Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Biol Chem ; 299(4): 103041, 2023 04.
Article in English | MEDLINE | ID: mdl-36803961

ABSTRACT

The U2AF Homology Motif Kinase 1 (UHMK1) is the only kinase that contains the U2AF homology motif, a common protein interaction domain among splicing factors. Through this motif, UHMK1 interacts with the splicing factors SF1 and SF3B1, known to participate in the 3' splice site recognition during the early steps of spliceosome assembly. Although UHMK1 phosphorylates these splicing factors in vitro, the involvement of UHMK1 in RNA processing has not previously been demonstrated. Here, we identify novel putative substrates of this kinase and evaluate UHMK1 contribution to overall gene expression and splicing, by integrating global phosphoproteomics, RNA-seq, and bioinformatics approaches. Upon UHMK1 modulation, 163 unique phosphosites were differentially phosphorylated in 117 proteins, of which 106 are novel potential substrates of this kinase. Gene Ontology analysis showed enrichment of terms previously associated with UHMK1 function, such as mRNA splicing, cell cycle, cell division, and microtubule organization. The majority of the annotated RNA-related proteins are components of the spliceosome but are also involved in several steps of gene expression. Comprehensive analysis of splicing showed that UHMK1 affected over 270 alternative splicing events. Moreover, splicing reporter assay further supported UHMK1 function on splicing. Overall, RNA-seq data demonstrated that UHMK1 knockdown had a minor impact on transcript expression and pointed to UHMK1 function in epithelial-mesenchymal transition. Functional assays demonstrated that UHMK1 modulation affects proliferation, colony formation, and migration. Taken together, our data implicate UHMK1 as a splicing regulatory kinase, connecting protein regulation through phosphorylation and gene expression in key cellular processes.


Subject(s)
Protein Serine-Threonine Kinases , RNA Splicing , Alternative Splicing , RNA Splicing Factors/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Splicing Factor U2AF/chemistry , Transcription Factors/metabolism , Epithelial-Mesenchymal Transition , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
2.
Nature ; 613(7945): 759-766, 2023 01.
Article in English | MEDLINE | ID: mdl-36631611

ABSTRACT

Protein phosphorylation is one of the most widespread post-translational modifications in biology1,2. With advances in mass-spectrometry-based phosphoproteomics, 90,000 sites of serine and threonine phosphorylation have so far been identified, and several thousand have been associated with human diseases and biological processes3,4. For the vast majority of phosphorylation events, it is not yet known which of the more than 300 protein serine/threonine (Ser/Thr) kinases encoded in the human genome are responsible3. Here we used synthetic peptide libraries to profile the substrate sequence specificity of 303 Ser/Thr kinases, comprising more than 84% of those predicted to be active in humans. Viewed in its entirety, the substrate specificity of the kinome was substantially more diverse than expected and was driven extensively by negative selectivity. We used our kinome-wide dataset to computationally annotate and identify the kinases capable of phosphorylating every reported phosphorylation site in the human Ser/Thr phosphoproteome. For the small minority of phosphosites for which the putative protein kinases involved have been previously reported, our predictions were in excellent agreement. When this approach was applied to examine the signalling response of tissues and cell lines to hormones, growth factors, targeted inhibitors and environmental or genetic perturbations, it revealed unexpected insights into pathway complexity and compensation. Overall, these studies reveal the intrinsic substrate specificity of the human Ser/Thr kinome, illuminate cellular signalling responses and provide a resource to link phosphorylation events to biological pathways.


Subject(s)
Phosphoproteins , Protein Serine-Threonine Kinases , Proteome , Serine , Threonine , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Substrate Specificity , Threonine/metabolism , Proteome/chemistry , Proteome/metabolism , Datasets as Topic , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Cell Line , Phosphoserine/metabolism , Phosphothreonine/metabolism
3.
FEBS J ; 289(3): 682-698, 2022 02.
Article in English | MEDLINE | ID: mdl-34520118

