Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
iScience ; 27(3): 109302, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38450154

ABSTRACT

Protein phosphatase 2A (PP2A) is an essential Ser/Thr phosphatase. The PP2A holoenzyme complex comprises a scaffolding (A), regulatory (B), and catalytic (C) subunit, with PPP2CA being the principal catalytic subunit. The full scope of PP2A substrates in cells remains to be defined. To address this, we employed dTAG proteolysis-targeting chimeras to efficiently and selectively degrade dTAG-PPP2CA in homozygous knock-in HEK293 cells. Unbiased global phospho-proteomics identified 2,204 proteins with significantly increased phosphorylation upon dTAG-PPP2CA degradation, implicating them as potential PPP2CA substrates. A vast majority of these are novel. Bioinformatic analyses revealed involvement of the potential PPP2CA substrates in spliceosome function, cell cycle, RNA transport, and ubiquitin-mediated proteolysis. We identify a pSP/pTP motif as a predominant target for PPP2CA and confirm some of our phospho-proteomic data with immunoblotting. We provide an in-depth atlas of potential PPP2CA substrates and establish targeted degradation as a robust tool to unveil phosphatase substrates in cells.

2.
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
3.
Nat Struct Mol Biol ; 31(3): 523-535, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38238586

ABSTRACT

Histone chaperones control nucleosome density and chromatin structure. In yeast, the H3-H4 chaperone Spt2 controls histone deposition at active genes but its roles in metazoan chromatin structure and organismal physiology are not known. Here we identify the Caenorhabditis elegans ortholog of SPT2 (CeSPT-2) and show that its ability to bind histones H3-H4 is important for germline development and transgenerational epigenetic gene silencing, and that spt-2 null mutants display signatures of a global stress response. Genome-wide profiling showed that CeSPT-2 binds to a range of highly expressed genes, and we find that spt-2 mutants have increased chromatin accessibility at a subset of these loci. We also show that SPT2 influences chromatin structure and controls the levels of soluble and chromatin-bound H3.3 in human cells. Our work reveals roles for SPT2 in controlling chromatin structure and function in Metazoa.


Subject(s)
DNA-Binding Proteins , Histone Chaperones , Animals , Humans , Histone Chaperones/genetics , Histone Chaperones/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Chromatin/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae/metabolism
4.
Sci Adv ; 9(50): eadj1205, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38091401

ABSTRACT

We demonstrate that the Parkinson's VPS35[D620N] mutation alters the expression of ~220 lysosomal proteins and stimulates recruitment and phosphorylation of Rab proteins at the lysosome. This recruits the phospho-Rab effector protein RILPL1 to the lysosome where it binds to the lysosomal integral membrane protein TMEM55B. We identify highly conserved regions of RILPL1 and TMEM55B that interact and design mutations that block binding. In mouse fibroblasts, brain, and lung, we demonstrate that the VPS35[D620N] mutation reduces RILPL1 levels, in a manner reversed by LRRK2 inhibition and proteasome inhibitors. Knockout of RILPL1 enhances phosphorylation of Rab substrates, and knockout of TMEM55B increases RILPL1 levels. The lysosomotropic agent LLOMe also induced LRRK2 kinase-mediated association of RILPL1 to the lysosome, but to a lower extent than the D620N mutation. Our study uncovers a pathway through which dysfunctional lysosomes resulting from the VPS35[D620N] mutation recruit and activate LRRK2 on the lysosomal surface, driving assembly of the RILPL1-TMEM55B complex.


Subject(s)
Parkinson Disease , Animals , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Mice, Knockout , Mutation , Lysosomes/metabolism , Lysosomal Membrane Proteins
5.
Cell Chem Biol ; 30(10): 1261-1276.e7, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37591251

ABSTRACT

Targeted protein degradation (TPD), induced by enforcing target proximity to an E3 ubiquitin ligase using small molecules has become an important drug discovery approach for targeting previously undruggable disease-causing proteins. However, out of over 600 E3 ligases encoded by the human genome, just over 10 E3 ligases are currently utilized for TPD. Here, using the affinity-directed protein missile (AdPROM) system, in which an anti-GFP nanobody was linked to an E3 ligase, we screened over 30 E3 ligases for their ability to degrade 4 target proteins, K-RAS, STK33, ß-catenin, and FoxP3, which were endogenously GFP-tagged. Several new E3 ligases, including CUL2 diGly receptor KLHDC2, emerged as effective degraders, suggesting that these E3 ligases can be taken forward for the development of small-molecule degraders, such as proteolysis targeting chimeras (PROTACs). As a proof of concept, we demonstrate that a KLHDC2-recruiting peptide-based PROTAC connected to chloroalkane is capable of degrading HALO-GFP protein in cells.


