Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Nature ; 627(8003): 437-444, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383789

ABSTRACT

Stalled ribosomes at the endoplasmic reticulum (ER) are covalently modified with the ubiquitin-like protein UFM1 on the 60S ribosomal subunit protein RPL26 (also known as uL24)1,2. This modification, which is known as UFMylation, is orchestrated by the UFM1 ribosome E3 ligase (UREL) complex, comprising UFL1, UFBP1 and CDK5RAP3 (ref. 3). However, the catalytic mechanism of UREL and the functional consequences of UFMylation are unclear. Here we present cryo-electron microscopy structures of UREL bound to 60S ribosomes, revealing the basis of its substrate specificity. UREL wraps around the 60S subunit to form a C-shaped clamp architecture that blocks the tRNA-binding sites at one end, and the peptide exit tunnel at the other. A UFL1 loop inserts into and remodels the peptidyl transferase centre. These features of UREL suggest a crucial function for UFMylation in the release and recycling of stalled or terminated ribosomes from the ER membrane. In the absence of functional UREL, 60S-SEC61 translocon complexes accumulate at the ER membrane, demonstrating that UFMylation is necessary for releasing SEC61 from 60S subunits. Notably, this release is facilitated by a functional switch of UREL from a 'writer' to a 'reader' module that recognizes its product-UFMylated 60S ribosomes. Collectively, we identify a fundamental role for UREL in dissociating 60S subunits from the SEC61 translocon and the basis for UFMylation in regulating protein homeostasis at the ER.


Subject(s)
Endoplasmic Reticulum , Protein Processing, Post-Translational , Ribosome Subunits, Large, Eukaryotic , Ubiquitin-Protein Ligases , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/ultrastructure , Cryoelectron Microscopy , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Homeostasis , Intracellular Membranes/metabolism , Peptidyl Transferases/chemistry , Peptidyl Transferases/metabolism , Peptidyl Transferases/ultrastructure , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , RNA, Transfer/metabolism , SEC Translocation Channels/chemistry , SEC Translocation Channels/metabolism , SEC Translocation Channels/ultrastructure , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/ultrastructure , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosome Subunits, Large, Eukaryotic/ultrastructure
2.
Mol Cell Proteomics ; 23(8): 100802, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38880245

ABSTRACT

The ATR kinase protects cells against DNA damage and replication stress and represents a promising anti-cancer drug target. The ATR inhibitors (ATRi) berzosertib and gartisertib are both in clinical trials for the treatment of advanced solid tumors as monotherapy or in combination with genotoxic agents. We carried out quantitative phospho-proteomic screening for ATR biomarkers that are highly sensitive to berzosertib and gartisertib, using an optimized mass spectrometry pipeline. Screening identified a range of novel ATR-dependent phosphorylation events, which were grouped into three broad classes: (i) targets whose phosphorylation is highly sensitive to ATRi and which could be the next generation of ATR biomarkers; (ii) proteins with known genome maintenance roles not previously known to be regulated by ATR; (iii) novel targets whose cellular roles are unclear. Class iii targets represent candidate DNA damage response proteins and, with this in mind, proteins in this class were subjected to secondary screening for recruitment to DNA damage sites. We show that one of the proteins recruited, SCAF1, interacts with RNAPII in a phospho-dependent manner and recruitment requires PARP activity and interaction with RNAPII. We also show that SCAF1 deficiency partly rescues RAD51 loading in cells lacking the BRCA1 tumor suppressor. Taken together these data reveal potential new ATR biomarkers and new genome maintenance factors.

3.
Cell ; 142(1): 65-76, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20603015

ABSTRACT

DNA interstrand crosslinks (ICLs) are highly toxic because they block the progression of replisomes. The Fanconi Anemia (FA) proteins, encoded by genes that are mutated in FA, are important for repair of ICLs. The FA core complex catalyzes the monoubiquitination of FANCD2, and this event is essential for several steps of ICL repair. However, how monoubiquitination of FANCD2 promotes ICL repair at the molecular level is unknown. Here, we describe a highly conserved protein, KIAA1018/MTMR15/FAN1, that interacts with, and is recruited to sites of DNA damage by, the monoubiquitinated form of FANCD2. FAN1 exhibits endonuclease activity toward 5' flaps and has 5' exonuclease activity, and these activities are mediated by an ancient VRR_nuc domain. Depletion of FAN1 from human cells causes hypersensitivity to ICLs, defects in ICL repair, and genome instability. These data at least partly explain how ubiquitination of FANCD2 promotes DNA repair.