ABSTRACT

Splicing factor mutations are frequent in myeloid neoplasms, blood cancers, and solid tumors. Cancer cells harboring these mutations present a particular vulnerability to drugs that target splicing factors such as SF3b155 or CAPERα. Still, the arsenal of chemical probes that target the spliceosome is very limited. U2AF homology motifs (UHMs) are common protein interaction domains among splicing factors. They present a hydrophobic pocket ideally suited to anchor small molecules with the aim to inhibit protein-protein interaction. Here, we combined a virtual screening of a small molecules database and an in vitro competition assay and identified a small molecule, we named UHMCP1 that prevents the SF3b155/U2AF65 interaction. NMR analyses and molecular dynamics simulations confirmed the binding of this molecule in the hydrophobic pocket of the U2AF65 UHM domain. We further provide evidence that UHMCP1 impacts RNA splicing and cell viability and is therefore an interesting novel compound targeting an UHM domain with potential anticancer properties.


Subject(s)
Neoplasms/genetics , Phosphoproteins/genetics , RNA Splicing Factors/genetics , RNA-Binding Proteins/genetics , Splicing Factor U2AF/genetics , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Magnetic Resonance Spectroscopy , Mass Screening , Molecular Dynamics Simulation , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs/genetics , RNA Splicing/drug effects , Small Molecule Libraries/chemistry , Spliceosomes/drug effects , User-Computer Interface
4.
Nucleic Acids Res ; 49(17): 10061-10081, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34469566

ABSTRACT

In the absence of the scanning ribosomes that unwind mRNA coding sequences and 5'UTRs, mRNAs are likely to form secondary structures and intermolecular bridges. Intermolecular base pairing of non polysomal mRNAs is involved in stress granule (SG) assembly when the pool of mRNAs freed from ribosomes increases during cellular stress. Here, we unravel the structural mechanisms by which a major partner of dormant mRNAs, YB-1 (YBX1), unwinds mRNA secondary structures without ATP consumption by using its conserved cold-shock domain to destabilize RNA stem/loops and its unstructured C-terminal domain to secure RNA unwinding. At endogenous levels, YB-1 facilitates SG disassembly during arsenite stress recovery. In addition, overexpression of wild-type YB-1 and to a lesser extent unwinding-defective mutants inhibit SG assembly in HeLa cells. Through its mRNA-unwinding activity, YB-1 may thus inhibit SG assembly in cancer cells and package dormant mRNA in an unfolded state, thus preparing mRNAs for translation initiation.


Subject(s)
Inverted Repeat Sequences/genetics , Peptide Chain Initiation, Translational/genetics , RNA, Messenger/genetics , Stress Granules/metabolism , Y-Box-Binding Protein 1/metabolism , Adenosine Triphosphate/metabolism , Arsenites/toxicity , Base Pairing/genetics , Cell Line, Tumor , HeLa Cells , Humans , Ribosomes/metabolism
5.
Commun Biol ; 4(1): 359, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742080

ABSTRACT

The RNA-binding protein Lin28 (Lin28a) is an important pluripotency factor that reprograms translation and promotes cancer progression. Although Lin28 blocks let-7 microRNA maturation, Lin28 also binds to a large set of cytoplasmic mRNAs directly. However, how Lin28 regulates the processing of many mRNAs to reprogram global translation remains unknown. We show here, using a structural and cellular approach, a mixing of Lin28 with YB-1 (YBX1) in the presence of mRNA owing to their cold-shock domain, a conserved ß-barrel structure that binds to ssRNA cooperatively. In contrast, the other RNA binding-proteins without cold-shock domains tested, HuR, G3BP-1, FUS and LARP-6, did not mix with YB-1. Given that YB-1 is the core component of dormant mRNPs, a model in which Lin28 gains access to mRNPs through its co-association with YB-1 to mRNA may provide a means for Lin28 to reprogram translation. We anticipate that the translational plasticity provided by mRNPs may contribute to Lin28 functions in development and adaptation of cancer cells to an adverse environment.