Subject(s)
Transcription Factors , beta Catenin , Humans , beta Catenin/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Proteolysis , Drug Discovery , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
6.
Life Sci Alliance ; 6(7)2023 07.
Article in English | MEDLINE | ID: mdl-37188479

ABSTRACT

The NEK1 kinase controls ciliogenesis, mitosis, and DNA repair, and NEK1 mutations cause human diseases including axial spondylometaphyseal dysplasia and amyotrophic lateral sclerosis. C21ORF2 mutations cause a similar pattern of human diseases, suggesting close functional links with NEK1 Here, we report that endogenous NEK1 and C21ORF2 form a tight complex in human cells. A C21ORF2 interaction domain "CID" at the C-terminus of NEK1 is necessary for its association with C21ORF2 in cells, and pathogenic mutations in this region disrupt the complex. AlphaFold modelling predicts an extended binding interface between a leucine-rich repeat domain in C21ORF2 and the NEK1-CID, and our model may explain why pathogenic mutations perturb the complex. We show that NEK1 mutations that inhibit kinase activity or weaken its association with C21ORF2 severely compromise ciliogenesis, and that C21ORF2, like NEK1 is required for homologous recombination. These data enhance our understanding of how the NEK1 kinase is regulated, and they shed light on NEK1-C21ORF2-associated diseases.


Subject(s)
DNA Repair , Osteochondrodysplasias , Humans , Mutation/genetics , NIMA-Related Kinase 1/genetics , Osteochondrodysplasias/genetics , Phosphorylation
7.
Methods Enzymol ; 681: 61-79, 2023.
Article in English | MEDLINE | ID: mdl-36764764

ABSTRACT

Targeted protein degradation (TPD) is a useful approach in dissecting protein function and therapeutics. Technologies such as RNA interference or gene knockout that are routinely used rely on protein turnover. However, RNA interference takes a long time to deplete target proteins and is not suitable for long-lived proteins, while a genetic knockout is irreversible, takes a long time to achieve and is not suitable for essential genes. TPD has the potential to overcome the limitations of RNA interference and gene editing approaches. We have established the Affinity directed PROtein Missile (AdPROM) system, which harnesses nanobodies or binders of target proteins to redirect E3 ubiquitin ligase activity to the target protein to induce TPD through the ubiquitin proteasome system. Here we provide a step-by-step protocol for using the AdPROM system for targeted proteolysis of endogenously GFP-tagged K-RAS through an anti-GFP nanobody. This protocol can be amended to target a wide range of different proteins of interest (POIs) either by replacing the anti-GFP nanobody with a nanobody recognising the POI or by endogenously tagging the POI with GFP through CRISPR/Cas9 genome editing.


Subject(s)
Single-Domain Antibodies , Proteolysis , Single-Domain Antibodies/genetics , Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitination , Ubiquitin-Protein Ligases/metabolism
8.
Cell Rep ; 40(5): 111168, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35926457

ABSTRACT

An essential first step in the post-translational modification of proteins with UFM1, UFMylation, is the proteolytic cleavage of pro-UFM1 to expose a C-terminal glycine. Of the two UFM1-specific proteases (UFSPs) identified in humans, only UFSP2 is reported to be active, since the annotated sequence of UFSP1 lacks critical catalytic residues. Nonetheless, efficient UFM1 maturation occurs in cells lacking UFSP2, suggesting the presence of another active protease. We herein identify UFSP1 translated from a non-canonical start site to be this protease. Cells lacking both UFSPs show complete loss of UFMylation resulting from an absence of mature UFM1. While UFSP2, but not UFSP1, removes UFM1 from the ribosomal subunit RPL26, UFSP1 acts earlier in the pathway to mature UFM1 and cleave a potential autoinhibitory modification on UFC1, thereby controlling activation of UFMylation. In summary, our studies reveal important distinctions in substrate specificity and localization-dependent functions for the two proteases in regulating UFMylation.