Subject(s)
DNA Repair , Exodeoxyribonucleases/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Amino Acid Sequence , Apoptosis Regulatory Proteins , BRCA2 Protein/metabolism , Cell Line , Cross-Linking Reagents/pharmacology , DNA Damage/drug effects , Endodeoxyribonucleases , Endonucleases/chemistry , Endonucleases/metabolism , Exodeoxyribonucleases/chemistry , Humans , Molecular Sequence Data , Multifunctional Enzymes , Protein Structure, Tertiary , Sequence Alignment , Ubiquitination
4.
EMBO J ; 37(24)2018 12 14.
Article in English | MEDLINE | ID: mdl-30266825

ABSTRACT

Mutations in the gene encoding the protein kinase CDKL5 cause a debilitating neurodevelopmental disease termed CDKL5 disorder. The impact of these mutations on CDKL5 function is poorly understood because the substrates and cellular processes controlled by CDKL5 are unclear. Here, we describe a quantitative phosphoproteomic screening which identified MAP1S, CEP131 and DLG5-regulators of microtubule and centrosome function-as cellular substrates of CDKL5. Antibodies against MAP1S phospho-Ser900 and CEP131 phospho-Ser35 confirmed CDKL5-dependent phosphorylation of these targets in human cells. The phospho-acceptor serine residues in MAP1S, CEP131 and DLG5 lie in the motif RPXSA, although CDKL5 can tolerate residues other than Ala immediately C-terminal to the phospho-acceptor serine. We provide insight into the control of CDKL5 activity and show that pathogenic mutations in CDKL5 cause a major reduction in CDKL5 activity in vitro and in cells. These data reveal the first cellular substrates of CDKL5, which may represent important biomarkers in the diagnosis and treatment of CDKL5 disorder, and illuminate the functions of this poorly characterized kinase.


Subject(s)
Epileptic Syndromes/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Spasms, Infantile/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Cell Cycle Proteins/genetics , Cell Line, Tumor , Centrosome/metabolism , Cytoskeletal Proteins , Epileptic Syndromes/genetics , Epileptic Syndromes/pathology , HEK293 Cells , Humans , Membrane Proteins/genetics , Microtubule Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Mutation , Protein Serine-Threonine Kinases/genetics , Proteomics , Spasms, Infantile/genetics , Spasms, Infantile/pathology , Tumor Suppressor Proteins/genetics
5.
EMBO Rep ; 21(3): e48412, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32009292

ABSTRACT

The intracellular trafficking pathway, macroautophagy, is a recycling and disposal service that can be upregulated during periods of stress to maintain cellular homeostasis. An essential phase is the elongation and closure of the phagophore to seal and isolate unwanted cargo prior to lysosomal degradation. Human ATG2A and ATG2B proteins, through their interaction with WIPI proteins, are thought to be key players during phagophore elongation and closure, but little mechanistic detail is known about their function. We have identified a highly conserved motif driving the interaction between human ATG2 and GABARAP proteins that is in close proximity to the ATG2-WIPI4 interaction site. We show that the ATG2A-GABARAP interaction mutants are unable to form and close phagophores resulting in blocked autophagy, similar to ATG2A/ATG2B double-knockout cells. In contrast, the ATG2A-WIPI4 interaction mutant fully restored phagophore formation and autophagy flux, similar to wild-type ATG2A. Taken together, we provide new mechanistic insights into the requirements for ATG2 function at the phagophore and suggest that an ATG2-GABARAP/GABARAP-L1 interaction is essential for phagophore formation, whereas ATG2-WIPI4 interaction is dispensable.


