207 results on '"Rachel E. Klevit"'
Search Results
2. Legionella effector MavC targets the Ube2N~Ub conjugate for noncanonical ubiquitination
- Author
-
Kedar Puvar, Shalini Iyer, Jiaqi Fu, Sebastian Kenny, Kristos I. Negrón Terón, Zhao-Qing Luo, Peter S. Brzovic, Rachel E. Klevit, and Chittaranjan Das
- Subjects
Science - Abstract
The Legionella pneumophila effector MavC inhibits the human ubiquitin-conjugating enzyme Ube2N. Here, the authors combine NMR, X-ray crystallography and biochemical assays and show that MavC catalyses the intramolecular transglutaminase reaction between the Ube2N and Ub subunits of the Ube2N∼Ub conjugate and present the substrate- and product-bound MavC crystal structures.
- Published
- 2020
- Full Text
- View/download PDF
3. Gcn4-Mediator Specificity Is Mediated by a Large and Dynamic Fuzzy Protein-Protein Complex
- Author
-
Lisa M. Tuttle, Derek Pacheco, Linda Warfield, Jie Luo, Jeff Ranish, Steven Hahn, and Rachel E. Klevit
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Transcription activation domains (ADs) are inherently disordered proteins that often target multiple coactivator complexes, but the specificity of these interactions is not understood. Efficient transcription activation by yeast Gcn4 requires its tandem ADs and four activator-binding domains (ABDs) on its target, the Mediator subunit Med15. Multiple ABDs are a common feature of coactivator complexes. We find that the large Gcn4-Med15 complex is heterogeneous and contains nearly all possible AD-ABD interactions. Gcn4-Med15 forms via a dynamic fuzzy protein-protein interface, where ADs bind the ABDs in multiple orientations via hydrophobic regions that gain helicity. This combinatorial mechanism allows individual low-affinity and specificity interactions to generate a biologically functional, specific, and higher affinity complex despite lacking a defined protein-protein interface. This binding strategy is likely representative of many activators that target multiple coactivators, as it allows great flexibility in combinations of activators that can cooperate to regulate genes with variable coactivator requirements. : Tuttle et al. report a “fuzzy free-for-all” interaction mechanism that explains how seemingly unrelated transcription activators converge on a limited number of coactivator targets. The mechanism provides a rationale for the observation that individually weak and low-specificity interactions can combine to produce biologically critical function without requiring highly ordered structure. Keywords: transcription activation, intrinsically disordered proteins, fuzzy binding
- Published
- 2018
- Full Text
- View/download PDF
4. A native chemical chaperone in the human eye lens
- Author
-
Eugene Serebryany, Sourav Chowdhury, Christopher N Woods, David C Thorn, Nicki E Watson, Arthur A McClelland, Rachel E Klevit, and Eugene I Shakhnovich
- Subjects
protein aggregation ,cataract ,eye lens ,crystallin ,chemical chaperone ,disulfide ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Cataract is one of the most prevalent protein aggregation disorders and still the most common cause of vision loss worldwide. The metabolically quiescent core region of the human lens lacks cellular or protein turnover; it has therefore evolved remarkable mechanisms to resist light-scattering protein aggregation for a lifetime. We now report that one such mechanism involves an unusually abundant lens metabolite, myo-inositol, suppressing aggregation of lens crystallins. We quantified aggregation suppression using our previously well-characterized in vitro aggregation assays of oxidation-mimicking human γD-crystallin variants and investigated myo-inositol’s molecular mechanism of action using solution NMR, negative-stain TEM, differential scanning fluorometry, thermal scanning Raman spectroscopy, turbidimetry in redox buffers, and free thiol quantitation. Unlike many known chemical chaperones, myo-inositol’s primary target was not the native, unfolded, or final aggregated states of the protein; rather, we propose that it was the rate-limiting bimolecular step on the aggregation pathway. Given recent metabolomic evidence that it is severely depleted in human cataractous lenses compared to age-matched controls, we suggest that maintaining or restoring healthy levels of myo-inositol in the lens may be a simple, safe, and globally accessible strategy to prevent or delay lens opacification due to age-onset cataract.
- Published
- 2022
- Full Text
- View/download PDF
5. Indirect sexual selection drives rapid sperm protein evolution in abalone
- Author
-
Damien Beau Wilburn, Lisa M Tuttle, Rachel E Klevit, and Willie J Swanson
- Subjects
sexual selection ,NMR spectroscopy ,fertilization ,abalone ,rapid evolution ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Sexual selection can explain the rapid evolution of fertilization proteins, yet sperm proteins evolve rapidly even if not directly involved in fertilization. In the marine mollusk abalone, sperm secrete enormous quantities of two rapidly evolving proteins, lysin and sp18, that are stored at nearly molar concentrations. We demonstrate that this extraordinary packaging is achieved by associating into Fuzzy Interacting Transient Zwitterion (FITZ) complexes upon binding the intrinsically disordered FITZ Anionic Partner (FITZAP). FITZ complexes form at intracellular ionic strengths and, upon exocytosis into seawater, lysin and sp18 are dispersed to drive fertilization. NMR analyses revealed that lysin uses a common molecular interface to bind both FITZAP and its egg receptor VERL. As sexual selection alters the lysin-VERL interface, FITZAP coevolves rapidly to maintain lysin binding. FITZAP-lysin interactions exhibit a similar species-specificity as lysin-VERL interactions. Thus, tethered molecular arms races driven by sexual selection can generally explain rapid sperm protein evolution.
- Published
- 2019
- Full Text
- View/download PDF
6. Interplay of disordered and ordered regions of a human small heat shock protein yields an ensemble of ‘quasi-ordered’ states
- Author
-
Amanda F Clouser, Hannah ER Baughman, Benjamin Basanta, Miklos Guttman, Abhinav Nath, and Rachel E Klevit
- Subjects
small heat shock protein ,intrinsically disordered protein ,HSPB1 ,HDXMS ,NMR ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Small heat shock proteins (sHSPs) are nature’s ‘first responders’ to cellular stress, interacting with affected proteins to prevent their aggregation. Little is known about sHSP structure beyond its structured α-crystallin domain (ACD), which is flanked by disordered regions. In the human sHSP HSPB1, the disordered N-terminal region (NTR) represents nearly 50% of the sequence. Here, we present a hybrid approach involving NMR, hydrogen-deuterium exchange mass spectrometry, and modeling to provide the first residue-level characterization of the NTR. The results support a model in which multiple grooves on the ACD interact with specific NTR regions, creating an ensemble of ‘quasi-ordered’ NTR states that can give rise to the known heterogeneity and plasticity of HSPB1. Phosphorylation-dependent interactions inform a mechanism by which HSPB1 is activated under stress conditions. Additionally, we examine the effects of disease-associated NTR mutations on HSPB1 structure and dynamics, leveraging our emerging structural insights.
- Published
- 2019
- Full Text
- View/download PDF
7. Supplementary Figures S1-S13 from Interaction of BARD1 and HP1 Is Required for BRCA1 Retention at Sites of DNA Damage
- Author
-
Tomohiko Ohta, Rachel E. Klevit, Yasuo Miyoshi, Masahide Asano, Vinayak Vittal, Takayo Fukuda, Hiroyuki Nishikawa, and Wenwen Wu
- Abstract
Supplementary Figures S1-S13. Interactions of exogenously overexpressed proteins in vivo (S1); Purified recombinant proteins used in the experiments shown in Figure 2 (S2); SPR analyses of the HP1α, β, or HP1γ− chromoshadow domain binding to BARD1-BRCT (S3); SPR analyses of the BRCA1 fragments binding to HP1α, β, or γ (S4); Inhibition of HP1γ only mildly decreases the stable retention of BRCA1/BARD1 at sites of DNA damage (S5); Inhibition of HP1γ increases the interaction of BARD1 with HP1α (S6); BARD1 is dispensable for recruitment of HP1γ at DSB sites (S7); ATM-dependent interaction of BARD1 with HP1γ/H3K9me2 (S8); Ectopic accumulation of RIF1 and inhibition of RAD51 retention at DSB sites by depletion of all three isoforms of HP1 or BARD1 mutant that does not bind HP1 (S9); Dissolution kinetics of γH2AX after DSB damage in G2 cells (S10); Cell cycle progression of cells treated with UNC0638 (S11); An H3K9-specific HKMT inhibitor UNC0638 disrupts BRCA1/BARD1 retention at sites of DNA damage (S12); A model for the role of BRCA1/BARD1/HP1 complex in the DSB repair pathways (S13).
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Interaction of BARD1 and HP1 Is Required for BRCA1 Retention at Sites of DNA Damage
- Author
-
Tomohiko Ohta, Rachel E. Klevit, Yasuo Miyoshi, Masahide Asano, Vinayak Vittal, Takayo Fukuda, Hiroyuki Nishikawa, and Wenwen Wu
- Abstract
Stable retention of BRCA1/BARD1 complexes at sites of DNA damage is required for the proper response to DNA double-strand breaks (DSB). Here, we demonstrate that the BRCT domain of BARD1 is crucial for its retention through interaction with HP1. In response to DNA damage, BARD1 interacts with Lys9-dimethylated histone H3 (H3K9me2) in an ATM-dependent but RNF168-independent manner. This interaction is mediated primarily by HP1γ. A conserved HP1-binding motif in the BARD1 BRCT domain directly interacted with the chromoshadow domain of HP1 in vitro. Mutations in this motif (or simultaneous depletion of all three HP1 isoforms) disrupted retention of BARD1, BRCA1, and CtIP at DSB sites and allowed ectopic accumulation of RIF1, an effector of nonhomologous end-joining, at damaged loci in S-phase. UNC0638, a small-molecule inhibitor of histone lysine methyltransferase (HKMT), abolished retention and cooperated with the PARP inhibitor olaparib to block cancer cell growth. Taken together, our findings show how BARD1 promotes retention of the BRCA1/BARD1 complex at damaged DNA sites and suggest the use of HKMT inhibitors to leverage the application of PARP inhibitors to treat breast cancer. Cancer Res; 75(7); 1311–21. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure Legends from Interaction of BARD1 and HP1 Is Required for BRCA1 Retention at Sites of DNA Damage
- Author
-
Tomohiko Ohta, Rachel E. Klevit, Yasuo Miyoshi, Masahide Asano, Vinayak Vittal, Takayo Fukuda, Hiroyuki Nishikawa, and Wenwen Wu
- Abstract
Legends for Supplementary Figures S1-S13.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemenatary Methods and References from Interaction of BARD1 and HP1 Is Required for BRCA1 Retention at Sites of DNA Damage
- Author
-
Tomohiko Ohta, Rachel E. Klevit, Yasuo Miyoshi, Masahide Asano, Vinayak Vittal, Takayo Fukuda, Hiroyuki Nishikawa, and Wenwen Wu
- Abstract
Supplemenatary Methods and References. Description of additional methods and procedures used in the study. Also includes Supplementary References.