Subject(s)
Peptide Hydrolases , Protein Processing, Post-Translational , Humans , Cysteine Endopeptidases/metabolism , Peptide Hydrolases/metabolism , Proteins/metabolism , Ribosomal Proteins/metabolism , Substrate Specificity
9.
Sci Signal ; 15(742): eabm5995, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35857630

ABSTRACT

The E3 ubiquitin ligase RNF12 plays essential roles during development, and the gene encoding it, RLIM, is mutated in the X-linked human developmental disorder Tonne-Kalscheuer syndrome (TOKAS). Substrates of RNF12 include transcriptional regulators such as the pluripotency-associated transcriptional repressor REX1. Using global quantitative proteomics in male mouse embryonic stem cells, we identified the deubiquitylase USP26 as a putative downstream target of RNF12 activity. RNF12 relieved REX1-mediated repression of Usp26, leading to an increase in USP26 abundance and the formation of RNF12-USP26 complexes. Interaction with USP26 prevented RNF12 autoubiquitylation and proteasomal degradation, thereby establishing a transcriptional feed-forward loop that amplified RNF12-dependent derepression of REX1 targets. We showed that the RNF12-USP26 axis operated specifically in mouse testes and was required for the expression of gametogenesis genes and for germ cell differentiation in vitro. Furthermore, this RNF12-USP26 axis was disrupted by RLIM and USP26 variants found in TOKAS and infertility patients, respectively. This work reveals synergy within the ubiquitylation cycle that controls a key developmental process in gametogenesis and that is disrupted in human genetic disorders.


Subject(s)
Transcription Factors , Ubiquitin-Protein Ligases , Animals , Cysteine Endopeptidases/genetics , Germ Cells/metabolism , Humans , Male , Mice , Mutation , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
10.
Open Biol ; 12(1): 210264, 2022 01.
Article in English | MEDLINE | ID: mdl-35042401

ABSTRACT

Autosomal recessive mutations in the PINK1 gene are causal for Parkinson's disease (PD). PINK1 encodes a mitochondrial localized protein kinase that is a master-regulator of mitochondrial quality control pathways. Structural studies to date have elaborated the mechanism of how mutations located within the kinase domain disrupt PINK1 function; however, the molecular mechanism of PINK1 mutations located upstream and downstream of the kinase domain is unknown. We have employed mutagenesis studies to define the minimal region of human PINK1 required for optimal ubiquitin phosphorylation, beginning at residue Ile111. Inspection of the AlphaFold human PINK1 structure model predicts a conserved N-terminal α-helical extension (NTE) domain forming an intramolecular interaction with the C-terminal extension (CTE), which we corroborate using hydrogen/deuterium exchange mass spectrometry of recombinant insect PINK1 protein. Cell-based analysis of human PINK1 reveals that PD-associated mutations (e.g. Q126P), located within the NTE : CTE interface, markedly inhibit stabilization of PINK1; autophosphorylation at Serine228 (Ser228) and Ubiquitin Serine65 (Ser65) phosphorylation. Furthermore, we provide evidence that NTE and CTE domain mutants disrupt PINK1 stabilization at the mitochondrial Translocase of outer membrane complex. The clinical relevance of our findings is supported by the demonstration of defective stabilization and activation of endogenous PINK1 in human fibroblasts of a patient with early-onset PD due to homozygous PINK1 Q126P mutations. Overall, we define a functional role of the NTE : CTE interface towards PINK1 stabilization and activation and show that loss of NTE : CTE interactions is a major mechanism of PINK1-associated mutations linked to PD.


Subject(s)
Protein Kinases , Ubiquitin , Enzyme Activation , Humans , Phosphorylation , Protein Conformation, alpha-Helical , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
11.
J Med Chem ; 64(20): 15477-15502, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34652918

ABSTRACT

Small-molecule-induced protein depletion technologies, also called inducible degrons, allow degradation of genetically engineered target proteins within cells and animals. Here, we design and develop the BromoTag, a new inducible degron system comprising a Brd4 bromodomain L387A variant as a degron tag that allows direct recruitment by heterobifunctional bumped proteolysis targeting chimeras (PROTACs) to hijack the VHL E3 ligase. We describe extensive optimization and structure-activity relationships of our bump-and-hole-PROTACs using a CRISPR knock-in cell line expressing model target BromoTag-Brd2 at endogenous levels. Collectively, our cellular and mechanistic data qualifies bumped PROTAC AGB1 as a potent, fast, and selective degrader of BromoTagged proteins, with a favorable pharmacokinetic profile in mice. The BromoTag adds to the arsenal of chemical genetic degradation tools allowing us to manipulate protein levels to interrogate the biological function and therapeutic potential in cells and in vivo.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Development , Proteolysis/drug effects , Transcription Factors/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship , Transcription Factors/metabolism
12.
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
13.
Life Sci Alliance ; 4(2)2021 02.
Article in English | MEDLINE | ID: mdl-33361109