Subject(s)
Autophagosomes , Membrane Proteins , Apoptosis Regulatory Proteins/metabolism , Autophagosomes/metabolism , Autophagy , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Protein Transport , Vesicular Transport Proteins/metabolism
6.
Biochem J ; 478(14): 2825-2842, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34195792

ABSTRACT

Cullin ubiquitin ligases drive replisome disassembly during DNA replication termination. In worm, frog and mouse cells, CUL2LRR1 is required to ubiquitylate the MCM7 subunit of the CMG helicase. Here, we show that cullin ligases also drive CMG-MCM7 ubiquitylation in human cells, thereby making the helicase into a substrate for the p97 unfoldase. Using purified human proteins, including a panel of E2 ubiquitin-conjugating enzymes, we have reconstituted CMG helicase ubiquitylation, dependent upon neddylated CUL2LRR1. The reaction is highly specific to CMG-MCM7 and requires the LRR1 substrate targeting subunit, since replacement of LRR1 with the alternative CUL2 adaptor VHL switches ubiquitylation from CMG-MCM7 to HIF1. CUL2LRR1 firstly drives monoubiquitylation of CMG-MCM7 by the UBE2D class of E2 enzymes. Subsequently, CUL2LRR1 activates UBE2R1/R2 or UBE2G1/G2 to extend a single K48-linked ubiquitin chain on CMG-MCM7. Thereby, CUL2LRR1 converts CMG into a substrate for p97, which disassembles the ubiquitylated helicase during DNA replication termination.


Subject(s)
Cullin Proteins/metabolism , DNA Helicases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Cell Line , Cloning, Molecular/methods , Cullin Proteins/genetics , DNA Helicases/genetics , Humans , Immunoblotting , Lysine/metabolism , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sf9 Cells , Spodoptera , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
7.
EMBO Rep ; 20(9): e47495, 2019 09.
Article in English | MEDLINE | ID: mdl-31338967

ABSTRACT

The concerted action of many protein kinases helps orchestrate the error-free progression through mitosis of mammalian cells. The roles and regulation of some prominent mitotic kinases, such as cyclin-dependent kinases, are well established. However, these and other known mitotic kinases alone cannot account for the extent of protein phosphorylation that has been reported during mammalian mitosis. Here we demonstrate that CK1α, of the casein kinase 1 family of protein kinases, localises to the spindle and is required for proper spindle positioning and timely cell division. CK1α is recruited to the spindle by FAM83D, and cells devoid of FAM83D, or those harbouring CK1α-binding-deficient FAM83DF283A/F283A knockin mutations, display pronounced spindle positioning defects, and a prolonged mitosis. Restoring FAM83D at the endogenous locus in FAM83D-/- cells, or artificially delivering CK1α to the spindle in FAM83DF283A/F283A cells, rescues these defects. These findings implicate CK1α as new mitotic kinase that orchestrates the kinetics and orientation of cell division.


Subject(s)
Casein Kinase I/metabolism , Cell Cycle Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Spindle Apparatus/metabolism , Animals , Casein Kinase I/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Flow Cytometry , HeLa Cells , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Mitosis/genetics , Mitosis/physiology
8.
Mol Cell ; 52(2): 221-33, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24076219

ABSTRACT

Holliday junctions (HJs) are X-shaped DNA structures that arise during homologous recombination, which must be removed to enable chromosome segregation. The SLX1 and MUS81-EME1 nucleases can both process HJs in vitro, and they bind in close proximity on the SLX4 scaffold, hinting at possible cooperation. However, the cellular roles of mammalian SLX1 are not yet known. Here, we use mouse genetics and structure function analysis to investigate SLX1 function. Disrupting the murine Slx1 and Slx4 genes revealed that they are essential for HJ resolution in mitotic cells. Moreover, SLX1 and MUS81-EME1 act together to resolve HJs in a manner that requires tethering to SLX4. We also show that SLX1, like MUS81-EME1, is required for repair of DNA interstrand crosslinks, but this role appears to be independent of HJ cleavage, at least in mouse cells. These findings shed light on HJ resolution in mammals and on maintenance of genome stability.


Subject(s)
DNA Repair , DNA, Cruciform , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Endonucleases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Endodeoxyribonucleases/genetics , Endonucleases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Molecular Sequence Data , Protein Binding , RNA Interference , Recombinases/genetics , Recombinases/metabolism , Sequence Homology, Amino Acid
9.
J Cell Sci ; 131(1)2018 01 10.
Article in English | MEDLINE | ID: mdl-29175910