- Published
- 2023
- Full Text
- View/download PDF
11. Disordered region encodes α-crystallin chaperone activity toward lens client γD-crystallin
- Author
-
Christopher N. Woods, Lindsey D. Ulmer, Miklos Guttman, Matthew F. Bush, and Rachel E. Klevit
- Subjects
Multidisciplinary - Abstract
Small heat-shock proteins (sHSPs) are a widely expressed family of ATP-independent molecular chaperones that are among the first responders to cellular stress. Mechanisms by which sHSPs delay aggregation of client proteins remain undefined. sHSPs have high intrinsic disorder content of up to ~60% and assemble into large, polydisperse homo- and hetero-oligomers, making them challenging structural and biochemical targets. Two sHSPs, HSPB4 and HSPB5, are present at millimolar concentrations in eye lens, where they are responsible for maintaining lens transparency over the lifetime of an organism. Together, HSPB4 and HSPB5 compose the hetero-oligomeric chaperone known as α-crystallin. To identify the determinants of sHSP function, we compared the effectiveness of HSPB4 and HSPB5 homo-oligomers and HSPB4/HSPB5 hetero-oligomers in delaying the aggregation of the lens protein γD-crystallin. In chimeric versions of HSPB4 and HSPB5, chaperone activity tracked with the identity of the 60-residue disordered N-terminal regions (NTR). A short 10-residue stretch in the middle of the NTR (“Critical sequence”) contains three residues that are responsible for high HSPB5 chaperone activity toward γD-crystallin. These residues affect structure and dynamics throughout the NTR. Abundant interactions involving the NTR Critical sequence reveal it to be a hub for a network of interactions within oligomers. We propose a model whereby the NTR critical sequence influences local structure and NTR dynamics that modulate accessibility of the NTR, which in turn modulates chaperone activity.
- Published
- 2023
- Full Text
- View/download PDF
12. Proof of principle for epitope-focused vaccine design.
- Author
-
Bruno E. Correia, John T. Bates, Rebecca J. Loomis, Gretchen Baneyx, Chris Carrico, Joseph G. Jardine, Peter Rupert, Colin E. Correnti, Oleksandr Kalyuzhniy, Vinayak Vittal, Mary J. Connell, Eric Stevens, Alexandria Schroeter, Man Chen, Skye MacPherson, Andreia M. Serra, Yumiko Adachi, Margaret A. Holmes, Yuxing Li, Rachel E. Klevit, Barney S. Graham, Richard T. Wyatt, David Baker 0001, Roland K. Strong, James E. Crowe Jr., Philip R. Johnson, and William R. Schief
- Published
- 2014
- Full Text
- View/download PDF
13. Conservation of transcriptional regulation by BRCA1 and BARD1 in Caenorhabditis elegans
- Author
-
Ishor Thapa, Russell Vahrenkamp, Samuel R Witus, Caitlin Lightle, Owen Falkenberg, Marlo K Sellin Jeffries, Rachel E Klevit, and Mikaela D Stewart
- Subjects
Genetics - Abstract
The tumor-suppressor proteins BRCA1 and BARD1 function as an E3 ubiquitin ligase to facilitate transcriptional repression and DNA damage repair. This is mediated in-part through its ability to mono-ubiquitylate histone H2A in nucleosomes. Studies in Caenorhabditis elegans have been used to elucidate numerous functions of BRCA1 and BARD1; however, it has not been established that the C. elegans orthologs, BRC-1 and BRD-1, retain all the functions of their human counterparts. Here we explore the conservation of enzymatic activity toward nucleosomes which leads to repression of estrogen-metabolizing cytochrome P450 (cyp) genes in humans. Biochemical assays establish that BRC-1 and BRD-1 contribute to ubiquitylation of histone H2A in the nucleosome. Mutational analysis shows that while BRC-1 likely binds the nucleosome using a conserved interface, BRD-1 and BARD1 have evolved different modes of binding, resulting in a difference in the placement of ubiquitin on H2A. Gene expression analysis reveals that in spite of this difference, BRC-1 and BRD-1 also contribute to cyp gene repression in C. elegans. Establishing conservation of these functions in C. elegans allows for use of this powerful model organism to address remaining questions regarding regulation of gene expression by BRCA1 and BARD1.
- Published
- 2022
14. BRCA1/BARD1 intrinsically disordered regions facilitate chromatin recruitment and ubiquitylation
- Author
-
Samuel R. Witus, Lisa M. Tuttle, Wenjing Li, Alex Zelter, Meiling Wang, Klaiten E. Kermoade, Damien B. Wilburn, Trisha N. Davis, Peter S. Brzovic, Weixing Zhao, and Rachel E. Klevit
- Abstract
BRCA1/BARD1 is a tumor suppressor E3 ubiquitin (Ub) ligase with roles in DNA damage repair and in transcriptional regulation. BRCA1/BARD1 RING domains interact with nucleosomes to facilitate mono-ubiquitylation of distinct residues on the C-terminal tail of histone H2A. These enzymatic domains constitute a small fraction of the heterodimer, raising the possibility of functional chromatin interactions involving other regions such as the BARD1 C-terminal domains that bind nucleosomes containing the DNA damage signal H2A K15-Ub and H4 K20me0, or portions of the expansive intrinsically disordered regions found in both subunits. Herein, we reveal novel interactions that support robust H2A ubiquitylation activity mediated through a high-affinity, intrinsically disordered DNA-binding region of BARD1. These interactions support BRCA1/BARD1 recruitment to chromatin and sites of DNA damage in cells and contribute to their survival. We also reveal distinct BRCA1/BARD1 complexes that depend on the presence of H2A K15-Ub, including a complex where a single BARD1 subunit spans adjacent nucleosome units. Our findings identify an extensive network of multivalent BARD1- nucleosome interactions that serve as a platform for BRCA1/BARD1-associated functions on chromatin.
- Published
- 2022
- Full Text
- View/download PDF
15. Abstract 6096: BRCA1-BARD1 ubiquitylates histones for genome maintenance
- Author
-
Wenjing Li, Samuel R. Witus, Meiling Wang, Peter S. Brzovic, Rachel E. Klevit, and Weixing Zhao
- Subjects
Cancer Research ,Oncology - Abstract
Background: Breast cancer type 1 susceptibility protein (BRCA1) is a tumor suppressor gene involved in DNA double strand break repair with well-known cancer implications. BRCA1 heterodimerizes with BRCA1 associated Ring domain 1 (BARD1) to form a complex with DNA binding and ubiquitin E3 ligase function capable of interacting with proteins of diverse biological processes, most notably homology-directed DNA repair. During DNA repair, BRCA1-BARD1 directly interfaces with nucleosomes and transfers mono ubiquitin (Ub) to lysine residues on the C-terminal tail of histone H2A. Although truncation of the enzymatic BRCA1-BARD1 RING-RING domain retains H2A ubiquitylating activity, full-length BRCA1-BARD1 binds more tightly with nucleosomes and displays higher H2A-Ub activity. However, the molecular basis and biological significance for this enhanced nucleosome binding and H2A-Ub activity is uncharacterized. Methods: Full length BRCA1-BARD1 or truncated mutants and histones were purified from E. coli. or insect cells. Nucleosomes were assembled for in vitro ubiquitylation reaction and binding assays. To determine the biological significance, mammalian cell lines that stably express wild type or mutant forms of BARD1 were established for cellular fractionation, foci analysis, and clonogenic survival studies alongside various DNA damage agents. Results: Our results show multiple interaction sites exist between BRCA1-BARD1 and nucleosomes which allow high-affinity chromatin binding and promote increased histone H2A ubiquitylation activity. Multivalent BARD1-nucleosome interactions, namely those using strong binding motifs located in the intrinsically disordered region (IDR) of BARD1, and the weak “kiss” interaction mediated by the RING domains of both BRCA1 and BARD1, are essential for H2A ubiquitylation by BRCA1-BARD1. Further, we isolated two types of specific histone binding and/or ubiquitylation-defective mutants of BARD1: a BARD1-IDR mutant with disrupted nucleosome binding withe retained H2A ubiquitylation ability, and a RING mutant that solely impairs H2A ubiquitylation. In both cases, we demonstrate that these mutants are hypersensitive to DNA damage agents, including polyADP-ribose polymerase (PARP) inhibitors, and demonstrate reduced capacity of BARD1 to associate with chromatin and foci formation owing to attenuated repair capacity. Conclusion: Our studies provide convincing evidence BRCA1-BARD1 interacts with nucleosomes and ubiquitylates histones via its E3 ligase activity. Further, it plays a critical role in DNA damage response and repair that contributes to genome stability, which when disrupted sensitizes them to DNA damage agents. Our results open new avenues towards understanding whether and how these mutations in BRCA1-BARD1 affect its tumor suppression functions and their implications clinically, ultimately with the goal to translate these findings for the benefit of cancer patients. Citation Format: Wenjing Li, Samuel R. Witus, Meiling Wang, Peter S. Brzovic, Rachel E. Klevit, Weixing Zhao. BRCA1-BARD1 ubiquitylates histones for genome maintenance [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6096.
- Published
- 2023
- Full Text
- View/download PDF
16. HSPB5 disease-associated mutations have long-range effects on structure and dynamics through networks of quasi-ordered interactions
- Author
-
Christopher N Woods, Lindsey D Ulmer, Maria K Janowska, Natalie L Stone, Ellie I James, Miklos Guttman, Matthew F Bush, and Rachel E Klevit
- Abstract
Small heat shock proteins (sHSPs) are chaperones whose importance in protein homeostasis is exemplified by dozens of missense mutations associated with tissue-specific disease states. Despite decades of studies, the structure, dynamics, and mechanism of chaperone activity remain unclear. Here we show that the human sHSP HSPB5 distinguishes native lens protein γD-crystallin from damaged γD-crystallin even though the mutant/damaged client is folded. The disordered N-terminal region of HSPB5 (NTR) is essential for its chaperone activity, whereas the structured domain (ACD) has no intrinsic activity. Nevertheless, two sHSP mutational hotspots associated with disease, D109 and R120, are located in the ACD. Our studies on wild-type HSPB5 oligomers reveal that distinct regions within the NTR interact with specific grooves presented on the ACD dimer and/or with other NTR sub-regions and that the number of binding partners is greater than the number of binding sites, leading to a large, but finite number of potential combinations of interactions at any given time. The ACD mutations result in increased dynamics and accessibility of the disordered NTR and enhanced chaperone activity in vitro. Our findings reveal that HSPB5 quasi-order is delicately balanced and that perturbations arising from mutations within the structured core cause alterations that contribute to misbalance in eye lens protein homeostasis that lead to cataract formation.