ABSTRACT

The function of the FAM83F protein, like the functions of many members of the FAM83 family, is poorly understood. Here, we show that injection of Fam83f mRNA into Xenopus embryos causes axis duplication, a phenotype indicative of enhanced Wnt signalling. Consistent with this, overexpression of FAM83F activates Wnt signalling, whereas ablation of FAM83F from human colorectal cancer (CRC) cells attenuates it. We demonstrate that FAM83F is farnesylated and interacts and co-localises with CK1α at the plasma membrane. This interaction with CK1α is essential for FAM83F to activate Wnt signalling, and FAM83F mutants that do not interact with CK1α fail to induce axis duplication in Xenopus embryos and to activate Wnt signalling in cells. FAM83F acts upstream of GSK-3ß because the attenuation of Wnt signalling caused by loss of FAM83F can be rescued by GSK-3 inhibition. Introduction of a farnesyl-deficient mutant of FAM83F in cells through CRISPR/Cas9 genome editing redirects the FAM83F-CK1α complex away from the plasma membrane and significantly attenuates Wnt signalling, indicating that FAM83F exerts its effects on Wnt signalling at the plasma membrane.


Subject(s)
Casein Kinase Ialpha/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Wnt Signaling Pathway , Animals , Cell Line , Cell Membrane/metabolism , Embryonic Development/genetics , Fluorescent Antibody Technique , Gene Expression Regulation , Gene Knockdown Techniques , Genes, Reporter , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Prenylation , Protein Binding , Protein Transport , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
14.
Life Sci Alliance ; 4(2)2021 02.
Article in English | MEDLINE | ID: mdl-33361334

ABSTRACT

Immunomodulatory imide drugs (IMiDs) bind CRBN, a substrate receptor of the Cul4A E3 ligase complex, enabling the recruitment of neo-substrates, such as CK1α, and their degradation via the ubiquitinproteasome system. Here, we report FAM83F as such a neo-substrate. The eight FAM83 proteins (A-H) interact with and regulate the subcellular distribution of CK1α. We demonstrate that IMiD-induced FAM83F degradation requires its association with CK1α. However, no other FAM83 protein is degraded by IMiDs. We have recently identified FAM83F as a mediator of the canonical Wnt signalling pathway. The IMiD-induced degradation of FAM83F attenuated Wnt signalling in colorectal cancer cells and removed CK1α from the plasma membrane, mirroring the phenotypes observed with genetic ablation of FAM83F. Intriguingly, the expression of FAM83G, which also binds to CK1α, appears to attenuate the IMiD-induced degradation of CK1α, suggesting a protective role for FAM83G on CK1α. Our findings reveal that the efficiency and extent of target protein degradation by IMiDs depends on the nature of inherent multiprotein complex in which the target protein is part of.


Subject(s)
Casein Kinase Ialpha/metabolism , Imides/pharmacology , Immunologic Factors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Wnt Signaling Pathway/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Base Sequence , Cell Line, Tumor , Cell Membrane/metabolism , Gene Knock-In Techniques , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proteolysis/drug effects , Ubiquitin-Protein Ligases/metabolism
15.
Dev Cell ; 55(5): 629-647.e7, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33080171

ABSTRACT

Conserved protein kinases with core cellular functions have been frequently redeployed during metazoan evolution to regulate specialized developmental processes. The Ser/Arg (SR)-rich splicing factor (SRSF) protein kinase (SRPK), which is implicated in splicing regulation, is one such conserved eukaryotic kinase. Surprisingly, we show that SRPK has acquired the capacity to control a neurodevelopmental ubiquitin signaling pathway. In mammalian embryonic stem cells and cultured neurons, SRPK phosphorylates Ser-Arg motifs in RNF12/RLIM, a key developmental E3 ubiquitin ligase that is mutated in an intellectual disability syndrome. Processive phosphorylation by SRPK stimulates RNF12-dependent ubiquitylation of nuclear transcription factor substrates, thereby acting to restrain a neural gene expression program that is aberrantly expressed in intellectual disability. SRPK family genes are also mutated in intellectual disability disorders, and patient-derived SRPK point mutations impair RNF12 phosphorylation. Our data reveal unappreciated functional diversification of SRPK to regulate ubiquitin signaling that ensures correct regulation of neurodevelopmental gene expression.