ABSTRACT

Our previous studies of PAWS1 (protein associated with SMAD1; also known as FAM83G) have suggested that this molecule has roles beyond BMP signalling. To investigate these roles, we have used CRISPR/Cas9 to generate PAWS1-knockout U2OS osteosarcoma cells. Here, we show that PAWS1 plays a role in the regulation of the cytoskeletal machinery, including actin and focal adhesion dynamics, and cell migration. Confocal microscopy and live cell imaging of actin in U2OS cells indicate that PAWS1 is also involved in cytoskeletal dynamics and organization. Loss of PAWS1 causes severe defects in F-actin organization and distribution as well as in lamellipodial organization, resulting in impaired cell migration. PAWS1 interacts in a dynamic fashion with the actin/cytoskeletal regulator CD2AP at lamellae, suggesting that its association with CD2AP controls actin organization and cellular migration. Genetic ablation of CD2AP from U2OS cells instigates actin and cell migration defects reminiscent of those seen in PAWS1-knockout cells.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Focal Adhesions/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Signal Transduction
10.
EMBO Rep ; 19(4)2018 04.
Article in English | MEDLINE | ID: mdl-29514862

ABSTRACT

The BMP and Wnt signalling pathways determine axis specification during embryonic development. Our previous work has shown that PAWS1 (also known as FAM83G) interacts with SMAD1 and modulates BMP signalling. Here, surprisingly, we show that overexpression of PAWS1 in Xenopus embryos activates Wnt signalling and causes complete axis duplication. Consistent with these observations in Xenopus, Wnt signalling is diminished in U2OS osteosarcoma cells lacking PAWS1, while BMP signalling is unaffected. We show that PAWS1 interacts and co-localises with the α isoform of casein kinase 1 (CK1), and that PAWS1 mutations incapable of binding CK1 fail both to activate Wnt signalling and to elicit axis duplication in Xenopus embryos.


Subject(s)
Casein Kinase Ialpha/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Wnt Signaling Pathway , Animals , Axin Protein/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Line, Tumor , Cell Nucleus , Ectopic Gene Expression , Gene Expression , Gene Knockout Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Protein Binding , Protein Transport , Xenopus , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , beta Catenin/metabolism
11.
Biochem J ; 476(20): 3081-3107, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31665227

ABSTRACT

The serum- and glucocorticoid-regulated kinase (SGK) isoforms contribute resistance to cancer therapies targeting the PI3K pathway. SGKs are homologous to Akt and these kinases display overlapping specificity and phosphorylate several substrates at the same residues, such as TSC2 to promote tumor growth by switching on the mTORC1 pathway. The SGK3 isoform is up-regulated in breast cancer cells treated with PI3K or Akt inhibitors and recruited and activated at endosomes, through its phox homology domain binding to PtdIns(3)P. We undertook genetic and pharmacological phosphoproteomic screens to uncover novel SGK3 substrates. We identified 40 potential novel SGK3 substrates, including four endosomal proteins STX7 (Ser126) and STX12 (Ser139), RFIP4 (Ser527) and WDR44 (Ser346) that were efficiently phosphorylated in vitro by SGK3 at the sites identified in vivo, but poorly by Akt. We demonstrate that these substrates are inefficiently phosphorylated by Akt as they possess an n + 1 residue from the phosphorylation site that is unfavorable for Akt phosphorylation. Phos-tag analysis revealed that stimulation of HEK293 cells with IGF1 to activate SGK3, promoted phosphorylation of a significant fraction of endogenous STX7 and STX12, in a manner that was blocked by knock-out of SGK3 or treatment with a pan SGK inhibitor (14H). SGK3 phosphorylation of STX12 enhanced interaction with the VAMP4/VTI1A/STX6 containing the SNARE complex and promoted plasma membrane localization. Our data reveal novel substrates for SGK3 and suggest a mechanism by which STX7 and STX12 SNARE complexes are regulated by SGK3. They reveal new biomarkers for monitoring SGK3 pathway activity.


Subject(s)
Class III Phosphatidylinositol 3-Kinases/metabolism , Endosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Qa-SNARE Proteins/metabolism , Biomarkers/metabolism , Cell Membrane/metabolism , Gene Knockout Techniques , HEK293 Cells , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Qa-SNARE Proteins/genetics , Substrate Specificity , Transfection
12.
Proc Natl Acad Sci U S A ; 114(17): E3481-E3489, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28404732