- Published
- 2022
- Full Text
- View/download PDF
17. Author response: A native chemical chaperone in the human eye lens
- Author
-
Eugene Serebryany, Sourav Chowdhury, Christopher N Woods, David C Thorn, Nicki E Watson, Arthur A McClelland, Rachel E Klevit, and Eugene I Shakhnovich
- Published
- 2022
- Full Text
- View/download PDF
18. Hemi-methylated DNA regulates DNA methylation inheritance through allosteric activation of H3 ubiquitylation by UHRF1
- Author
-
Joseph S Harrison, Evan M Cornett, Dennis Goldfarb, Paul A DaRosa, Zimeng M Li, Feng Yan, Bradley M Dickson, Angela H Guo, Daniel V Cantu, Lilia Kaustov, Peter J Brown, Cheryl H Arrowsmith, Dorothy A Erie, Michael B Major, Rachel E Klevit, Krzysztof Krajewski, Brian Kuhlman, Brian D Strahl, and Scott B Rothbart
- Subjects
DNA methylation ,ubiquitin ,histone post-translational modifications ,UHRF1 ,ubiquitylation ,epigenetics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The epigenetic inheritance of DNA methylation requires UHRF1, a histone- and DNA-binding RING E3 ubiquitin ligase that recruits DNMT1 to sites of newly replicated DNA through ubiquitylation of histone H3. UHRF1 binds DNA with selectivity towards hemi-methylated CpGs (HeDNA); however, the contribution of HeDNA sensing to UHRF1 function remains elusive. Here, we reveal that the interaction of UHRF1 with HeDNA is required for DNA methylation but is dispensable for chromatin interaction, which is governed by reciprocal positive cooperativity between the UHRF1 histone- and DNA-binding domains. HeDNA recognition activates UHRF1 ubiquitylation towards multiple lysines on the H3 tail adjacent to the UHRF1 histone-binding site. Collectively, our studies are the first demonstrations of a DNA-protein interaction and an epigenetic modification directly regulating E3 ubiquitin ligase activity. They also define an orchestrated epigenetic control mechanism involving modifications both to histones and DNA that facilitate UHRF1 chromatin targeting, H3 ubiquitylation, and DNA methylation inheritance.
- Published
- 2016
- Full Text
- View/download PDF
19. UbcH5 Interacts with Substrates to Participate in Lysine Selection with the E3 Ubiquitin Ligase CHIP
- Author
-
Holly N Haver, Theodore Keppel, Kenneth Matthew Scaglione, Adam J. Kanack, Rachel E. Klevit, Rebekah L. Gundry, and Vinayak Vittal
- Subjects
Models, Molecular ,biology ,HSC70-INTERACTING PROTEIN ,Chemistry ,Lysine ,Ubiquitin-Protein Ligases ,Ubiquitination ,Substrate (chemistry) ,Chip ,complex mixtures ,Biochemistry ,Article ,Ubiquitin ligase ,Cell biology ,Residue (chemistry) ,Ubiquitin ,Ubiquitin-Conjugating Enzymes ,biology.protein ,Humans ,bacteria ,Protein folding - Abstract
The E3 ubiquitin ligase C-terminus of Hsc70 interacting protein (CHIP) plays a critical role in regulating the ubiquitin-dependent degradation of misfolded proteins. CHIP mediates the ubiquitination of the α-amino-terminus of substrates with the E2 Ube2w and facilitates the ubiquitination of lysine residues with the E2 UbcH5. While it is known that Ube2w directly interacts with the disordered regions at the N-terminus of its substrates, it is unclear how CHIP and UbcH5 mediate substrate lysine selection. Here, we have decoupled the contributions of the E2, UbcH5, and the E3, CHIP, in ubiquitin transfer. We show that UbcH5 selects substrate lysine residues independent of CHIP, and that CHIP participates in lysine selection by fine-tuning the subset of substrate lysines that are ubiquitinated. We also identify lysine 128 near the C-terminus of UbcH5 as a critical residue for the efficient ubiquitin transfer by UbcH5 in both the presence and absence of CHIP. Together, these data demonstrate an important role of the UbcH5/substrate interactions in mediating the efficient ubiquitin transfer by the CHIP/UbcH5 complex.
- Published
- 2020
- Full Text
- View/download PDF
20. Peeking from behind the veil of enigma: emerging insights on small heat shock protein structure and function
- Author
-
Rachel E. Klevit
- Subjects
Models, Molecular ,0301 basic medicine ,Physics ,030102 biochemistry & molecular biology ,Short paper ,PERSPECTIVES ON sHSPs ,Cell Biology ,Computational biology ,Biochemistry ,Heat-Shock Proteins, Small ,Structure and function ,03 medical and health sciences ,030104 developmental biology ,Structural biology ,Heat shock protein ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Sequence space (evolution) ,Small Heat-Shock Proteins ,Function (biology) ,Dynamic equilibrium - Abstract
This is a short paper on new ways to think about the structure and function of small heat shock proteins (sHSPs), perhaps the most enigmatic family among protein chaperones. The goal is to incorporate new observations regarding the disordered regions of small heat shock proteins (sHSPs) into the large body of structural information on the conserved structural alpha-crystallin domains (ACD) that define the sHSP family. Disordered regions (N-terminal region and C-terminal region or NTR and CTR, respectively) represent over 50% of the sHSP sequence space in the human genome and are refractory to traditional structural biology approaches, posing a roadblock on the path towards a mechanistic understanding of how sHSPs function. A model in which an ACD dimer serves as a template that presents three grooves into which other proteins or other segments of sHSPs can bind is presented. Short segments within the disordered regions are observed to bind into the ACD grooves. There are more binding segments than there are grooves, and each binding event is weak and transient, creating a dynamic equilibrium of tethered and untethered disordered regions. The ability of an NTR to be in dynamic equilibrium between tethered/sequestered and untethered states suggests several mechanistic alternatives that need not be mutually exclusive. New ways of thinking about (and approaching) the intrinsic properties of sHSPs may finally allow the veil of enigma to be removed from sHSPs.
- Published
- 2020
- Full Text
- View/download PDF
21. Cullin-independent recognition of HHARI substrates by a dynamic RBR catalytic domain
- Author
-
Katherine H. Reiter, Alex Zelter, Maria K. Janowska, Michael Riffle, Nicholas Shulman, Brendan X. MacLean, Kaipo Tamura, Matthew C. Chambers, Michael J. MacCoss, Trisha N. Davis, Miklos Guttman, Peter S. Brzovic, and Rachel E. Klevit
- Subjects
Structural Biology ,Ubiquitin ,Catalytic Domain ,Ubiquitin-Protein Ligases ,Ubiquitination ,Cullin Proteins ,Molecular Biology - Abstract
RING-between-RING (RBR) E3 ligases mediate ubiquitin transfer through an obligate E3-ubiquitin thioester intermediate prior to substrate ubiquitination. Although RBRs share a conserved catalytic module, substrate recruitment mechanisms remain enigmatic, and the relevant domains have yet to be identified for any member of the class. Here we characterize the interaction between the auto-inhibited RBR, HHARI (AriH1), and its target protein, 4EHP, using a combination of XL-MS, HDX-MS, NMR, and biochemical studies. The results show that (1) a di-aromatic surface on the catalytic HHARI Rcat domain forms a binding platform for substrates and (2) a phosphomimetic mutation on the auto-inhibitory Ariadne domain of HHARI promotes release and reorientation of Rcat for transthiolation and substrate modification. The findings identify a direct binding interaction between a RING-between-RING ligase and its substrate and suggest a general model for RBR substrate recognition.
- Published
- 2022
22. A conserved histidine modulates HSPB5 structure to trigger chaperone activity in response to stress-related acidosis
- Author
-
Ponni Rajagopal, Eric Tse, Andrew J Borst, Scott P Delbecq, Lei Shi, Daniel R Southworth, and Rachel E Klevit
- Subjects
protein chaperone ,heat shock protein ,protein aggregation ,electron microscopy ,NMR ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Small heat shock proteins (sHSPs) are essential ‘holdase’ chaperones that form large assemblies and respond dynamically to pH and temperature stresses to protect client proteins from aggregation. While the alpha-crystallin domain (ACD) dimer of sHSPs is the universal building block, how the ACD transmits structural changes in response to stress to promote holdase activity is unknown. We found that the dimer interface of HSPB5 is destabilized over physiological pHs and a conserved histidine (His-104) controls interface stability and oligomer structure in response to acidosis. Destabilization by pH or His-104 mutation shifts the ACD from dimer to monomer but also results in a large expansion of HSPB5 oligomer states. Remarkably, His-104 mutant-destabilized oligomers are efficient holdases that reorganize into structurally distinct client–bound complexes. Our data support a model for sHSP function wherein cell stress triggers small perturbations that alter the ACD building blocks to unleash a cryptic mode of chaperone action.
- Published
- 2015
- Full Text
- View/download PDF
23. Acidic pH and divalent cation sensing by PhoQ are dispensable for systemic salmonellae virulence
- Author
-
Kevin G Hicks, Scott P Delbecq, Enea Sancho-Vaello, Marie-Pierre Blanc, Katja K Dove, Lynne R Prost, Margaret E Daley, Kornelius Zeth, Rachel E Klevit, and Samuel I Miller
- Subjects
Salmonella ,bacterial pathogenesis ,intracellular virulence ,innate immunity ,signal transduction ,sensor kinase ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Salmonella PhoQ is a histidine kinase with a periplasmic sensor domain (PD) that promotes virulence by detecting the macrophage phagosome. PhoQ activity is repressed by divalent cations and induced in environments of acidic pH, limited divalent cations, and cationic antimicrobial peptides (CAMP). Previously, it was unclear which signals are sensed by salmonellae to promote PhoQ-mediated virulence. We defined conformational changes produced in the PhoQ PD on exposure to acidic pH that indicate structural flexibility is induced in α-helices 4 and 5, suggesting this region contributes to pH sensing. Therefore, we engineered a disulfide bond between W104C and A128C in the PhoQ PD that restrains conformational flexibility in α-helices 4 and 5. PhoQW104C-A128C is responsive to CAMP, but is inhibited for activation by acidic pH and divalent cation limitation. phoQW104C-A128C Salmonella enterica Typhimurium is virulent in mice, indicating that acidic pH and divalent cation sensing by PhoQ are dispensable for virulence.
- Published
- 2015
- Full Text
- View/download PDF
24. The BRCA1/BARD1 ubiquitin ligase and its substrates
- Author
-
Samuel R. Witus, Rachel E. Klevit, and Mikaela D. Stewart
- Subjects
DNA Repair ,Carcinogenesis ,Ubiquitin-Protein Ligases ,Breast Neoplasms ,medicine.disease_cause ,Biochemistry ,Article ,Histones ,Ubiquitin ,BARD1 ,Cell Line, Tumor ,Transcriptional regulation ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Ligase activity ,Molecular Biology ,chemistry.chemical_classification ,Ovarian Neoplasms ,DNA ligase ,biology ,Chemistry ,BRCA1 Protein ,Tumor Suppressor Proteins ,Estrogen Receptor alpha ,Cell Biology ,Cell biology ,Ubiquitin ligase ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Ubiquitins ,Mutation ,biology.protein ,Female - Abstract
Mutations in breast cancer type 1 susceptibility protein (BRCA1) and its heterodimeric binding partner BARD1 confer a high risk for the development of breast and ovarian cancers. The sole enzymatic function of the BRCA1/BARD1 complex is as a RING-type E3 ubiquitin (Ub) ligase, leading to the deposition of Ub signals onto a variety of substrate proteins. Distinct types of Ub signals deposited by BRCA1/BARD1 (i.e. degradative vs. non-degradative; mono-Ub vs. poly-Ub chains) on substrate proteins mediate aspects of its function in DNA double-stranded break repair, cell-cycle regulation, and transcriptional regulation. While cancer-predisposing mutations in both subunits lead to the inactivation of BRCA1/BARD1 ligase activity, controversy remains as to whether its Ub ligase activity directly inhibits tumorigenesis. Investigation of BRCA1/BARD1 substrates using rigorous, well-validated mutants and experimental systems will ultimately clarify the role of its ligase activity in cancer and possibly establish prognostic and diagnostic metrics for patients with mutations. In this review, we discuss the Ub ligase function of BRCA1/BARD1, highlighting experimental approaches, mechanistic considerations, and reagents that are useful in the study of substrate ubiquitylation. We also discuss the current understanding of two well-established BRCA1/BARD1 substrates (nucleosomal H2A and estrogen receptor α) and several recently discovered substrates (p50, NF2, Oct1, and LARP7). Lessons from the current body of work should provide a road map to researchers examining novel substrates and biological functions attributed to BRCA1/BARD1 Ub ligase activity.