Subject(s)
Nervous System/embryology , Nervous System/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Gene Expression Regulation, Developmental , Humans , Intellectual Disability/genetics , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Mutation/genetics , Neurons/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Proteolysis , Substrate Specificity , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
16.
Neuroscience ; 448: 234-254, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32890664

ABSTRACT

Both rare, high risk, loss-of-function mutations and common, low risk, genetic variants in the CUL3 gene are strongly associated with neuropsychiatric disorders. Network analyses of neuropsychiatric risk genes have shown high CUL3 expression in the prenatal human brain and an enrichment in neural precursor cells (NPCs) and cortical neurons. The role of CUL3 in human neurodevelopment however, is poorly understood. In the present study, we used CRISPR/Cas9 nickase to knockout CUL3 in human induced pluripotent stem cells (iPSCs). iPSCs were subsequently differentiated into cortical glutamatergic neurons using two different protocols and tested for structural/functional alterations. Immunocytochemical analysis and transcriptomic profiling revealed that pluripotency of heterozygous CUL3 knockout (KO) iPSCs remained unchanged compared to isogenic control iPSCs. Following small molecule-mediated differentiation into cortical glutamatergic neurons however, we detected a significant delay in transition from proliferating radial glia cells/NPCs to postmitotic neurons in CUL3 KO cultures. Notably, direct neural conversion of CUL3 KO iPSCs by lentiviral expression of Neurogenin-2 massively attenuated the neurodevelopmental delay. However, both optogenetic and electrical stimulation of induced neurons revealed decreased excitability in Cullin-3 deficient cultures, while basal synaptic transmission remained unchanged. Analysis of target gene expression pointed to alterations in FGF signaling in CUL3 KO NPCs, which is required for NPC proliferation and self-renewal, while RhoA and Notch signaling appeared unaffected. Our data provide first evidence for a major role of Cullin-3 in neuronal differentiation, and for neurodevelopmental deficits underlying neuropsychiatric disorders associated with CUL3 mutations.


Subject(s)
Induced Pluripotent Stem Cells , Mental Disorders , Neural Stem Cells , Cell Differentiation , Cullin Proteins/genetics , Female , Humans , Mutation , Pregnancy
17.
Cell Chem Biol ; 27(9): 1151-1163.e6, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32668202

ABSTRACT

K-RAS is known as the most frequently mutated oncogene. However, the development of conventional K-RAS inhibitors has been extremely challenging, with a mutation-specific inhibitor reaching clinical trials only recently. Targeted proteolysis has emerged as a new modality in drug discovery to tackle undruggable targets. Our laboratory has developed a system for targeted proteolysis using peptidic high-affinity binders, called "AdPROM." Here, we used CRISPR/Cas9 technology to knock in a GFP tag on the native K-RAS gene in A549 adenocarcinoma (A549GFPKRAS) cells and constructed AdPROMs containing high-affinity GFP or H/K-RAS binders. Expression of GFP-targeting AdPROM in A549GFPKRAS led to robust proteasomal degradation of endogenous GFP-K-RAS, while expression of anti-HRAS-targeting AdPROM in different cell lines resulted in the degradation of both GFP-tagged and untagged K-RAS, and untagged H-RAS. Our findings imply that endogenous RAS proteins can be targeted for proteolysis, supporting the idea of an alternative therapeutic approach to these undruggable targets.


Subject(s)
Proteolysis , Proto-Oncogene Proteins p21(ras)/metabolism , A549 Cells , Affinity Labels , CRISPR-Cas Systems/genetics , Cell Line , Cell Proliferation , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Peptides/chemistry , Peptides/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
Cell Chem Biol ; 27(9): 1164-1180.e5, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32668203

