202 results on '"Schulman BA"'
Search Results
2. Structural basis for C-degron selectivity across KLHDCX family E3 ubiquitin ligases.
- Author
-
Scott DC, Chittori S, Purser N, King MT, Maiwald SA, Churion K, Nourse A, Lee C, Paulo JA, Miller DJ, Elledge SJ, Harper JW, Kleiger G, and Schulman BA
- Subjects
- Substrate Specificity, Humans, Crystallography, X-Ray, Cryoelectron Microscopy, Protein Binding, Models, Molecular, Ubiquitin metabolism, Amino Acid Motifs, Degrons, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases genetics, Ubiquitination
- Abstract
Specificity of the ubiquitin-proteasome system depends on E3 ligase-substrate interactions. Many such pairings depend on E3 ligases binding to peptide-like sequences - termed N- or C-degrons - at the termini of substrates. However, our knowledge of structural features distinguishing closely related C-degron substrate-E3 pairings is limited. Here, by systematically comparing ubiquitylation activities towards a suite of common model substrates, and defining interactions by biochemistry, crystallography, and cryo-EM, we reveal principles of C-degron recognition across the KLHDCX family of Cullin-RING ligases (CRLs). First, a motif common across these E3 ligases anchors a substrate's C-terminus. However, distinct locations of this C-terminus anchor motif in different blades of the KLHDC2, KLHDC3, and KLHDC10 β-propellers establishes distinct relative positioning and molecular environments for substrate C-termini. Second, our structural data show KLHDC3 has a pre-formed pocket establishing preference for an Arg or Gln preceding a C-terminal Gly, whereas conformational malleability contributes to KLHDC10's recognition of varying features adjacent to substrate C-termini. Finally, additional non-consensus interactions, mediated by C-degron binding grooves and/or by distal propeller surfaces and substrate globular domains, can substantially impact substrate binding and ubiquitylatability. Overall, the data reveal combinatorial mechanisms determining specificity and plasticity of substrate recognition by KLDCX-family C-degron E3 ligases., Competing Interests: Competing interests D.C.S. and B.A.S. are co-inventors of intellectual property that is unrelated to this work (DCN1 inhibitors licensed to Cinsano). J.W.H. is a founder and consultant for Caraway Therapeutics and is a scientific advisory board member for Lyterian Therapeutics. SJE is a founder of, and holds equity in TScan Therapeutics and Immune ID. S.J.E. is also founder of MAZE Therapeutics, and Mirimus and serves on the scientific advisory board of TSCAN Therapeutics, and MAZE Therapeutics. In accordance with Partners HealthCare’s conflict of interest policies, the Partners Office for Interactions with Industry has reviewed SJE’s financial interest in TSCAN and determined that it creates no significant risk to the welfare of participants in this study or to the integrity of this research. B.A.S. is a member of the scientific advisory boards of Biotheryx and Proxygen. The remaining authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. Ubiquitin is a chemist's playground.
- Author
-
Farnung J and Schulman BA
- Published
- 2024
- Full Text
- View/download PDF
4. Global cellular proteo-lipidomic profiling of diverse lysosomal storage disease mutants using nMOST.
- Author
-
Kraus F, He Y, Swarup S, Overmyer KA, Jiang Y, Brenner J, Capitanio C, Bieber A, Jen A, Nightingale NM, Anderson BJ, Lee C, Paulo JA, Smith IR, Plitzko JM, Gygi SP, Schulman BA, Wilfling F, Coon JJ, and Harper JW
- Abstract
Lysosomal storage diseases (LSDs) comprise ~50 monogenic disorders marked by the buildup of cellular material in lysosomes, yet systematic global molecular phenotyping of proteins and lipids is lacking. We present a nanoflow-based multi-omic single-shot technology (nMOST) workflow that quantifies HeLa cell proteomes and lipidomes from over two dozen LSD mutants. Global cross-correlation analysis between lipids and proteins identified autophagy defects, notably the accumulation of ferritinophagy substrates and receptors, especially in NPC1
-/- and NPC2-/- mutants, where lysosomes accumulate cholesterol. Autophagic and endocytic cargo delivery failures correlated with elevated lyso-phosphatidylcholine species and multi-lamellar structures visualized by cryo-electron tomography. Loss of mitochondrial cristae, MICOS-complex components, and OXPHOS components rich in iron-sulfur cluster proteins in NPC2-/- cells was largely alleviated when iron was provided through the transferrin system. This study reveals how lysosomal dysfunction affects mitochondrial homeostasis and underscores nMOST as a valuable discovery tool for identifying molecular phenotypes across LSDs., Competing Interests: DECLARATION OF INTERESTS J.W.H. is a consultant and founder of Caraway Therapeutics (a wholly owned subsidiary of Merck & Co, Inc) and is a member of the scientific advisory board for Lyterian Therapeutics. B.A.S. is a co-founding scientific advisory board member of Interline Therapeutics and on the scientific advisory boards of Biotheryx and Proxygen. J.M.P. holds a position on the advisory board of Thermo Fisher Scientific. J.J.C. is a consultant for Thermo Fischer Scientific. Other authors declare no competing interests. S.P.G. is on the advisory board for Thermo Fisher Scientific, Cedilla Therapeutics, Casma Therapeutics, Cell Signaling Technology, and Frontier Medicines.- Published
- 2024
- Full Text
- View/download PDF
5. Principles of paralog-specific targeted protein degradation engaging the C-degron E3 KLHDC2.
- Author
-
Scott DC, Dharuman S, Griffith E, Chai SC, Ronnebaum J, King MT, Tangallapally R, Lee C, Gee CT, Yang L, Li Y, Loudon VC, Lee HW, Ochoada J, Miller DJ, Jayasinghe T, Paulo JA, Elledge SJ, Harper JW, Chen T, Lee RE, and Schulman BA
- Subjects
- Humans, HEK293 Cells, Binding Sites, Ligands, Ubiquitination, Substrate Specificity, Protein Binding, Triazoles chemistry, Triazoles pharmacology, Triazoles metabolism, Ubiquitin metabolism, Azepines pharmacology, Azepines chemistry, Azepines metabolism, Cell Cycle Proteins metabolism, Cell Cycle Proteins genetics, Degrons, Proteolysis, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Transcription Factors metabolism, Transcription Factors genetics
- Abstract
PROTAC® (proteolysis-targeting chimera) molecules induce proximity between an E3 ligase and protein-of-interest (POI) to target the POI for ubiquitin-mediated degradation. Cooperative E3-PROTAC-POI complexes have potential to achieve neo-substrate selectivity beyond that established by POI binding to the ligand alone. Here, we extend the collection of ubiquitin ligases employable for cooperative ternary complex formation to include the C-degron E3 KLHDC2. Ligands were identified that engage the C-degron binding site in KLHDC2, subjected to structure-based improvement, and linked to JQ1 for BET-family neo-substrate recruitment. Consideration of the exit vector emanating from the ligand engaged in KLHDC2's U-shaped degron-binding pocket enabled generation of SJ46421, which drives formation of a remarkably cooperative, paralog-selective ternary complex with BRD3
BD2 . Meanwhile, screening pro-drug variants enabled surmounting cell permeability limitations imposed by acidic moieties resembling the KLHDC2-binding C-degron. Selectivity for BRD3 compared to other BET-family members is further manifested in ubiquitylation in vitro, and prodrug version SJ46420-mediated degradation in cells. Selectivity is also achieved for the ubiquitin ligase, overcoming E3 auto-inhibition to engage KLHDC2, but not the related KLHDC1, KLHDC3, or KLHDC10 E3s. In sum, our study establishes neo-substrate-specific targeted protein degradation via KLHDC2, and provides a framework for developing selective PROTAC protein degraders employing C-degron E3 ligases., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. Visualizing chaperone-mediated multistep assembly of the human 20S proteasome.
- Author
-
Adolf F, Du J, Goodall EA, Walsh RM Jr, Rawson S, von Gronau S, Harper JW, Hanna J, and Schulman BA
- Subjects
- Humans, Protein Conformation, Proteasome Endopeptidase Complex metabolism, Proteasome Endopeptidase Complex chemistry, Cryoelectron Microscopy, Molecular Chaperones metabolism, Molecular Chaperones chemistry, Models, Molecular
- Abstract
Dedicated assembly factors orchestrate the stepwise production of many molecular machines, including the 28-subunit proteasome core particle (CP) that mediates protein degradation. Here we report cryo-electron microscopy reconstructions of seven recombinant human subcomplexes that visualize all five chaperones and the three active site propeptides across a wide swath of the assembly pathway. Comparison of these chaperone-bound intermediates and a matching mature CP reveals molecular mechanisms determining the order of successive subunit additions, as well as how proteasome subcomplexes and assembly factors structurally adapt upon progressive subunit incorporation to stabilize intermediates, facilitate the formation of subsequent intermediates and ultimately rearrange to coordinate proteolytic activation with gated access to active sites. This work establishes a methodologic approach for structural analysis of multiprotein complex assembly intermediates, illuminates specific functions of assembly factors and reveals conceptual principles underlying human proteasome biogenesis, thus providing an explanation for many previous biochemical and genetic observations., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Monovalent Pseudo-Natural Product Degraders Supercharge the Native Degradation of IDO1 by KLHDC3.
