Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Mol Cells ; 45(6): 425-434, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35680375

ABSTRACT

The post-translational modification (e.g., phosphorylation) of estrogen receptor α (ERα) plays a role in controlling the expression and subcellular localization of ERα as well as its sensitivity to hormone response. Here, we show that ERα is also modified by UFM1 and this modification (ufmylation) plays a crucial role in promoting the stability and transactivity of ERα, which in turn promotes breast cancer development. The elevation of ufmylation via the knockdown of UFSP2 (the UFM1-deconjugating enzyme in humans) dramatically increases ERα stability by inhibiting ubiquitination. In contrast, ERα stability is decreased by the prevention of ufmylation via the silencing of UBA5 (the UFM1-activating E1 enzyme). Lys171 and Lys180 of ERα were identified as the major UFM1 acceptor sites, and the replacement of both Lys residues by Arg (2KR mutation) markedly reduced ERα stability. Moreover, the 2KR mutation abrogated the 17ß-estradiol-induced transactivity of ERα and the expression of its downstream target genes, including pS2, cyclin D1, and c-Myc; this indicates that ERα ufmylation is required for its transactivation function. In addition, the 2KR mutation prevented anchorage-independent colony formation by MCF7 cells. Most notably, the expression of UFM1 and its conjugating machinery (i.e., UBA5, UFC1, UFL1, and UFBP1) were dramatically upregulated in ERα-positive breast cancer cell lines and tissues. Collectively, these findings implicate a critical role attributed to ERα ufmylation in breast cancer development by ameliorating its stability and transactivity.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , MCF-7 Cells , Proteins/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination
2.
J Struct Biol ; 213(4): 107796, 2021 12.
Article in English | MEDLINE | ID: mdl-34508858

ABSTRACT

Ubiquitin fold modifier 1 (UFM1) is an ubiquitin-like protein (Ubl) involved especially in endoplasmic stress response. Activation occurs via a three-step mechanism like other Ubls. Data obtained reveal that UFM1 regulates the oligomeric state of ubiquitin activating enzyme 5 (UBA5) to initiate the activation step. Mixtures of homodimers and heterotrimers are observed in solution at the equilibrium state, demonstrating that the UBA5-UFM1 complex undergoes several concentration dependent oligomeric translational states to form a final functional complex. The oligomerization state of unbound UBA5 is also concentration dependent and shifts from the monomeric to the dimeric state. Data describing different oligomeric states are complemented with binding studies that reveal a negative cooperativity for the complex formation and thereby provide more detailed insights into the complex formation mechanism.


Subject(s)
Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Protein Conformation , Protein Multimerization , Proteins/chemistry , Ubiquitin-Activating Enzymes/chemistry , Humans , Models, Molecular , Multiprotein Complexes/metabolism , Protein Binding , Proteins/genetics , Proteins/metabolism , Scattering, Small Angle , Spectrometry, Fluorescence , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , X-Ray Diffraction
3.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34299007

ABSTRACT

Ubiquitin fold modifier 1 (UFM1) is a member of the ubiquitin-like protein family. UFM1 undergoes a cascade of enzymatic reactions including activation by UBA5 (E1), transfer to UFC1 (E2) and selective conjugation to a number of target proteins via UFL1 (E3) enzymes. Despite the importance of ufmylation in a variety of cellular processes and its role in the pathogenicity of many human diseases, the molecular mechanisms of the ufmylation cascade remains unclear. In this study we focused on the biophysical and biochemical characterization of the interaction between UBA5 and UFC1. We explored the hypothesis that the unstructured C-terminal region of UBA5 serves as a regulatory region, controlling cellular localization of the elements of the ufmylation cascade and effective interaction between them. We found that the last 20 residues in UBA5 are pivotal for binding to UFC1 and can accelerate the transfer of UFM1 to UFC1. We solved the structure of a complex of UFC1 and a peptide spanning the last 20 residues of UBA5 by NMR spectroscopy. This structure in combination with additional NMR titration and isothermal titration calorimetry experiments revealed the mechanism of interaction and confirmed the importance of the C-terminal unstructured region in UBA5 for the ufmylation cascade.