ABSTRACT

It is widely accepted that the essential role of TRAF6 in vivo is to generate the Lys63-linked ubiquitin (K63-Ub) chains needed to activate the "master" protein kinase TAK1. Here, we report that TRAF6 E3 ligase activity contributes to but is not essential for the IL-1-dependent formation of K63-Ub chains, TAK1 activation, or IL-8 production in human cells, because Pellino1 and Pellino2 generate the K63-Ub chains required for signaling in cells expressing E3 ligase-inactive TRAF6 mutants. The IL-1-induced formation of K63-Ub chains and ubiquitylation of IRAK1, IRAK4, and MyD88 was abolished in TRAF6/Pellino1/Pellino2 triple-knockout (KO) cells, but not in TRAF6 KO or Pellino1/2 double-KO cells. The reexpression of E3 ligase-inactive TRAF6 mutants partially restored IL-1 signaling in TRAF6 KO cells, but not in TRAF6/Pellino1/Pellino2 triple-KO cells. Pellino1-generated K63-Ub chains activated the TAK1 complex in vitro with similar efficiently to TRAF6-generated K63-Ub chains. The early phase of TLR signaling and the TLR-dependent secretion of IL-10 (controlled by IRAKs 1 and 2) was only reduced modestly in primary macrophages from knockin mice expressing the E3 ligase-inactive TRAF6[L74H] mutant, but the late-phase production of IL-6, IL-12, and TNFα (controlled only by the pseudokinase IRAK2) was abolished. RANKL-induced signaling in macrophages and the differentiation of bone marrow to osteoclasts was similar in TRAF6[L74H] and wild-type cells, explaining why the bone structure and teeth of the TRAF6[L74H] mice was normal, unlike TRAF6 KO mice. We identify two essential roles of TRAF6 that are independent of its E3 ligase activity.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Nuclear Proteins/metabolism , RANK Ligand/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Substitution , Animals , Gene Knockdown Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Nuclear Proteins/genetics , Polyubiquitin/genetics , Polyubiquitin/metabolism , RANK Ligand/genetics , TNF Receptor-Associated Factor 6/genetics , Ubiquitin-Protein Ligases/genetics
13.
EMBO J ; 34(3): 326-43, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25538220

ABSTRACT

Reversible protein ubiquitylation plays important roles in various processes including DNA repair. Here, we identify the deubiquitylase USP45 as a critical DNA repair regulator. USP45 associates with ERCC1, a subunit of the DNA repair endonuclease XPF-ERCC1, via a short acidic motif outside of the USP45 catalytic domain. Wild-type USP45, but not a USP45 mutant defective in ERCC1 binding, efficiently deubiquitylates ERCC1 in vitro, and the levels of ubiquitylated ERCC1 are markedly enhanced in USP45 knockout cells. Cells lacking USP45 are hypersensitive specifically to UV irradiation and DNA interstrand cross-links, similar to cells lacking ERCC1. Furthermore, the repair of UV-induced DNA damage is markedly reduced in USP45-deficient cells. ERCC1 translocation to DNA damage-induced subnuclear foci is markedly impaired in USP45 knockout cells, possibly accounting for defective DNA repair. Finally, USP45 localises to sites of DNA damage in a manner dependent on its deubiquitylase activity, but independent of its ability to bind ERCC1-XPF. Together, these results establish USP45 as a new regulator of XPF-ERCC1 crucial for efficient DNA repair.


Subject(s)
Cell Nucleus/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Endopeptidases/metabolism , Ubiquitination , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/radiation effects , Animals , Cell Line, Tumor , Cell Nucleus/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Endopeptidases/genetics , Humans , Mice , Mice, Knockout , Ultraviolet Rays/adverse effects
14.
EMBO J ; 34(22): 2840-61, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26471730