- Published
- 2021
25. The ubiquitin ligase SspH1 from Salmonella uses a modular and dynamic E3 domain to catalyze substrate ubiquitylation
- Author
-
Rachel E. Klevit, Peter S. Brzovic, Matthew Cook, Scott P Delbecq, Thomas P. Schweppe, and Miklos Guttman
- Subjects
0301 basic medicine ,Conformational change ,030102 biochemistry & molecular biology ,biology ,Effector ,Chemistry ,Active site ,Cell Biology ,Biochemistry ,Ubiquitin ligase ,03 medical and health sciences ,030104 developmental biology ,Ubiquitin ,biology.protein ,Biophysics ,Hydrogen–deuterium exchange ,Molecular Biology ,Function (biology) ,Cysteine - Abstract
SspH/IpaH bacterial effector E3 ubiquitin (Ub) ligases, unrelated in sequence or structure to eukaryotic E3s, are utilized by a wide variety of Gram-negative bacteria during pathogenesis. These E3s function in a eukaryotic environment, utilize host cell E2 ubiquitin-conjugating enzymes of the Ube2D family, and target host proteins for ubiquitylation. Despite several crystal structures, details of Ube2D∼Ub binding and the mechanism of ubiquitin transfer are poorly understood. Here, we show that the catalytic E3 ligase domain of SspH1 can be divided into two subdomains: an N-terminal subdomain that harbors the active-site cysteine and a C-terminal subdomain containing the Ube2D∼Ub-binding site. SspH1 mutations designed to restrict subdomain motions show rapid formation of an E3∼Ub intermediate, but impaired Ub transfer to substrate. NMR experiments using paramagnetic spin labels reveal how SspH1 binds Ube2D∼Ub and targets the E2∼Ub active site. Unexpectedly, hydrogen/deuterium exchange MS shows that the E2∼Ub-binding region is dynamic but stabilized in the E3∼Ub intermediate. Our results support a model in which both subunits of an Ube2D∼Ub clamp onto a dynamic region of SspH1, promoting an E3 conformation poised for transthiolation. A conformational change is then required for Ub transfer from E3∼Ub to substrate.
- Published
- 2019
- Full Text
- View/download PDF
26. Toggle switch residues control allosteric transitions in bacterial adhesins by participating in a concerted repacking of the protein core
- Author
-
Hovhannes Avagyan, Rachel E. Klevit, Laura A. Carlucci, Evgeni V. Sokurenko, Benjamin Basanta, Dagmara I. Kisiela, Angelo Ramos, Wendy E. Thomas, Ronald E. Stenkamp, Pearl Magala, Veronika Tchesnokova, Vladimir Yarov-Yarovoy, Anahit Hovhannisyan, Gianluca Interlandi, and Francetic, Olivera
- Subjects
Models, Molecular ,Hydrolases ,Mannose ,Catch bond ,Pathology and Laboratory Medicine ,Spectrum analysis techniques ,Biochemistry ,Epitope ,Bacterial Adhesion ,chemistry.chemical_compound ,0302 clinical medicine ,Electronics Engineering ,Models ,Microbial Physiology ,Lectins ,Medicine and Health Sciences ,Bacterial Physiology ,Amino Acids ,Enzyme-Linked Immunoassays ,Biology (General) ,Alanine ,0303 health sciences ,Adhesins, Escherichia coli ,Chemistry ,Organic Compounds ,Mannose binding ,Bacterial ,Adhesins ,Enzymes ,Medical Microbiology ,Physical Sciences ,Engineering and Technology ,Fimbriae Proteins ,Cellular Structures and Organelles ,Pathogens ,Research Article ,Protein Binding ,Steric effects ,Pathogen Motility ,Stereochemistry ,Virulence Factors ,Nucleases ,QH301-705.5 ,Allosteric regulation ,Immunology ,Microbiology ,Fimbriae ,03 medical and health sciences ,Ribonucleases ,NMR spectroscopy ,Virology ,DNA-binding proteins ,Genetics ,Toggle Switches ,Escherichia coli ,Adhesins, Bacterial ,Immunoassays ,Molecular Biology ,030304 developmental biology ,Organic Chemistry ,Chemical Compounds ,Biology and Life Sciences ,Proteins ,Molecular ,Bacteriology ,Cell Biology ,RC581-607 ,Bacterial adhesin ,Research and analysis methods ,Pili and Fimbriae ,Aliphatic Amino Acids ,Fimbriae, Bacterial ,Enzymology ,Immunologic Techniques ,Parasitology ,Generic health relevance ,Immunologic diseases. Allergy ,030217 neurology & neurosurgery - Abstract
Critical molecular events that control conformational transitions in most allosteric proteins are ill-defined. The mannose-specific FimH protein of Escherichia coli is a prototypic bacterial adhesin that switches from an ‘inactive’ low-affinity state (LAS) to an ‘active’ high-affinity state (HAS) conformation allosterically upon mannose binding and mediates shear-dependent catch bond adhesion. Here we identify a novel type of antibody that acts as a kinetic trap and prevents the transition between conformations in both directions. Disruption of the allosteric transitions significantly slows FimH’s ability to associate with mannose and blocks bacterial adhesion under dynamic conditions. FimH residues critical for antibody binding form a compact epitope that is located away from the mannose-binding pocket and is structurally conserved in both states. A larger antibody-FimH contact area is identified by NMR and contains residues Leu-34 and Val-35 that move between core-buried and surface-exposed orientations in opposing directions during the transition. Replacement of Leu-34 with a charged glutamic acid stabilizes FimH in the LAS conformation and replacement of Val-35 with glutamic acid traps FimH in the HAS conformation. The antibody is unable to trap the conformations if Leu-34 and Val-35 are replaced with a less bulky alanine. We propose that these residues act as molecular toggle switches and that the bound antibody imposes a steric block to their reorientation in either direction, thereby restricting concerted repacking of side chains that must occur to enable the conformational transition. Residues homologous to the FimH toggle switches are highly conserved across a diverse family of fimbrial adhesins. Replacement of predicted switch residues reveals that another E. coli adhesin, galactose-specific FmlH, is allosteric and can shift from an inactive to an active state. Our study shows that allosteric transitions in bacterial adhesins depend on toggle switch residues and that an antibody that blocks the switch effectively disables adhesive protein function., Author summary To bind their ligands, allosteric proteins shift between ‘inactive’ and ‘active’ states, but molecular details of the conformational changes during the transition are often unclear. We describe a monoclonal antibody against the mannose-specific bacterial adhesin, FimH, that blocks the conformational transition in both directions. The antibody-trapped LAS and HAS conformations of FimH are unable to mediate bacterial adhesion under dynamic shear conditions. We propose that the conformational trapping involves a steric block of the core-to-surface switching of certain residues which is critical for the allosteric transitions. Furthermore, we demonstrate that the allosteric switches are structurally and functionally conserved across a broad spectrum of bacterial fimbrial adhesins.
- Published
- 2021
27. Mediator subunit Med15 dictates the conserved 'fuzzy' binding mechanism of yeast transcription activators Gal4 and Gcn4
- Author
-
Linda Warfield, Rachel E. Klevit, Lisa M. Tuttle, Steven Hahn, Damien B. Wilburn, and Derek Pacheco
- Subjects
0301 basic medicine ,Models, Molecular ,Saccharomyces cerevisiae Proteins ,Molecular biology ,Protein subunit ,Science ,Activation function ,General Physics and Astronomy ,Repressor ,Sequence (biology) ,Saccharomyces cerevisiae ,Intrinsically disordered proteins ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Mediator ,NMR spectroscopy ,Transcription (biology) ,Protein Interaction Domains and Motifs ,Transcription factor ,Multidisciplinary ,Mediator Complex ,Chemistry ,General Chemistry ,Methyltransferases ,DNA-Binding Proteins ,Repressor Proteins ,030104 developmental biology ,Basic-Leucine Zipper Transcription Factors ,Biophysics ,Transcription ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
The acidic activation domain (AD) of yeast transcription factor Gal4 plays a dual role in transcription repression and activation through binding to Gal80 repressor and Mediator subunit Med15. The activation function of Gal4 arises from two hydrophobic regions within the 40-residue AD. We show by NMR that each AD region binds the Mediator subunit Med15 using a “fuzzy” protein interface. Remarkably, comparison of chemical shift perturbations shows that Gal4 and Gcn4, two intrinsically disordered ADs of different sequence, interact nearly identically with Med15. The finding that two ADs of different sequence use an identical fuzzy binding mechanism shows a common sequence-independent mechanism for AD-Mediator binding, similar to interactions within a hydrophobic cloud. In contrast, the same region of Gal4 AD interacts strongly with Gal80 via a distinct structured complex, implying that the structured binding partner of an intrinsically disordered protein dictates the type of protein–protein interaction., The intrinsically disordered acidic activation domain (AD) of the yeast transcription factor Gal4 acts through binding to the Med15 subunit of the Mediator complex. Here, the authors show that Gal4 interacts with Med15 through an identical fuzzy binding mechanism as Gcn4 AD, which has a different sequence, revealing a common sequence-independent mechanism for AD-Mediator binding. In contrast, Gal4 AD binds to the Gal80 repressor as a structured polypeptide, which strongly suggests that the structured binding partner dictates the type of protein–protein interaction for an intrinsically disordered protein.
- Published
- 2021
28. A native chemical chaperone in the human eye lens
- Author
-
Eugene Serebryany, Sourav Chowdhury, Christopher N. Woods, David C. Thorn, Nicki E. Watson, Arthur McClelland, Rachel E. Klevit, and Eugene I. Shakhnovich
- Subjects
General Immunology and Microbiology ,Chemistry ,General Neuroscience ,General Medicine ,Protein aggregation ,Cataract ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Lens (optics) ,Lens protein ,Protein Aggregates ,Metabolomics ,medicine.anatomical_structure ,law ,Lens, Crystalline ,medicine ,Biophysics ,Humans ,Human eye ,Electron microscope ,Chemical chaperone ,Inositol ,Molecular Chaperones - Abstract
Cataract is one of the most prevalent protein aggregation disorders and still the most common cause of vision loss worldwide. The metabolically quiescent core region of the human lens lacks cellular or protein turnover; it has therefore evolved remarkable mechanisms to resist light-scattering protein aggregation for a lifetime. We now report that one such mechanism involves an unusually abundant lens metabolite, myo-inositol, suppressing aggregation of lens crystallins. We quantified aggregation suppression using our previously well-characterized in vitro aggregation assays of oxidation-mimicking human γD-crystallin variants and investigated myo-inositol’s molecular mechanism of action using solution NMR, negative-stain TEM, differential scanning fluorometry, thermal scanning Raman spectroscopy, turbidimetry in redox buffers, and free thiol quantitation. Unlike many known chemical chaperones, myo-inositol’s primary target was neither the native nor the unfolded state of the protein, nor the final aggregated state, but rather the rate-limiting bimolecular step on the aggregation pathway. Given recent metabolomic evidence that it is severely depleted in human cataractous lenses compared to age-matched controls, we suggest that maintaining or restoring healthy levels of myo-inositol in the lens may be a simple, safe, and globally accessible strategy to prevent or delay lens opacification due to age-onset cataract.