Subject(s)
Cytoplasmic Granules/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Uterine Cervical Neoplasms/metabolism , Y-Box-Binding Protein 1/metabolism , Binding Sites , Cell Proliferation , Cytoplasmic Granules/genetics , Cytoplasmic Granules/pathology , Female , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Domains and Motifs , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Y-Box-Binding Protein 1/genetics
6.
EMBO Rep ; 20(8): e47604, 2019 08.
Article in English | MEDLINE | ID: mdl-31271494

ABSTRACT

The essential splicing factor U2AF65 is known to help anchoring U2 snRNP at the branch site. Its C-terminal UHM domain interacts with ULM motifs of SF3b155, an U2 snRNP protein. Here, we report a cooperative binding of U2AF65 and the related protein CAPERα to the multi-ULM domain of SF3b155. In addition, we show that the RS domain of U2AF65 drives a liquid-liquid phase separation that is amplified by intronic RNA with repeated pyrimidine tracts. In cells, knockdown of either U2AF65 or CAPERα improves the inclusion of cassette exons that are preceded by such repeated pyrimidine-rich motifs. These results support a model in which liquid-like assemblies of U2AF65 and CAPERα on repetitive pyrimidine-rich RNA sequences are driven by their RS domains, and facilitate the recruitment of the multi-ULM domain of SF3b155. We anticipate that posttranslational modifications and proteins recruited in dynamical U2AF65 and CAPERα condensates may further contribute to the complex mechanisms leading to specific splice site choice that occurs in cells.


Subject(s)
Alternative Splicing , Phosphoproteins/genetics , RNA Splicing Factors/genetics , RNA-Binding Proteins/genetics , Spliceosomes/genetics , Splicing Factor U2AF/genetics , Cloning, Molecular , Computational Biology , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression , Gene Expression Profiling , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HeLa Cells , Humans , Nucleotide Motifs , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism , Spliceosomes/metabolism , Splicing Factor U2AF/antagonists & inhibitors , Splicing Factor U2AF/metabolism
7.
Nucleic Acids Res ; 47(6): 3127-3141, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30605522

ABSTRACT

The structural rearrangements accompanying mRNA during translation in mammalian cells remain poorly understood. Here, we discovered that YB-1 (YBX1), a major partner of mRNAs in the cytoplasm, forms a linear nucleoprotein filament with mRNA, when part of the YB-1 unstructured C-terminus has been truncated. YB-1 possesses a cold-shock domain (CSD), a remnant of bacterial cold shock proteins that have the ability to stimulate translation under the low temperatures through an RNA chaperone activity. The structure of the nucleoprotein filament indicates that the CSD of YB-1 preserved its chaperone activity also in eukaryotes and shows that mRNA is channeled between consecutive CSDs. The energy benefit needed for the formation of stable nucleoprotein filament relies on an electrostatic zipper mediated by positively charged amino acid residues in the YB-1 C-terminus. Thus, YB-1 displays a structural plasticity to unfold structured mRNAs into extended linear filaments. We anticipate that our findings will shed the light on the scanning of mRNAs by ribosomes during the initiation and elongation steps of mRNA translation.


Subject(s)
Nucleoproteins/chemistry , RNA-Binding Proteins/ultrastructure , Y-Box-Binding Protein 1/ultrastructure , Amino Acid Sequence/genetics , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Escherichia coli/genetics , Humans , Nucleoproteins/genetics , Nucleoproteins/ultrastructure , Protein Binding/genetics , Protein Biosynthesis/genetics , Protein Folding , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribosomes/chemistry , Ribosomes/genetics , Y-Box-Binding Protein 1/chemistry , Y-Box-Binding Protein 1/genetics
8.
J Cell Sci ; 131(11)2018 06 11.
Article in English | MEDLINE | ID: mdl-29728455

ABSTRACT

Liquid-liquid phase separation enables compartmentalization of biomolecules in cells, notably RNA and associated proteins in the nucleus. Besides having critical functions in RNA processing, there is a major interest in deciphering the molecular mechanisms of compartmentalization orchestrated by RNA-binding proteins such as TDP-43 (also known as TARDBP) and FUS because of their link to neuron diseases. However, tools for probing compartmentalization in cells are lacking. Here, we developed a method to analyze the mixing and demixing of two different phases in a cellular context. The principle is the following: RNA-binding proteins are confined on microtubules and quantitative parameters defining their spatial segregation are measured along the microtubule network. Through this approach, we found that four mRNA-binding proteins, HuR (also known as ELAVL1), G3BP1, TDP-43 and FUS form mRNA-rich liquid-like compartments on microtubules. TDP-43 is partly miscible with FUS but immiscible with either HuR or G3BP1. We also demonstrate that mRNA is essential to capture the mixing and demixing behavior of mRNA-binding proteins in cells. Taken together, we show that microtubules can be used as platforms to understand the mechanisms underlying liquid-liquid phase separation and their deregulation in human diseases.