ABSTRACT

Mutations in the PTEN-induced kinase 1 (PINK1) are causative of autosomal recessive Parkinson's disease (PD). We have previously reported that PINK1 is activated by mitochondrial depolarisation and phosphorylates serine 65 (Ser(65)) of the ubiquitin ligase Parkin and ubiquitin to stimulate Parkin E3 ligase activity. Here, we have employed quantitative phosphoproteomics to search for novel PINK1-dependent phosphorylation targets in HEK (human embryonic kidney) 293 cells stimulated by mitochondrial depolarisation. This led to the identification of 14,213 phosphosites from 4,499 gene products. Whilst most phosphosites were unaffected, we strikingly observed three members of a sub-family of Rab GTPases namely Rab8A, 8B and 13 that are all phosphorylated at the highly conserved residue of serine 111 (Ser(111)) in response to PINK1 activation. Using phospho-specific antibodies raised against Ser(111) of each of the Rabs, we demonstrate that Rab Ser(111) phosphorylation occurs specifically in response to PINK1 activation and is abolished in HeLa PINK1 knockout cells and mutant PINK1 PD patient-derived fibroblasts stimulated by mitochondrial depolarisation. We provide evidence that Rab8A GTPase Ser(111) phosphorylation is not directly regulated by PINK1 in vitro and demonstrate in cells the time course of Ser(111) phosphorylation of Rab8A, 8B and 13 is markedly delayed compared to phosphorylation of Parkin at Ser(65). We further show mechanistically that phosphorylation at Ser(111) significantly impairs Rab8A activation by its cognate guanine nucleotide exchange factor (GEF), Rabin8 (by using the Ser111Glu phosphorylation mimic). These findings provide the first evidence that PINK1 is able to regulate the phosphorylation of Rab GTPases and indicate that monitoring phosphorylation of Rab8A/8B/13 at Ser(111) may represent novel biomarkers of PINK1 activity in vivo. Our findings also suggest that disruption of Rab GTPase-mediated signalling may represent a major mechanism in the neurodegenerative cascade of Parkinson's disease.


Subject(s)
Oncogene Proteins/metabolism , Parkinsonian Disorders/metabolism , Protein Kinases/metabolism , rab GTP-Binding Proteins/metabolism , Amino Acid Substitution , Enzyme Activation/genetics , Germinal Center Kinases , HEK293 Cells , HeLa Cells , Humans , Mutation, Missense , Oncogene Proteins/genetics , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Phosphorylation/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , rab GTP-Binding Proteins/genetics
15.
Biochem J ; 475(1): 117-135, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29150437

ABSTRACT

Derailment of the PI3K-AGC protein kinase signalling network contributes to many human diseases including cancer. Recent work has revealed that the poorly studied AGC kinase family member, SGK3, promotes resistance to cancer therapies that target the Class 1 PI3K pathway, by substituting for loss of Akt kinase activity. SGK3 is recruited and activated at endosomes, by virtue of its phox homology domain binding to PtdIns(3)P. Here, we demonstrate that endogenous SGK3 is rapidly activated by growth factors such as IGF1, through pathways involving both Class 1 and Class 3 PI3Ks. We provide evidence that IGF1 enhances endosomal PtdIns(3)P levels via a pathway involving the UV-RAG complex of hVPS34 Class 3 PI3K. Our data point towards IGF1-induced activation of Class 1 PI3K stimulating SGK3 through enhanced production of PtdIns(3)P resulting from the dephosphorylation of PtdIns(3,4,5)P3 Our findings are also consistent with activation of Class 1 PI3K promoting mTORC2 phosphorylation of SGK3 and with oncogenic Ras-activating SGK3 solely through the Class 1 PI3K pathway. Our results highlight the versatility of upstream pathways that activate SGK3 and help explain how SGK3 substitutes for Akt following inhibition of Class 1 PI3K/Akt pathways. They also illustrate robustness of SGK3 activity that can remain active and counteract physiological conditions or stresses where either Class 1 or Class 3 PI3K pathways are inhibited.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/genetics , Insulin-Like Growth Factor I/pharmacology , Mechanistic Target of Rapamycin Complex 2/genetics , Protein Serine-Threonine Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Endosomes/drug effects , Endosomes/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Insulin-Like Growth Factor I/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation/drug effects , Plasmids/chemistry , Plasmids/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transfection
16.
Biochem J ; 475(11): 1861-1883, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29743203