- Author
-
Hennes E, Lucas B, Scholes NS, Cheng XF, Scott DC, Bischoff M, Reich K, Gasper R, Lucas M, Xu TT, Pulvermacher LM, Dötsch L, Imrichova H, Brause A, Naredla KR, Sievers S, Kumar K, Janning P, Gersch M, Murray PJ, Schulman BA, Winter GE, Ziegler S, and Waldmann H
- Abstract
Targeted protein degradation (TPD) modulates protein function beyond inhibition of enzyme activity or protein-protein interactions. Most degraders function by proximity induction, and directly bridge an E3 ligase with the target to be degraded. However, many proteins might not be addressable via proximity-based degraders, and other challenges, such as resistance acquisition, exist. Here, we identified pseudo-natural products derived from (-)-myrtanol, termed iDegs, that inhibit and induce degradation of the immunomodulatory enzyme indoleamine-2,3-dioxygenase 1 (IDO1) by a distinct mechanism. iDegs induce a unique conformational change and, thereby, boost IDO1 ubiquitination and degradation by the cullin-RING E3 ligase CRL2
KLHDC3 , which we identified to also mediate native IDO1 degradation. Therefore, iDegs supercharge the native proteolytic pathway of IDO1, rendering this mechanism of action distinct from traditional degrader approaches involving proteolysis-targeting chimeras (PROTACs) or molecular-glue degraders (MGDs). In contrast to clinically explored IDO1 inhibitors, iDegs reduce formation of kynurenine by both inhibition and induced degradation of the enzyme and should also modulate non-enzymatic functions of IDO1. This unique mechanism of action may open up new therapeutic opportunities for the treatment of cancer beyond classical inhibition of IDO1.- Published
- 2024
- Full Text
- View/download PDF
8. Noncanonical assembly, neddylation and chimeric cullin-RING/RBR ubiquitylation by the 1.8 MDa CUL9 E3 ligase complex.
- Author
-
Horn-Ghetko D, Hopf LVM, Tripathi-Giesgen I, Du J, Kostrhon S, Vu DT, Beier V, Steigenberger B, Prabu JR, Stier L, Bruss EM, Mann M, Xiong Y, and Schulman BA
- Subjects
- Humans, Carrier Proteins metabolism, Carrier Proteins chemistry, Carrier Proteins genetics, HEK293 Cells, Models, Molecular, NEDD8 Protein metabolism, NEDD8 Protein genetics, NEDD8 Protein chemistry, Protein Binding, Protein Multimerization, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases genetics, Cryoelectron Microscopy, Cullin Proteins metabolism, Cullin Proteins chemistry, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitination
- Abstract
Ubiquitin ligation is typically executed by hallmark E3 catalytic domains. Two such domains, 'cullin-RING' and 'RBR', are individually found in several hundred human E3 ligases, and collaborate with E2 enzymes to catalyze ubiquitylation. However, the vertebrate-specific CUL9 complex with RBX1 (also called ROC1), of interest due to its tumor suppressive interaction with TP53, uniquely encompasses both cullin-RING and RBR domains. Here, cryo-EM, biochemistry and cellular assays elucidate a 1.8-MDa hexameric human CUL9-RBX1 assembly. Within one dimeric subcomplex, an E2-bound RBR domain is activated by neddylation of its own cullin domain and positioning from the adjacent CUL9-RBX1 in trans. Our data show CUL9 as unique among RBX1-bound cullins in dependence on the metazoan-specific UBE2F neddylation enzyme, while the RBR domain protects it from deneddylation. Substrates are recruited to various upstream domains, while ubiquitylation relies on both CUL9's neddylated cullin and RBR domains achieving self-assembled and chimeric cullin-RING/RBR E3 ligase activity., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Alkylamine-tethered molecules recruit FBXO22 for targeted protein degradation.
- Author
-
Kagiou C, Cisneros JA, Farnung J, Liwocha J, Offensperger F, Dong K, Yang K, Tin G, Horstmann CS, Hinterndorfer M, Paulo JA, Scholes NS, Sanchez Avila J, Fellner M, Andersch F, Hannich JT, Zuber J, Kubicek S, Gygi SP, Schulman BA, and Winter GE
- Subjects
- Humans, HEK293 Cells, Tacrolimus Binding Protein 1A metabolism, Tacrolimus Binding Protein 1A genetics, Ubiquitin-Protein Ligases metabolism, Amines metabolism, Amines chemistry, Proteasome Endopeptidase Complex metabolism, Ligands, Receptors, Cytoplasmic and Nuclear, Proteolysis, Ubiquitination, F-Box Proteins metabolism, F-Box Proteins chemistry
- Abstract
Targeted protein degradation (TPD) relies on small molecules to recruit proteins to E3 ligases to induce their ubiquitylation and degradation by the proteasome. Only a few of the approximately 600 human E3 ligases are currently amenable to this strategy. This limits the actionable target space and clinical opportunities and thus establishes the necessity to expand to additional ligases. Here we identify and characterize SP3N, a specific degrader of the prolyl isomerase FKBP12. SP3N features a minimal design, where a known FKBP12 ligand is appended with a flexible alkylamine tail that conveys degradation properties. We found that SP3N is a precursor and that the alkylamine is metabolized to an active aldehyde species that recruits the SCF
FBXO22 ligase for FKBP12 degradation. Target engagement occurs via covalent adduction of Cys326 in the FBXO22 C-terminal domain, which is critical for ternary complex formation, ubiquitylation and degradation. This mechanism is conserved for two recently reported alkylamine-based degraders of NSD2 and XIAP, thus establishing alkylamine tethering and covalent hijacking of FBXO22 as a generalizable TPD strategy., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
10. mTORC1-CTLH E3 ligase regulates the degradation of HMG-CoA synthase 1 through the Pro/N-degron pathway.
- Author
-
Yi SA, Sepic S, Schulman BA, Ordureau A, and An H
- Subjects
- Humans, HEK293 Cells, Proteasome Endopeptidase Complex metabolism, Proteasome Endopeptidase Complex genetics, TOR Serine-Threonine Kinases metabolism, TOR Serine-Threonine Kinases genetics, Mevalonic Acid metabolism, Multiprotein Complexes metabolism, Multiprotein Complexes genetics, Signal Transduction, Degrons, Adaptor Proteins, Signal Transducing, Mechanistic Target of Rapamycin Complex 1 metabolism, Mechanistic Target of Rapamycin Complex 1 genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Proteolysis, Hydroxymethylglutaryl-CoA Synthase metabolism, Hydroxymethylglutaryl-CoA Synthase genetics, Ubiquitination, Cell Proliferation
- Abstract
Mammalian target of rapamycin (mTOR) senses changes in nutrient status and stimulates the autophagic process to recycle amino acids. However, the impact of nutrient stress on protein degradation beyond autophagic turnover is incompletely understood. We report that several metabolic enzymes are proteasomal targets regulated by mTOR activity based on comparative proteome degradation analysis. In particular, 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) synthase 1 (HMGCS1), the initial enzyme in the mevalonate pathway, exhibits the most significant half-life adaptation. Degradation of HMGCS1 is regulated by the C-terminal to LisH (CTLH) E3 ligase through the Pro/N-degron motif. HMGCS1 is ubiquitylated on two C-terminal lysines during mTORC1 inhibition, and efficient degradation of HMGCS1 in cells requires a muskelin adaptor. Importantly, modulating HMGCS1 abundance has a dose-dependent impact on cell proliferation, which is restored by adding a mevalonate intermediate. Overall, our unbiased degradomics study provides new insights into mTORC1 function in cellular metabolism: mTORC1 regulates the stability of limiting metabolic enzymes through the ubiquitin system., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
11. Non-canonical substrate recognition by the human WDR26-CTLH E3 ligase regulates prodrug metabolism.
- Author
-
Gottemukkala KV, Chrustowicz J, Sherpa D, Sepic S, Vu DT, Karayel Ö, Papadopoulou EC, Gross A, Schorpp K, von Gronau S, Hadian K, Murray PJ, Mann M, Schulman BA, and Alpi AF
- Subjects
- Humans, Cryoelectron Microscopy, HEK293 Cells, Protein Binding, Substrate Specificity, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Nicotinamide-Nucleotide Adenylyltransferase metabolism, Nicotinamide-Nucleotide Adenylyltransferase genetics, Prodrugs metabolism, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitination
- Abstract
The yeast glucose-induced degradation-deficient (GID) E3 ubiquitin ligase forms a suite of complexes with interchangeable receptors that selectively recruit N-terminal degron motifs of metabolic enzyme substrates. The orthologous higher eukaryotic C-terminal to LisH (CTLH) E3 complex has been proposed to also recognize substrates through an alternative subunit, WDR26, which promotes the formation of supramolecular CTLH E3 assemblies. Here, we discover that human WDR26 binds the metabolic enzyme nicotinamide/nicotinic-acid-mononucleotide-adenylyltransferase 1 (NMNAT1) and mediates its CTLH E3-dependent ubiquitylation independently of canonical GID/CTLH E3-family substrate receptors. The CTLH subunit YPEL5 inhibits NMNAT1 ubiquitylation and cellular turnover by WDR26-CTLH E3, thereby affecting NMNAT1-mediated metabolic activation and cytotoxicity of the prodrug tiazofurin. Cryoelectron microscopy (cryo-EM) structures of NMNAT1- and YPEL5-bound WDR26-CTLH E3 complexes reveal an internal basic degron motif of NMNAT1 essential for targeting by WDR26-CTLH E3 and degron mimicry by YPEL5's N terminus antagonizing substrate binding. Thus, our data provide a mechanistic understanding of how YPEL5-WDR26-CTLH E3 acts as a modulator of NMNAT1-dependent metabolism., Competing Interests: Declaration of interests B.A.S. is a member of the scientific advisory boards of Proxygen and BioTheryX and a co-inventor of intellectual property licensed to Cinsano. P.J.M. is a member of the scientific advisory boards of Palleon Pharmaceuticals and ImCheck Pharma. These relationships have no bearing on or relevance to this work., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Skraban-Deardorff intellectual disability syndrome-associated mutations in WDR26 impair CTLH E3 complex assembly.
- Author
-
Gross A, Müller J, Chrustowicz J, Strasser A, Gottemukkala KV, Sherpa D, Schulman BA, Murray PJ, and Alpi AF
- Subjects
- Humans, HEK293 Cells, Models, Molecular, Adaptor Proteins, Signal Transducing genetics, Intellectual Disability genetics, Intellectual Disability metabolism, Mutation, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases chemistry
- Abstract
Patients with Skraban-Deardorff syndrome (SKDEAS), a neurodevelopmental syndrome associated with a spectrum of developmental and intellectual delays and disabilities, harbor diverse mutations in WDR26, encoding a subunit of the multiprotein CTLH E3 ubiquitin ligase complex. Structural studies revealed that homodimers of WDR26 bridge two core-CTLH E3 complexes to generate giant, hollow oval-shaped supramolecular CTLH E3 assemblies. Additionally, WDR26 mediates CTLH E3 complex binding to subunit YPEL5 and functions as substrate receptor for the transcriptional repressor HBP1. Here, we mapped SKDEAS-associated mutations on a WDR26 structural model and tested their functionality in complementation studies using genetically engineered human cells lacking CTLH E3 supramolecular assemblies. Despite the diversity of mutations, 15 of 16 tested mutants impaired at least one CTLH E3 complex function contributing to complex assembly and interactions, thus providing first mechanistic insights into SKDEAS pathology., (© 2024 The Authors. FEBS Letters published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