Subject(s)
Cells/metabolism , Microscopy, Fluorescence/methods , Microtubules/metabolism , RNA-Binding Proteins/metabolism , Cells/chemistry , Cytoplasmic Granules/chemistry , Cytoplasmic Granules/metabolism , HeLa Cells , Humans , Microtubules/chemistry , Protein Binding , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry
9.
Biophys J ; 111(12): 2570-2586, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-28002734

ABSTRACT

Splicing factor 1 (SF1) recognizes 3' splice sites of the major class of introns as a ternary complex with U2AF65 and U2AF35 splicing factors. A conserved SPSP motif in a coiled-coil domain of SF1 is highly phosphorylated in proliferating human cells and is required for cell proliferation. The UHM kinase 1 (UHMK1), also called KIS, double-phosphorylates both serines of this SF1 motif. Here, we use isothermal titration calorimetry to demonstrate that UHMK1 phosphorylation of the SF1 SPSP motif slightly enhances specific binding of phospho-SF1 to its cognate U2AF65 protein partner. Conversely, quantitative fluorescence anisotropy RNA binding assays and isothermal titration calorimetry experiments establish that double-SPSP phosphorylation reduces phospho-SF1 and phospho-SF1-U2AF65 binding affinities for either optimal or suboptimal splice-site RNAs. Domain-substitution and mutagenesis experiments further demonstrate that arginines surrounding the phosphorylated SF1 loop are required for cooperative 3' splice site recognition by the SF1-U2AF65 complex (where cooperativity is defined as a nonadditive increase in RNA binding by the protein complex relative to the individual proteins). In the context of local, intracellular concentrations, the subtle effects of SF1 phosphorylation on its associations with U2AF65 and splice-site RNAs are likely to influence pre-mRNA splicing. However, considering roles for SF1 in pre-mRNA retention and transcriptional repression, as well as in splicing, future comprehensive investigations are needed to fully explain the requirement for SF1 SPSP phosphorylation in proliferating human cells.


Subject(s)
RNA Splice Sites , RNA Splicing Factors/chemistry , RNA Splicing Factors/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , Base Sequence , Humans , Models, Molecular , Mutation , Phosphorylation , Protein Domains , RNA Splice Sites/genetics , RNA Splicing Factors/genetics
10.
J Biol Chem ; 289(25): 17325-37, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24795046

ABSTRACT

U2AF homology motifs (UHMs) mediate protein-protein interactions with U2AF ligand motifs (ULMs) of pre-mRNA splicing factors. The UHM-containing alternative splicing factor CAPERα regulates splicing of tumor-promoting VEGF isoforms, yet the molecular target of the CAPERα UHM is unknown. Here we present structures of the CAPERα UHM bound to a representative SF3b155 ULM at 1.7 Å resolution and, for comparison, in the absence of ligand at 2.2 Å resolution. The prototypical UHM/ULM interactions authenticate CAPERα as a bona fide member of the UHM family of proteins. We identify SF3b155 as the relevant ULM-containing partner of full-length CAPERα in human cell extracts. Isothermal titration calorimetry comparisons of the purified CAPERα UHM binding known ULM-containing proteins demonstrate that high affinity interactions depend on the presence of an intact, intrinsically unstructured SF3b155 domain containing seven ULM-like motifs. The interplay among bound CAPERα molecules gives rise to the appearance of two high affinity sites in the SF3b155 ULM-containing domain. In conjunction with the previously identified, UHM/ULM-mediated complexes of U2AF(65) and SPF45 with SF3b155, this work demonstrates the capacity of SF3b155 to offer a platform for coordinated recruitment of UHM-containing splicing factors.