- Published
- 2020
- Full Text
- View/download PDF
29. BRCA1/BARD1 site-specific ubiquitylation of nucleosomal H2A is directed by BARD1
- Author
-
Weixing Zhao, Champak Chatterjee, Anika L. Burrell, Lisa M. Tuttle, Samuel R. Witus, Rachel E. Klevit, Peter S. Brzovic, Jianming Kang, Meiling Wang, Frank DiMaio, Daniel P. Farrell, Mikaela D. Stewart, Jesse M Hansen, Justin M. Kollman, and Alex Pravat
- Subjects
Models, Molecular ,Ubiquitin-Protein Ligases ,Lysine ,Article ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Structural Biology ,BARD1 ,Catalytic Domain ,Nucleosome ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Binding Sites ,biology ,Chemistry ,BRCA1 Protein ,Tumor Suppressor Proteins ,Cryoelectron Microscopy ,Ubiquitination ,Reproducibility of Results ,Methylation ,Ubiquitin ligase ,Cell biology ,Nucleosomes ,BMI1 ,Ubiquitin-Conjugating Enzymes ,biology.protein ,PRC1 ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Mutations in the E3 ubiquitin ligase RING domains of BRCA1/BARD1 predispose carriers to breast and ovarian cancers. We present the structure of the BRCA1/BARD1 RING heterodimer with the E2 enzyme UbcH5c bound to its cellular target, the nucleosome, along with biochemical data that explain how the complex selectively ubiquitylates lysines 125, 127 and 129 in the flexible C-terminal tail of H2A in a fully human system. The structure reveals that a novel BARD1-histone interface couples to a repositioning of UbcH5c compared to the structurally similar PRC1 E3 ligase Ring1b/Bmi1 that ubiquitylates H2A Lys119 in nucleosomes. This interface is sensitive to both H3 Lys79 methylation status and mutations found in individuals with cancer. Furthermore, NMR reveals an unexpected mode of E3-mediated substrate regulation through modulation of dynamics in the C-terminal tail of H2A. Our findings provide insight into how E3 ligases preferentially target nearby lysine residues in nucleosomes by a steric occlusion and distancing mechanism.
- Published
- 2020
30. Legionella effector MavC targets the Ube2N~Ub conjugate for noncanonical ubiquitination
- Author
-
Shalini Iyer, Rachel E. Klevit, Kedar Puvar, Jiaqi Fu, Kristos I. Negrón Terón, Sebastian Kenny, Zhao-Qing Luo, Peter S. Brzovic, and Chittaranjan Das
- Subjects
Models, Molecular ,0301 basic medicine ,Tissue transglutaminase ,Science ,General Physics and Astronomy ,Plasma protein binding ,Crystallography, X-Ray ,Article ,General Biochemistry, Genetics and Molecular Biology ,Legionella pneumophila ,Substrate Specificity ,03 medical and health sciences ,Protein structure ,Bacterial Proteins ,Ubiquitin ,Catalytic Domain ,Cloning, Molecular ,lcsh:Science ,Deamidation ,X-ray crystallography ,Transglutaminases ,Multidisciplinary ,030102 biochemistry & molecular biology ,biology ,Effector ,Chemistry ,Ubiquitination ,Substrate (chemistry) ,General Chemistry ,Recombinant Proteins ,Protein Structure, Tertiary ,030104 developmental biology ,Ubiquitin-Conjugating Enzymes ,Mutagenesis, Site-Directed ,biology.protein ,Biophysics ,lcsh:Q ,Structural biology ,Protein Binding ,Conjugate - Abstract
The bacterial effector MavC modulates the host immune response by blocking Ube2N activity employing an E1-independent ubiquitin ligation, catalyzing formation of a γ-glutamyl-ε-Lys (Gln40Ub-Lys92Ube2N) isopeptide crosslink using a transglutaminase mechanism. Here we provide biochemical evidence in support of MavC targeting the activated, thioester-linked Ube2N~ubiquitin conjugate, catalyzing an intramolecular transglutamination reaction, covalently crosslinking the Ube2N and Ub subunits effectively inactivating the E2~Ub conjugate. Ubiquitin exhibits weak binding to MavC alone, but shows an increase in affinity when tethered to Ube2N in a disulfide-linked substrate that mimics the charged E2~Ub conjugate. Crystal structures of MavC in complex with the substrate mimic and crosslinked product provide insights into the reaction mechanism and underlying protein dynamics that favor transamidation over deamidation, while revealing a crucial role for the structurally unique insertion domain in substrate recognition. This work provides a structural basis of ubiquitination by transglutamination and identifies this enzyme’s true physiological substrate., The Legionella pneumophila effector MavC inhibits the human ubiquitin-conjugating enzyme Ube2N. Here, the authors combine NMR, X-ray crystallography and biochemical assays and show that MavC catalyses the intramolecular transglutaminase reaction between the Ube2N and Ub subunits of the Ube2N∼Ub conjugate and present the substrate- and product-bound MavC crystal structures.
- Published
- 2020
- Full Text
- View/download PDF
31. Who with whom: functional coordination of E2 enzymes by RING E3 ligases during poly-ubiquitylation
- Author
-
Christian, Lips, Tobias, Ritterhoff, Annika, Weber, Maria K, Janowska, Mandy, Mustroph, Thomas, Sommer, and Rachel E, Klevit
- Subjects
Saccharomyces cerevisiae Proteins ,Ubiquitin ,Ubiquitin-Protein Ligases ,Ubiquitination ,Post-translational Modifications, Proteolysis & Proteomics ,Saccharomyces cerevisiae ,Articles ,Article ,ER‐associated protein degradation ,Structural Biology ,Proteolysis ,Ubiquitin-Conjugating Enzymes ,Humans ,RING E3 ligase ,E2 conjugating enzyme ,Poly A ,Polyubiquitin ,Protein Processing, Post-Translational ,linchpin - Abstract
Protein modification with poly‐ubiquitin chains is a crucial process involved in a myriad of cellular pathways. Chain synthesis requires two steps: substrate modification with ubiquitin (priming) followed by repetitive ubiquitin‐to‐ubiquitin attachment (elongation). RING‐type E3 ligases catalyze both reactions in collaboration with specific priming and elongating E2 enzymes. We provide kinetic insight into poly‐ubiquitylation during protein quality control by showing that priming is the rate‐determining step in protein degradation as directed by the yeast ERAD RING E3 ligases, Hrd1 and Doa10. Doa10 cooperates with the dedicated priming E2, Ubc6, while both E3s use Ubc7 for elongation. Here, we provide direct evidence that Hrd1 uses Ubc7 also for priming. We found that Ubc6 has an unusually high basal activity that does not require strong stimulation from an E3. Doa10 exploits this property to pair with Ubc6 over Ubc7 during priming. Our work not only illuminates the mechanisms of specific E2/E3 interplay in ERAD, but also offers a basis to understand how RING E3s may have properties that are tailored to pair with their preferred E2s., Kinetic analyses of ERAD E3 (Hrd1, Doa10) and E2 (Ubc6, Ubc7) enzymes reveal the rate‐limiting nature of the ubiquitin chain priming step, and the molecular basis for respective E2 preferences of these E3s during priming.
- Published
- 2020
32. Mechanistic insights revealed by a UBE2A mutation linked to intellectual disability
- Author
-
Juliana Ferreira de Oliveira, Camila Canateli, Carla Rosenberg, Paulo Alberto Otto, Ana Cristina Victorino Krepischi, Rachel E. Klevit, Mariana Maschietto, Americo Tavares Ranzani, Paulo S. Oliveira, Kleber G. Franchini, Silvia S. Costa, Paula Favoretti Vital do Prado, Maurício L. Sforça, and UNIVERSIDADE ESTADUAL DE CAMPINAS
- Subjects
Adult ,Male ,Magnetic Resonance Spectroscopy ,Lysine ,Mutation, Missense ,Intellectual disability ,Crystallography, X-Ray ,medicine.disease_cause ,03 medical and health sciences ,Ubiquitin ,Catalytic Domain ,Intellectual Disability ,Proliferating Cell Nuclear Antigen ,medicine ,Humans ,Artigo original ,Missense mutation ,Deficiência intelectual ,Ubiquitins ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,DNA ligase ,Mutation ,biology ,Chemistry ,030302 biochemistry & molecular biology ,Ubiquitination ,Cell Biology ,Hydrogen-Ion Concentration ,Ubiquitina ,PESSOAS COM DEFICIÊNCIA INTELECTUAL ,Protein ubiquitination ,Ubiquitin ligase ,Biochemistry ,Ubiquitin-Conjugating Enzymes ,biology.protein ,Female ,Cysteine - Abstract
Agradecimentos: LNBio/CNPEM; Brazilian National Council for Scientific and Technological Development (CNPq)National Council for Scientific and Technological Development (CNPq) [306879/2014-0, 310536/2014-6, 422790/2016-8]; Sao Paulo Research Foundation (FAPESP)Fundacao de Amparo a Pesquisa do Estado de Sao Paulo (FAPESP) [2012/50981-5, 2013/08028-1, 2015/06281-7]; NIH/NIGMSUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Institute of General Medical Sciences (NIGMS) [R01 GM088055] Ubiquitin-conjugating enzymes (E2) enable protein ubiquitination by conjugating ubiquitin to their catalytic cysteine for subsequent transfer to a target lysine side chain. Deprotonation of the incoming lysine enables its nucleophilicity, but determinants of lysine activation remain poorly understood. We report a novel pathogenic mutation in the E2 UBE2A, identified in two brothers with mild intellectual disability. The pathogenic Q93E mutation yields UBE2A with impaired aminolysis activity but no loss of the ability to be conjugated with ubiquitin. Importantly, the low intrinsic reactivity of UBE2A Q93E was not overcome by a cognate ubiquitin E3 ligase, RAD18, with the UBE2A target PCNA. However, UBE2A Q93E was reactive at high pH or with a lowpK a amine as the nucleophile, thus providing the first evidence of reversion of a defective UBE2A mutation. We propose that Q93E substitution perturbs the UBE2A catalytic microenvironment essential for lysine deprotonation during ubiquitin transfer, thus generating an enzyme that is disabled but not dead FUNDAÇÃO DE AMPARO À PESQUISA DO ESTADO DE SÃO PAULO - FAPESP CONSELHO NACIONAL DE DESENVOLVIMENTO CIENTÍFICO E TECNOLÓGICO - CNPQ Fechado
- Published
- 2018
- Full Text
- View/download PDF
33. The mechanism of phosphoribosyl-ubiquitination mediated by a single Legionella effector
- Author
-
Katherine H. Reiter, Yong Zhang, Peter S. Brzovic, David J. Wasilko, Xiaochun Wu, Jiazhang Qiu, Yao Liu, Rachel E. Klevit, Anil Akturk, Yuxin Mao, and Zhao-Qing Luo
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,Effector ,Chemistry ,C-terminus ,Protein domain ,Lysine ,Phosphodiesterase ,Article ,Cell biology ,Serine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Ubiquitin ,biology.protein ,Target protein ,030217 neurology & neurosurgery - Abstract
Summary Ubiquitination is a post-translational modification that regulates a myriad of cellular processes in eukaryotes1–4. The conventional ubiquitination cascade culminates in a covalent linkage between the C-terminus of ubiquitin (Ub) and a target protein, most often on a lysine sidechain1,5. Recent studies of the Legionella pneumophila SidE family of effector proteins revealed a novel mode of ubiquitination in which a phosphoribosylated ubiquitin (PR-Ub) is conjugated to a serine residue on substrates via a phosphodiester bond6–8. To uncover the molecular mechanism of this unique post-translational modification, we determined the crystal structure of a fragment of the SidE family member SdeA that retains ubiquitination activity. The structure reveals that the catalytic module contains two distinct functional units: a phosphodiesterase domain (PDE) and a mono-ADP-ribosyltransferase (mART) domain. Biochemical analysis shows that the mART domain-mediated conversion of Ub to ADP-ribosylated Ub (ADPR-Ub) and the PDE domain-mediated ligation of PR-Ub to substrates are two independent activities of SdeA. Furthermore, we present two crystal structures of a homologous PDE domain from the SidE family member SdeD9 in complex with Ub or ADPR-Ub. The structures suggest an intriguing mechanism for how SdeA processes ADPR-Ub to PR-Ub plus AMP and conjugates PR-Ub to a serine residue in substrates. Our study establishes the molecular mechanism of phosphoribosyl-ubiquitination (PR-ubiquitination) and paves the way for future studies of this unusual type of ubiquitination in eukaryotes.