13. Cullin-RING ligases employ geometrically optimized catalytic partners for substrate targeting.
- Author
-
Li J, Purser N, Liwocha J, Scott DC, Byers HA, Steigenberger B, Hill S, Tripathi-Giesgen I, Hinkle T, Hansen FM, Prabu JR, Radhakrishnan SK, Kirkpatrick DS, Reichermeier KM, Schulman BA, and Kleiger G
- Subjects
- Humans, Ubiquitination, Ubiquitin metabolism, Polyubiquitin metabolism, Carrier Proteins metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Cullin Proteins genetics, Cullin Proteins metabolism
- Abstract
Cullin-RING ligases (CRLs) ubiquitylate specific substrates selected from other cellular proteins. Substrate discrimination and ubiquitin transferase activity were thought to be strictly separated. Substrates are recognized by substrate receptors, such as Fbox or BCbox proteins. Meanwhile, CRLs employ assorted ubiquitin-carrying enzymes (UCEs, which are a collection of E2 and ARIH-family E3s) specialized for either initial substrate ubiquitylation (priming) or forging poly-ubiquitin chains. We discovered specific human CRL-UCE pairings governing substrate priming. The results reveal pairing of CUL2-based CRLs and UBE2R-family UCEs in cells, essential for efficient PROTAC-induced neo-substrate degradation. Despite UBE2R2's intrinsic programming to catalyze poly-ubiquitylation, CUL2 employs this UCE for geometrically precise PROTAC-dependent ubiquitylation of a neo-substrate and for rapid priming of substrates recruited to diverse receptors. Cryo-EM structures illuminate how CUL2-based CRLs engage UBE2R2 to activate substrate ubiquitylation. Thus, pairing with a specific UCE overcomes E2 catalytic limitations to drive substrate ubiquitylation and targeted protein degradation., Competing Interests: Declaration of interests B.A.S. is a member of the scientific advisory boards of Interline Therapeutics and BioTheryX and co-inventor of intellectual property licensed to Cinsano. D.S.K. and K.M.R. were employees of Genentech/Roche at the time the work was completed. T.H. is currently an employee of Genentech/Roche. D.S.K. is an employee and shareholder of Orion Medicines. S.H. is currently an employee and shareholder of Amgen., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Combinatorial selective ER-phagy remodels the ER during neurogenesis.
- Author
-
Hoyer MJ, Capitanio C, Smith IR, Paoli JC, Bieber A, Jiang Y, Paulo JA, Gonzalez-Lozano MA, Baumeister W, Wilfling F, Schulman BA, and Harper JW
- Subjects
- Humans, Endoplasmic Reticulum metabolism, Autophagy physiology, Endoplasmic Reticulum Stress, Carrier Proteins metabolism, Neurogenesis, Proteome, Proteomics
- Abstract
The endoplasmic reticulum (ER) employs a diverse proteome landscape to orchestrate many cellular functions, ranging from protein and lipid synthesis to calcium ion flux and inter-organelle communication. A case in point concerns the process of neurogenesis, where a refined tubular ER network is assembled via ER shaping proteins into the newly formed neuronal projections to create highly polarized dendrites and axons. Previous studies have suggested a role for autophagy in ER remodelling, as autophagy-deficient neurons in vivo display axonal ER accumulation within synaptic boutons, and the membrane-embedded ER-phagy receptor FAM134B has been genetically linked with human sensory and autonomic neuropathy. However, our understanding of the mechanisms underlying selective removal of the ER and the role of individual ER-phagy receptors is limited. Here we combine a genetically tractable induced neuron (iNeuron) system for monitoring ER remodelling during in vitro differentiation with proteomic and computational tools to create a quantitative landscape of ER proteome remodelling via selective autophagy. Through analysis of single and combinatorial ER-phagy receptor mutants, we delineate the extent to which each receptor contributes to both the magnitude and selectivity of ER protein clearance. We define specific subsets of ER membrane or lumenal proteins as preferred clients for distinct receptors. Using spatial sensors and flux reporters, we demonstrate receptor-specific autophagic capture of ER in axons, and directly visualize tubular ER membranes within autophagosomes in neuronal projections by cryo-electron tomography. This molecular inventory of ER proteome remodelling and versatile genetic toolkit provide a quantitative framework for understanding the contributions of individual ER-phagy receptors for reshaping ER during cell state transitions., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. Structural snapshots along K48-linked ubiquitin chain formation by the HECT E3 UBR5.
- Author
-
Hehl LA, Horn-Ghetko D, Prabu JR, Vollrath R, Vu DT, Pérez Berrocal DA, Mulder MPC, van der Heden van Noort GJ, and Schulman BA
- Subjects
- Humans, Cryoelectron Microscopy, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitins metabolism, Ubiquitination, Ubiquitin chemistry, Ubiquitin-Protein Ligases metabolism
- Abstract
Ubiquitin (Ub) chain formation by homologous to E6AP C-terminus (HECT)-family E3 ligases regulates vast biology, yet the structural mechanisms remain unknown. We used chemistry and cryo-electron microscopy (cryo-EM) to visualize stable mimics of the intermediates along K48-linked Ub chain formation by the human E3, UBR5. The structural data reveal a ≈ 620 kDa UBR5 dimer as the functional unit, comprising a scaffold with flexibly tethered Ub-associated (UBA) domains, and elaborately arranged HECT domains. Chains are forged by a UBA domain capturing an acceptor Ub, with its K48 lured into the active site by numerous interactions between the acceptor Ub, manifold UBR5 elements and the donor Ub. The cryo-EM reconstructions allow defining conserved HECT domain conformations catalyzing Ub transfer from E2 to E3 and from E3. Our data show how a full-length E3, ubiquitins to be adjoined, E2 and intermediary products guide a feed-forward HECT domain conformational cycle establishing a highly efficient, broadly targeting, K48-linked Ub chain forging machine., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Mechanism of millisecond Lys48-linked poly-ubiquitin chain formation by cullin-RING ligases.
- Author
-
Liwocha J, Li J, Purser N, Rattanasopa C, Maiwald S, Krist DT, Scott DC, Steigenberger B, Prabu JR, Schulman BA, and Kleiger G
- Subjects
- Ubiquitin-Protein Ligases metabolism, Ubiquitination, Ubiquitin metabolism, Polyubiquitin metabolism, Cullin Proteins metabolism, Ubiquitin-Conjugating Enzymes metabolism
- Abstract
E3 ubiquitin ligases, in collaboration with E2 ubiquitin-conjugating enzymes, modify proteins with poly-ubiquitin chains. Cullin-RING ligase (CRL) E3s use Cdc34/UBE2R-family E2s to build Lys48-linked poly-ubiquitin chains to control an enormous swath of eukaryotic biology. Yet the molecular mechanisms underlying this exceptional linkage specificity and millisecond kinetics of poly-ubiquitylation remain unclear. Here we obtain cryogenic-electron microscopy (cryo-EM) structures that provide pertinent insight into how such poly-ubiquitin chains are forged. The CRL RING domain not only activates the E2-bound ubiquitin but also shapes the conformation of a distinctive UBE2R2 loop, positioning both the ubiquitin to be transferred and the substrate-linked acceptor ubiquitin within the active site. The structures also reveal how the ubiquitin-like protein NEDD8 uniquely activates CRLs during chain formation. NEDD8 releases the RING domain from the CRL, but unlike previous CRL-E2 structures, does not contact UBE2R2. These findings suggest how poly-ubiquitylation may be accomplished by many E2s and E3s., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. Multisite phosphorylation dictates selective E2-E3 pairing as revealed by Ubc8/UBE2H-GID/CTLH assemblies.
- Author
-
Chrustowicz J, Sherpa D, Li J, Langlois CR, Papadopoulou EC, Vu DT, Hehl LA, Karayel Ö, Beier V, von Gronau S, Müller J, Prabu JR, Mann M, Kleiger G, Alpi AF, and Schulman BA
- Subjects
- Humans, Phosphorylation, Cryoelectron Microscopy, Ubiquitin-Protein Ligases metabolism, Ubiquitination, Ubiquitin-Conjugating Enzymes metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism
- Abstract
Ubiquitylation is catalyzed by coordinated actions of E3 and E2 enzymes. Molecular principles governing many important E3-E2 partnerships remain unknown, including those for RING-family GID/CTLH E3 ubiquitin ligases and their dedicated E2, Ubc8/UBE2H (yeast/human nomenclature). GID/CTLH-Ubc8/UBE2H-mediated ubiquitylation regulates biological processes ranging from yeast metabolic signaling to human development. Here, cryoelectron microscopy (cryo-EM), biochemistry, and cell biology reveal this exquisitely specific E3-E2 pairing through an unconventional catalytic assembly and auxiliary interactions 70-100 Å away, mediated by E2 multisite phosphorylation. Rather than dynamic polyelectrostatic interactions reported for other ubiquitylation complexes, multiple Ubc8/UBE2H phosphorylation sites within acidic CK2-targeted sequences specifically anchor the E2 C termini to E3 basic patches. Positions of phospho-dependent interactions relative to the catalytic domains correlate across evolution. Overall, our data show that phosphorylation-dependent multivalency establishes a specific E3-E2 partnership, is antagonistic with dephosphorylation, rigidifies the catalytic centers within a flexing GID E3-substrate assembly, and facilitates substrate collision with ubiquitylation active sites., Competing Interests: Declaration of interests B.A.S. is adjunct faculty at St. Jude Children’s Research Hospital, member of the scientific advisory boards of BioTheryX and Proxygen, and co-inventor of intellectual property licensed to Cinsano., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Doa10/MARCH6 architecture interconnects E3 ligase activity with lipid-binding transmembrane channel to regulate SQLE.
- Author
-
Botsch JJ, Junker R, Sorgenfrei M, Ogger PP, Stier L, von Gronau S, Murray PJ, Seeger MA, Schulman BA, and Bräuning B
- Subjects
- Ubiquitination, Binding Sites, Saccharomyces cerevisiae genetics, Lipids, Ubiquitin-Protein Ligases genetics, Biological Assay
- Abstract
Transmembrane E3 ligases play crucial roles in homeostasis. Much protein and organelle quality control, and metabolic regulation, are determined by ER-resident MARCH6 E3 ligases, including Doa10 in yeast. Here, we present Doa10/MARCH6 structural analysis by cryo-EM and AlphaFold predictions, and a structure-based mutagenesis campaign. The majority of Doa10/MARCH6 adopts a unique circular structure within the membrane. This channel is established by a lipid-binding scaffold, and gated by a flexible helical bundle. The ubiquitylation active site is positioned over the channel by connections between the cytosolic E3 ligase RING domain and the membrane-spanning scaffold and gate. Here, by assaying 95 MARCH6 variants for effects on stability of the well-characterized substrate SQLE, which regulates cholesterol levels, we reveal crucial roles of the gated channel and RING domain consistent with AlphaFold-models of substrate-engaged and ubiquitylation complexes. SQLE degradation further depends on connections between the channel and RING domain, and lipid binding sites, revealing how interconnected Doa10/MARCH6 elements could orchestrate metabolic signals, substrate binding, and E3 ligase activity., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
19. Recovering from the stress of the COVID-19 pandemic.
- Author
-
Lewis SC, Jourdain AA, Schulman BA, Vousden KH, Fabius JM, and Liu H
- Subjects
- Humans, Pandemics, COVID-19 epidemiology
- Abstract
For our special issue on stress, we asked scientists about recovering from the stress of the pandemic, including some who shared insights with us in mid-2020. They discuss the importance of teamwork, reassessing priorities, and the added stresses of the cost-of-living crisis, funding cuts, and retaining scientists in academia., Competing Interests: Declaration of interests B.A.S. is a member of the scientific advisory board of Biotheryx and is a co-inventor of intellectual property related to DCN1 inhibitors licensed to Cinsano. K.H.V. is a member of the Molecular Cell Advisory Board and is funded by and an advisor for Cancer Research UK., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. K6-linked ubiquitylation marks formaldehyde-induced RNA-protein crosslinks for resolution.