Subject(s)
Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , Amino Acid Motifs , HEK293 Cells , Humans , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA Splicing Factors , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Ribonucleoprotein, U2 Small Nuclear/chemistry , Ribonucleoprotein, U2 Small Nuclear/genetics
11.
Biochim Biophys Acta ; 1833(5): 1269-79, 2013 May.
Article in English | MEDLINE | ID: mdl-23419774

ABSTRACT

The CATS protein (also known as FAM64A and RCS1) was first identified as a novel CALM (PICALM) interactor that influences the subcellular localization of the leukemogenic fusion protein CALM/AF10. CATS is highly expressed in cancer cell lines in a cell cycle dependent manner and is induced by mitogens. CATS is considered a marker for proliferation, known to control the metaphase-to-anaphase transition during the cell division. Using CATS as a bait in a yeast two-hybrid screen we identified the Kinase Interacting Stathmin (KIS or UHMK1) protein as a CATS interacting partner. The interaction between CATS and KIS was confirmed by GST pull-down, co-immunoprecipitation and co-localization experiments. Using kinase assay we showed that CATS is a substrate of KIS and mapped the phosphorylation site to CATS serine 131 (S131). Protein expression analysis revealed that KIS levels changed in a cell cycle-dependent manner and in the opposite direction to CATS levels. In a reporter gene assay KIS was able to enhance the transcriptional repressor activity of CATS, independent of CATS phophorylation at S131. Moreover, we showed that CATS and KIS antagonize the transactivation capacity of CALM/AF10.In summary, our results show that CATS interacts with and is a substrate for KIS, suggesting that KIS regulates CATS function.


Subject(s)
Carrier Proteins , Intracellular Signaling Peptides and Proteins , Oncogene Proteins, Fusion , Protein Serine-Threonine Kinases , Binding Sites , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Protein Binding , Protein Interaction Maps , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
12.
Structure ; 21(2): 197-208, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23273425

ABSTRACT

The essential splicing factors U2AF65 and SF1 cooperatively bind consensus sequences at the 3' end of introns. Phosphorylation of SF1 on a highly conserved "SPSP" motif enhances its interaction with U2AF65 and the pre-mRNA. Here, we reveal that phosphorylation induces essential conformational changes in SF1 and in the SF1/U2AF65/3' splice site complex. Crystal structures of the phosphorylated (P)SF1 domain bound to the C-terminal domain of U2AF65 at 2.29 Å resolution and of the unphosphorylated SF1 domain at 2.48 Å resolution demonstrate that phosphorylation induces a disorder-to-order transition within a previously unknown SF1/U2AF65 interface. We find by small-angle X-ray scattering that the local folding of the SPSP motif transduces into global conformational changes in the nearly full-length (P)SF1/U2AF65/3' splice site assembly. We further determine that SPSP phosphorylation and the SF1/U2AF65 interface are essential in vivo. These results offer a structural prototype for phosphorylation-dependent control of pre-mRNA splicing factors.


Subject(s)
DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Protein Processing, Post-Translational , Ribonucleoproteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Base Sequence , Cell Proliferation , Crystallography, X-Ray , DNA-Binding Proteins/physiology , HEK293 Cells , HeLa Cells , Humans , Hydrogen Bonding , Mice , Models, Molecular , Molecular Sequence Data , NIH 3T3 Cells , Nuclear Proteins/physiology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Secondary , RNA Splice Sites , RNA Splicing Factors , Ribonucleoproteins/physiology , Splicing Factor U2AF , Transcription Factors/physiology
13.
PLoS One ; 7(8): e43946, 2012.
Article in English | MEDLINE | ID: mdl-22937132