- Published
- 2018
34. <scp>S</scp> tructural basis for tankyrase‐RNF146 interaction reveals noncanonical tankyrase‐binding motifs
- Author
-
Rachel E. Klevit, Paul A. DaRosa, and Wenqing Xu
- Subjects
Ankyrins ,Models, Molecular ,0301 basic medicine ,Poly Adenosine Diphosphate Ribose ,Ubiquitin-Protein Ligases ,Plasma protein binding ,Biochemistry ,03 medical and health sciences ,Tankyrases ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Wnt signaling pathway ,Articles ,Ubiquitin ligase ,Cell biology ,Vesicular transport protein ,030104 developmental biology ,Hippo signaling ,biology.protein ,Ankyrin repeat ,Protein Binding ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Poly(ADP‐ribosyl)ation (PARylation) catalyzed by the tankyrase enzymes (Tankyrase‐1 and ‐2; a.k.a. PARP‐5a and ‐5b) is involved in mitosis, telomere length regulation, GLUT‐4 vesicle transport, and cell growth and differentiation. Together with the E3 ubiquitin ligase RNF146 (a.k.a. Iduna), tankyrases regulate the cellular levels of several important proteins including Axin, 3BP2, and angiomotins, which are key regulators of Wnt, Src and Hippo signaling, respectively. These tankyrase substrates are first PARylated and then ubiquitylated by RNF146, which is allosterically activated by binding to PAR polymer. Each tankyrase substrate is recognized by a tankyrase‐binding motif (TBM). Here we show that RNF146 binds directly to tankyrases via motifs in its C‐terminal region. Four of these RNF146 motifs represent novel, extended TBMs, that have one or two additional amino acids between the most conserved Arg and Gly residues. The individual RNF146 motifs display weak binding, but together mediate a strong multivalent interaction with the substrate‐binding region of TNKS, forming a robust one‐to‐one complex. A crystal structure of the first RNF146 noncanonical TBM in complex with the second ankyrin repeat domain of TNKS shows how an extended motif can be accommodated in a peptide‐binding groove on tankyrases. Overall, our work demonstrates the existence of a new class of extended TBMs that exist in previously uncharacterized tankyrase‐binding proteins including those of IF4A1 and NELFE.
- Published
- 2018
- Full Text
- View/download PDF
35. Gcn4-Mediator Specificity Is Mediated by a Large and Dynamic Fuzzy Protein-Protein Complex
- Author
-
Rachel E. Klevit, Lisa M. Tuttle, Jie Luo, Linda Warfield, Steven Hahn, Jeff Ranish, and Derek Pacheco
- Subjects
Transcriptional Activation ,0301 basic medicine ,Mediator Complex ,Saccharomyces cerevisiae Proteins ,Chemistry ,Protein subunit ,Membrane Proteins ,Saccharomyces cerevisiae ,Computational biology ,Intrinsically disordered proteins ,Fuzzy logic ,Transcription Activation ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Basic-Leucine Zipper Transcription Factors ,030104 developmental biology ,Mediator ,Protein Domains ,lcsh:Biology (General) ,Transcription (biology) ,Coactivator ,lcsh:QH301-705.5 ,Gene - Abstract
Summary Transcription activation domains (ADs) are inherently disordered proteins that often target multiple coactivator complexes, but the specificity of these interactions is not understood. Efficient transcription activation by yeast Gcn4 requires its tandem ADs and four activator-binding domains (ABDs) on its target, the Mediator subunit Med15. Multiple ABDs are a common feature of coactivator complexes. We find that the large Gcn4-Med15 complex is heterogeneous and contains nearly all possible AD-ABD interactions. Gcn4-Med15 forms via a dynamic fuzzy protein-protein interface, where ADs bind the ABDs in multiple orientations via hydrophobic regions that gain helicity. This combinatorial mechanism allows individual low-affinity and specificity interactions to generate a biologically functional, specific, and higher affinity complex despite lacking a defined protein-protein interface. This binding strategy is likely representative of many activators that target multiple coactivators, as it allows great flexibility in combinations of activators that can cooperate to regulate genes with variable coactivator requirements., Graphical abstract In Brief: Tuttle et al. report a “fuzzy free-for-all” interaction mechanism that explains how seemingly unrelated transcription activators converge on a limited number of coactivator targets. The mechanism provides a rationale for the observation that individually weak and low-specificity interactions can combine to produce biologically critical function without requiring highly ordered structure.
- Published
- 2018
- Full Text
- View/download PDF
36. De novo mutation in RING1 with epigenetic effects on neurodevelopment
- Author
-
Tom Walsh, Rachel E. Klevit, Jon McClellan, Sarah B. Pierce, Abhinav Dhall, Mary Claire King, Suleyman Gulsuner, and Mikaela D. Stewart
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,Mutant ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Ubiquitin ,Histone H2A ,Animals ,Humans ,Missense mutation ,Nucleosome ,Amino Acid Sequence ,Epigenetics ,Allele ,Caenorhabditis elegans ,Polycomb Repressive Complex 1 ,Multidisciplinary ,biology ,Ubiquitination ,Gene Expression Regulation, Developmental ,Biological Sciences ,biology.organism_classification ,Nucleosomes ,Cell biology ,030104 developmental biology ,Neurodevelopmental Disorders ,Case-Control Studies ,Mutation ,biology.protein - Abstract
RING1 is an E3-ubiquitin ligase that is involved in epigenetic control of transcription during development. It is a component of the polycomb repressive complex 1, and its role in that complex is to ubiquitylate histone H2A. In a 13-year-old girl with syndromic neurodevelopmental disabilities, we identified a de novo mutation, RING1 p.R95Q, which alters a conserved arginine residue in the catalytic RING domain. In vitro assays demonstrated that the mutant RING1 retains capacity to catalyze ubiquitin chain formation, but is defective in its ability to ubiquitylate histone H2A in nucleosomes. Consistent with this in vitro effect, cells of the patient showed decreased monoubiquitylation of histone H2A. We modeled the mutant RING1 in Caenorhabditis elegans by editing the comparable amino acid change into spat-3, the suggested RING1 ortholog. Animals with either the missense mutation or complete knockout of spat-3 were defective in monoubiquitylation of histone H2A and had defects in neuronal migration and axon guidance. Relevant to our patient, animals heterozygous for either the missense or knockout allele also showed neuronal defects. Our results support three conclusions: mutation of RING1 is the likely cause of a human neurodevelopmental syndrome, mutation of RING1 can disrupt histone H2A ubiquitylation without disrupting RING1 catalytic activity, and the comparable mutation in C. elegans spat-3 both recapitulates the effects on histone H2A ubiquitylation and leads to neurodevelopmental abnormalities. This role for RING1 adds to our understanding of the importance of aberrant epigenetic effects as causes of human neurodevelopmental disorders.
- Published
- 2018
- Full Text
- View/download PDF
37. BARD1 is necessary for ubiquitylation of nucleosomal histone H2A and for transcriptional regulation of estrogen metabolism genes
- Author
-
Champak Chatterjee, Abhinav Dhall, Jacob E. Corn, Rachel E. Klevit, Elena Zelin, Mary Claire King, Mikaela D. Stewart, Esha Upadhyay, and Tom Walsh
- Subjects
Male ,0301 basic medicine ,endocrine system diseases ,Tumor suppressor gene ,Ubiquitin-Protein Ligases ,Mutant ,Mutation, Missense ,Breast Neoplasms ,Histones ,03 medical and health sciences ,Protein Domains ,Ubiquitin ,BARD1 ,Histone H2A ,Cytochrome P-450 CYP1A1 ,Transcriptional regulation ,Cytochrome P-450 CYP3A ,Humans ,Nucleosome ,skin and connective tissue diseases ,Multidisciplinary ,biology ,BRCA1 Protein ,Chemistry ,Tumor Suppressor Proteins ,Ubiquitination ,Estrogens ,Biological Sciences ,Nucleosomes ,Cell biology ,Ubiquitin ligase ,030104 developmental biology ,Gene Expression Regulation ,biology.protein ,Female - Abstract
Missense mutations that disrupt the RING domain of the tumor suppressor gene BRCA1 lead to increased risk of breast and ovarian cancer. The BRCA1 RING domain is a ubiquitin ligase, whose structure and function rely critically on forming a heterodimer with BARD1, which also harbors a RING domain. The function of the BARD1 RING domain is unknown. In families severely affected with breast cancer, we identified inherited BARD1 missense mutations Cys53Trp, Cys71Tyr, and Cys83Arg that alter three zinc-binding residues of the BARD1 RING domain. Each of these mutant BARD1 proteins retained the ability to form heterodimeric complexes with BRCA1 to make an active ubiquitin ligase, but the mutant BRCA1/BARD1 complexes were deficient in binding to nucleosomes and in ubiquitylating histone H2A. The BARD1 mutations also caused loss of transcriptional repression of BRCA1-regulated estrogen metabolism genes CYP1A1 and CYP3A4; breast epithelial cells edited to create heterozygous loss of BARD1 showed significantly higher expression of CYP1A1 and CYP3A4. Reintroduction of wild-type BARD1 into these cells restored CYP1A1 and CYP3A4 transcription to normal levels, but introduction of the cancer-predisposing BARD1 RING mutants failed to do so. These results indicate that an intact BARD1 RING domain is critical to BRCA1/BARD1 binding to nucleosomes and hence to ubiquitylation of histone H2A and also critical to transcriptional repression of BRCA1-regulated genes active in estrogen metabolism.