ABSTRACT

Missense mutations in the LRRK2 (Leucine-rich repeat protein kinase-2) and VPS35 genes result in autosomal dominant Parkinson's disease. The VPS35 gene encodes for the cargo-binding component of the retromer complex, while LRRK2 modulates vesicular trafficking by phosphorylating a subgroup of Rab proteins. Pathogenic mutations in LRRK2 increase its kinase activity. It is not known how the only thus far described pathogenic VPS35 mutation, [p.D620N] exerts its effects. We reveal that the VPS35[D620N] knock-in mutation strikingly elevates LRRK2-mediated phosphorylation of Rab8A, Rab10, and Rab12 in mouse embryonic fibroblasts. The VPS35[D620N] mutation also increases Rab10 phosphorylation in mouse tissues (the lung, kidney, spleen, and brain). Furthermore, LRRK2-mediated Rab10 phosphorylation is increased in neutrophils as well as monocytes isolated from three Parkinson's patients with a heterozygous VPS35[D620N] mutation compared with healthy donors and idiopathic Parkinson's patients. LRRK2-mediated Rab10 phosphorylation is significantly suppressed by knock-out or knock-down of VPS35 in wild-type, LRRK2[R1441C], or VPS35[D620N] cells. Finally, VPS35[D620N] mutation promotes Rab10 phosphorylation more potently than LRRK2 pathogenic mutations. Available data suggest that Parkinson's patients with VPS35[D620N] develop the disease at a younger age than those with LRRK2 mutations. Our observations indicate that VPS35 controls LRRK2 activity and that the VPS35[D620N] mutation results in a gain of function, potentially causing PD through hyperactivation of the LRRK2 kinase. Our findings suggest that it may be possible to elaborate compounds that target the retromer complex to suppress LRRK2 activity. Moreover, patients with VPS35[D620N] associated Parkinson's might benefit from LRRK2 inhibitor treatment that have entered clinical trials in humans.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/metabolism , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/metabolism , Animals , Gene Knock-In Techniques , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Mice, Inbred C57BL , Mutation, Missense , Parkinson Disease/genetics , Phosphorylation , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/genetics
17.
Mol Cell ; 40(4): 632-44, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21055984

ABSTRACT

Budding yeast Mms22 is required for homologous recombination (HR)-mediated repair of stalled or broken DNA replication forks. Here we identify a human Mms22-like protein (MMS22L) and an MMS22L-interacting protein, NFκBIL2/TONSL. Depletion of MMS22L or TONSL from human cells causes a high level of double-strand breaks (DSBs) during DNA replication. Both proteins accumulate at stressed replication forks, and depletion of MMS22L or TONSL from cells causes hypersensitivity to agents that cause S phase-associated DSBs, such as topoisomerase (TOP) inhibitors. In this light, MMS22L and TONSL are required for the HR-mediated repair of replication fork-associated DSBs. In cells depleted of either protein, DSBs induced by the TOP1 inhibitor camptothecin are resected normally, but the loading of the RAD51 recombinase is defective. Therefore, MMS22L and TONSL are required for the maintenance of genome stability when unscheduled DSBs occur in the vicinity of DNA replication forks.


Subject(s)
DNA-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Recombination, Genetic , Amino Acid Sequence , Cell Cycle Proteins/metabolism , Cell Line , Cell Survival , Computational Biology , DNA Breaks, Double-Stranded , DNA-Binding Proteins/chemistry , DNA-Directed DNA Polymerase , Drug Resistance , Humans , Models, Biological , Molecular Chaperones , Molecular Sequence Data , Multienzyme Complexes , NF-kappa B/chemistry , Nuclear Proteins/chemistry , Protein Binding , Rad51 Recombinase/metabolism , S Phase
18.
Biochem J ; 474(13): 2235-2248, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28507161

ABSTRACT

Interleukin-1 (IL-1) signaling induces the formation of Lys63-linked ubiquitin (K63-Ub) chains, which are thought to activate the 'master' protein kinase TGFß-activated kinase 1 (TAK1) by interacting with its TAK1-binding 2 (TAB2) and TAB3 subunits. Here, we report that IL-1ß can also activate the TAB1-TAK1 heterodimer present in TAB2/TAB3 double knockout (DKO) IL-1 receptor-expressing cells. The IL-1ß-dependent activation of the TAB1-TAK1 heterodimer in TAB2/3 DKO cells is required for the expression and E3 ligase activity of tumor necrosis factor receptor-associated factor 6 (TRAF6) and is reduced by the small interfering RNA (siRNA) knockdown of ubiquitin conjugating 13 (Ubc13), an E2-conjugating enzyme that directs the formation of K63-Ub chains. IL-1ß signaling was restored to TAB1/2/3 triple KO cells by the re-expression of either TAB1 or TAB2, but not by an ubiquitin binding-defective mutant of TAB2. We conclude that IL-1ß can induce the activation of TAK1 in two ways, only one of which requires the binding of K63-Ub chains to TAB2/3. The early IL-1ß-stimulated, TAK1-dependent activation of p38α mitogen-activated protein (MAP) kinase and the canonical IκB kinase (IKK) complex, as well as the NF-κB-dependent transcription of immediate early genes, was similar in TAB2/3 DKO cells and TAB2/3-expressing cells. However, in contrast with TAB2/3-expressing cells, IL-1ß signaling was transient in TAB2/3 DKO cells, and the activation of c-Jun N-terminal kinase 1 (JNK1), JNK2 and p38γ was greatly reduced at all times. These observations indicate a role for TAB2/3 in directing the TAK1-dependent activation of MAP kinase kinases that switch on JNK1/2 and p38γ MAP kinases. These observations and the transient activation of the TAB1-TAK1 heterodimer may explain why IL-1ß-dependent IL-8 mRNA formation was abolished in TAB2/3 DKO cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation/drug effects , Interleukin-1/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , TNF Receptor-Associated Factor 6/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , CRISPR-Cas Systems , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , Phosphorylation , Protein Binding , Signal Transduction/drug effects , Ubiquitin/metabolism , Ubiquitination
19.
Hum Mol Genet ; 24(16): 4545-58, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25994507