ABSTRACT

The affinity-directed protein missile (AdPROM) system utilizes specific polypeptide binders of intracellular proteins of interest (POIs) conjugated to an E3 ubiquitin ligase moiety to enable targeted proteolysis of the POI. However, a chemically tuneable AdPROM system is more desirable. Here, we use Halo-tag/VHL-recruiting proteolysis-targeting chimera (HaloPROTAC) technology to develop a ligand-inducible AdPROM (L-AdPROM) system. When we express an L-AdPROM construct consisting of an anti-GFP nanobody conjugated to the Halo-tag, we achieve robust degradation of GFP-tagged POIs only upon treatment of cells with the HaloPROTAC. For GFP-tagged POIs, ULK1, FAM83D, and SGK3 were knocked in with a GFP-tag using CRISPR/Cas9. By substituting the anti-GFP nanobody for a monobody that binds H- and K-RAS, we achieve robust degradation of unmodified endogenous RAS proteins only in the presence of the HaloPROTAC. Through substitution of the polypeptide binder, the highly versatile L-AdPROM system is useful for the inducible degradation of potentially any intracellular POI.


Subject(s)
Proteolysis , Single-Chain Antibodies/metabolism , Affinity Labels , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , CRISPR-Cas Systems/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Green Fluorescent Proteins/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Ubiquitination , ras Proteins/metabolism
19.
Sci Rep ; 10(1): 6580, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313105

ABSTRACT

Tumor invasion requires efficient cell migration, which is achieved by the generation of persistent and polarized lamellipodia. The generation of lamellipodia is supported by actin dynamics at the leading edge where a complex of proteins known as the WAVE regulatory complex (WRC) promotes the required assembly of actin filaments to push the front of the cell ahead. By using an U2OS osteosarcoma cell line with high metastatic potential, proven by a xenotransplant in zebrafish larvae, we have studied the role of the plasma membrane Ca2+ channel ORAI1 in this process. We have found that epidermal growth factor (EGF) triggered an enrichment of ORAI1 at the leading edge, where colocalized with cortactin (CTTN) and other members of the WRC, such as CYFIP1 and ARP2/3. ORAI1-CTTN co-precipitation was sensitive to the inhibition of the small GTPase RAC1, an upstream activator of the WRC. RAC1 potentiated ORAI1 translocation to the leading edge, increasing the availability of surface ORAI1 and increasing the plasma membrane ruffling. The role of ORAI1 at the leading edge was studied in genetically engineered U2OS cells lacking ORAI1 expression that helped us to prove the key role of this Ca2+ channel on lamellipodia formation, lamellipodial persistence, and cell directness, which are required for tumor cell invasiveness in vivo.


Subject(s)
Cortactin/genetics , ORAI1 Protein/genetics , Osteosarcoma/genetics , Pseudopodia/genetics , rac1 GTP-Binding Protein/genetics , Actin Cytoskeleton/genetics , Actin-Related Protein 2-3 Complex/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Pseudopodia/metabolism
20.
Cell Signal ; 72: 109632, 2020 08.
Article in English | MEDLINE | ID: mdl-32289446

ABSTRACT

The majority of mutations identified in patients with amelogenesis imperfecta have been mapped to FAM83H. As FAM83H expression is not limited to the enamel, how FAM83H contributes to amelogenesis is still largely unknown. We previously reported that members of the FAM83 family of proteins interact with and regulate the subcellular distribution of the promiscuous serine-threonine protein kinase CK1 family, through their shared N-terminal DUF1669 domains. FAM83H co-localises with CK1 isoforms to speckle-like structures in both the cytoplasm and nucleus. In this report, we show FAM83H, unlike other FAM83 proteins, interacts and colocalises with NCK1/2 tyrosine kinase adaptor proteins. This interaction is mediated by proline-rich motifs within the C-terminus of FAM83H, specifically interacting with the second and third SH3 domains of NCK1/2. Moreover, FAM83H pathogenic AI mutant proteins, which trigger C-terminal truncations of FAM83H, retain their interactions with CK1 isoforms but lose interaction with NCK1/2. These AI mutant FAM83H proteins acquire a nuclear localisation, and recruit CK1 isoforms to the nucleus where CK1 retains its kinase activity. As understanding the constituents of the FAM83H-localised speckles may hold the key to unravelling potential substrates of FAM83H-associated CK1 substrates, we employed a TurboID-based proximity labelling approach and uncovered several proteins including Iporin and BAG3 as potential constituents of the speckles.


Subject(s)
Amelogenesis Imperfecta/genetics , Mutation/genetics , Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , HEK293 Cells , Humans , Oncogene Proteins/metabolism , Protein Binding , Protein Domains , Proteins/chemistry , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...