- Published
- 2018
- Full Text
- View/download PDF
38. Solution structure of sperm lysin yields novel insights into molecular dynamics of rapid protein evolution
- Author
-
Willie J. Swanson, Damien B. Wilburn, Lisa M. Tuttle, and Rachel E. Klevit
- Subjects
Male ,Models, Molecular ,0106 biological sciences ,0301 basic medicine ,Magnetic Resonance Spectroscopy ,Gastropoda ,Lysin ,Mutagenesis (molecular biology technique) ,Vitelline membrane ,Computational biology ,Molecular Dynamics Simulation ,Biology ,complex mixtures ,010603 evolutionary biology ,01 natural sciences ,Protein evolution ,Evolution, Molecular ,03 medical and health sciences ,Negative selection ,Molecular dynamics ,Mucoproteins ,Animals ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Multidisciplinary ,Chemistry ,Ecology ,Positive selection ,030302 biochemistry & molecular biology ,Biological Sciences ,Spermatozoa ,Solution structure ,Sperm ,030104 developmental biology ,Mutagenesis, Site-Directed ,bacteria ,Protein Multimerization - Abstract
Protein evolution is driven by the sum of different physiochemical and genetic processes that usually results in strong purifying selection to maintain biochemical functions. However, proteins that are part of systems under arms race dynamics often evolve at unparalleled rates that can produce atypical biochemical properties. In the marine mollusk abalone, lysin and VERL are a pair of rapidly coevolving proteins that are essential for species-specific interactions between sperm and egg. Despite extensive biochemical characterization of lysin, including crystal structures of multiple orthologs, it was unclear how sites under positive selection may facilitate recognition of VERL. Using a combination of targeted mutagenesis and multidimensional NMR, we present a high-definition solution structure of sperm lysin from red abalone (Haliotis rufescens). Unapparent from the crystallography data, multiple NMR-based analyses conducted in solution reveal clustering of the N-and C-termini to form a nexus of 13 positively selected sites that constitute a VERL binding interface. Evolutionary rate was found to be a significant predictor of backbone flexibility, which may be critical for lysin bioactivity and / or accelerated evolution. These flexible, rapidly evolving segments that constitute the VERL binding interface were also the most distorted regions of the crystal structure relative to what was observed in solution. While lysin has been the subject of extensive biochemical and evolutionary analyses for more than 30 years, this study highlights the enhanced insights gained from applying NMR approaches to rapidly evolving proteins.SignificanceThe fertilization of eggs by sperm is a critical biological process for nearly all sexually reproducing organisms to propagate their genetic information. Despite the importance of fertilization, the molecules that mediate egg-sperm interactions have been characterized for only a few species, and the biochemical mechanisms underlying these interactions are even less well understood. In the marine mollusk abalone, sperm lysin interacts with egg VERL in a species-specific manner to facilitate fertilization. In this report, we characterized the solution structure and molecular evolution of sperm lysin from red abalone (Haliotis rufescens), and identified the VERL binding interface as well as important lysin dimer interactions that have emerged as part of an incessant sexual arms race.
- Published
- 2018
- Full Text
- View/download PDF
39. Edmond Fischer (1920–2021)
- Author
-
John D. Scott, Rachel E. Klevit, Trisha N. Davis, and William A. Catterall
- Subjects
medicine.medical_specialty ,Multidisciplinary ,business.industry ,General surgery ,medicine ,MEDLINE ,business - Abstract
A biochemical virtuoso
- Published
- 2021
- Full Text
- View/download PDF
40. RING-Between-RING E3 Ligases: Emerging Themes amid the Variations
- Author
-
Katja K. Dove and Rachel E. Klevit
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,Computational biology ,Ring (chemistry) ,Article ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Structural Biology ,Animals ,Humans ,Molecular Biology ,chemistry.chemical_classification ,DNA ligase ,biology ,Mechanism (biology) ,Ubiquitination ,Active site ,Eukaryotic Cells ,030104 developmental biology ,chemistry ,Biochemistry ,Domain (ring theory) ,biology.protein ,030217 neurology & neurosurgery ,Cullin - Abstract
Covalent, reversible, post-translational modification of cellular proteins with the small modifier, ubiquitin (Ub), regulates virtually every known cellular process in eukaryotes. The process is carried out by a trio of enzymes: a Ub-activating (E1) enzyme, a Ub-conjugating (E2) enzyme, and a Ub ligase (E3) enzyme. RING-in-Between-RING (RBR) E3s constitute one of three classes of E3 ligases and are defined by a RING-HECT-hybrid mechanism that utilizes a E2-binding RING domain and a second domain (called RING2) that contains an active site Cys required for the formation of an obligatory E3~Ub intermediate. Albeit a small class, RBR E3s in humans regulate diverse cellular process. This review focuses on non-Parkin members such as HOIP/HOIL-1L (the only E3s known to generate linear Ub chains), HHARI and TRIAD1, both of which have been recently demonstrated to work together with Cullin RING E3 ligases. We provide a brief historical background and highlight, summarize, and discuss recent developments in the young field of RBR E3s. Insights reviewed here include new understandings of the RBR Ub-transfer mechanism, specifically the role of RING1 and various Ub-binding sites, brief structural comparisons among members, and different modes of auto-inhibition and activation.
- Published
- 2017
- Full Text
- View/download PDF
41. Tuning BRCA1 and BARD1 activity to investigate RING ubiquitin ligase mechanisms
- Author
-
Rachel E. Klevit, Paul A. DaRosa, Ernesto Coronado, Jonathan N. Pruneda, Emily D Duncan, Peter S. Brzovic, and Mikaela D. Stewart
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,030102 biochemistry & molecular biology ,biology ,Ring (chemistry) ,Biochemistry ,In vitro ,Ubiquitin ligase ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Enzyme ,Ubiquitin ,chemistry ,BARD1 ,biology.protein ,Ring domain ,Molecular Biology ,Function (biology) - Abstract
The tumor-suppressor protein BRCA1 works with BARD1 to catalyze the transfer of ubiquitin onto protein substrates. The N-terminal regions of BRCA1 and BARD1 that contain their RING domains are responsible for dimerization and ubiquitin ligase activity. This activity is a common feature among hundreds of human RING domain-containing proteins. RING domains bind and activate E2 ubiquitin-conjugating enzymes to promote ubiquitin transfer to substrates. We show that the identity of residues at specific positions in the RING domain can tune activity levels up or down. We report substitutions that create a structurally intact BRCA1/BARD1 heterodimer that is inactive in vitro with all E2 enzymes. Other substitutions in BRCA1 or BARD1 RING domains result in hyperactivity, revealing that both proteins have evolved attenuated activity. Loss of attenuation results in decreased product specificity, providing a rationale for why nature has tuned BRCA1 activity. The ability to tune BRCA1 provides powerful tools for understanding its biological functions and provides a basis to assess mechanisms for rescuing the activity of cancer-associated variations. Beyond the applicability to BRCA1, we show the identity of residues at tuning positions that can be used to predict and modulate the activity of an unrelated RING E3 ligase. These findings provide valuable insights into understanding the mechanism and function of RING E3 ligases like BRCA1.
- Published
- 2017
- Full Text
- View/download PDF
42. Author response: Indirect sexual selection drives rapid sperm protein evolution in abalone
- Author
-
Willie J. Swanson, Rachel E. Klevit, Lisa M. Tuttle, and Damien B. Wilburn
- Subjects
Abalone ,Sexual selection ,Zoology ,Biology ,Sperm protein - Published
- 2019
- Full Text
- View/download PDF
43. Indirect sexual selection drives rapid sperm protein evolution in abalone
- Author
-
Willie J. Swanson, Rachel E. Klevit, Damien B. Wilburn, and Lisa M. Tuttle
- Subjects
0301 basic medicine ,Male ,Magnetic Resonance Spectroscopy ,QH301-705.5 ,Structural Biology and Molecular Biophysics ,Science ,Gastropoda ,Lysin ,complex mixtures ,General Biochemistry, Genetics and Molecular Biology ,Exocytosis ,Evolution, Molecular ,03 medical and health sciences ,0302 clinical medicine ,Human fertilization ,Mucoproteins ,NMR spectroscopy ,Animals ,sexual selection ,Amino Acid Sequence ,Selection, Genetic ,Biology (General) ,rapid evolution ,Evolutionary Biology ,abalone ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,Molecular biophysics ,Egg Proteins ,Proteins ,General Medicine ,Sperm ,Spermatozoa ,Cell biology ,030104 developmental biology ,Structural biology ,fertilization ,Sexual selection ,bacteria ,Medicine ,Female ,Other ,030217 neurology & neurosurgery ,Intracellular ,Research Article - Abstract
Sexual selection can explain the rapid evolution of fertilization proteins, yet sperm proteins evolve rapidly even if not directly involved in fertilization. In the marine mollusk abalone, sperm secrete enormous quantities of two rapidly evolving proteins, lysin and sp18, that are stored at nearly molar concentrations. We demonstrate that this extraordinary packaging is achieved by associating into Fuzzy Interacting Transient Zwitterion (FITZ) complexes upon binding the intrinsically disordered FITZ Anionic Partner (FITZAP). FITZ complexes form at intracellular ionic strengths and, upon exocytosis into seawater, lysin and sp18 are dispersed to drive fertilization. NMR analyses revealed that lysin uses a common molecular interface to bind both FITZAP and its egg receptor VERL. As sexual selection alters the lysin-VERL interface, FITZAP coevolves rapidly to maintain lysin binding. FITZAP-lysin interactions exhibit a similar species-specificity as lysin-VERL interactions. Thus, tethered molecular arms races driven by sexual selection can generally explain rapid sperm protein evolution.
- Published
- 2019
44. Mediator subunit Med15 dictates the conserved 'fuzzy' binding mechanism of yeast transcription activators Gal4 and Gcn4
- Author
-
Steven Hahn, Linda Warfield, Rachel E. Klevit, Lisa M. Tuttle, and Derek Pacheco
- Subjects
Mediator ,Transcription (biology) ,Chemistry ,Protein subunit ,Activation function ,Biophysics ,Transcription Repression ,Repressor ,Transcription factor ,Yeast - Abstract
SUMMARYThe acidic activation domain (AD) of yeast transcription factor Gal4 plays a dual role in both transcription repression and activation through sequence-dependent binding to Gal80 repressor and sequence-independent binding to Mediator subunit Med15. The activation function of Gal4 arises from two hydrophobic regions within the 40-residue AD. We show by NMR that each AD region binds the Mediator subunit Med15 using a “fuzzy” protein interface. Remarkably, comparison of chemical shift perturbations shows that Gal4 and Gcn4, two ADs of different sequence, interact nearly identically with Med15. The findings that two ADs of different sequence use an identical fuzzy binding mechanism shows a common sequence-independent mechanism for AD-Mediator binding, similar to interactions within a hydrophobic cloud. In contrast, the same region of Gal4 AD interacts with Gal80 via a tight structured complex, implying that the structured binding partner of an intrinsically disordered protein dictates the type of protein interaction.
