remain unexplored in the centrosome. In this report, we show clearly that a spindle-associated protein, Astrin, interacts and co-localizes with hNinein at the centrosome during the S and G2 phases, and this complex may dissociate in the M phase. We also demonstrate that the truncated forms of hNinein, which could interfere with gamma-tubulin and function as dominant-negative mutants, are able to affect Astrin localization to the centrosome. Moreover, siRNA-mediated knockdown of hNinein in HeLa cells causes Astrin to fail to target to the centrosome, whereas hNinein can localize at the centrosome in the absence of Astrin. In addition, reduction in hNinein protein levels causes mislocalization of Astrin with the spindle apparatus and results in the formation of an aberrant mitotic spindle. Collectively, these data suggest that hNinein is required for targeting Astrin to the centrosome during the S and G2 phases. We therefore propose a model wherein hNinein regulates the dynamic movement of Astrin throughout the cell cycle and this interaction, in turn, is required for maintenance of centrosome/spindle pole integrity.


Cell Cycle Proteins/metabolism , Centrosome/physiology , Cytoskeletal Proteins/metabolism , G2 Phase , Nuclear Proteins/metabolism , S Phase , Spindle Apparatus/metabolism , Cell Cycle Proteins/genetics , Cell Line , Cytoskeletal Proteins/genetics , Humans , Models, Biological , Nuclear Proteins/genetics , Protein Binding , Tubulin/metabolism , Two-Hybrid System Techniques
20.
Cancer Res ; 66(17): 8484-91, 2006 Sep 01.
Article En | MEDLINE | ID: mdl-16951160

Human p29 is a newly identified nuclear protein whose function is largely undetermined. We found that p29 associated with chromatin, interacted with MCM3, and localized with proliferating cell nuclear antigen foci in the S phase. Silencing of p29 using small interfering RNA duplexes reduced DNA synthesis and increased the expression of p107, a member of the RB family, and of cyclin-dependent kinase inhibitor p21, accompanied with a decreased expression of DNA polymerase alpha. Lethal events consisting of premature chromatin condensation with a reduced Chk1 phosphorylation were observed in p29-depleted cells in response to UV irradiation. Intriguingly, the phosphorylation of ataxia telangectasia-mutated kinases at S1981 was suppressed in p29-depleted HeLa cells with UV irradiation, but not in hydroxyurea- and ionizing radiation-treated cells. Taken together, these results reveal a novel function of p29 in the regulation of DNA replication checkpoint responses.


DNA Damage , DNA/genetics , Gene Silencing , Nuclear Proteins/genetics , Ultraviolet Rays , Apoptosis/radiation effects , DNA Replication/radiation effects , HeLa Cells , Humans , Mitosis/radiation effects , Nuclear Proteins/deficiency , RNA, Small Interfering/genetics , RNA, Small Interfering/radiation effects , RNA-Binding Proteins
...