- Author
-
Suryo Rahmanto A, Blum CJ, Scalera C, Heidelberger JB, Mesitov M, Horn-Ghetko D, Gräf JF, Mikicic I, Hobrecht R, Orekhova A, Ostermaier M, Ebersberger S, Möckel MM, Krapoth N, Da Silva Fernandes N, Mizi A, Zhu Y, Chen JX, Choudhary C, Papantonis A, Ulrich HD, Schulman BA, König J, and Beli P
- Subjects
- Humans, Ubiquitination, Formaldehyde toxicity, Aldehydes toxicity, RNA, Messenger genetics, RNA, Messenger metabolism, RNA metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Reactive aldehydes are produced by normal cellular metabolism or after alcohol consumption, and they accumulate in human tissues if aldehyde clearance mechanisms are impaired. Their toxicity has been attributed to the damage they cause to genomic DNA and the subsequent inhibition of transcription and replication. However, whether interference with other cellular processes contributes to aldehyde toxicity has not been investigated. We demonstrate that formaldehyde induces RNA-protein crosslinks (RPCs) that stall the ribosome and inhibit translation in human cells. RPCs in the messenger RNA (mRNA) are recognized by the translating ribosomes, marked by atypical K6-linked ubiquitylation catalyzed by the RING-in-between-RING (RBR) E3 ligase RNF14, and subsequently resolved by the ubiquitin- and ATP-dependent unfoldase VCP. Our findings uncover an evolutionary conserved formaldehyde-induced stress response pathway that protects cells against RPC accumulation in the cytoplasm, and they suggest that RPCs contribute to the cellular and tissue toxicity of reactive aldehydes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Insights into the ISG15 transfer cascade by the UBE1L activating enzyme.
- Author
-
Wallace I, Baek K, Prabu JR, Vollrath R, von Gronau S, Schulman BA, and Swatek KN
- Subjects
- Ubiquitin-Activating Enzymes metabolism, Ubiquitin metabolism, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Viral Proteins, Cytokines metabolism, Ubiquitins genetics, Ubiquitins metabolism
- Abstract
The attachment of the ubiquitin-like protein ISG15 to substrates by specific E1-E2-E3 enzymes is a well-established signalling mechanism of the innate immune response. Here, we present a 3.45 Å cryo-EM structure of a chemically trapped UBE1L-UBE2L6 complex bound to activated ISG15. This structure reveals the details of the first steps of ISG15 recognition and UBE2L6 recruitment by UBE1L (also known as UBA7). Taking advantage of viral effector proteins from severe acute respiratory coronavirus 2 (SARS-CoV-2) and influenza B virus (IBV), we validate the structure and confirm the importance of the ISG15 C-terminal ubiquitin-like domain in the adenylation reaction. Moreover, biochemical characterization of the UBE1L-ISG15 and UBE1L-UBE2L6 interactions enables the design of ISG15 and UBE2L6 mutants with altered selectively for the ISG15 and ubiquitin conjugation pathways. Together, our study helps to define the molecular basis of these interactions and the specificity determinants that ensure the fidelity of ISG15 signalling during the antiviral response., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
22. Activity-based profiling of cullin-RING E3 networks by conformation-specific probes.
- Author
-
Henneberg LT, Singh J, Duda DM, Baek K, Yanishevski D, Murray PJ, Mann M, Sidhu SS, and Schulman BA
- Subjects
- Ubiquitination, Ubiquitin metabolism, Ubiquitins metabolism, NEDD8 Protein metabolism, Cullin Proteins genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
The cullin-RING ubiquitin ligase (CRL) network comprises over 300 unique complexes that switch from inactive to activated conformations upon site-specific cullin modification by the ubiquitin-like protein NEDD8. Assessing cellular repertoires of activated CRL complexes is critical for understanding eukaryotic regulation. However, probes surveying networks controlled by site-specific ubiquitin-like protein modifications are lacking. We developed a synthetic antibody recognizing the active conformation of NEDD8-linked cullins. Implementing the probe to profile cellular networks of activated CUL1-, CUL2-, CUL3- and CUL4-containing E3s revealed the complexes responding to stimuli. Profiling several cell types showed their baseline neddylated CRL repertoires vary, and prime efficiency of targeted protein degradation. Our probe also unveiled differential rewiring of CRL networks across distinct primary cell activation pathways. Thus, conformation-specific probes can permit nonenzymatic activity-based profiling across a system of numerous multiprotein complexes, which in the case of neddylated CRLs reveals widespread regulation and could facilitate the development of degrader drugs., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
23. Catalysis of non-canonical protein ubiquitylation by the ARIH1 ubiquitin ligase.
- Author
-
Purser N, Tripathi-Giesgen I, Li J, Scott DC, Horn-Ghetko D, Baek K, Schulman BA, Alpi AF, and Kleiger G
- Subjects
- Ubiquitination, Proteins metabolism, Catalysis, Ubiquitin genetics, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Protein ubiquitylation typically involves isopeptide bond formation between the C-terminus of ubiquitin to the side-chain amino group on Lys residues. However, several ubiquitin ligases (E3s) have recently been identified that ubiquitylate proteins on non-Lys residues. For instance, HOIL-1 belongs to the RING-in-between RING (RBR) class of E3s and has an established role in Ser ubiquitylation. Given the homology between HOIL-1 and ARIH1, an RBR E3 that functions with the large superfamily of cullin-RING E3 ligases (CRLs), a biochemical investigation was undertaken, showing ARIH1 catalyzes Ser ubiquitylation to CRL-bound substrates. However, the efficiency of ubiquitylation was exquisitely dependent on the location and chemical environment of the Ser residue within the primary structure of the substrate. Comprehensive mutagenesis of the ARIH1 Rcat domain identified residues whose mutation severely impacted both oxyester and isopeptide bond formation at the preferred site for Ser ubiquitylation while only modestly affecting Lys ubiquitylation at the physiological site. The results reveal dual isopeptide and oxyester protein ubiquitylation activities of ARIH1 and set the stage for physiological investigations into this function of emerging importance., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
24. Combinatorial selective ER-phagy remodels the ER during neurogenesis.
- Author
-
Hoyer MJ, Capitanio C, Smith IR, Paoli JC, Bieber A, Jiang Y, Paulo JA, Gonzalez-Lozano MA, Baumeister W, Wilfling F, Schulman BA, and Harper JW
- Abstract
The endoplasmic reticulum (ER) employs a diverse proteome landscape to orchestrate many cellular functions ranging from protein and lipid synthesis to calcium ion flux and inter-organelle communication. A case in point concerns the process of neurogenesis: a refined tubular ER network is assembled via ER shaping proteins into the newly formed neuronal projections to create highly polarized dendrites and axons. Previous studies have suggested a role for autophagy in ER remodeling, as autophagy-deficient neurons in vivo display axonal ER accumulation within synaptic boutons, and the membrane-embedded ER-phagy receptor FAM134B has been genetically linked with human sensory and autonomic neuropathy. However, our understanding of the mechanisms underlying selective removal of ER and the role of individual ER-phagy receptors is limited. Here, we combine a genetically tractable induced neuron (iNeuron) system for monitoring ER remodeling during in vitro differentiation with proteomic and computational tools to create a quantitative landscape of ER proteome remodeling via selective autophagy. Through analysis of single and combinatorial ER-phagy receptor mutants, we delineate the extent to which each receptor contributes to both magnitude and selectivity of ER protein clearance. We define specific subsets of ER membrane or lumenal proteins as preferred clients for distinct receptors. Using spatial sensors and flux reporters, we demonstrate receptor-specific autophagic capture of ER in axons, and directly visualize tubular ER membranes within autophagosomes in neuronal projections by cryo-electron tomography. This molecular inventory of ER proteome remodeling and versatile genetic toolkit provides a quantitative framework for understanding contributions of individual ER-phagy receptors for reshaping ER during cell state transitions.
- Published
- 2023
- Full Text
- View/download PDF
25. A Pro-Fluorescent Ubiquitin-Based Probe to Monitor Cysteine-Based E3 Ligase Activity.
- Author
-
Pérez Berrocal DA, Vishwanatha TM, Horn-Ghetko D, Botsch JJ, Hehl LA, Kostrhon S, Misra M, Ðikić I, Geurink PP, van Dam H, Schulman BA, and Mulder MPC
- Subjects
- Cysteine metabolism, Ubiquitination, Ubiquitin-Protein Ligases metabolism, Ubiquitin metabolism, Fluorescent Dyes
- Abstract
Protein post-translational modification with ubiquitin (Ub) is a versatile signal regulating almost all aspects of cell biology, and an increasing range of diseases is associated with impaired Ub modification. In this light, the Ub system offers an attractive, yet underexplored route to the development of novel targeted treatments. A promising strategy for small molecule intervention is posed by the final components of the enzymatic ubiquitination cascade, E3 ligases, as they determine the specificity of the protein ubiquitination pathway. Here, we present UbSRhodol, an autoimmolative Ub-based probe, which upon E3 processing liberates the pro-fluorescent dye, amenable to profile the E3 transthiolation activity for recombinant and in cell-extract E3 ligases. UbSRhodol enabled detection of changes in transthiolation efficacy evoked by enzyme key point mutations or conformational changes, and offers an excellent assay reagent amenable to a high-throughput screening setup allowing the identification of small molecules modulating E3 activity., (© 2023 The Authors. Angewandte Chemie International Edition published by Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
26. To be (in a transcriptional complex) or not to be (promoting UBR5 ubiquitylation): That is an answer to how degradation controls gene expression.
- Author
-
Hehl LA and Schulman BA
- Subjects
- Ubiquitination, Gene Expression, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism
- Abstract
Tsai et al.