- Published
- 2019
- Full Text
- View/download PDF
45. Author response: Interplay of disordered and ordered regions of a human small heat shock protein yields an ensemble of ‘quasi-ordered’ states
- Author
-
Benjamin Basanta, Amanda F. Clouser, Abhinav Nath, Miklos Guttman, Rachel E. Klevit, and Hannah E.R. Baughman
- Subjects
Physics ,Condensed matter physics ,Heat shock protein - Published
- 2019
- Full Text
- View/download PDF
46. Interplay of disordered and ordered regions of a human small heat shock protein yields an ensemble of 'quasi-ordered' states
- Author
-
Hannah E.R. Baughman, Abhinav Nath, Amanda F. Clouser, Rachel E. Klevit, Benjamin Basanta, and Miklos Guttman
- Subjects
Models, Molecular ,animal structures ,QH301-705.5 ,Protein Conformation ,Science ,Structural Biology and Molecular Biophysics ,General Biochemistry, Genetics and Molecular Biology ,Mass Spectrometry ,Protein Aggregates ,03 medical and health sciences ,Heat shock protein ,Scattering, Small Angle ,None ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Biology (General) ,HDXMS ,Small Heat-Shock Proteins ,Heat-Shock Proteins ,030304 developmental biology ,Sequence (medicine) ,Physics ,HSPB1 ,0303 health sciences ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,Molecular biophysics ,030302 biochemistry & molecular biology ,General Medicine ,intrinsically disordered protein ,Hybrid approach ,small heat shock protein ,NMR ,Heat-Shock Proteins, Small ,Structural biology ,Biophysics ,Medicine ,Stress conditions ,Protein Multimerization ,Molecular Chaperones ,Research Article - Abstract
Small heat shock proteins (sHPSs) are nature’s “first responders” to cellular stress, interacting with affected proteins to prevent their aggregation. Little is known about sHSP structure beyond its structured α-crystallin domain (ACD), which is flanked by disordered regions. In the human sHSP HSPB1, the disordered N-terminal region (NTR) represents nearly 50% of the sequence. Here, we present a hybrid approach involving NMR, hydrogen-deuterium exchange mass spectrometry, and modeling to provide the first residue-level characterization of the NTR. The results support a model in which multiple grooves on the ACD interact with specific NTR regions, creating an ensemble of “quasi-ordered” NTR states that can give rise to the known heterogeneity and plasticity of HSPB1. Phosphorylation-dependent interactions inform a mechanism by which HSPB1 is activated under stress conditions. Additionally, we examine the effects of disease-associated NTR mutations on HSPB1 structure and dynamics, leveraging our emerging structural insights.
- Published
- 2019
- Full Text
- View/download PDF
47. RMSD analysis of structures of the bacterial protein FimH identifies five conformations of its lectin domain
- Author
-
Wendy E. Thomas, Pearl Magala, Evgeni V. Sokurenko, Rachel E. Klevit, and Ronald E. Stenkamp
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Allosteric regulation ,Genetic Vectors ,Mannose ,Gene Expression ,Catch bond ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Bacterial Adhesion ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Allosteric Regulation ,Structural Biology ,Lectins ,Escherichia coli ,Protein Interaction Domains and Motifs ,Cloning, Molecular ,Molecular Biology ,030304 developmental biology ,Mannan-binding lectin ,0303 health sciences ,Adhesins, Escherichia coli ,Binding Sites ,biology ,Chemistry ,030302 biochemistry & molecular biology ,computer.file_format ,Protein Data Bank ,Ligand (biochemistry) ,Recombinant Proteins ,Bacterial adhesin ,Pilin ,Fimbriae, Bacterial ,biology.protein ,Biophysics ,Protein Conformation, beta-Strand ,Fimbriae Proteins ,computer ,Protein Binding - Abstract
FimH is a bacterial adhesin protein located at the tip of Escherichia coli fimbria that functions to adhere bacteria to host cells. Thus, FimH is a critical factor in bacterial infections such as urinary tract infections and is of interest in drug development. It is also involved in vaccine development and as a model for understanding shear-enhanced catch bond cell adhesion. To date, over 60 structures have been deposited in the Protein Data Bank showing interactions between FimH and mannose ligands, potential inhibitors, and other fimbrial proteins. In addition to providing insights about ligand recognition and fimbrial assembly, these structures provide insights into conformational changes in the two domains of FimH that are critical for its function. To gain further insights into these structural changes, we have superposed FimH's mannose binding lectin domain in all these structures and categorized the structures into five groups of lectin domain conformers using RMSD as a metric. Many structures also include the pilin domain, which anchors FimH to the fimbriae and regulates the conformation and function of the lectin domain. For these structures, we have also compared the relative orientations of the two domains. These structural analyses enhance our understanding of the conformational changes associated with FimH ligand binding and domain-domain interactions, including its catch bond behavior through allosteric action of force in bacterial adhesion.
- Published
- 2019
48. Indirect Sexual Selection Drives Rapid Sperm Protein Evolution
- Author
-
Damien B. Wilburn, Willie J. Swanson, Rachel E. Klevit, and Lisa M. Tuttle
- Subjects
Protein interface ,0303 health sciences ,Chemistry ,030302 biochemistry & molecular biology ,Lysin ,Sperm protein ,complex mixtures ,Sperm ,Cell biology ,03 medical and health sciences ,Human fertilization ,Sexual selection ,bacteria ,Receptor ,Intracellular ,030304 developmental biology - Abstract
Sexual selection can explain rapid evolution of fertilization proteins, yet sperm proteins evolve rapidly even if they are not directly involved in fertilization. Here we demonstrate that FITZAP, an intrinsically disordered sperm protein in the marine mollusk abalone, exploits differences in the intracellular and oceanic ionic environments to package the fertilization protein lysin at extraordinary concentrations inside sperm by forming Fuzzy Interacting Transient Zwitterion (FITZ) complexes. FITZAP binds lysin at the same protein interface as its egg receptor VERL, and as sexual selection rapidly alters the lysin-VERL interface, FITZAP coevolves rapidly to maintain lysin binding. Consequently, FITZAP-lysin interactions exhibit a similar species-specificity as lysin-VERL interactions. Thus, tethered molecular arms races driven by sexual selection can generally explain rapid sperm protein evolution.One Sentence SummaryStructural study of sperm proteins reveals a novel protein packaging/dispersion system embedded in a coevolutionary arms race.
- Published
- 2019
- Full Text
- View/download PDF
49. Cbl interacts with multiple E2s in vitro and in cells
- Author
-
Donna Voeller, Rachel E. Klevit, Ke Ma, Mariya S. Liyasova, Jinqiu Chen, Philip E. Ryan, and Stanley Lipkowitz
- Subjects
Cultured tumor cells ,Syk ,Plasma protein binding ,environment and public health ,Biochemistry ,Receptor tyrosine kinase ,Ubiquitin ,hemic and lymphatic diseases ,Yeast Two-Hybrid Assays ,Two-Hybrid Screening ,Monoubiquitination ,Small interfering RNAs ,Proto-Oncogene Proteins c-cbl ,Phosphorylation ,Post-Translational Modification ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,030302 biochemistry & molecular biology ,Cell biology ,Precipitation Techniques ,ErbB Receptors ,Nucleic acids ,Medicine ,Cell lines ,biological phenomena, cell phenomena, and immunity ,Biological cultures ,Protein Interaction Assays ,Proto-oncogene tyrosine-protein kinase Src ,Protein Binding ,Research Article ,Science ,Immunoblotting ,Molecular Probe Techniques ,Library Screening ,Research and Analysis Methods ,03 medical and health sciences ,Two-Hybrid System Techniques ,Genetics ,Humans ,Immunoprecipitation ,Gene Silencing ,HeLa cells ,Molecular Biology Techniques ,Non-coding RNA ,Molecular Biology ,030304 developmental biology ,Molecular Biology Assays and Analysis Techniques ,fungi ,Ubiquitination ,Biology and Life Sciences ,Proteins ,Cell cultures ,Gene regulation ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,Gene Expression Regulation ,Mutation ,Ubiquitin-Conjugating Enzymes ,biology.protein ,RNA ,UBE2E Family ,Gene expression - Abstract
Many receptor tyrosine kinases (RTKs, such as EGFR, MET) are negatively regulated by ubiquitination and degradation mediated by Cbl proteins, a family of RING finger (RF) ubiquitin ligases (E3s). Loss of Cbl protein function is associated with malignant transformation driven by increased RTK activity. RF E3s, such as the Cbl proteins, interact with a ubiquitin-conjugating enzyme (E2) to confer specificity to the ubiquitination process and direct the transfer of ubiquitin from the E2 to one or more lysines on the target proteins. Using in vitro E3 assays and yeast two-hybrid screens, we found that Ube2d, Ube2e families, Ube2n/2v1, and Ube2w catalyze autoubiquitination of the Cbl protein and Ube2d2, Ube2e1, and Ube 2n/2v1 catalyze Cbl-mediated substrate ubiquitination of the EGFR and SYK. Phosphorylation of the Cbl protein by by Src resulted in increased E3 activity compared to unphosphorylated cbl or Cbl containing a phosphomimetic Y371E mutation. Ubiquitin chain formation depended on the E2 tested with Cbl with Ube2d2 forming both K48 and K63 linked chains, Ube2n/2v1 forming only K63 linked chains, and Ube2w inducing monoubiquitination. In cells, the Ube2d family, Ube2e family, and Ube2n/2v1 contributed to EGFR ubiquitination. Our data suggest that multiple E2s can interact with Cbl and modulate its E3 activity in vitro and in cells.
- Published
- 2019
50. Mechanisms of Small Heat Shock Proteins
- Author
-
Rachel E. Klevit, Hannah E.R. Baughman, Maria K. Janowska, and Christopher N. Woods
- Subjects
0301 basic medicine ,030102 biochemistry & molecular biology ,Protein Conformation ,fungi ,Oxidation reduction ,Protein aggregation ,Protein Homeostasis ,Biology ,Hydrogen-Ion Concentration ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Heat-Shock Proteins, Small ,03 medical and health sciences ,Oxidative Stress ,030104 developmental biology ,Protein structure ,PERSPECTIVES ,Metals ,Heat shock protein ,Humans ,Chaperone activity ,Phosphorylation ,Small Heat-Shock Proteins ,Oxidation-Reduction ,Function (biology) - Abstract
Small heat shock proteins (sHSPs) are ATP-independent chaperones that delay formation of harmful protein aggregates. sHSPs' role in protein homeostasis has been appreciated for decades, but their mechanisms of action remain poorly understood. This gap in understanding is largely a consequence of sHSP properties that make them recalcitrant to detailed study. Multiple stress-associated conditions including pH acidosis, oxidation, and unusual availability of metal ions, as well as reversible stress-induced phosphorylation can modulate sHSP chaperone activity. Investigations of sHSPs reveal that sHSPs can engage in transient or long-lived interactions with client proteins depending on solution conditions and sHSP or client identity. Recent advances in the field highlight both the diversity of function within the sHSP family and the exquisite sensitivity of individual sHSPs to cellular and experimental conditions. Here, we will present and highlight current understanding, recent progress, and future challenges.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.