ABSTRACT

The brain-enriched protein kinase KIS (product of the gene UHMK1) has been shown to phosphorylate the human splicing factor SF1 in vitro. This phosphorylation in turn favors the formation of a U2AF(65)-SF1-RNA complex which occurs at the 3' end of introns at an early stage of spliceosome assembly. Here, we analyzed the effects of KIS knockout on mouse SF1 phosphorylation, physiology, adult behavior, and gene expression in the neonate brain. We found SF1 isoforms are differently expressed in KIS-ko mouse brains and fibroblasts. Re-expression of KIS in fibroblasts restores a wild type distribution of SF1 isoforms, confirming the link between KIS and SF1. Microarray analysis of transcripts in the neonate brain revealed a subtle down-regulation of brain specific genes including cys-loop ligand-gated ion channels and metabolic enzymes. Q-PCR analyses confirmed these defects and point to an increase of pre-mRNA over mRNA ratios, likely due to changes in splicing efficiency. While performing similarly in prepulse inhibition and most other behavioral tests, KIS-ko mice differ in spontaneous activity and contextual fear conditioning. This difference suggests that disregulation of gene expression due to KIS inactivation affects specific brain functions.


Subject(s)
Brain/metabolism , Conditioning, Psychological/physiology , Fear/physiology , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Behavior, Animal/physiology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Hyperkinesis/genetics , Hyperkinesis/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Motor Activity/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
14.
Psychiatr Genet ; 21(6): 315-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21399567

ABSTRACT

In two recent papers, polymorphisms located in U2AF homology motif kinase 1 (UHMK1) gene have been associated to schizophrenia. This gene encodes the serine/threonine kinase, kinase interacting with Stathmin, and has been functionally related to RNA metabolism and neurite outgrowth. In this study, we explored the contribution of this gene in schizophrenia susceptibility, using a case-control association study, a mutation screening, a transcription level analysis, and by the investigation of the phosphorylation status of the splicing factor, SF1, in B-lymphoblastoid cell lines of patients and controls. No association was observed in our French cohort, and no amino acid substitution was predicted in the subsample studied for mutation screening. No difference was observed in expression level or in SF1 phosphorylation between patients and controls. Despite a slight difference persisting in the meta-analysis carried out using four European populations, these data suggest, altogether, that UHMK1 does not play a major role in susceptibility to schizophrenia.


Subject(s)
Genetic Predisposition to Disease , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Schizophrenia/enzymology , Schizophrenia/genetics , France , Gene Frequency/genetics , Humans , Meta-Analysis as Topic , Polymorphism, Single Nucleotide/genetics , White People/genetics
15.
J Mol Biol ; 381(3): 748-62, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18588901

ABSTRACT

The protein kinase KIS is made by the juxtaposition of a unique kinase domain and a C-terminal domain with a U2AF homology motif (UHM), a sequence motif for protein interaction initially identified in the heterodimeric pre-mRNA splicing factor U2AF. This domain of KIS is closely related to the C-terminal UHM domain of the U2AF large subunit, U2AF(65). KIS phosphorylates the splicing factor SF1, which in turn enhances SF1 binding to U2AF(65) and the 3' splice site, an event known to take place at an early step of spliceosome assembly. Here, the analysis of the subcellular localization of mutated forms of KIS indicates that the kinase domain of KIS is the necessary domain for its nuclear localization. As in the case of U2AF(65), the UHM-containing C-terminal domain of KIS is required for binding to the splicing factors SF1 and SF3b155. The efficiency of KIS binding to SF1 and SF3b155 is similar to that of U2AF(65) in pull-down assays. These results further support the functional link of KIS with splicing factors. Interestingly, when compared to other UHM-containing proteins, KIS presents a different specificity for the UHM docking sites that are present in the N-terminal region of SF3b155, thus providing a new insight into the variety of interactions mediated by UHM domains.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleoproteins/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Animals , CHO Cells , Cricetinae , Cricetulus , Cytoplasm/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , RNA Splicing Factors , Rats , Splicing Factor U2AF
16.
FEBS J ; 273(3): 577-87, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16420481