1 in this issue and Mark et al.2 in Cell reveal how the E3 ligase UBR5 mediates broad regulation by selectively targeting agonist-bound nuclear hormone receptors, MYC, and other transcriptional regulators not incorporated into active gene expression complexes., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
27. PARK15/FBXO7 is dispensable for PINK1/Parkin mitophagy in iNeurons and HeLa cell systems.
- Author
-
Kraus F, Goodall EA, Smith IR, Jiang Y, Paoli JC, Adolf F, Zhang J, Paulo JA, Schulman BA, and Harper JW
- Subjects
- Humans, HeLa Cells, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Protein Kinases genetics, Protein Kinases metabolism, Ubiquitin metabolism, Mitophagy genetics, F-Box Proteins genetics, F-Box Proteins metabolism
- Abstract
The protein kinase PINK1 and ubiquitin ligase Parkin promote removal of damaged mitochondria via a feed-forward mechanism involving ubiquitin (Ub) phosphorylation (pUb), Parkin activation, and ubiquitylation of mitochondrial outer membrane proteins to support the recruitment of mitophagy receptors. The ubiquitin ligase substrate receptor FBXO7/PARK15 is mutated in an early-onset parkinsonian-pyramidal syndrome. Previous studies have proposed a role for FBXO7 in promoting Parkin-dependent mitophagy. Here, we systematically examine the involvement of FBXO7 in depolarization and
mt UPR-dependent mitophagy in the well-established HeLa and induced-neurons cell systems. We find that FBXO7-/- cells have no demonstrable defect in: (i) kinetics of pUb accumulation, (ii) pUb puncta on mitochondria by super-resolution imaging, (iii) recruitment of Parkin and autophagy machinery to damaged mitochondria, (iv) mitophagic flux, and (v) mitochondrial clearance as quantified by global proteomics. Moreover, global proteomics of neurogenesis in the absence of FBXO7 reveals no obvious alterations in mitochondria or other organelles. These results argue against a general role for FBXO7 in Parkin-dependent mitophagy and point to the need for additional studies to define how FBXO7 mutations promote parkinsonian-pyramidal syndrome., (© 2023 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)- Published
- 2023
- Full Text
- View/download PDF
28. Ubiquitination regulates ER-phagy and remodelling of endoplasmic reticulum.
- Author
-
González A, Covarrubias-Pinto A, Bhaskara RM, Glogger M, Kuncha SK, Xavier A, Seemann E, Misra M, Hoffmann ME, Bräuning B, Balakrishnan A, Qualmann B, Dötsch V, Schulman BA, Kessels MM, Hübner CA, Heilemann M, Hummer G, and Dikić I
- Subjects
- Intracellular Signaling Peptides and Proteins metabolism, Ubiquitins metabolism, Microtubule-Associated Proteins metabolism, Receptors, Autocrine Motility Factor metabolism, Autophagy physiology, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum Stress, Ubiquitination
- Abstract
The endoplasmic reticulum (ER) undergoes continuous remodelling via a selective autophagy pathway, known as ER-phagy
1 . ER-phagy receptors have a central role in this process2 , but the regulatory mechanism remains largely unknown. Here we report that ubiquitination of the ER-phagy receptor FAM134B within its reticulon homology domain (RHD) promotes receptor clustering and binding to lipidated LC3B, thereby stimulating ER-phagy. Molecular dynamics (MD) simulations showed how ubiquitination perturbs the RHD structure in model bilayers and enhances membrane curvature induction. Ubiquitin molecules on RHDs mediate interactions between neighbouring RHDs to form dense receptor clusters that facilitate the large-scale remodelling of lipid bilayers. Membrane remodelling was reconstituted in vitro with liposomes and ubiquitinated FAM134B. Using super-resolution microscopy, we discovered FAM134B nanoclusters and microclusters in cells. Quantitative image analysis revealed a ubiquitin-mediated increase in FAM134B oligomerization and cluster size. We found that the E3 ligase AMFR, within multimeric ER-phagy receptor clusters, catalyses FAM134B ubiquitination and regulates the dynamic flux of ER-phagy. Our results show that ubiquitination enhances RHD functions via receptor clustering, facilitates ER-phagy and controls ER remodelling in response to cellular demands., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
29. Systemwide disassembly and assembly of SCF ubiquitin ligase complexes.
- Author
-
Baek K, Scott DC, Henneberg LT, King MT, Mann M, and Schulman BA
- Published
- 2023
- Full Text
- View/download PDF
30. An expanded lexicon for the ubiquitin code.
- Author
-
Dikic I and Schulman BA
- Subjects
- Humans, SARS-CoV-2, Ubiquitination, Proteins metabolism, Ubiquitin-Protein Ligases metabolism, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism, COVID-19
- Abstract
Our understanding of the ubiquitin code has greatly evolved from conventional E1, E2 and E3 enzymes that modify Lys residues on specific substrates with a single type of ubiquitin chain to more complex processes that regulate and mediate ubiquitylation. In this Review, we discuss recently discovered endogenous mechanisms and unprecedented pathways by which pathogens rewrite the ubiquitin code to promote infection. These processes include unconventional ubiquitin modifications involving ester linkages with proteins, lipids and sugars, or ubiquitylation through a phosphoribosyl bridge involving Arg42 of ubiquitin. We also introduce the enzymatic pathways that write and reverse these modifications, such as the papain-like proteases of severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. Furthermore, structural studies have revealed that the ultimate functions of ubiquitin are mediated not simply by straightforward recognition by ubiquitin-binding domains. Instead, elaborate multivalent interactions between ubiquitylated targets or ubiquitin chains and their readers (for example, the proteasome, the MLL1 complex or DOT1L) can elicit conformational changes that regulate protein degradation or transcription. The newly discovered mechanisms provide opportunities for innovative therapeutic interventions for diseases such as cancer and infectious diseases., (© 2022. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
31. In situ snapshots along a mammalian selective autophagy pathway.
- Author
-
Li M, Tripathi-Giesgen I, Schulman BA, Baumeister W, and Wilfling F
- Subjects
- Humans, Autophagy, Endoplasmic Reticulum metabolism, HeLa Cells, Autophagosomes metabolism, Macroautophagy
- Abstract
Selective macroautophagy (hereafter referred to as autophagy) describes a process in which cytosolic material is engulfed in a double membrane organelle called an autophagosome. Autophagosomes are carriers responsible for delivering their content to a lytic compartment for destruction. The cargo can be of diverse origin, ranging from macromolecular complexes to protein aggregates, organelles, and even invading pathogens. Each cargo is unique in composition and size, presenting different challenges to autophagosome biogenesis. Among the largest cargoes targeted by the autophagy machinery are intracellular bacteria, which can, in the case of Salmonella, range from 2 to 5 μm in length and 0.5 to 1.5 μm in width. How phagophores form and expand on such a large cargo remains mechanistically unclear. Here, we used HeLa cells infected with an auxotrophic Salmonella to study the process of phagophore biogenesis using in situ correlative cryo-ET. We show that host cells generate multiple phagophores at the site of damaged Salmonella -containing vacuoles (SCVs). The observed double membrane structures range from disk-shaped to expanded cup-shaped phagophores, which have a thin intermembrane lumen with a dilating rim region and expand using the SCV, the outer membrane of Salmonella , or existing phagophores as templates. Phagophore rims establish different forms of contact with the endoplasmic reticulum (ER) via structurally distinct molecular entities for membrane formation and expansion. Early omegasomes correlated with the marker Double-FYVE domain-Containing Protein 1 (DFCP1) are observed in close association with the ER without apparent membrane continuity. Our study provides insights into the formation of phagophores around one of the largest selective cargoes.
- Published
- 2023
- Full Text
- View/download PDF
32. E3 ligase autoinhibition by C-degron mimicry maintains C-degron substrate fidelity.
- Author
-
Scott DC, King MT, Baek K, Gee CT, Kalathur R, Li J, Purser N, Nourse A, Chai SC, Vaithiyalingam S, Chen T, Lee RE, Elledge SJ, Kleiger G, and Schulman BA
- Subjects
- Humans, Carrier Proteins, Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
E3 ligase recruitment of proteins containing terminal destabilizing motifs (degrons) is emerging as a major form of regulation. How those E3s discriminate bona fide substrates from other proteins with terminal degron-like sequences remains unclear. Here, we report that human KLHDC2, a CRL2 substrate receptor targeting C-terminal Gly-Gly degrons, is regulated through interconversion between two assemblies. In the self-inactivated homotetramer, KLHDC2's C-terminal Gly-Ser motif mimics a degron and engages the substrate-binding domain of another protomer. True substrates capture the monomeric CRL2
KLHDC2 , driving E3 activation by neddylation and subsequent substrate ubiquitylation. Non-substrates such as NEDD8 bind KLHDC2 with high affinity, but its slow on rate prevents productive association with CRL2KLHDC2 . Without substrate, neddylated CRL2KLHDC2 assemblies are deactivated via distinct mechanisms: the monomer by deneddylation and the tetramer by auto-ubiquitylation. Thus, substrate specificity is amplified by KLHDC2 self-assembly acting like a molecular timer, where only bona fide substrates may bind before E3 ligase inactivation., Competing Interests: Declaration of interests B.A.S. is a member of the scientific advisory boards of Interline Therapeutics and BioTheryX. B.A.S. and D.C.S. are co-inventors of intellectual property licensed to Cinsano., (Copyright © 2023 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
33. A central role for regulated protein stability in the control of TFE3 and MITF by nutrients.
- Author
-
Nardone C, Palanski BA, Scott DC, Timms RT, Barber KW, Gu X, Mao A, Leng Y, Watson EV, Schulman BA, Cole PA, and Elledge SJ
- Subjects
- Animals, Mechanistic Target of Rapamycin Complex 1 metabolism, Nutrients, Protein Stability, Lysosomes genetics, Lysosomes metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Mammals metabolism, Microphthalmia-Associated Transcription Factor genetics, Microphthalmia-Associated Transcription Factor metabolism, Amino Acids metabolism
- Abstract
The TFE3 and MITF master transcription factors maintain metabolic homeostasis by regulating lysosomal, melanocytic, and autophagy genes. Previous studies posited that their cytosolic retention by 14-3-3, mediated by the Rag GTPases-mTORC1, was key for suppressing transcriptional activity in the presence of nutrients. Here, we demonstrate using mammalian cells that regulated protein stability plays a fundamental role in their control. Amino acids promote the recruitment of TFE3 and MITF to the lysosomal surface via the Rag GTPases, activating an evolutionarily conserved phospho-degron and leading to ubiquitination by CUL1
β-TrCP and degradation. Elucidation of the minimal functional degron revealed a conserved alpha-helix required for interaction with RagA, illuminating the molecular basis for a severe neurodevelopmental syndrome caused by missense mutations in TFE3 within the RagA-TFE3 interface. Additionally, the phospho-degron is recurrently lost in TFE3 genomic translocations that cause kidney cancer. Therefore, two divergent pathologies converge on the loss of protein stability regulation by nutrients., Competing Interests: Declaration of interests S.J.E. is a founder of TSCAN Therapeutics, MAZE Therapeutics, ImmuneID, and Mirimus and serves on the scientific advisory boards of Homology Medicines, ImmuneID, MAZE Therapeutics, TSCAN Therapeutics, and Molecular Cell; P.A.C. is a consultant at Scorpion Therapeutics. None of these affect this work., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