Subject(s)
Proteins/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Calorimetry, Differential Scanning , Gene Expression , Magnetic Resonance Spectroscopy , Mutation , Peptides/chemistry , Protein Binding , Protein Domains , Proteins/genetics , Proteins/metabolism , Recombinant Proteins , Thermodynamics , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
4.
J Med Chem ; 64(11): 7533-7543, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34060310

ABSTRACT

An analysis of the rotatable bond geometry of drug-like ligand models is reported for high-resolution (<1.1 Å) crystallographic protein-ligand complexes. In cases where the ligand fit to the electron density is very good, unusual torsional geometry is rare and, most often, though not exclusively, associated with strong polar, metal, or covalent ligand-protein interactions. It is rarely associated with a torsional strain of greater than 2 kcal mol-1 by calculation. An unusual torsional geometry is more prevalent where the fit to electron density is not perfect. Multiple low-strain conformer bindings were observed in 21% of the set and, it is suggested, may also lie behind many of the 35% of single-occupancy cases, where a poor fit to the e-density was found. It is concluded that multiple conformer ligand binding is an under-recognized phenomenon in structure-based drug design and that there is a need for more robust crystallographic refinement methods to better handle such cases.


Subject(s)
Ligands , Proteins/chemistry , Crystallography, X-Ray , Density Functional Theory , Models, Theoretical , Proteins/metabolism , Substrate Specificity , Thermodynamics , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism
5.
Cell Chem Biol ; 28(10): 1394-1406.e10, 2021 10 21.
Article in English | MEDLINE | ID: mdl-33979648

ABSTRACT

Natural products (NPs) encompass a rich source of bioactive chemical entities. Here, we used human cancer stem cells (CSCs) in a chemical genomics campaign with NP chemical space to interrogate extracts from diverse strains of actinomycete for anti-cancer properties. We identified a compound (McM25044) capable of selectively inhibiting human CSC function versus normal stem cell counterparts. Biochemical and molecular studies revealed that McM025044 exerts inhibition on human CSCs through the small ubiquitin-like modifier (SUMO) cascade, found to be hyperactive in a variety of human cancers. McM025044 impedes the SUMOylation pathway via direct targeting of the SAE1/2 complex. Treatment of patient-derived CSCs resulted in reduced levels of SUMOylated proteins and suppression of progenitor and stem cell capacity measured in vitro and in vivo. Our study overcomes a barrier in chemically inhibiting oncogenic SUMOylation activity and uncovers a unique role for SAE2 in the biology of human cancers.


Subject(s)
Neoplastic Stem Cells/metabolism , Ubiquitin-Activating Enzymes/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Biological Products/chemistry , Biological Products/metabolism , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Line, Tumor , Cell Self Renewal , Cell Survival/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Docking Simulation , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Sumoylation/drug effects , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics
6.
Biochemistry ; 60(16): 1276-1285, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33848125

ABSTRACT

Ubiquitin (Ub) signaling requires the covalent passage of Ub among E1, E2, and E3 enzymes. The choice of E2 and E3 enzymes combined with multiple rounds of the cascade leads to the formation of polyubiquitin chains linked through any one of the seven lysines on Ub. The linkage type and length act as a signal to trigger important cellular processes such as protein degradation or the DNA damage response. Recently, proteomics studies have identified that Ub can be acetylated at six of its seven lysine residues under various cell stress conditions. To understand the potential differences in Ub signaling caused by acetylation, we synthesized all possible acetylated ubiquitin (acUb) variants and examined the E1-mediated formation of the corresponding E2∼acUb conjugates in vitro using kinetic methods. A Förster resonance energy transfer assay was optimized in which the Ub constructs were labeled with a CyPet fluorophore and the E2 UBE2D1 was labeled with a YPet fluorophore to monitor the formation of E2∼Ub conjugates. Our methods enable the detection of small differences that may otherwise be concealed in steady-state ubiquitination experiments. We determined that Ub, acetylated at K11, K27, K33, K48, or K63, has altered turnover numbers for E2∼Ub conjugate formation by the E1 enzyme Uba1. This work provides evidence that acetylation of Ub can alter the catalysis of ubiquitination early on in the pathway.


Subject(s)
Ubiquitin-Activating Enzymes/metabolism , Ubiquitin/metabolism , Acetylation , Catalytic Domain , Models, Molecular , Ubiquitin-Activating Enzymes/chemistry , Ubiquitination
7.
Nat Commun ; 12(1): 2370, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888705

ABSTRACT

E1 enzymes function as gatekeepers of ubiquitin (Ub) signaling by catalyzing activation and transfer of Ub to tens of cognate E2 conjugating enzymes in a process called E1-E2 transthioesterification. The molecular mechanisms of transthioesterification and the overall architecture of the E1-E2-Ub complex during catalysis are unknown. Here, we determine the structure of a covalently trapped E1-E2-ubiquitin thioester mimetic. Two distinct architectures of the complex are observed, one in which the Ub thioester (Ub(t)) contacts E1 in an open conformation and another in which Ub(t) instead contacts E2 in a drastically different, closed conformation. Altogether our structural and biochemical data suggest that these two conformational states represent snapshots of the E1-E2-Ub complex pre- and post-thioester transfer, and are consistent with a model in which catalysis is enhanced by a Ub(t)-mediated affinity switch that drives the reaction forward by promoting productive complex formation or product release depending on the conformational state.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Catalytic Domain , Crystallography, X-Ray , Esterification/physiology , Models, Molecular , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Ubiquitin/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/isolation & purification , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/isolation & purification
8.
J Cell Physiol ; 236(4): 2934-2949, 2021 04.
Article in English | MEDLINE | ID: mdl-33037615

ABSTRACT

Transient receptor potential melastatin member 8 (TRPM8), a Ca2+ -permeable nonselective cation channel activated by cold and cooling agents, mediates allodynia. Dysfunction or abnormal expression of TRPM8 has been found in several human cancers. The role of ubiquitination in the regulation of TRPM8 function remains poorly understood. Here, we identified the ubiquitin (Ub)-ligase E3, tripartite motif-containing 4 (TRIM4), as a novel interaction partner of TRPM8 and confirmed that the TRIM4-TRPM8 interaction was mediated through the SPRY domain of TRIM4. Patch-clamp assays showed that TRIM4 negatively regulates TRPM8-mediated currents in HEK293 cells. Moreover, TRIM4 reduced the expression of TRPM8 on the cell surface by promoting the K63-linked ubiquitination of TRPM8. Further analyses revealed that the TRPM8 N-terminal lysine residue at 423 was the major ubiquitination site that mediates its functional regulation by TRIM4. A Ub-activating enzyme E1, Ub-like modifier-activating enzyme 1 (UBA1), was also found to interact with TRPM8, thereby regulating its channel function and ubiquitination. In addition, knockdown of UBA1 impaired the regulation of TRPM8 ubiquitination and function by TRIM4. Thus, this study demonstrates that TRIM4 downregulates TRPM8 via K423-mediated TRPM8 ubiquitination and requires UBA1 to regulate TRPM8.


Subject(s)
Lysine/metabolism , TRPM Cation Channels/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitination , Amino Acid Sequence , Animals , HEK293 Cells , Humans , MCF-7 Cells , Protein Binding , Protein Domains , Rats , Sequence Deletion , Tripartite Motif Proteins/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism
9.
J Comput Chem ; 41(29): 2459-2468, 2020 11.
Article in English | MEDLINE | ID: mdl-32882060

ABSTRACT

The domain-based local pair natural orbital coupled-cluster with single, double, and perturbative triples excitation (DLPNO-CCSD(T)) method was employed to portray the activation and reaction energies of four ubiquitous enzymatic reactions, and its performance was confronted to CCSD(T)/complete basis set (CBS) to assess its accuracy and robustness in this specific field. The DLPNO-CCSD(T) results were also confronted to those of a set of density functionals (DFs) to understand the benefit of implementing this technique in enzymatic quantum mechanics/molecular mechanics (QM/MM) calculations as a second QM component, which is often treated with DF theory (DFT). On average, the DLPNO-CCSD(T)/aug-cc-pVTZ results were 0.51 kcal·mol-1 apart from the canonic CCSD(T)/CBS, without noticeable biases toward any of the reactions under study. All DFs fell short to the DLPNO-CCSD(T), both in terms of accuracy and robustness, which suggests that this method is advantageous to characterize enzymatic reactions and that its use in QM/MM calculations, either alone or in conjugation with DFT, in a two-region QM layer (DLPNO-CCSD(T):DFT), should enhance the quality and faithfulness of the results.


Subject(s)
Ubiquitin-Activating Enzymes/chemistry , Ubiquitins/chemistry , Amino Acids/chemistry , Catalysis , Density Functional Theory , Enzyme Activation , Models, Molecular , Molecular Conformation , Thermodynamics
10.
Cell Biochem Biophys ; 78(3): 309-319, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32583128

ABSTRACT

Ubiquitin-activating enzyme (E1) is the first enzyme of the ubiquitination pathway and is required to activate ubiquitin. E1 of Saccharomyces cerevisiae is a large multidomain monomeric protein. There are no studies available on the domains of yeast E1 as independent entities. Four domains of E1 namely, first catalytic cysteine half-domain (FCCH), four-helix bundle (4HB), second catalytic cysteine half-domain (SCCH) and ubiquitin fold domain (UFD) were characterised to understand their structural and functional independence vis-a-vis full length E1. Spectroscopic characterisation using circular dichroism and fluorescence suggested that these domains can act as independent folding units and attain native-like secondary structure. The structural features obtained with the peptides SCCH and FCCH of S. cerevisiae bear a high degree of structural similarity to the corresponding fragments of mouse from literature. Nearly 50% of the residues of the 4HB domain of the S. cerevisiae sample showed helical conformation. They displayed a high degree of conservation when compared with 4HB of mouse with respect to their identity and arrangement. The fragment UFD of yeast formed an α/ß domain as in the whole protein and exhibited 45% homology with that of mouse, showing a similar arrangement of α and ß elements in its secondary structure. Overexpression of the domains in vivo indicated that the SCCH domain and to some extent UFD apparently interfere with cellular functions such as survival under various stresses.


Subject(s)
Saccharomyces cerevisiae/enzymology , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin/chemistry , Animals , Cell Proliferation , Circular Dichroism , Escherichia coli/metabolism , Mice , Microbial Sensitivity Tests , Protein Domains , Protein Structure, Secondary , Spectrometry, Fluorescence
11.
Biomolecules ; 10(5)2020 05 21.
Article in English | MEDLINE | ID: mdl-32455657

ABSTRACT

The 26S proteasome is a large (~2.5 MDa) protein complex consisting of at least 33 different subunits and many other components, which form the ubiquitin proteasomal system (UPS), an ATP-dependent protein degradation system in the cell. UPS serves as an essential component of the cellular protein surveillance machinery, and its dysfunction leads to cancer, neurodegenerative and immunological disorders. Importantly, the functions and regulations of proteins are governed by the combination of ordered regions, intrinsically disordered protein regions (IDPRs) and molecular recognition features (MoRFs). The structure-function relationships of UPS components have not been identified completely; therefore, in this study, we have carried out the functional intrinsic disorder and MoRF analysis for potential neurodegenerative disease and anti-cancer targets of this pathway. Our report represents the presence of significant intrinsic disorder and disorder-based binding regions in several UPS proteins, such as extraproteasomal polyubiquitin receptors (UBQLN1 and UBQLN2), proteasome-associated polyubiquitin receptors (ADRM1 and PSMD4), deubiquitinating enzymes (DUBs) (ATXN3 and USP14), and ubiquitinating enzymes (E2 (UBE2R2) and E3 (STUB1) enzyme). We believe this study will have implications for the conformation-specific roles of different regions of these proteins. This will lead to a better understanding of the molecular basis of UPS-associated diseases.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Proteasome Endopeptidase Complex/chemistry , Ubiquitination , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Ataxin-3/chemistry , Ataxin-3/metabolism , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Intrinsically Disordered Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Folding , Protein Interaction Maps , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
12.
Int J Biochem Cell Biol ; 123: 105746, 2020 06.
Article in English | MEDLINE | ID: mdl-32315770

ABSTRACT

Ubiquitin-activating enzyme E1, UBA1, functions at the apex of the enzymatic ubiquitylation cascade, catalysing ubiquitin activation. UBA1 is thus of fundamental importance to the modulation of ubiquitin homeostasis and to all downstream ubiquitylation-dependent cellular processes, including proteolysis through the ubiquitin-proteasome system and selective autophagy. The proteasome-dependent and -independent functions of UBA1 contribute significantly to a range of processes crucial to neuronal health. The significance of UBA1 activity to neuronal health is clear in light of accumulating evidence implicating impaired UBA1 activity in a range of neurodegenerative conditions, including Parkinson's disease, Alzheimer's disease, Huntington's disease and spinal muscular atrophy. Moreover, ubiquitylation-independent functions of UBA1 of importance to neuronal functioning have been proposed. Here, we summarise findings supporting the significant role of UBA1 in regulating neuronal functioning, and discuss the detrimental consequences of UBA1 impairment that contribute to neuronal dysfunction and degeneration.


Subject(s)
Neurodegenerative Diseases/enzymology , Neurons/enzymology , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination , Animals , Autophagy/genetics , Humans , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/therapy , Neurons/metabolism , Signal Transduction/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology , Ubiquitination/genetics
13.
Angew Chem Int Ed Engl ; 59(32): 13496-13501, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32346954

ABSTRACT

Triazole-based deubiquitylase (DUB)-resistant ubiquitin (Ub) probes have recently emerged as effective tools for the discovery of Ub chain-specific interactors in proteomic studies, but their structural diversity is limited. A new family of DUB-resistant Ub probes is reported based on isopeptide-N-ethylated dimeric or polymeric Ub chains, which can be efficiently prepared by a one-pot, ubiquitin-activating enzyme (E1)-catalyzed condensation reaction of recombinant Ub precursors to give various homotypic and even branched Ub probes at multi-milligram scale. Proteomic studies using label-free quantitative (LFQ) MS indicated that the isopeptide-N-ethylated Ub probes may complement the triazole-based probes in the study of Ub interactome. Our study highlights the utility of modern protein synthetic chemistry to develop structurally and new families of tool molecules needed for proteomic studies.


Subject(s)
Molecular Probes/chemistry , Polyubiquitin/chemistry , Ubiquitin-Activating Enzymes/chemistry , Cyclin B1/chemistry , Cyclin B1/genetics , HEK293 Cells , HeLa Cells , Histones/chemistry , Histones/genetics , Humans , Molecular Probes/chemical synthesis , Mutation , Polyubiquitin/chemical synthesis , Proteomics
14.
Chemistry ; 26(28): 6279-6284, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32154932

ABSTRACT

Simple and robust assays to monitor enzymatic ATP cleavage with high efficiency in real-time are scarce. To address this shortcoming, we developed fluorescently labelled adenosine tri-, tetra- and pentaphosphate analogues of ATP. The novel ATP analogues bear - in contrast to earlier reports - only a single acridone-based dye at the terminal phosphate group. The dye's fluorescence is quenched by the adenine component of the ATP analogue and is restored upon cleavage of the phosphate chain and dissociation of the dye from the adenosine moiety. Thereby the activity of ATP-cleaving enzymes can be followed in real-time. We demonstrate this proficiency for ubiquitin activation by the ubiquitin-activating enzymes UBA1 and UBA6 which represents the first step in an enzymatic cascade leading to the covalent attachment of ubiquitin to substrate proteins, a process that is highly conserved from yeast to humans. We found that the efficiency to serve as cofactor for UBA1/UBA6 very much depends on the length of the phosphate chain of the ATP analogue: triphosphates are used poorly while pentaphosphates are most efficiently processed. Notably, the novel pentaphosphate-harbouring ATP analogue supersedes the efficiency of recently reported dual-dye labelled analogues and thus, is a promising candidate for broad applications.


Subject(s)
Adenosine Triphosphate/chemistry , Fluorescent Dyes/chemistry , Ubiquitin-Activating Enzymes/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Fluorescent Dyes/metabolism , Humans , Ubiquitin-Activating Enzymes/chemistry
15.
Biochemistry ; 59(7): 851-861, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31951392

ABSTRACT

The ubiquitin (Ub) system regulates a wide range of cellular signaling pathways. Several hundred E1, E2, and E3 enzymes are together responsible for protein ubiquitination, thereby controlling cellular activities. Due to the numerous enzymes and processes involved, studies of ubiquitination activities have been challenging. We here report a novel Förster resonance energy transfer (FRET)-based assay for studying the in vitro kinetics of ubiquitination. FRET is established upon binding of fluorophore-labeled Ub to eGFP-tagged ZnUBP, a domain that exclusively binds unconjugated Ub. We name this assay the free Ub sensor system (FUSS). Using Uba1, UbcH5, and CHIP as model E1, E2, and E3 enzymes, respectively, we demonstrate that ubiquitination results in decreasing FRET efficiency, from which reaction rates can be determined. Further treatment with USP21, a deubiquitinase, leads to increased FRET efficiency, confirming the reversibility of the assay. We subsequently use this assay to show that increasing the concentration of CHIP or UbcH5 but not Uba1 enhances ubiquitination rates and develop a novel machine learning approach to model ubiquitination. The overall ubiquitination activity is also increased upon incubation with tau, a substrate of CHIP. Our data together demonstrate the versatile applications of a novel ubiquitination assay that does not require labeling of E1, E2, E3, or substrates and is thus likely compatible with any E1-E2-E3 combinations.


Subject(s)
Endopeptidases/chemistry , Enzyme Assays/methods , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitins/chemistry , Animals , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , Humans , Kinetics , Mice , Peptide Fragments/chemistry , Protein Domains , Ubiquitin Thiolesterase/chemistry , Ubiquitination , tau Proteins/chemistry
16.
Autophagy ; 16(2): 256-270, 2020 02.
Article in English | MEDLINE | ID: mdl-30990354

ABSTRACT

Short linear motifs, known as LC3-interacting regions (LIRs), interact with mactoautophagy/autophagy modifiers (Atg8/LC3/GABARAP proteins) via a conserved universal mechanism. Typically, this includes the occupancy of 2 hydrophobic pockets on the surface of Atg8-family proteins by 2 specific aromatic and hydrophobic residues within the LIR motifs. Here, we describe an alternative mechanism of Atg8-family protein interaction with the non-canonical UBA5 LIR, an E1-like enzyme of the ufmylation pathway that preferentially interacts with GABARAP but not LC3 proteins. By solving the structures of both GABARAP and GABARAPL2 in complex with the UBA5 LIR, we show that in addition to the binding to the 2 canonical hydrophobic pockets (HP1 and HP2), a conserved tryptophan residue N-terminal of the LIR core sequence binds into a novel hydrophobic pocket on the surface of GABARAP proteins, which we term HP0. This mode of action is unique for UBA5 and accompanied by large rearrangements of key residues including the side chains of the gate-keeping K46 and the adjacent K/R47 in GABARAP proteins. Swapping mutations in LC3B and GABARAPL2 revealed that K/R47 is the key residue in the specific binding of GABARAP proteins to UBA5, with synergetic contributions of the composition and dynamics of the loop L3. Finally, we elucidate the physiological relevance of the interaction and show that GABARAP proteins regulate the localization and function of UBA5 on the endoplasmic reticulum membrane in a lipidation-independent manner.Abbreviations: ATG: AuTophaGy-related; EGFP: enhanced green fluorescent protein; GABARAP: GABA-type A receptor-associated protein; ITC: isothermal titration calorimetry; KO: knockout; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; NMR: nuclear magnetic resonance; RMSD: root-mean-square deviation of atomic positions; TKO: triple knockout; UBA5: ubiquitin like modifier activating enzyme 5.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 8 Family/metabolism , Intracellular Membranes/metabolism , Microtubule-Associated Proteins/metabolism , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoptosis Regulatory Proteins/chemistry , Autophagy-Related Protein 8 Family/chemistry , Autophagy-Related Protein 8 Family/genetics , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Lysine/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , Mutation/genetics , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Structure-Activity Relationship
17.
Sci Rep ; 9(1): 16195, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31700050

ABSTRACT

Novel strategies to target Trypanosoma brucei, Trypanosoma cruzi and Leishmania are urgently needed to generate better and safer drugs against Human African Trypanosomiasis, Chagas disease and Leishmaniasis, respectively. Here, we investigated the feasibility of selectively targeting in trypanosomatids the ubiquitin E1 activating enzyme (UBA1), an essential eukaryotic protein required for protein ubiquitination. Trypanosomatids contain two UBA1 genes in contrast to mammals and yeast that only have one, and using T. brucei as a model system, we show that both are active in vitro. Surprisingly, neither protein is inhibited by TAK-243, a potent inhibitor of human UBA1. This resistance stems from differences with the human protein at key amino acids, which includes a residue termed the gatekeeper because its mutation in E1s leads to resistance to TAK-243 and related compounds. Importantly, our results predict that trypanosomatid selective UBA1 inhibition is feasible and suggest ways to design novel compounds to achieve this.


Subject(s)
Enzyme Inhibitors/chemistry , Protozoan Proteins , Trypanosoma brucei brucei/enzymology , Ubiquitin-Activating Enzymes , Ubiquitin/chemistry , Humans , Mutation , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Ubiquitin/genetics , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics
18.
Biomolecules ; 9(10)2019 10 08.
Article in English | MEDLINE | ID: mdl-31597257

ABSTRACT

Bio-guided fractionation performed on the leaves-derived ethanol extract of Esenbeckia alata (Rutaceae), a plant used in traditional medicine, led to the isolation of two alkaloids, kokusaginine 1 and flindersiamine 2, as main cytotoxic agents. Primary ethanolic extract and raw fractions exhibited cell inhibition against five cancer cell lines at different levels (25-97% inhibition at 50 µg/mL) as well as isolated alkaloids 1-2 (30-90% inhibition at 20 µM). Although alkaloid 2 generally was the most active compound, both alkaloids showed a selective effect on K562, a human chronic myelogenous leukemia cell line. The E1-like ubiquitin-activating enzymes (e.g., UBA5) have been recently described as important targets for future treatment of cancer progression, such as leukemia, among others. Therefore, as a rationale to the observed cytotoxic selectivity, an in-silico evaluation by molecular docking and molecular dynamics was also explored. Compounds 1-2 exhibited good performance on the interaction within the active site of UBA5.


Subject(s)
Alkaloids/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Quinolines/chemistry , Rutaceae/chemistry , Ubiquitin-Activating Enzymes/metabolism , Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemical Fractionation , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Plant Extracts/chemistry , Plant Leaves/chemistry , Quinolines/pharmacology , Ubiquitin-Activating Enzymes/chemistry
19.
Sci Rep ; 9(1): 12801, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31488855

ABSTRACT

We developed and implemented a reconstituted system to screen for modulators of the ubiquitination of proliferating cell nuclear antigen, a process that activates pathways of DNA damage tolerance and drug resistance. We identified the primary putatively health-beneficial green tea polyphenol epigallocatechin gallate (EGCG) and certain related small molecules as potent inhibitors of ubiquitination. EGCG directly and reversibly targets the ubiquitin-activating enzyme Uba1, blocking formation of the Uba1~ubiquitin thioester conjugate and thus ubiquitination and in the cell. Structure-activity relationship profiles across multiple biochemical and cellular assays for a battery of EGCG analogues revealed distinct chemical and mechanism-of-action clusters of molecules, with catechin gallates, alkyl gallates, and myricetin potently inhibiting ubiquitination. This study defines a number of related though distinct first-in-class inhibitors of ubiquitination, each series with its own unique activity pattern and mechanistic signature.


Subject(s)
Catechin/analogs & derivatives , Tea/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitination , Catechin/chemistry , Catechin/pharmacology , Flavonoids/chemistry , Flavonoids/pharmacology , HEK293 Cells , Humans , Proliferating Cell Nuclear Antigen/chemistry , Structure-Activity Relationship , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitination/drug effects
20.
Nat Commun ; 10(1): 3296, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31341161

ABSTRACT

Ubiquitin (Ub) signaling requires the sequential interactions and activities of three enzymes, E1, E2, and E3. Cdc34 is an E2 that plays a key role in regulating cell cycle progression and requires unique structural elements to function. The molecular basis by which Cdc34 engages its E1 and the structural mechanisms by which its unique C-terminal extension functions in Cdc34 activity are unknown. Here, we present crystal structures of Cdc34 alone and in complex with E1, and a Cdc34~Ub thioester mimetic that represents the product of Uba1-Cdc34 Ub transthiolation. These structures reveal conformational changes in Uba1 and Cdc34 and a unique binding mode that are required for transthiolation. The Cdc34~Ub structure reveals contacts between the Cdc34 C-terminal extension and Ub that stabilize Cdc34~Ub in a closed conformation and are critical for Ub discharge. Altogether, our structural, biochemical, and cell-based studies provide insights into the molecular mechanisms by which Cdc34 function in cells.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Cloning, Molecular , Crystallography, X-Ray , Humans , Protein Domains , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/physiology , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...