ABSTRACT

Protein phosphorylation ensures the accurate and controlled expression of the genome, for instance by regulating the activities of pre-mRNA splicing factors. Here we report that splicing factor 1 (SF1), which is involved in an early step of intronic sequence recognition, is highly phosphorylated in mammalian cells on two serines within an SPSP motif at the junction between its U2AF65 and RNA binding domains. We show that SF1 interacts in vitro with the protein kinase KIS, which possesses a 'U2AF homology motif' (UHM) domain. The UHM domain of KIS is required for KIS and SF1 to interact, and for KIS to efficiently phosphorylate SF1 on the SPSP motif. Importantly, SPSP phosphorylation by KIS increases binding of SF1 to U2AF65, and enhances formation of the ternary SF1-U2AF65-RNA complex. These results further suggest that this phosphorylation event has an important role for the function of SF1, and possibly for the structural rearrangements associated with spliceosome assembly and function.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proline/metabolism , Ribonucleoproteins/metabolism , Serine/metabolism , Transcription Factors/metabolism , Amino Acid Motifs/physiology , HeLa Cells , Humans , In Vitro Techniques , Phosphorylation , Protein Binding , RNA/metabolism , RNA Splicing Factors , Splicing Factor U2AF
17.
Brain Res Mol Brain Res ; 114(1): 55-64, 2003 May 26.
Article in English | MEDLINE | ID: mdl-12782393

ABSTRACT

KIS is the only known protein kinase that possesses an RNA recognition motif. This original structure indicates a role for KIS in the maturation of RNAs possibly by phosphorylating and regulating the activities of RNA associated factors. Another function of KIS has recently been unravelled--it negatively regulates the cdk inhibitor p27Kip1 and thus promotes cell cycle progression through G1. In order to explore the functional expression of this kinase, we quantified its mRNA in a wide range of rat and human tissues, during development and in tumors. In both species, the highest level of KIS gene expression was in adult neural tissues. Interestingly, within the adult rat brain, KIS mRNA is enriched in several areas including the substantia nigra compacta and nuclei of the brain stem. Furthermore, KIS gene expression increases dramatically during brain development. Altogether our results point to a ubiquitous function for KIS together with a particular implication during neural differentiation or in the function of mature neural cells. No dysregulation of KIS gene expression was detected in human tumors from breast, bladder, prostate, liver and kidney origins. On the other hand, the KIS gene was overexpressed in NF1-associated plexiform neurofibromas and malignant peripheral nerve sheath tumors (MPNSTs) as compared to dermal neurofibroma which suggests a possible implication of KIS in the genesis of NF1-associated tumors.


Subject(s)
Brain Stem/enzymology , Gene Expression Regulation, Enzymologic , Protein Serine-Threonine Kinases/genetics , Substantia Nigra/enzymology , Age Factors , Amino Acid Sequence , Animals , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Gene Expression Regulation, Developmental , Humans , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Neurofibromatoses/genetics , Neurofibromin 1/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/metabolism
18.
Genomics ; 81(4): 400-10, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12676564

ABSTRACT

The stathmin family consists of phosphoproteins highly conserved in vertebrates and thought to be implicated in the development and functional regulation of various organs, most notably the nervous system. This family includes stathmin, SCG10, SCLIP, and RB3, phosphoproteins that are related by structural and functional homologies. They all sequester tubulin and interfere with microtubule dynamics, a property due to their shared stathmin-like domain. Little is known about the expression of the stathmin gene family in humans. Herein, we describe for the first time, for a collection of human tissues, the expression of each member of this family, using real-time quantitative RT-PCR. We found that stathmin is ubiquitously expressed, whereas SCG10 and RB3 are neural enriched, expression patterns similar to those reported for other mammals. Surprisingly, SCLIP, whose expression is thought to be neural-specific, exhibits a broader tissue distribution. Analyses of the SCLIP gene (approved symbol STMN3) show that it contains several NRSE-like elements that display low or no affinity for the cognate binding protein NRSF. The substantial expression of SCLIP in most tissues points out a novel function for this protein outside the nervous system and raises the possibility that its coexpression with stathmin could provide some degree of functional redundancy.


Subject(s)
Gene Expression Profiling , Microtubule Proteins , Multigene Family/genetics , Nerve Growth Factors/genetics , Phosphoproteins/genetics , Antibodies, Monoclonal , Carrier Proteins , DNA Primers , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation , HeLa Cells , Humans , Membrane Proteins , Nervous System/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Stathmin
SELECTION OF CITATIONS
SEARCH DETAIL