34. Modular UBE2H-CTLH E2-E3 complexes regulate erythroid maturation.
- Author
-
Sherpa D, Mueller J, Karayel Ö, Xu P, Yao Y, Chrustowicz J, Gottemukkala KV, Baumann C, Gross A, Czarnecki O, Zhang W, Gu J, Nilvebrant J, Sidhu SS, Murray PJ, Mann M, Weiss MJ, Schulman BA, and Alpi AF
- Subjects
- Humans, Erythrocytes, Proteome, Ubiquitin, Ubiquitin-Conjugating Enzymes genetics, Microtubule-Associated Proteins, Guanine Nucleotide Exchange Factors, Proteomics, Erythropoiesis
- Abstract
The development of haematopoietic stem cells into mature erythrocytes - erythropoiesis - is a controlled process characterized by cellular reorganization and drastic reshaping of the proteome landscape. Failure of ordered erythropoiesis is associated with anaemias and haematological malignancies. Although the ubiquitin system is a known crucial post-translational regulator in erythropoiesis, how the erythrocyte is reshaped by the ubiquitin system is poorly understood. By measuring the proteomic landscape of in vitro human erythropoiesis models, we found dynamic differential expression of subunits of the CTLH E3 ubiquitin ligase complex that formed maturation stage-dependent assemblies of topologically homologous RANBP9- and RANBP10-CTLH complexes. Moreover, protein abundance of CTLH's cognate E2 ubiquitin conjugating enzyme UBE2H increased during terminal differentiation, and UBE2H expression depended on catalytically active CTLH E3 complexes. CRISPR-Cas9-mediated inactivation of CTLH E3 assemblies or UBE2H in erythroid progenitors revealed defects, including spontaneous and accelerated erythroid maturation as well as inefficient enucleation. Thus, we propose that dynamic maturation stage-specific changes of UBE2H-CTLH E2-E3 modules control the orderly progression of human erythropoiesis., Competing Interests: DS, JM, ÖK, PX, YY, JC, KG, CB, AG, OC, WZ, JG, JN, SS, PM, MM, MW, AA No competing interests declared, BS B.A.S. is adjunct faculty at St. Jude Children's Research Hospital, honorary professor at Technical University of Munich, a member of the scientific advisory bards of Interline Therapeutics and BioTheryX, and a convector of intellectual property related to DCN1 inhibitors licensed to Cinsano, (© 2022, Sherpa, Mueller, Karayel et al.)
- Published
- 2022
- Full Text
- View/download PDF
35. In situ structural analysis reveals membrane shape transitions during autophagosome formation.
- Author
-
Bieber A, Capitanio C, Erdmann PS, Fiedler F, Beck F, Lee CW, Li D, Hummer G, Schulman BA, Baumeister W, and Wilfling F
- Subjects
- Cell Membrane, Endoplasmic Reticulum metabolism, Saccharomyces cerevisiae, Autophagosomes metabolism, Macroautophagy, Vacuoles metabolism
- Abstract
Autophagosomes are unique organelles that form de novo as double-membrane vesicles engulfing cytosolic material for destruction. Their biogenesis involves membrane transformations of distinctly shaped intermediates whose ultrastructure is poorly understood. Here, we combine cell biology, correlative cryo-electron tomography (cryo-ET), and extensive data analysis to reveal the step-by-step structural progression of autophagosome biogenesis at high resolution directly within yeast cells. The analysis uncovers an unexpectedly thin intermembrane distance that is dilated at the phagophore rim. Mapping of individual autophagic structures onto a timeline based on geometric features reveals a dynamical change of membrane shape and curvature in growing phagophores. Moreover, our tomograms show the organelle interactome of growing autophagosomes, highlighting a polar organization of contact sites between the phagophore and organelles, such as the vacuole and the endoplasmic reticulum (ER). Collectively, these findings have important implications for the contribution of different membrane sources during autophagy and for the forces shaping and driving phagophores toward closure without a templating cargo.
- Published
- 2022
- Full Text
- View/download PDF
36. Structure of CRL7 FBXW8 reveals coupling with CUL1-RBX1/ROC1 for multi-cullin-RING E3-catalyzed ubiquitin ligation.
- Author
-
Hopf LVM, Baek K, Klügel M, von Gronau S, Xiong Y, and Schulman BA
- Subjects
- Carrier Proteins metabolism, Catalysis, Humans, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism, Ubiquitination, Cullin Proteins chemistry, Cullin Proteins metabolism, F-Box Proteins metabolism
- Abstract
Most cullin-RING ubiquitin ligases (CRLs) form homologous assemblies between a neddylated cullin-RING catalytic module and a variable substrate-binding receptor (for example, an F-box protein). However, the vertebrate-specific CRL7
FBXW8 is of interest because it eludes existing models, yet its constituent cullin CUL7 and F-box protein FBXW8 are essential for development, and CUL7 mutations cause 3M syndrome. In this study, cryo-EM and biochemical analyses reveal the CRL7FBXW8 assembly. CUL7's exclusivity for FBXW8 among all F-box proteins is explained by its unique F-box-independent binding mode. In CRL7FBXW8 , the RBX1 (also known as ROC1) RING domain is constrained in an orientation incompatible with binding E2~NEDD8 or E2~ubiquitin intermediates. Accordingly, purified recombinant CRL7FBXW8 lacks auto-neddylation and ubiquitination activities. Instead, our data indicate that CRL7 serves as a substrate receptor linked via SKP1-FBXW8 to a neddylated CUL1-RBX1 catalytic module mediating ubiquitination. The structure reveals a distinctive CRL-CRL partnership, and provides a framework for understanding CUL7 assemblies safeguarding human health., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
37. Workplace Wellness Program Interest and Barriers Among Workers With Work-Related Permanent Impairments.
- Author
-
Sears JM, Edmonds AT, Hannon PA, Schulman BA, and Fulton-Kehoe D
- Subjects
- Health Promotion methods, Humans, Obesity, Workers' Compensation, Disabled Persons, Workplace
- Abstract
Background: Nearly half of U.S. workers have access to workplace wellness programs (WWPs), 58% of workers with access participate. The aim of this study was to assess interest in WWP participation and identify reasons for lack of interest among workers with work-related permanent impairments-a population at elevated risk of adverse health outcomes., Methods: Workers who returned to work after a work-related permanent impairment were interviewed 11 to 15 months after workers' compensation claim closure. Qualitative content analysis methods were used to code open-ended responses., Findings: Of 560 respondents, 51.4% expressed interest in WWP participation. Numerous adverse health and economic characteristics were associated with WWP interest, for example, interest was expressed by 63.3% of workers reporting fair/poor health status versus 47.1% reporting good/excellent; 56.9% of workers reporting moderate/severe pain versus 41.4% reporting mild/no pain; 64.7% of workers without health insurance versus 50.1% with health insurance; 69.0% of workers reporting depression versus 47.2% without depression; 70.4% of workers reporting obesity versus 48.0% without obesity; and 63.2% of workers often worried about expenses versus 46.9% reporting sometimes/never worried. Specific participation barriers were described by 34.2% of the 272 workers who were not interested., Conclusions/applications to Practice: A majority of workers with work-related permanent impairments-particularly those with adverse health and economic characteristics-were interested in WWPs. Many workers who reported no interest cited participation barriers. Further research is needed to determine whether addressing such barriers would enhance equitable access. Those undertaking WWP planning, implementation, and outreach should ensure that WWPs are inclusive and serve workers with disabilities.
- Published
- 2022
- Full Text
- View/download PDF
38. A GID E3 ligase assembly ubiquitinates an Rsp5 E3 adaptor and regulates plasma membrane transporters.
- Author
-
Langlois CR, Beier V, Karayel O, Chrustowicz J, Sherpa D, Mann M, and Schulman BA
- Subjects
- Cell Membrane metabolism, Membrane Transport Proteins genetics, Membrane Transport Proteins metabolism, Proteomics, Ubiquitin metabolism, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitination, Endosomal Sorting Complexes Required for Transport genetics, Endosomal Sorting Complexes Required for Transport metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Ubiquitin-Protein Ligase Complexes genetics, Ubiquitin-Protein Ligase Complexes metabolism
- Abstract
Cells rapidly remodel their proteomes to align their cellular metabolism to environmental conditions. Ubiquitin E3 ligases enable this response, by facilitating rapid and reversible changes to protein stability, localization, or interaction partners. In Saccharomyces cerevisiae, the GID E3 ligase regulates the switch from gluconeogenic to glycolytic conditions through induction and incorporation of the substrate receptor subunit Gid4, which promotes the degradation of gluconeogenic enzymes. Here, we show an alternative substrate receptor, Gid10, which is induced in response to changes in temperature, osmolarity, and nutrient availability, regulates the ART-Rsp5 ubiquitin ligase pathway, a component of plasma membrane quality control. Proteomic studies reveal that the levels of the adaptor protein Art2 are elevated upon GID10 deletion. A crystal structure shows the basis for Gid10-Art2 interactions, and we demonstrate that Gid10 directs a GID E3 ligase complex to ubiquitinate Art2. Our data suggest that the GID E3 ligase affects Art2-dependent amino acid transport. This study reveals GID as a system of E3 ligases with metabolic regulatory functions outside of glycolysis and gluconeogenesis, controlled by distinct stress-specific substrate receptors., (© 2021 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
39. Cryo-EM structures of Gid12-bound GID E3 reveal steric blockade as a mechanism inhibiting substrate ubiquitylation.
- Author
-
Qiao S, Lee CW, Sherpa D, Chrustowicz J, Cheng J, Duennebacke M, Steigenberger B, Karayel O, Vu DT, von Gronau S, Mann M, Wilfling F, and Schulman BA
- Subjects
- Cryoelectron Microscopy, Gluconeogenesis physiology, Saccharomyces cerevisiae metabolism, Ubiquitination, Saccharomyces cerevisiae Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Protein degradation, a major eukaryotic response to cellular signals, is subject to numerous layers of regulation. In yeast, the evolutionarily conserved GID E3 ligase mediates glucose-induced degradation of fructose-1,6-bisphosphatase (Fbp1), malate dehydrogenase (Mdh2), and other gluconeogenic enzymes. "GID" is a collection of E3 ligase complexes; a core scaffold, RING-type catalytic core, and a supramolecular assembly module together with interchangeable substrate receptors select targets for ubiquitylation. However, knowledge of additional cellular factors directly regulating GID-type E3s remains rudimentary. Here, we structurally and biochemically characterize Gid12 as a modulator of the GID E3 ligase complex. Our collection of cryo-EM reconstructions shows that Gid12 forms an extensive interface sealing the substrate receptor Gid4 onto the scaffold, and remodeling the degron binding site. Gid12 also sterically blocks a recruited Fbp1 or Mdh2 from the ubiquitylation active sites. Our analysis of the role of Gid12 establishes principles that may more generally underlie E3 ligase regulation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
40. The structural context of posttranslational modifications at a proteome-wide scale.
- Author
-
Bludau I, Willems S, Zeng WF, Strauss MT, Hansen FM, Tanzer MC, Karayel O, Schulman BA, and Mann M
- Subjects
- Humans, Mass Spectrometry methods, Phosphorylation, Proteomics methods, Protein Processing, Post-Translational, Proteome
- Abstract
The recent revolution in computational protein structure prediction provides folding models for entire proteomes, which can now be integrated with large-scale experimental data. Mass spectrometry (MS)-based proteomics has identified and quantified tens of thousands of posttranslational modifications (PTMs), most of them of uncertain functional relevance. In this study, we determine the structural context of these PTMs and investigate how this information can be leveraged to pinpoint potential regulatory sites. Our analysis uncovers global patterns of PTM occurrence across folded and intrinsically disordered regions. We found that this information can help to distinguish regulatory PTMs from those marking improperly folded proteins. Interestingly, the human proteome contains thousands of proteins that have large folded domains linked by short, disordered regions that are strongly enriched in regulatory phosphosites. These include well-known kinase activation loops that induce protein conformational changes upon phosphorylation. This regulatory mechanism appears to be widespread in kinases but also occurs in other protein families such as solute carriers. It is not limited to phosphorylation but includes ubiquitination and acetylation sites as well. Furthermore, we performed three-dimensional proximity analysis, which revealed examples of spatial coregulation of different PTM types and potential PTM crosstalk. To enable the community to build upon these first analyses, we provide tools for 3D visualization of proteomics data and PTMs as well as python libraries for data accession and processing., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
41. How the ends signal the end: Regulation by E3 ubiquitin ligases recognizing protein termini.
- Author
-
Sherpa D, Chrustowicz J, and Schulman BA
- Subjects
- Amino Acid Motifs, Proteins metabolism, Proteolysis, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Specificity of eukaryotic protein degradation is determined by E3 ubiquitin ligases and their selective binding to protein motifs, termed "degrons," in substrates for ubiquitin-mediated proteolysis. From the discovery of the first substrate degron and the corresponding E3 to a flurry of recent studies enabled by modern systems and structural methods, it is clear that many regulatory pathways depend on E3s recognizing protein termini. Here, we review the structural basis for recognition of protein termini by E3s and how this recognition underlies biological regulation. Diverse E3s evolved to harness a substrate's N and/or C terminus (and often adjacent residues as well) in a sequence-specific manner. Regulation is achieved through selective activation of E3s and also through generation of degrons at ribosomes or by posttranslational means. Collectively, many E3 interactions with protein N and C termini enable intricate control of protein quality and responses to cellular signals., Competing Interests: Declaration of interests B.A.S. is an honorary professor at the Technical University of Munich, Germany and adjunct faculty at St. Jude Children’s Research Hospital, Memphis, TN, USA. She is on the scientific advisory boards of Interline Therapeutics and BioTheryX and is a coinventor of intellectual property related to DCN1 small-molecule inhibitors licensed to Cinsano., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
42. New classes of E3 ligases illuminated by chemical probes.
- Author
-
Horn-Ghetko D and Schulman BA
- Subjects
- Catalytic Domain, Humans, Ubiquitin metabolism, Ubiquitination, Cullin Proteins genetics, Cullin Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Specificity in the ubiquitin system depends on E3 ligases, largely belonging to a handful of families discovered more than a decade ago. However, the last two years brought a quantum leap in the identification and/or mechanistic characterization of eukaryotic ubiquitin ligases, in part through implementation of activity-based chemical probes and cryo-EM. Here, we survey recent discoveries of RING-Cys-Relay, RZ-finger, and neddylated cullin-RING-ARIH RBR E3-E3 ubiquitin ligase mechanisms. These ligases transfer ubiquitin through unprecedented mechanisms-via novel catalytic domains or domain combinations-and collectively modify unconventional amino acids, non-proteinaceous bacterial lipid targets, and structurally-diverse substrates recruited to numerous cullin-RING ligases. We anticipate major expansion of the types, features, and mechanisms of E3 ligases will emerge from such chemical and structural approaches in the coming years., Competing Interests: Conflict of interest statement B.A.S. is on the scientific advisory boards of Interline Therapeutics and BioTheryX, and is co-inventor of intellectual property related to DCN1 small molecule inhibitors licensed to Cinsano., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
43. Targeted protein degradation: from small molecules to complex organelles-a Keystone Symposia report.
- Author
-
Cable J, Weber-Ban E, Clausen T, Walters KJ, Sharon M, Finley DJ, Gu Y, Hanna J, Feng Y, Martens S, Simonsen A, Hansen M, Zhang H, Goodwin JM, Reggio A, Chang C, Ge L, Schulman BA, Deshaies RJ, Dikic I, Harper JW, Wertz IE, Thomä NH, Słabicki M, Frydman J, Jakob U, David DC, Bennett EJ, Bertozzi CR, Sardana R, Eapen VV, and Carra S
- Subjects
- Autophagy physiology, Humans, Organelles, Proteins metabolism, Proteolysis, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism
- Abstract
Targeted protein degradation is critical for proper cellular function and development. Protein degradation pathways, such as the ubiquitin proteasomes system, autophagy, and endosome-lysosome pathway, must be tightly regulated to ensure proper elimination of misfolded and aggregated proteins and regulate changing protein levels during cellular differentiation, while ensuring that normal proteins remain unscathed. Protein degradation pathways have also garnered interest as a means to selectively eliminate target proteins that may be difficult to inhibit via other mechanisms. On June 7 and 8, 2021, several experts in protein degradation pathways met virtually for the Keystone eSymposium "Targeting protein degradation: from small molecules to complex organelles." The event brought together researchers working in different protein degradation pathways in an effort to begin to develop a holistic, integrated vision of protein degradation that incorporates all the major pathways to understand how changes in them can lead to disease pathology and, alternatively, how they can be leveraged for novel therapeutics., (© 2022 New York Academy of Sciences.)
- Published
- 2022
- Full Text
- View/download PDF
44. Multifaceted N-Degron Recognition and Ubiquitylation by GID/CTLH E3 Ligases.
- Author
-
Chrustowicz J, Sherpa D, Teyra J, Loke MS, Popowicz GM, Basquin J, Sattler M, Prabu JR, Sidhu SS, and Schulman BA
- Subjects
- Humans, Protein Binding, Protein Domains, Protein Interaction Domains and Motifs, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Ubiquitin metabolism, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases metabolism, Ubiquitination
- Abstract
N-degron E3 ubiquitin ligases recognize specific residues at the N-termini of substrates. Although molecular details of N-degron recognition are known for several E3 ligases, the range of N-terminal motifs that can bind a given E3 substrate binding domain remains unclear. Here, we discovered capacity of Gid4 and Gid10 substrate receptor subunits of yeast "GID"/human "CTLH" multiprotein E3 ligases to tightly bind a wide range of N-terminal residues whose recognition is determined in part by the downstream sequence context. Screening of phage displaying peptide libraries with exposed N-termini identified novel consensus motifs with non-Pro N-terminal residues binding Gid4 or Gid10 with high affinity. Structural data reveal that conformations of flexible loops in Gid4 and Gid10 complement sequences and folds of interacting peptides. Together with analysis of endogenous substrate degrons, the data show that degron identity, substrate domains harboring targeted lysines, and varying E3 ligase higher-order assemblies combinatorially determine efficiency of ubiquitylation and degradation., Competing Interests: Declaration of Competing Interest B.A.S. is an honorary professor at Technical University of Munich, Germany and adjunct faculty at St. Jude Children’s Research Hospital, Memphis, TN, USA, is on the scientific advisory boards of Interline Therapeutics and BioTheryX, and is co-inventor of intellectual property related to DCN1 small molecule inhibitors licensed to Cinsano., (Copyright © 2021 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
45. APC7 mediates ubiquitin signaling in constitutive heterochromatin in the developing mammalian brain.
- Author
-
Ferguson CJ, Urso O, Bodrug T, Gassaway BM, Watson ER, Prabu JR, Lara-Gonzalez P, Martinez-Chacin RC, Wu DY, Brigatti KW, Puffenberger EG, Taylor CM, Haas-Givler B, Jinks RN, Strauss KA, Desai A, Gabel HW, Gygi SP, Schulman BA, Brown NG, and Bonni A
- Subjects
- Adolescent, Animals, Antigens, CD, Apc7 Subunit, Anaphase-Promoting Complex-Cyclosome genetics, Behavior, Animal, Brain growth & development, Cadherins genetics, Cadherins metabolism, Cell Line, Child, Child, Preschool, Disease Models, Animal, Female, Heterochromatin genetics, Humans, Infant, Intellectual Disability pathology, Intellectual Disability physiopathology, Intellectual Disability psychology, Intelligence, Ki-67 Antigen genetics, Ki-67 Antigen metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Mitosis, Mutation, Neural Stem Cells pathology, Proteolysis, Signal Transduction, Syndrome, Ubiquitination, Young Adult, Mice, Apc7 Subunit, Anaphase-Promoting Complex-Cyclosome metabolism, Brain enzymology, Heterochromatin metabolism, Intellectual Disability enzymology, Neural Stem Cells enzymology, Neurogenesis
- Abstract
Neurodevelopmental cognitive disorders provide insights into mechanisms of human brain development. Here, we report an intellectual disability syndrome caused by the loss of APC7, a core component of the E3 ubiquitin ligase anaphase promoting complex (APC). In mechanistic studies, we uncover a critical role for APC7 during the recruitment and ubiquitination of APC substrates. In proteomics analyses of the brain from mice harboring the patient-specific APC7 mutation, we identify the chromatin-associated protein Ki-67 as an APC7-dependent substrate of the APC in neurons. Conditional knockout of the APC coactivator protein Cdh1, but not Cdc20, leads to the accumulation of Ki-67 protein in neurons in vivo, suggesting that APC7 is required for the function of Cdh1-APC in the brain. Deregulated neuronal Ki-67 upon APC7 loss localizes predominantly to constitutive heterochromatin. Our findings define an essential function for APC7 and Cdh1-APC in neuronal heterochromatin regulation, with implications for understanding human brain development and disease., Competing Interests: Declaration of interests The authors declare no competing interests. A.B. is an employee of Roche. B.S. is on the scientific advisory board of BioTheryX and Interline Therapeutics, a shareholder of Interline Therapeutics, and a co-inventor of intellectual property licensed to Cinsano., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
46. CUL5-ARIH2 E3-E3 ubiquitin ligase structure reveals cullin-specific NEDD8 activation.
- Author
-
Kostrhon S, Prabu JR, Baek K, Horn-Ghetko D, von Gronau S, Klügel M, Basquin J, Alpi AF, and Schulman BA
- Subjects
- Animals, Cell Line, Cloning, Molecular, Cryoelectron Microscopy, Crystallization, Cullin Proteins genetics, Gene Expression Regulation, Humans, Insecta, Models, Molecular, NEDD8 Protein genetics, Protein Conformation, Ubiquitin-Protein Ligases genetics, Ubiquitination, Cullin Proteins metabolism, NEDD8 Protein metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
An emerging mechanism of ubiquitylation involves partnering of two distinct E3 ligases. In the best-characterized E3-E3 pathways, ARIH-family RING-between-RING (RBR) E3s ligate ubiquitin to substrates of neddylated cullin-RING E3s. The E3 ARIH2 has been implicated in ubiquitylation of substrates of neddylated CUL5-RBX2-based E3s, including APOBEC3-family substrates of the host E3 hijacked by HIV-1 virion infectivity factor (Vif). However, the structural mechanisms remained elusive. Here structural and biochemical analyses reveal distinctive ARIH2 autoinhibition, and activation on assembly with neddylated CUL5-RBX2. Comparison to structures of E3-E3 assemblies comprising ARIH1 and neddylated CUL1-RBX1-based E3s shows cullin-specific regulation by NEDD8. Whereas CUL1-linked NEDD8 directly recruits ARIH1, CUL5-linked NEDD8 does not bind ARIH2. Instead, the data reveal an allosteric mechanism. NEDD8 uniquely contacts covalently linked CUL5, and elicits structural rearrangements that unveil cryptic ARIH2-binding sites. The data reveal how a ubiquitin-like protein induces protein-protein interactions indirectly, through allostery. Allosteric specificity of ubiquitin-like protein modifications may offer opportunities for therapeutic targeting., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
47. Decoding the messaging of the ubiquitin system using chemical and protein probes.
- Author
-
Henneberg LT and Schulman BA
- Subjects
- Humans, Protein Processing, Post-Translational, Ubiquitination, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Post-translational modification of proteins by ubiquitin is required for nearly all aspects of eukaryotic cell function. The numerous targets of ubiquitylation, and variety of ubiquitin modifications, are often likened to a code, where the ultimate messages are diverse responses to target ubiquitylation. E1, E2, and E3 multiprotein enzymatic assemblies modify specific targets and thus function as messengers. Recent advances in chemical and protein tools have revolutionized our ability to explore the ubiquitin system, through enabling new high-throughput screening methods, matching ubiquitylation enzymes with their cellular targets, revealing intricate allosteric mechanisms regulating ubiquitylating enzymes, facilitating structural revelation of transient assemblies determined by multivalent interactions, and providing new paradigms for inhibiting and redirecting ubiquitylation in vivo as new therapeutics. Here we discuss the development of methods that control, disrupt, and extract the flow of information across the ubiquitin system and have enabled elucidation of the underlying molecular and cellular biology., Competing Interests: Declaration of interests B.A.S. is on the Scientific Advisory Board of Interline Therapeutics, and is Adjunct Faculty at St. Jude Children's Research Hospital, and Honorary Professor at Technical University of Munich., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
48. Cullin-RING Ubiquitin Ligase Regulatory Circuits: A Quarter Century Beyond the F-Box Hypothesis.
- Author
-
Harper JW and Schulman BA
- Subjects
- F-Box Proteins chemistry, Feedback, Physiological, Host-Pathogen Interactions physiology, Humans, NEDD8 Protein metabolism, Plant Growth Regulators metabolism, Protein Domains, Protein Processing, Post-Translational, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Protein Ligases chemistry, Ubiquitination, Cullin Proteins chemistry, Cullin Proteins metabolism, F-Box Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Cullin-RING ubiquitin ligases (CRLs) are dynamic modular platforms that regulate myriad biological processes through target-specific ubiquitylation. Our knowledge of this system emerged from the F-box hypothesis, posited a quarter century ago: Numerous interchangeable F-box proteins confer specific substrate recognition for a core CUL1-based RING E3 ubiquitin ligase. This paradigm has been expanded through the evolution of a superfamily of analogous modular CRLs, with five major families and over 200 different substrate-binding receptors in humans. Regulation is achieved by numerous factors organized in circuits that dynamically control CRL activation and substrate ubiquitylation. CRLs also serve as a vast landscape for developing small molecules that reshape interactions and promote targeted ubiquitylation-dependent turnover of proteins of interest. Here, we review molecular principles underlying CRL function, the role of allosteric and conformational mechanisms in controlling substrate timing and ubiquitylation, and how the dynamics of substrate receptor interchange drives the turnover of selected target proteins to promote cellular decision-making.
- Published
- 2021
- Full Text
- View/download PDF
49. Workplace Organizational and Psychosocial Factors Associated with Return-to-Work Interruption and Reinjury Among Workers with Permanent Impairment.
- Author
-
Sears JM, Schulman BA, Fulton-Kehoe D, and Hogg-Johnson S
- Subjects
- Humans, Retrospective Studies, Return to Work, Workplace, Occupational Exposure, Reinjuries
- Abstract
Objectives: Roughly 10% of occupational injuries result in permanent impairment and a permanent partial disability (PPD) award. After initial return to work (RTW) following a work injury, many workers with permanent impairment face RTW interruption (breaks in ongoing employment due to reinjury, poor health, disability, lay-off, etc.). Most RTW and reinjury research has focused on worker-level risk factors, and less is known about contextual factors that may be amenable to workplace or workers' compensation (WC)-based interventions. The aim of this study was to identify modifiable organizational and psychosocial workplace factors associated with (i) RTW interruption and (ii) reinjury among workers with a permanent impairment., Methods: This retrospective cohort study included WC claims data and survey data for 567 injured workers who RTW at least briefly after a work-related injury that resulted in permanent impairment. Workers were interviewed once by phone, 11-15 months after WC claim closure with a PPD award. Logistic regression models were used to estimate associations between each workplace factor of interest and each outcome, controlling for whole body impairment percentage, gender, age, nativity, educational level, State Fund versus self-insured WC coverage, employer size, union membership, industry sector, and employment duration of current/most recent job., Results: Twelve percent of workers had been reinjured in their current or most recent job, 12% of workers were no longer working at the time of interview, and <1% of workers reported both outcomes. The most frequently reported reason for RTW interruption was impairment, disability, and/or pain from the previous work injury. Lower reported levels of safety climate, supervisor support, and ability to take time off work for personal/family matters were significantly associated with both RTW interruption and reinjury. Inadequate employer/health care provider communication, perceived stigmatization from supervisors and/or coworkers, and lower levels of coworker support were significantly associated with RTW interruption but not with reinjury. Discomfort with reporting an unsafe situation at work, absence of a health and safety committee, and higher job strain were significantly associated with reinjury, but not with RTW interruption. Inadequate safety training and lack of needed job accommodations were not significantly associated with either outcome. There were no notable or statistically significant interactions between workplace factors and degree of impairment, and no consistent direction of association., Conclusions: This study provides evidence that several potentially modifiable organizational and psychosocial factors are associated with safe and sustained RTW among injured workers with work-related permanent impairment. The lack of interaction between any of these workplace factors and degree of impairment suggests that these findings may be generalizable to all workers, and further suggests that workplace interventions based on these findings might be useful for both primary and secondary prevention. Though primary prevention is key, secondary prevention efforts to sustain RTW and prevent reinjury may reduce the considerable health, economic, and social burden of occupational injury and illness., (© The Author(s) 2021. Published by Oxford University Press on behalf of the British Occupational Hygiene Society.)
- Published
- 2021
- Full Text
- View/download PDF
50. GID E3 ligase supramolecular chelate assembly configures multipronged ubiquitin targeting of an oligomeric metabolic enzyme.
- Author
-
Sherpa D, Chrustowicz J, Qiao S, Langlois CR, Hehl LA, Gottemukkala KV, Hansen FM, Karayel O, von Gronau S, Prabu JR, Mann M, Alpi AF, and Schulman BA
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Binding Sites, Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Cryoelectron Microscopy, Fructose-Bisphosphatase genetics, Fructose-Bisphosphatase metabolism, Gene Expression, Gluconeogenesis genetics, Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, K562 Cells, Kinetics, Models, Molecular, Multienzyme Complexes genetics, Multienzyme Complexes metabolism, Promoter Regions, Genetic, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Saccharomyces cerevisiae, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Sf9 Cells, Spodoptera, Structural Homology, Protein, Substrate Specificity, Ubiquitin genetics, Ubiquitin metabolism, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitination, Adaptor Proteins, Signal Transducing chemistry, Cell Adhesion Molecules chemistry, Fructose-Bisphosphatase chemistry, Intracellular Signaling Peptides and Proteins chemistry, Multienzyme Complexes chemistry, Saccharomyces cerevisiae Proteins chemistry, Ubiquitin chemistry, Ubiquitin-Conjugating Enzymes chemistry
- Abstract
How are E3 ubiquitin ligases configured to match substrate quaternary structures? Here, by studying the yeast GID complex (mutation of which causes deficiency in glucose-induced degradation of gluconeogenic enzymes), we discover supramolecular chelate assembly as an E3 ligase strategy for targeting an oligomeric substrate. Cryoelectron microscopy (cryo-EM) structures show that, to bind the tetrameric substrate fructose-1,6-bisphosphatase (Fbp1), two minimally functional GID E3s assemble into the 20-protein Chelator-GID
SR4 , which resembles an organometallic supramolecular chelate. The Chelator-GIDSR4 assembly avidly binds multiple Fbp1 degrons so that multiple Fbp1 protomers are simultaneously ubiquitylated at lysines near the allosteric and substrate binding sites. Importantly, key structural and biochemical features, including capacity for supramolecular assembly, are preserved in the human ortholog, the CTLH E3. Based on our integrative structural, biochemical, and cell biological data, we propose that higher-order E3 ligase assembly generally enables multipronged targeting, capable of simultaneously incapacitating multiple protomers and functionalities of oligomeric substrates., Competing Interests: Declaration of interests B.A.S. is an honorary professor at Technical University of Munich, Germany and adjunct faculty at St. Jude Children’s Research Hospital, Memphis, TN, USA and is on the scientific advisory board of Interline Therapeutics., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.