ABSTRACT

The STE20/SPS1-related proline/alanine-rich kinase (SPAK) controls blood pressure (BP) by phosphorylating and stimulating the Na-Cl (NCC) and Na-K-2Cl (NKCC2) co-transporters, which regulate salt reabsorption in the kidney. SPAK possesses a conserved carboxy-terminal (CCT) domain, which recognises RFXV/I motifs present in its upstream activator [isoforms of the With-No-lysine (K) kinases (WNKs)] as well as its substrates (NCC and NKCC2). To define the physiological importance of the CCT domain, we generated knock-in mice in which the critical CCT domain Leu502 residue required for high affinity recognition of the RFXI/V motif was mutated to Alanine. The SPAK CCT domain defective knock-in animals are viable, and the Leu502Ala mutation abolished co-immunoprecipitation of SPAK with WNK1, NCC and NKCC2. The CCT domain defective animals displayed markedly reduced SPAK activity and phosphorylation of NCC and NKCC2 co-transporters at the residues phosphorylated by SPAK. This was also accompanied by a reduction in the expression of NCC and NKCC2 protein without changes in mRNA levels. The SPAK CCT domain knock-in mice showed typical features of Gitelman Syndrome with mild hypokalaemia, hypomagnesaemia, hypocalciuria and displayed salt wasting on switching to a low-Na diet. These observations establish that the CCT domain plays a crucial role in controlling SPAK activity and BP. Our results indicate that CCT domain inhibitors would be effective at reducing BP by lowering phosphorylation as well as expression of NCC and NKCC2.


Subject(s)
Blood Pressure , Gitelman Syndrome/metabolism , Mutation, Missense , Protein Serine-Threonine Kinases/metabolism , Amino Acid Substitution , Animals , Gitelman Syndrome/genetics , Gitelman Syndrome/physiopathology , HEK293 Cells , Humans , Mice , Mice, Mutant Strains , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Solute Carrier Family 12, Member 3/biosynthesis , Solute Carrier Family 12, Member 3/genetics
20.
Mol Cell ; 35(1): 116-27, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19595721

ABSTRACT

Budding yeast Slx4 interacts with the structure-specific endonuclease Slx1 to ensure completion of ribosomal DNA replication. Slx4 also interacts with the Rad1-Rad10 endonuclease to control cleavage of 3' flaps during repair of double-strand breaks (DSBs). Here we describe the identification of human SLX4, a scaffold for DNA repair nucleases XPF-ERCC1, MUS81-EME1, and SLX1. SLX4 immunoprecipitates show SLX1-dependent nuclease activity toward Holliday junctions and MUS81-dependent activity toward other branched DNA structures. Furthermore, SLX4 enhances the nuclease activity of SLX1, MUS81, and XPF. Consistent with a role in processing recombination intermediates, cells depleted of SLX4 are hypersensitive to genotoxins that cause DSBs and show defects in the resolution of interstrand crosslink-induced DSBs. Depletion of SLX4 causes a decrease in DSB-induced homologous recombination. These data show that SLX4 is a regulator of structure-specific nucleases and that SLX4 and SLX1 are important regulators of genome stability in human cells.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Endonucleases/metabolism , Recombinases/metabolism , Blotting, Western , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Protein Binding , RNA, Small Interfering/genetics , Recombinases/genetics , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL