3,408 results on '"Cell Cycle Proteins chemistry"'
Search Results
102. In silico identification of small molecule modulators for disruption of Hsp90-Cdc37 protein-protein interaction interface for cancer therapeutic application.
- Author
-
Dike PP, Bhowmick S, Eldesoky GE, Wabaidur SM, Patil PC, and Islam MA
- Subjects
- Cell Cycle Proteins chemistry, HSP90 Heat-Shock Proteins chemistry, Humans, Molecular Docking Simulation, Protein Binding, Protein Interaction Mapping, Triazines, Cell Cycle Proteins antagonists & inhibitors, Chaperonins antagonists & inhibitors, Chaperonins chemistry, HSP90 Heat-Shock Proteins antagonists & inhibitors, Neoplasms drug therapy
- Abstract
The protein-protein interactions (PPIs) in the biological systems are important to maintain a number of cellular processes. Several disorders including cancer may be developed due to dysfunction in the assembly of PPI networks. Hence, targeting intracellular PPIs can be considered as a crucial drug target for cancer therapy. Among the enormous and diverse group of cancer-enabling PPIs, the Hsp90-Cdc37 is prominent for cancer therapeutic development. The successful inhibition of Hsp90-Cdc37 PPI interface can be an important therapeutic option for cancer management. In the current study, a set of more than sixty thousand compounds belong to four databases were screened through a multi-steps molecular docking study in Glide against the Hsp90-Cdc37 interaction interface. The Glide-score and Prime-MM-GBSA based binding free energy of DCZ3112, standard Hsp90-Cdc37 inhibitor were found to be -6.96 and -40.46 kcal/mol, respectively. The above two parameters were used as cut-off score to reduce the chemical space from all successfully docked molecules. Furthermore, the in-silico pharmacokinetics parameters, common-feature pharmacophore analyses and the molecular binding interactions were used to wipe out the inactive molecules. Finally, four molecules were found to be important to modulate the Hsp90-Cdc37 interface. The potentiality of the final four molecules was checked through several drug-likeness characteristics. The molecular dynamics (MD) simulation study explained that all four molecules retained inside the interface of Hsp90-Cdc37. The binding free energy of each molecule obtained from the MD simulation trajectory was clearly explained the strong affection towards the Hsp90-Cdc37. Hence, the proposed molecule might be crucial for successful inhibition of the Hsp90-Cdc37 interface.Communicated by Ramaswamy H. Sarma.
- Published
- 2022
- Full Text
- View/download PDF
103. The binding mechanism of NHWD-870 to bromodomain-containing protein 4 based on molecular dynamics simulations and free energy calculation.
- Author
-
Shi M, He J, Weng T, Shi N, Qi W, Guo Y, Chen T, Chen L, and Xu D
- Subjects
- Cell Cycle Proteins chemistry, Molecular Docking Simulation, Nuclear Proteins chemistry, Protein Binding, Protein Domains, Molecular Dynamics Simulation, Transcription Factors chemistry
- Abstract
Bromodomain and extra-terminal (BET) proteins (BRD2, BRD3, BRD4, and BRDT) are epigenetic readers with tandem bromodomains. Small-molecule inhibitors of BET proteins are a promising treatment strategy against cancer. For example, NHWD-870 can inhibit BRD4 (BD1 + BD2). Presently, structural data on NHWD-870 bound BRD4 remain lacking. Herein, we investigate the interactions between NHWD-870 and BRD4 (BD1 and BD2) via molecular docking, molecular dynamics simulation, and binding free energy calculations. NHWD-870 showed a similar binding affinity for BD1 and BD2 of BRD4. Binding free energy calculations for the R / S conformations of NHWD-870 suggest that the chiral centre of NHWD-870 may confer similar roles upon the R and S conformations for binding with BRD4, facilitating the identification of novel BRD4 inhibitors.
- Published
- 2022
- Full Text
- View/download PDF
104. Whole exome sequencing identifies the potential role of genes involved in p53 pathway in Nasopharyngeal Carcinoma from Northeast India.
- Author
-
Laskar S, Das R, Kundu S, Saha A, Nandi N, Choudhury Y, and Kumar Ghosh S
- Subjects
- Adult, Amino Acid Substitution, Case-Control Studies, Cell Cycle Proteins chemistry, Female, Genetic Predisposition to Disease, High-Throughput Nucleotide Sequencing, Humans, India, Male, Middle Aged, Models, Molecular, Oxidoreductases chemistry, Polymorphism, Single Nucleotide, Protein Conformation, Protein Domains, Cell Cycle Proteins genetics, Jagged-1 Protein genetics, Nasopharyngeal Carcinoma genetics, Nasopharyngeal Neoplasms genetics, Nuclear Proteins genetics, Oxidoreductases genetics, Receptor, Transforming Growth Factor-beta Type I genetics, Exome Sequencing methods
- Abstract
Nasopharyngeal Carcinoma (NPC) found to be dependent on geographical and racial variation and is more prevalent in Northeast (NE) India. WES-based study was conducted in three states (tribes); Nagaland (Naga), Mizoram (Mizo) and Manipur (Manipuri), which provided an overview of germline variants involved inthemajor signaling pathways. Validation and recurrence assessment of WES data confirmed the risk effect of STEAP3_rs138941861 and JAG1_rs2273059, and the protective role of PARP4_rs17080653 and TGFBR1_rs11568778 variants, where STEAP3_rs138941861conferring Arg290His substitution was the only exonic non-synonymous variant and to be located in proximity to the linking region between the transmembrane and oxidoreductasedomainsof STEAP3 protein, andaffectedits structural and functional dynamics by altering the Electrostatic Potential around this connecting region. Moreover, these significantly associated variants having deleterious effect were observed to have interactions in p53 signaling pathway which emphasizes the importance of this pathway in the causation of NPC., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
105. Novel Macrocyclic Peptidomimetics Targeting the Polo-Box Domain of Polo-Like Kinase 1.
- Author
-
Ryu S, Park JE, Ham YJ, Lim DC, Kwiatkowski NP, Kim DH, Bhunia D, Kim ND, Yaffe MB, Son W, Kim N, Choi TI, Swain P, Kim CH, Lee JY, Gray NS, Lee KS, and Sim T
- Subjects
- Animals, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, HEK293 Cells, HeLa Cells, Humans, Molecular Docking Simulation, Molecular Structure, Peptides, Cyclic chemical synthesis, Peptides, Cyclic metabolism, Peptidomimetics chemical synthesis, Peptidomimetics metabolism, Protein Binding, Protein Domains, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors metabolism, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins chemistry, Proto-Oncogene Proteins metabolism, Structure-Activity Relationship, Zebrafish, Polo-Like Kinase 1, Cell Cycle Proteins antagonists & inhibitors, Peptides, Cyclic pharmacology, Peptidomimetics pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors
- Abstract
The polo-box domain (PBD) of Plk1 is a promising target for cancer therapeutics. We designed and synthesized novel phosphorylated macrocyclic peptidomimetics targeting PBD based on acyclic phosphopeptide PMQSpTPL. The inhibitory activities of 16e on Plk1-PBD is >30-fold higher than those of PMQSpTPL. Both 16a and 16e possess excellent selectivity for Plk1-PBD over Plk2/3-PBD. Analysis of the cocrystal structure of Plk1-PBD in complex with 16a reveals that the 3-(trifluoromethyl)benzoyl group in 16a interacts with Arg516 through a π-stacking interaction. This π-stacking interaction, which has not been reported previously, provides insight into the design of novel and potent Plk1-PBD inhibitors. Furthermore, 16h , a PEGlyated macrocyclic phosphopeptide derivative, induces Plk1 delocalization and mitotic failure in HeLa cells. Also, the number of phospho-H3-positive cells in a zebrafish embryo increases in proportion to the amount of 16a . Collectively, the novel macrocyclic peptidomimetics should serve as valuable templates for the design of potent and novel Plk1-PBD inhibitors.
- Published
- 2022
- Full Text
- View/download PDF
106. A Structure-based Design Approach for Generating High Affinity BRD4 D1-Selective Chemical Probes.
- Author
-
Cui H, Divakaran A, Hoell ZJ, Ellingson MO, Scholtz CR, Zahid H, Johnson JA, Griffith EC, Gee CT, Lee AL, Khanal S, Shi K, Aihara H, Shah VH, Lee RE, Harki DA, and Pomerantz WCK
- Subjects
- Binding Sites, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Tumor, Drug Design, Humans, Imidazoles chemistry, Imidazoles metabolism, Molecular Structure, Protein Binding, Protein Domains, Proto-Oncogene Proteins c-myc metabolism, Structure-Activity Relationship, Transcription Factors chemistry, Transcription Factors metabolism, Cell Cycle Proteins antagonists & inhibitors, Imidazoles pharmacology, Transcription Factors antagonists & inhibitors
- Abstract
Chemical probes for epigenetic proteins are essential tools for dissecting the molecular mechanisms for gene regulation and therapeutic development. The bromodomain and extra-terminal (BET) proteins are master transcriptional regulators. Despite promising therapeutic targets, selective small molecule inhibitors for a single bromodomain remain an unmet goal due to their high sequence similarity. Here, we address this challenge via a structure-activity relationship study using 1,4,5-trisubstituted imidazoles against the BRD4 N-terminal bromodomain (D1). Leading compounds 26 and 30 have 15 and 18 nM affinity against BRD4 D1 and over 500-fold selectivity against BRD2 D1 and BRD4 D2 via ITC. Broader BET selectivity was confirmed by fluorescence anisotropy, thermal shift, and CETSA. Despite BRD4 engagement, BRD4 D1 inhibition was unable to reduce c-Myc expression at low concentration in multiple myeloma cells. Conversely, for inflammation, IL-8 and chemokine downregulation were observed. These results provide new design rules for selective inhibitors of an individual BET bromodomain.
- Published
- 2022
- Full Text
- View/download PDF
107. SMC complexes: Lifting the lid on loop extrusion.
- Author
-
Higashi TL and Uhlmann F
- Subjects
- Chromosomes, DNA chemistry, Cell Cycle Proteins chemistry, Chromatin
- Abstract
Loop extrusion has emerged as a prominent hypothesis for how SMC complexes shape chromosomes - single molecule in vitro observations have yielded fascinating images of this process. When not extruding loops, SMC complexes are known to topologically entrap one or more DNAs. Here, we review how structural insight into the SMC complex cohesin has led to a molecular framework for both activities: a Brownian ratchet motion, associated with topological DNA entry, might repeat itself to elicit loop extrusion. After contrasting alternative loop extrusion models, we explore whether topological loading or loop extrusion is more adept at explaining in vivo SMC complex function. SMC variants that experimentally separate topological loading from loop extrusion will in the future probe their respective contributions to chromosome biology., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
108. Recognition of Dimethylarginine Analogues by Tandem Tudor Domain Protein Spindlin1.
- Author
-
Porzberg MRB, Moesgaard L, Johansson C, Oppermann U, Kongsted J, and Mecinović J
- Subjects
- Arginine chemistry, Arginine metabolism, Binding Sites, Cell Cycle Proteins chemistry, Histones metabolism, Humans, Microtubule-Associated Proteins chemistry, Molecular Dynamics Simulation, Phosphoproteins chemistry, Protein Binding, Protein Conformation, Thermodynamics, Arginine analogs & derivatives, Cell Cycle Proteins metabolism, Microtubule-Associated Proteins metabolism, Phosphoproteins metabolism, Tudor Domain
- Abstract
Epigenetic readout of the combinatorial posttranslational modification comprised of trimethyllysine and asymmetric dimethylarginine (H3K4me3R8me2a) takes place via biomolecular recognition of tandem Tudor-domain-containing protein Spindlin1. Through comparative thermodynamic data and molecular dynamics simulations, we sought to explore the binding scope of asymmetric dimethylarginine mimics by Spindlin1. Herein, we provide evidence that the biomolecular recognition of H3K4me2R8me2a is not significantly affected when R8me2a is replaced by dimethylarginine analogues, implying that the binding of K4me3 provides the major binding contribution. High-energy water molecules inside both aromatic cages of the ligand binding sites contribute to the reader-histone association upon displacement by histone peptide, with the K4me3 hydration site being lower in free energy due to a flip of Trp151.
- Published
- 2022
- Full Text
- View/download PDF
109. Crystal Structure of the Core Module of the Yeast Paf1 Complex.
- Author
-
Chen F, Liu B, Zeng J, Guo L, Ge X, Feng W, Li DF, Zhou H, and Long J
- Subjects
- Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Histones metabolism, Humans, Models, Molecular, Nuclear Proteins genetics, Protein Conformation, RNA Polymerase II metabolism, RNA-Binding Proteins metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Transcription Factors chemistry, Transcription Factors genetics, Transcription Factors metabolism, Transcriptional Elongation Factors chemistry, Transcriptional Elongation Factors metabolism, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins metabolism
- Abstract
The highly conserved multifunctional polymerase-associated factor 1 (Paf1) complex (PAF1C), which consists of five core subunits: Ctr9, Paf1, Leo1, Cdc73, and Rtf1, acts as a diverse hub that regulates all stages of RNA polymerase II-mediated transcription and various other cellular functions. However, the underlying mechanisms remain unclear. Here, we report the crystal structure of the core module derived from a quaternary Ctr9/Paf1/Cdc73/Rtf1 complex of S. cerevisiae PAF1C, which reveals interfaces between the tetratricopeptide repeat module in Ctr9 and Cdc73 or Rtf1, and find that the Ctr9/Paf1 subcomplex is the key scaffold for PAF1C assembly. Our study demonstrates that Cdc73 binds Ctr9/Paf1 subcomplex with a very similar conformation within thermophilic fungi or human PAF1C, and that the binding of Cdc73 to PAF1C is important for yeast growth. Importantly, our structure reveals for the first time that the extreme C-terminus of Rtf1 adopts an "L"-shaped structure, which interacts with Ctr9 specifically. In addition, disruption of the binding of either Cdc73 or Rtf1 to PAF1C greatly affects the normal level of histone H2B K123 monoubiquitination in vivo. Collectively, our results provide a structural insight into the architecture of the quaternary Ctr9/Paf1/Cdc73/Rtf1 complex and PAF1C functional regulation., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
110. Genetic screen for suppression of transcriptional interference reveals fission yeast 14-3-3 protein Rad24 as an antagonist of precocious Pol2 transcription termination.
- Author
-
Garg A, Shuman S, and Schwer B
- Subjects
- Acid Phosphatase genetics, Acid Phosphatase metabolism, Amino Acid Sequence, Cell Cycle Proteins chemistry, Chromosome Mapping, Gene Expression Profiling, Intracellular Signaling Peptides and Proteins chemistry, Models, Molecular, Mutagenesis, Mutation, Protein Subunits genetics, Protein Subunits metabolism, RNA Interference, RNA, Long Noncoding genetics, Schizosaccharomyces pombe Proteins chemistry, Schizosaccharomyces pombe Proteins genetics, Sequence Deletion, Structure-Activity Relationship, Synthetic Lethal Mutations, Transcription Termination, Genetic, Whole Genome Sequencing, 14-3-3 Proteins metabolism, Cell Cycle Proteins metabolism, Gene Expression Regulation, Fungal, Intracellular Signaling Peptides and Proteins metabolism, RNA Polymerase II metabolism, Schizosaccharomyces genetics, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins metabolism, Transcription, Genetic
- Abstract
Expression of fission yeast Pho1 acid phosphatase is repressed under phosphate-replete conditions by transcription of an upstream prt lncRNA that interferes with the pho1 mRNA promoter. lncRNA control of pho1 mRNA synthesis is influenced by inositol pyrophosphate (IPP) kinase Asp1, deletion of which results in pho1 hyper-repression. A forward genetic screen for ADS (Asp1 Deletion Suppressor) mutations identified the 14-3-3 protein Rad24 as a governor of phosphate homeostasis. Production of full-length interfering prt lncRNA was squelched in rad24Δ cells, concomitant with increased production of pho1 mRNA and increased Pho1 activity, while shorter precociously terminated non-interfering prt transcripts persisted. Epistasis analysis showed that pho1 de-repression by rad24Δ depends on: (i) 3'-processing and transcription termination factors CPF, Pin1, and Rhn1; and (ii) Threonine-4 of the Pol2 CTD. Combining rad24Δ with the IPP pyrophosphatase-dead asp1-H397A allele caused a severe synthetic growth defect that was ameliorated by loss-of-function mutations in CPF, Pin1, and Rhn1, and by CTD phospho-site mutations T4A and Y1F. Rad24 function in repressing pho1 was effaced by mutation of its phosphate-binding pocket. Our findings instate a new role for a 14-3-3 protein as an antagonist of precocious RNA 3'-processing/termination., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2022
- Full Text
- View/download PDF
111. Protein Dimerization via Tyr Residues: Highlight of a Slow Process with Co-Existence of Numerous Intermediates and Final Products.
- Author
-
Gatin A, Duchambon P, Rest GV, Billault I, and Sicard-Roselli C
- Subjects
- Humans, Oxidation-Reduction, Calcium-Binding Proteins chemistry, Calmodulin chemistry, Cell Cycle Proteins chemistry, Peptide Fragments chemistry, Protein Multimerization, Tyrosine chemistry
- Abstract
Protein dimerization via tyrosine residues is a crucial process in response to an oxidative attack, which has been identified in many ageing-related pathologies. Recently, it has been found that for isolated tyrosine amino acid, dimerization occurs through three types of tyrosine-tyrosine crosslinks and leads to at least four final products. Herein, considering two protected tyrosine residues, tyrosine-containing peptides and finally proteins, we investigate the dimerization behavior of tyrosine when embedded in a peptidic sequence. After azide radical oxidation and by combining UPLC-MS and H/D exchange analyzes, we were able to evidence: (i) the slow kinetics of Michael Addition Dimers (MAD) formation, i.e., more than 48 h; (ii) the co-existence of intermediates and final cyclized dimer products; and (iii) the probable involvement of amide functions to achieve Michael additions even in proteins. This raises the question of the possible in vivo existence of both intermediates and final entities as well as their toxicity and the potential consequences on protein structure and/or function.
- Published
- 2022
- Full Text
- View/download PDF
112. Structural mechanism for the selective phosphorylation of DNA-loaded MCM double hexamers by the Dbf4-dependent kinase.
- Author
-
Greiwe JF, Miller TCR, Locke J, Martino F, Howell S, Schreiber A, Nans A, Diffley JFX, and Costa A
- Subjects
- Amino Acid Sequence, Cell Cycle Proteins chemistry, Checkpoint Kinase 2 metabolism, Minichromosome Maintenance Complex Component 4 chemistry, Minichromosome Maintenance Complex Component 4 metabolism, Molecular Docking Simulation, Nucleotides metabolism, Phosphorylation, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Saccharomyces cerevisiae Proteins chemistry, Substrate Specificity, Cell Cycle Proteins metabolism, DNA, Fungal metabolism, Protein Multimerization, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Loading of the eukaryotic replicative helicase onto replication origins involves two MCM hexamers forming a double hexamer (DH) around duplex DNA. During S phase, helicase activation requires MCM phosphorylation by Dbf4-dependent kinase (DDK), comprising Cdc7 and Dbf4. DDK selectively phosphorylates loaded DHs, but how such fidelity is achieved is unknown. Here, we determine the cryogenic electron microscopy structure of Saccharomyces cerevisiae DDK in the act of phosphorylating a DH. DDK docks onto one MCM ring and phosphorylates the opposed ring. Truncation of the Dbf4 docking domain abrogates DH phosphorylation, yet Cdc7 kinase activity is unaffected. Late origin firing is blocked in response to DNA damage via Dbf4 phosphorylation by the Rad53 checkpoint kinase. DDK phosphorylation by Rad53 impairs DH phosphorylation by blockage of DDK binding to DHs, and also interferes with the Cdc7 active site. Our results explain the structural basis and regulation of the selective phosphorylation of DNA-loaded MCM DHs, which supports bidirectional replication., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
113. A paradigm shift in structural biology.
- Author
-
Subramaniam S and Kleywegt GJ
- Subjects
- Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cryoelectron Microscopy, Humans, Ligands, Protein Conformation, Protein Interaction Maps, Transcription Factors chemistry, Transcription Factors metabolism, Algorithms, Computational Biology methods, Databases, Protein statistics & numerical data, Models, Molecular, Proteins chemistry
- Published
- 2022
- Full Text
- View/download PDF
114. Dimerization underlies the aggregation propensity of intrinsically disordered coiled-coil domain-containing 124.
- Author
-
Tuzlakoğlu Öztürk M and Güllülü Ö
- Subjects
- HEK293 Cells, Humans, Intrinsically Disordered Proteins chemistry, Intrinsically Disordered Proteins metabolism, Molecular Docking Simulation, Protein Binding, Protein Structure, Tertiary, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins metabolism, Protein Multimerization physiology
- Abstract
Coiled-coil domain-containing 124 (CCDC124) is a recently discovered ribosome-binding protein conserved in eukaryotes. CCDC124 has regulatory functions on the mediation of reversible ribosomal hibernation and translational recovery by direct attachment to large subunit ribosomal protein uL5, 25S rRNA backbone, and tRNA-binding P/A-site major groove. Moreover, it independently mediates cell division and cellular stress response by facilitating cytokinetic abscission and disulfide stress-dependent transcriptional regulation, respectively. However, the structural characterization and intracellular physiological status of CCDC124 remain unknown. In this study, we employed advanced in silico protein modeling and characterization tools to generate a native-like tertiary structure of CCDC124 and examine the disorder, low sequence complexity, and aggregation propensities, as well as high-order dimeric/oligomeric states. Subsequently, dimerization of CCDC124 was investigated with co-immunoprecipitation (CO-IP) analysis, immunostaining, and a recent live-cell protein-protein interaction method, bimolecular fluorescence complementation (BiFC). Results revealed CCDC124 as a highly disordered protein consisting of low complexity regions at the N-terminus and an aggregation sequence (151-IAVLSV-156) located in the middle region. Molecular docking and post-docking binding free energy analyses highlighted a potential involvement of V153 residue on the generation of high-order dimeric/oligomeric structures. Co-IP, immunostaining, and BiFC analyses were used to further confirm the dimeric state of CCDC124 predominantly localized at the cytoplasm. In conclusion, our findings revealed in silico structural characterization and in vivo subcellular physiological state of CCDC124, suggesting low-complexity regions located at the N-terminus of disordered CCDC124 may regulate the formation of aggregates or high-order dimeric/oligomeric states., (© 2021 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
115. ZipA Uses a Two-Pronged FtsZ-Binding Mechanism Necessary for Cell Division.
- Author
-
Cameron TA, Vega DE, Yu C, Xiao H, and Margolin W
- Subjects
- Bacterial Proteins chemistry, Bacterial Proteins genetics, Carrier Proteins chemistry, Carrier Proteins genetics, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cell Division, Cytoskeletal Proteins chemistry, Cytoskeletal Proteins genetics, Escherichia coli chemistry, Escherichia coli genetics, Escherichia coli Proteins chemistry, Escherichia coli Proteins genetics, Mutation, Protein Binding, Protein Domains, Bacterial Proteins metabolism, Carrier Proteins metabolism, Cell Cycle Proteins metabolism, Cytoskeletal Proteins metabolism, Escherichia coli cytology, Escherichia coli metabolism, Escherichia coli Proteins metabolism
- Abstract
In most bacteria, cell division is centrally organized by the FtsZ protein, which assembles into dynamic filaments at the division site along the cell membrane that interact with other key cell division proteins. In gammaproteobacteria such as Escherichia coli, FtsZ filaments are anchored to the cell membrane by two essential proteins, FtsA and ZipA. Canonically, this interaction was believed to be mediated solely by the FtsZ C-terminal peptide (CTP) domain that interacts with these and several other regulatory proteins. However, we now provide evidence of a second interaction between FtsZ and ZipA. Using site-specific photoactivated cross-linking, we identified a noncanonical FtsZ-binding site on ZipA on the opposite side from the FtsZ CTP-binding pocket. Cross-linking at this site was unaffected by the truncation of the FtsZ linker and CTP domains, indicating that this noncanonical site must interact directly with the globular core domain of FtsZ. Mutations introduced into either the canonical or noncanonical binding sites on ZipA disrupted photo-cross-linking with FtsZ and normal ZipA function in cell division, suggesting that both binding modes are important for normal cell growth and division. One mutation at the noncanonical face was also found to suppress defects of several other canonical and noncanonical site mutations in ZipA, suggesting there is some interdependence between the two sites. Taken together, these results suggest that ZipA employs a two-pronged FtsZ-binding mechanism. IMPORTANCE The tubulin homolog FtsZ plays a central early role in organizing bacterial cell division proteins at the cytoplasmic membrane. However, FtsZ does not directly interact with the membrane itself, instead relying on proteins such as FtsA to tether it to the membrane. In gammaproteobacteria, ZipA serves as a second essential membrane anchor along with FtsA. Although FtsA has a unique role in activating synthesis of the cell division septum, and ZipA may in turn activate FtsA, it was thought that both proteins interacted only with the conserved C terminus of FtsZ and were essentially interchangeable in their ability to tether FtsZ to the membrane. Here we challenge this view, providing evidence that ZipA directly contacts both the C terminus and the core domain of FtsZ. Such a two-pronged interaction between ZipA and FtsZ suggests that ZipA and FtsA may serve distinct membrane-anchoring roles for FtsZ.
- Published
- 2021
- Full Text
- View/download PDF
116. Mechanisms of distinctive mismatch tolerance between Rad51 and Dmc1 in homologous recombination.
- Author
-
Xu J, Zhao L, Peng S, Chu H, Liang R, Tian M, Connell PP, Li G, Chen C, and Wang HW
- Subjects
- Amino Acid Sequence, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cryoelectron Microscopy, DNA chemistry, DNA genetics, DNA metabolism, DNA Breaks, Double-Stranded, DNA-Binding Proteins chemistry, DNA-Binding Proteins genetics, Humans, Molecular Dynamics Simulation, Multiprotein Complexes chemistry, Multiprotein Complexes ultrastructure, Nucleic Acid Conformation, Protein Conformation, Rad51 Recombinase chemistry, Rad51 Recombinase genetics, Sequence Homology, Amino Acid, Cell Cycle Proteins metabolism, DNA Repair, DNA-Binding Proteins metabolism, Homologous Recombination, Multiprotein Complexes metabolism, Rad51 Recombinase metabolism
- Abstract
Homologous recombination (HR) is a primary DNA double-strand breaks (DSBs) repair mechanism. The recombinases Rad51 and Dmc1 are highly conserved in the RecA family; Rad51 is mainly responsible for DNA repair in somatic cells during mitosis while Dmc1 only works during meiosis in germ cells. This spatiotemporal difference is probably due to their distinctive mismatch tolerance during HR: Rad51 does not permit HR in the presence of mismatches, whereas Dmc1 can tolerate certain mismatches. Here, the cryo-EM structures of Rad51-DNA and Dmc1-DNA complexes revealed that the major conformational differences between these two proteins are located in their Loop2 regions, which contain invading single-stranded DNA (ssDNA) binding residues and double-stranded DNA (dsDNA) complementary strand binding residues, stabilizing ssDNA and dsDNA in presynaptic and postsynaptic complexes, respectively. By combining molecular dynamic simulation and single-molecule FRET assays, we identified that V273 and D274 in the Loop2 region of human RAD51 (hRAD51), corresponding to P274 and G275 of human DMC1 (hDMC1), are the key residues regulating mismatch tolerance during strand exchange in HR. This HR accuracy control mechanism provides mechanistic insights into the specific roles of Rad51 and Dmc1 in DNA double-strand break repair and may shed light on the regulatory mechanism of genetic recombination in mitosis and meiosis., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
117. Tanshinone IIA suppresses the progression of lung adenocarcinoma through regulating CCNA2-CDK2 complex and AURKA/PLK1 pathway.
- Author
-
Li Z, Zhang Y, Zhou Y, Wang F, Yin C, Ding L, and Zhang S
- Subjects
- Abietanes chemistry, Adenocarcinoma of Lung drug therapy, Adenocarcinoma of Lung etiology, Adenocarcinoma of Lung pathology, Antineoplastic Agents, Phytogenic chemistry, Apoptosis drug effects, Aurora Kinase A chemistry, Biomarkers, Tumor, Cell Cycle drug effects, Cell Cycle Proteins chemistry, Cell Line, Tumor, Computational Biology methods, Cyclin A2 chemistry, Cyclin-Dependent Kinase 2 chemistry, Disease Susceptibility, Gene Expression Profiling, Humans, Models, Molecular, Protein Interaction Mapping, Protein Interaction Maps, Protein Serine-Threonine Kinases chemistry, Proto-Oncogene Proteins chemistry, Signal Transduction drug effects, Structure-Activity Relationship, Transcriptome, Polo-Like Kinase 1, Abietanes pharmacology, Adenocarcinoma of Lung metabolism, Antineoplastic Agents, Phytogenic pharmacology, Aurora Kinase A metabolism, Cell Cycle Proteins metabolism, Cyclin A2 metabolism, Cyclin-Dependent Kinase 2 metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Lung adenocarcinoma (LUAD) belongs to a subgroup of non-small cell lung cancer (NSCLC) with an increasing incidence all over the world. Tanshinone IIA (TSA), an active compound of Salvia miltiorrhiza Bunge., has been found to have anti-tumor effects on many tumors, but its anti-LUAD effect and its mechanism have not been reported yet. In this study, bio-information analysis was applied to characterize the potential mechanism of TSA on LUA, biological experiments were used to verify the mechanisms involved. TCGA, Pubchem, SwissTargetPrediction, Venny2.1.0, STRING, DAVID, Cytoscape 3.7.2, Omicshare, GEPIA, RSCBPDB, Chem Draw, AutoDockTools, and PyMOL were utilized for analysis in the bio-information analysis and network pharmacology. Our experiments in vitro focused on the anti-LUAD effects and mechanisms of TSA on LUAD cells (A549 and NCI-H1975 cells) via MTT, plate cloning, Annexin V-FITC and PI dual staining, flow cytometry, and western blot assays. A total of 64 differentially expressed genes (DEGs) of TSA for treatment of LUAD were screened out. Gene ontology and pathway analysis revealed characteristic of the DEGs network. After GEPIA-based DEGs confirmation, 46 genes were considered having significant differences. Further, 10 key DEGs (BTK, HSD11B1, ADAM33, TNNC1, THRA, CCNA2, AURKA, MIF, PLK1, and SORD) were identified as the most likely relevant genes from overall survival analysis. Molecular Docking results showed that CCNA2, CDK2 and PLK1 had the lowest docking energy. MTT and plate cloning assays results showed that TSA inhibited the proliferation of LUAD cells in a concentration-dependent manner. Annexin V-FITC and PI dual staining and flow cytometry assays results told that TSA promoted the apoptosis of the two LUAD cells in different degrees, and induced cycle arrest in the G1/S phase. Western blot results showed that TSA significantly down-regulated the expression of CCNA2, CDK2, AURKA, PLK1, and p-ERK. In summary, TSA could suppress the progression of LUAD by inducing cell apoptosis and arresting cell cycle, and these were done by regulating CCNA2-CDK2 complex and AURKA/PLK1 pathway. These findings are the first to demonstrate the molecular mechanism of TSA in treatment of LUAD combination of network bio-information analysis and biological experiments in vitro., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
118. Cryo-EM structure of MukBEF reveals DNA loop entrapment at chromosomal unloading sites.
- Author
-
Bürmann F, Funke LFH, Chin JW, and Löwe J
- Subjects
- Adenosine Triphosphatases chemistry, Cell Cycle Proteins chemistry, Chromosomes, Bacterial, DNA metabolism, DNA Repair, DNA-Binding Proteins chemistry, Dimerization, Escherichia coli metabolism, Genetic Techniques, Genome, Bacterial, Multiprotein Complexes chemistry, Photorhabdus, Protein Binding, Protein Conformation, Protein Domains, Cohesins, Chromosomal Proteins, Non-Histone chemistry, Chromosomes ultrastructure, Cryoelectron Microscopy methods, DNA chemistry, Escherichia coli Proteins chemistry, Repressor Proteins chemistry
- Abstract
The ring-like structural maintenance of chromosomes (SMC) complex MukBEF folds the genome of Escherichia coli and related bacteria into large loops, presumably by active DNA loop extrusion. MukBEF activity within the replication terminus macrodomain is suppressed by the sequence-specific unloader MatP. Here, we present the complete atomic structure of MukBEF in complex with MatP and DNA as determined by electron cryomicroscopy (cryo-EM). The complex binds two distinct DNA double helices corresponding to the arms of a plectonemic loop. MatP-bound DNA threads through the MukBEF ring, while the second DNA is clamped by the kleisin MukF, MukE, and the MukB ATPase heads. Combinatorial cysteine cross-linking confirms this topology of DNA loop entrapment in vivo. Our findings illuminate how a class of near-ubiquitous DNA organizers with important roles in genome maintenance interacts with the bacterial chromosome., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 MRC Laboratory of Molecular Biology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
119. Structure of a human replisome shows the organisation and interactions of a DNA replication machine.
- Author
-
Jones ML, Baris Y, Taylor MRG, and Yeeles JTP
- Subjects
- Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing genetics, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cytoskeletal Proteins chemistry, Cytoskeletal Proteins genetics, DNA Polymerase II chemistry, DNA Polymerase II genetics, DNA-Binding Proteins chemistry, DNA-Binding Proteins genetics, Humans, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins genetics, Models, Molecular, Poly-ADP-Ribose Binding Proteins chemistry, Poly-ADP-Ribose Binding Proteins genetics, Protein Conformation, Adaptor Proteins, Signal Transducing metabolism, Cell Cycle Proteins metabolism, Cytoskeletal Proteins metabolism, DNA Polymerase II metabolism, DNA Replication, DNA-Binding Proteins metabolism, Intracellular Signaling Peptides and Proteins metabolism, Poly-ADP-Ribose Binding Proteins metabolism
- Abstract
The human replisome is an elaborate arrangement of molecular machines responsible for accurate chromosome replication. At its heart is the CDC45-MCM-GINS (CMG) helicase, which, in addition to unwinding the parental DNA duplex, arranges many proteins including the leading-strand polymerase Pol ε, together with TIMELESS-TIPIN, CLASPIN and AND-1 that have key and varied roles in maintaining smooth replisome progression. How these proteins are coordinated in the human replisome is poorly understood. We have determined a 3.2 Å cryo-EM structure of a human replisome comprising CMG, Pol ε, TIMELESS-TIPIN, CLASPIN and AND-1 bound to replication fork DNA. The structure permits a detailed understanding of how AND-1, TIMELESS-TIPIN and Pol ε engage CMG, reveals how CLASPIN binds to multiple replisome components and identifies the position of the Pol ε catalytic domain. Furthermore, the intricate network of contacts contributed by MCM subunits and TIMELESS-TIPIN with replication fork DNA suggests a mechanism for strand separation., (© 2021 MRC Laboratory of Molecular Biology. Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
120. Synthesis, telomerase inhibitory and anticancer activity of new 2-phenyl-4H-chromone derivatives containing 1,3,4-oxadiazole moiety.
- Author
-
Han X, Yu YL, Ma D, Zhang ZY, and Liu XH
- Subjects
- Amides chemical synthesis, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Chromones metabolism, Chromones pharmacology, Drug Design, Drug Screening Assays, Antitumor, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, Humans, Inhibitory Concentration 50, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Staurosporine pharmacology, Structure-Activity Relationship, Telomerase chemistry, Telomerase metabolism, Antineoplastic Agents chemical synthesis, Cell Cycle Proteins antagonists & inhibitors, Chromones chemical synthesis, Enzyme Inhibitors chemical synthesis, Nuclear Proteins antagonists & inhibitors, Oxadiazoles chemistry, Telomerase antagonists & inhibitors
- Abstract
Based on previous studies, 66 2-phenyl-4H-chromone derivatives containing amide and 1,3,4-oxadiazole moieties were prepared as potential telomerase inhibitors. The results showed most of the title compounds exhibited significantly inhibitory activity on telomerase. Among them, some compounds demonstrated the most potent telomerase inhibitory activity (IC
50 < 1 µM), which was significantly superior to the staurosporine (IC50 = 6.41 µM). In addition, clear structure-activity relationships were summarised, indicating that the substitution of the methoxy group and the position, type and number of the substituents on the phenyl ring had significant effects on telomerase activity. Among them, compound A33 showed considerable inhibition against telomerase. Flow cytometric analysis showed that compound A33 could arrest MGC-803 cell cycle at G2/M phase and induce apoptosis in a concentration-dependent way. Meanwhile, Western blotting revealed that this compound could reduce the expression of dyskerin, which is a fragment of telomerase.- Published
- 2021
- Full Text
- View/download PDF
121. Theoretically exploring selective-binding mechanisms of BRD4 through integrative computational approaches.
- Author
-
Luo D, Tong JB, Xiao XC, Bian S, Zhang X, Wang J, and Xu HY
- Subjects
- Cell Cycle Proteins chemistry, Molecular Docking Simulation, Molecular Dynamics Simulation, Pteridines chemistry, Transcription Factors chemistry, Cell Cycle Proteins antagonists & inhibitors, Drug Design, Pteridines pharmacology, Quantitative Structure-Activity Relationship, Transcription Factors antagonists & inhibitors
- Abstract
The origin of cancer is related to the dysregulation of multiple signal pathways and of physiological processes. Bromodomain-containing protein 4 (BRD4) has become an attractive target for the development of anticancer and anti-inflammatory agents since it can epigenetically regulate the transcription of growth-promoting genes. The synthesized BRD4 inhibitors with new chemical structures can reduce the drug resistance, but their binding modes and the inhibitory mechanism remain unclear. Here, we initially constructed robust QSAR models based on 68 reported tetrahydropteridin analogues using topomer CoMFA and HQSAR. On the basis of QSAR results, we designed 16 novel tetrahydropteridin analogues with modified structures and carried out docking studies. Instead of significant hydrogen bondings with amino acid residue Asn140 as reported in previous research, the molecular docking modelling suggested a novel docking pose that involves the amino acid residues (Trp81, Pro82, Val87, Leu92, Leu94, Cys136, Asp144, and Ile146) at the active site of BRD4. The MD simulations, free energy calculations, and residual energy contributions all indicate that hydrophobic interactions are decisive factors affecting bindings between inhibitors and BRD4. The current study provides new insights that can aid the discovery of BRD4 inhibitors with enhanced anti-cancer ability.
- Published
- 2021
- Full Text
- View/download PDF
122. Plasmin generates vasoinhibin-like peptides by cleaving prolactin and placental lactogen.
- Author
-
Friedrich C, Neugebauer L, Zamora M, Robles JP, Martínez de la Escalera G, Clapp C, Bertsch T, and Triebel J
- Subjects
- Cell Cycle Proteins chemistry, Genetic Variation, HEK293 Cells, Humans, Mass Spectrometry, Placental Lactogen genetics, Prolactin genetics, Proteolysis, Fibrinolysin metabolism, Peptides analysis, Placental Lactogen chemistry, Prolactin chemistry
- Abstract
Vasoinhibin is an antiangiogenic, profibrinolytic peptide generated by the proteolytic cleavage of the pituitary hormone prolactin by cathepsin D, matrix metalloproteinases, and bone morphogenetic protein-1. Vasoinhibin can also be generated when placental lactogen or growth hormone are enzymatically cleaved. Here, it is investigated whether plasmin cleaves human prolactin and placental lactogen to generate vasoinhibin-like peptides. Co-incubation of prolactin and placental lactogen with plasmin was performed and analyzed by gel electrophoresis and Western blotting. Mass spectrometric analyses were carried out for sequence validation and precise cleavage site identification. The cleavage sites responsible for the generation of the vasoinhibin-like peptides were located at K170-E171 in prolactin and R160-T161 in placental lactogen. Various genetic variants of the human prolactin and placental lactogen genes are projected to affect proteolytic generation of the vasoinhibin-like peptides. The endogenous counterparts of the vasoinhibin-like peptides generated by plasmin may represent vasoinhibin-isoforms with inhibitory effects on vasculature and coagulation., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
123. Y06014 is a selective BET inhibitor for the treatment of prostate cancer.
- Author
-
Wu TB, Xiang QP, Wang C, Wu C, Zhang C, Zhang MF, Liu ZX, Zhang Y, Xiao LJ, and Xu Y
- Subjects
- Antineoplastic Agents chemical synthesis, Antineoplastic Agents metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Humans, Indoles chemical synthesis, Indoles metabolism, Isoxazoles chemical synthesis, Isoxazoles metabolism, Male, Molecular Docking Simulation, Molecular Structure, Protein Binding, Protein Domains, Small Molecule Libraries chemical synthesis, Small Molecule Libraries metabolism, Small Molecule Libraries pharmacology, Structure-Activity Relationship, Transcription Factors chemistry, Transcription Factors metabolism, Antineoplastic Agents pharmacology, Indoles pharmacology, Isoxazoles pharmacology, Prostatic Neoplasms drug therapy
- Abstract
Bromodomain and extra-terminal proteins (BETs) are potential targets for the therapeutic treatment of prostate cancer (PC). Herein, we report the design, the synthesis, and a structure-activity relationship study of 6-(3,5-dimethylisoxazol-4-yl)benzo[cd]indol-2(1H)-one derivative as novel selective BET inhibitors. One representative compound, 19 (Y06014), bound to BRD4(1) in the low micromolar range and demonstrated high selectivity for BRD4(1) over other non-BET bromodomain-containing proteins. This molecule also potently inhibited cell growth, colony formation, and mRNA expression of AR-regulated genes in PC cell lines. Y06014 also shows stronger activity than the second-generation antiandrogen enzalutamide. Y06014 may serve as a new small molecule probe for further validation of BET as a molecular target for PC drug development., (© 2021. The Author(s), under exclusive licence to CPS and SIMM.)
- Published
- 2021
- Full Text
- View/download PDF
124. Structural Characterization of Degrader-Induced Ternary Complexes Using Hydrogen-Deuterium Exchange Mass Spectrometry and Computational Modeling: Implications for Structure-Based Design.
- Author
-
Eron SJ, Huang H, Agafonov RV, Fitzgerald ME, Patel J, Michael RE, Lee TD, Hart AA, Shaulsky J, Nasveschuk CG, Phillips AJ, Fisher SL, and Good A
- Subjects
- Acetamides chemistry, Acetamides pharmacology, Gene Expression Regulation drug effects, HEK293 Cells, Humans, Indoles chemistry, Indoles pharmacology, Models, Chemical, Models, Molecular, Molecular Structure, Piperidines chemistry, Piperidines pharmacology, Protein Conformation, Cell Cycle Proteins chemistry, Computer Simulation, Deuterium Exchange Measurement, Mass Spectrometry methods, Transcription Factors chemistry
- Abstract
The field of targeted protein degradation (TPD) has grown exponentially over the past decade with the goal of developing therapies that mark proteins for destruction leveraging the ubiquitin-proteasome system. One common approach to achieve TPD is to employ a heterobifunctional molecule, termed as a degrader, to recruit the protein target of interest to the E3 ligase machinery. The resultant generation of an intermediary ternary complex (target-degrader-ligase) is pivotal in the degradation process. Understanding the ternary complex geometry offers valuable insight into selectivity, catalytic efficiency, linker chemistry, and rational degrader design. In this study, we utilize hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify degrader-induced protein-protein interfaces. We then use these data in conjunction with constrained protein docking to build three-dimensional models of the ternary complex. The approach was used to characterize complex formation between the E3 ligase CRBN and the first bromodomain of BRD4, a prominent oncology target. We show marked differences in the ternary complexes formed in solution based on distinct patterns of deuterium uptake for two degraders, CFT-1297 and dBET6. CFT-1297, which exhibited positive cooperativity, altered the deuterium uptake profile revealing the degrader-induced protein-protein interface of the ternary complex. For CFT-1297, the ternary complexes generated by the highest scoring HDX-constrained docking models differ markedly from those observed in the published crystal structures. These results highlight the potential utility of HDX-MS to provide rapidly accessible structural insights into degrader-induced protein-protein interfaces in solution. They further suggest that degrader ternary complexes exhibit significant conformation flexibility and that biologically relevant complexes may well not exhibit the largest interaction surfaces between proteins. Taken together, the results indicate that methods capable of incorporating linker conformation uncertainty may prove an important component in degrader design moving forward. In addition, the development of scoring functions modified to handle interfaces with no evolved complementarity, for example, through consideration of high levels of water infiltration, may prove valuable. Furthermore, the use of crystal structures as validation tools for novel degrader methods needs to be considered with caution.
- Published
- 2021
- Full Text
- View/download PDF
125. Differential BET Bromodomain Inhibition by Dihydropteridinone and Pyrimidodiazepinone Kinase Inhibitors.
- Author
-
Karim RM, Bikowitz MJ, Chan A, Zhu JY, Grassie D, Becker A, Berndt N, Gunawan S, Lawrence NJ, and Schönbrunn E
- Subjects
- Cell Cycle Proteins chemistry, Crystallography, X-Ray, HEK293 Cells, Humans, Molecular Docking Simulation, Protein Binding, Protein Conformation, Protein Domains, Transcription Factors chemistry, Cell Cycle Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Transcription Factors antagonists & inhibitors
- Abstract
BRD4 and other members of the bromodomain and extraterminal (BET) family of proteins are promising epigenetic targets for the development of novel therapeutics. Among the reported BRD4 inhibitors are dihydropteridinones and benzopyrimidodiazepinones originally designed to target the kinases PLK1, ERK5, and LRRK2. While these kinase inhibitors were identified as BRD4 inhibitors, little is known about their binding potential and structural details of interaction with the other BET bromodomains. We comprehensively characterized a series of known and newly identified dual BRD4-kinase inhibitors against all eight individual BET bromodomains. A detailed analysis of 23 novel cocrystal structures of BET-kinase inhibitor complexes in combination with direct binding assays and cell signaling studies revealed significant differences in molecular shape complementarity and inhibitory potential. Collectively, the data offer new insights into the action of kinase inhibitors across BET bromodomains, which may aid the development of drugs to inhibit certain BET proteins and kinases differentially.
- Published
- 2021
- Full Text
- View/download PDF
126. The role of distinct BRD4 isoforms and their contribution to high-grade serous ovarian carcinoma pathogenesis.
- Author
-
Drumond-Bock AL and Bieniasz M
- Subjects
- Animals, Biomarkers, Tumor, Cell Cycle Proteins antagonists & inhibitors, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cell Transformation, Neoplastic genetics, Cystadenocarcinoma, Serous pathology, Drug Resistance, Neoplasm genetics, Female, Gene Expression Regulation, Neoplastic, Genomic Instability, Humans, Molecular Targeted Therapy, Nerve Tissue Proteins metabolism, Ovarian Neoplasms pathology, Ovarian Neoplasms therapy, Protein Isoforms, Receptors, Cell Surface metabolism, Structure-Activity Relationship, Transcription Factors antagonists & inhibitors, Transcription Factors chemistry, Transcription Factors genetics, Cell Cycle Proteins metabolism, Cell Transformation, Neoplastic metabolism, Cystadenocarcinoma, Serous etiology, Cystadenocarcinoma, Serous metabolism, Disease Susceptibility, Ovarian Neoplasms etiology, Ovarian Neoplasms metabolism, Transcription Factors metabolism
- Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most aggressive type of ovarian cancer, often diagnosed at advanced stages. Molecularly, HGSOC shows high degree of genomic instability associated with large number of genetic alterations. BRD4 is the 4th most amplified gene in HGSOC, which correlates with poor patients' prognosis. BRD4 is constitutively expressed and generates two proteins, BRD4 long (BRD4-L) and BRD4 short (BRD4-S). Both isoforms contain bromodomains that bind to lysine-acetylated histones. Amongst other functions, BRD4 participates in chromatin organization, acetylation of histones, transcriptional control and DNA damage repair. In cancer patients with amplified BRD4, the increased activity of BRD4 is associated with higher expression of oncogenes, such as MYC, NOTCH3 and NRG1. BRD4-driven oncogenes promote increased tumor cells proliferation, genetic instability, epithelial-mesenchymal transition, metastasis and chemoresistance. Ablation of BRD4 activity can be successfully achieved with bromodomain inhibitors (BETi) and degraders, and it has been applied in pre-clinical and clinical settings. Inhibition of BRD4 function has an effective anti-cancer effect, reducing tumor growth whether ablated by single agents or in combination with other drugs. When combined with standard chemotherapy, BETi are capable of sensitizing highly resistant ovarian cancer cell lines to platinum drugs. Despite the evidence that BRD4 amplification in ovarian cancer contributes to poor patient prognosis, little is known about the specific mechanisms by which BRD4 drives tumor progression. In addition, newly emerging data revealed that BRD4 isoforms exhibit contradicting functions in cancer. Therefore, it is paramount to expand studies elucidating distinct roles of BRD4-L and BRD4-S in HGSOC, which has important implications on development of therapeutic approaches targeting BRD4., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
127. Oncogenic Truncations of ASXL1 Enhance a Motif for BRD4 ET-Domain Binding.
- Author
-
Burgess AE, Kleffmann T, and Mace PD
- Subjects
- Amino Acid Motifs, Binding Sites, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Epitopes, Frameshift Mutation, Gain of Function Mutation, HEK293 Cells, Humans, Multiprotein Complexes metabolism, Protein Domains, Repressor Proteins chemistry, Repressor Proteins immunology, Reproducibility of Results, Transcription Factors chemistry, Transcription Factors genetics, Cell Cycle Proteins metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Transcription Factors metabolism
- Abstract
Proper regulation of gene-expression relies on specific protein-protein interactions between a myriad of epigenetic regulators. As such, mutation of genes encoding epigenetic regulators often drive cancer and developmental disorders. Additional sex combs-like protein 1 (ASXL1) is a key example, where mutations frequently drive haematological cancers and can cause developmental disorders. It has been reported that nonsense mutations in ASXL1 promote an interaction with BRD4, another central epigenetic regulator. Here we provide a molecular mechanism for the BRD4-ASXL1 interaction, demonstrating that a motif near to common truncation breakpoints of ASXL1 contains an epitope that binds the ET domain within BRD4. Binding-studies show that this interaction is analogous to common ET-binding modes of BRD4-interactors, and that all three ASX-like protein orthologs (ASXL1-3) contain a functional ET domain-binding epitope. Crucially, we observe that BRD4-ASXL1 binding is markedly increased in the prevalent ASXL1
Y591X truncation that maintains the BRD4-binding epitope, relative to full-length ASXL1 or truncated proteins that delete the epitope. Together, these results show that ASXL1 truncation enhances BRD4 recruitment to transcriptional complexes via its ET domain, which could misdirect regulatory activity of either BRD4 or ASXL1 and may inform potential therapeutic interventions., Competing Interests: Declaration of interests The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2021 The Author(s). Published by Elsevier Ltd.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
128. Nematode CDC-37 and DNJ-13 form complexes and can interact with HSP-90.
- Author
-
Schmauder L, Absmeier E, Bepperling A, Barkovits K, Marcus K, and Richter K
- Subjects
- Animals, Caenorhabditis elegans chemistry, Caenorhabditis elegans Proteins chemistry, Cell Cycle Proteins chemistry, HSP40 Heat-Shock Proteins chemistry, HSP90 Heat-Shock Proteins chemistry, Models, Molecular, Protein Binding, Protein Folding, Protein Interaction Maps, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Cell Cycle Proteins metabolism, HSP40 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins metabolism
- Abstract
The molecular chaperones Hsc70 and Hsp90 are required for proteostasis control and specific folding of client proteins in eukaryotic and prokaryotic organisms. Especially in eukaryotes these ATP-driven molecular chaperones are interacting with cofactors that specify the client spectrum and coordinate the ATPase cycles. Here we find that a Hsc70-cofactor of the Hsp40 family from nematodes, DNJ-13, directly interacts with the kinase-specific Hsp90-cofactor CDC-37. The interaction is specific for DNJ-13, while DNJ-12 another DnaJ-like protein of C. elegans, does not bind to CDC-37 in a similar manner. Analytical ultracentrifugation is employed to show that one CDC-37 molecule binds to a dimeric DNJ-13 protein with low micromolar affinity. We perform cross-linking studies with mass spectrometry to identify the interaction site and obtain specific cross-links connecting the N-terminal J-domain of DNJ-13 with the N-terminal domain of CDC-37. Further AUC experiments reveal that both, the N-terminal part of CDC-37 and the C-terminal domain of CDC-37, are required for efficient interaction. Furthermore, the presence of DNJ-13 strengthens the complex formation between CDC-37 and HSP-90 and modulates the nucleotide-dependent effects. These findings on the interaction between Hsp40 proteins and Hsp90-cofactors provide evidence for a more intricate interaction between the two chaperone systems during client processing., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
129. Ribonucleotide reductase: In-vitro S-glutathionylation of R2 and p53R2 subunits of mammalian class I ribonucleotide reductase protein.
- Author
-
Chatterji A, Holmgren A, and Sengupta R
- Subjects
- Animals, Mice, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, DNA Replication, Glutathione chemistry, Glutathione metabolism, Protein Subunits chemistry, Protein Subunits metabolism, Ribonucleotide Reductases chemistry, Ribonucleotide Reductases metabolism, S Phase
- Abstract
Ribonucleotide reductases (RNR) catalyze the rate-limiting step in DNA synthesis during the S-phase of the cell cycle. Its constant activity in order to maintain dNTP homeostasis is a fascinating area of research and an attractive candidate for cancer research and antiviral drugs. Redox modification such as S-glutathionylation of the R1 subunit of mammalian RNR protein has been presumed to regulate the activity of RNR during catalytic cycles. Herein, we report S-glutathionylation of the R2 subunit. We have also shown Grx1 system can efficiently deglutathionylate the S-glutathionylated R2 subunit. Additionally, our data also showed for the very first time S-glutathionylation of mammalian p53R2 subunit that regulates DNA synthesis outside S-phase during DNA damage and repair. Taken together, these data will open new avenues for future research relating to exact physiological significance, target thiols, and/or overall RNR activity due to S-glutathionylation of R2 and p53R2 subunits and provide valuable insights for effective treatment regimes., (© 2021. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2021
- Full Text
- View/download PDF
130. Mechanistic roles of tyrosine phosphorylation in reversible amyloids, autoinhibition, and endosomal membrane association of ALIX.
- Author
-
Elias RD, Ramaraju B, and Deshmukh L
- Subjects
- Humans, Nuclear Magnetic Resonance, Biomolecular, Phosphorylation, Protein Domains, Structure-Activity Relationship, Tyrosine, Amyloid chemistry, Amyloid metabolism, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Endosomal Sorting Complexes Required for Transport chemistry, Endosomal Sorting Complexes Required for Transport metabolism, Endosomes chemistry, Endosomes metabolism, Intracellular Membranes chemistry, Intracellular Membranes metabolism
- Abstract
Human apoptosis-linked gene-2 interacting protein X (ALIX), a versatile adapter protein, regulates essential cellular processes by shuttling between late endosomal membranes and the cytosol, determined by its interactions with Src kinase. Here, we investigate the molecular basis of these transitions and the effects of tyrosine phosphorylation on the interplay between structure, assembly, and intramolecular and intermolecular interactions of ALIX. As evidenced by transmission electron microscopy, fluorescence and circular dichroism spectroscopy, the proline-rich domain of ALIX, which encodes binding epitopes of multiple cellular partners, formed rope-like β-sheet-rich reversible amyloid fibrils that dissolved upon Src-mediated phosphorylation and were restored on protein-tyrosine phosphatase 1B-mediated dephosphorylation of its conserved tyrosine residues. Analyses of the Bro1 domain of ALIX by solution NMR spectroscopy elucidated the conformational changes originating from its phosphorylation by Src and established that Bro1 binds to hyperphosphorylated proline-rich domain and to analogs of late endosomal membranes via its highly basic surface. These results uncover the autoinhibition mechanism that relocates ALIX to the cytosol and the diverse roles played by tyrosine phosphorylation in cellular and membrane functions of ALIX., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
131. Molecular modelling of the FOXO4-TP53 interaction to design senolytic peptides for the elimination of senescent cancer cells.
- Author
-
Le HH, Cinaroglu SS, Manalo EC, Ors A, Gomes MM, Duan Sahbaz B, Bonic K, Origel Marmolejo CA, Quentel A, Plaut JS, Kawashima TE, Ozdemir ES, Malhotra SV, Ahiska Y, Sezerman U, Bayram Akcapinar G, Saldivar JC, Timucin E, and Fischer JM
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Cell Cycle Proteins metabolism, Cellular Senescence drug effects, Disease Models, Animal, Female, Forkhead Transcription Factors metabolism, Humans, Male, Melanoma, Mice, Molecular Docking Simulation, Molecular Dynamics Simulation, Peptides pharmacology, Protein Conformation, Senotherapeutics pharmacology, Structure-Activity Relationship, Tumor Suppressor Protein p53 metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents chemistry, Cell Cycle Proteins chemistry, Drug Design, Forkhead Transcription Factors chemistry, Models, Molecular, Peptides chemistry, Senotherapeutics chemistry, Tumor Suppressor Protein p53 chemistry
- Abstract
Background: Senescent cells accumulate in tissues over time as part of the natural ageing process and the removal of senescent cells has shown promise for alleviating many different age-related diseases in mice. Cancer is an age-associated disease and there are numerous mechanisms driving cellular senescence in cancer that can be detrimental to recovery. Thus, it would be beneficial to develop a senolytic that acts not only on ageing cells but also senescent cancer cells to prevent cancer recurrence or progression., Methods: We used molecular modelling to develop a series of rationally designed peptides to mimic and target FOXO4 disrupting the FOXO4-TP53 interaction and releasing TP53 to induce apoptosis. We then tested these peptides as senolytic agents for the elimination of senescent cells both in cell culture and in vivo., Findings: Here we show that these peptides can act as senolytics for eliminating senescent human cancer cells both in cell culture and in orthotopic mouse models. We then further characterized one peptide, ES2, showing that it disrupts FOXO4-TP53 foci, activates TP53 mediated apoptosis and preferentially binds FOXO4 compared to TP53. Next, we show that intratumoural delivery of ES2 plus a BRAF inhibitor results in a significant increase in apoptosis and a survival advantage in mouse models of melanoma. Finally, we show that repeated systemic delivery of ES2 to older mice results in reduced senescent cell numbers in the liver with minimal toxicity., Interpretation: Taken together, our results reveal that peptides can be generated to specifically target and eliminate FOXO4+ senescent cancer cells, which has implications for eradicating residual disease and as a combination therapy for frontline treatment of cancer., Funding: This work was supported by the Cancer Early Detection Advanced Research Center at Oregon Health & Science University., Competing Interests: Declaration of Competing Interest S.S. Cinaroglu, Y. Ahiska, U. Sezerman, G. Bayram Akcapinar, E. Timucin have a patent for the ES2 structure and are members of a company, Eternans Ltd. that is working on ES2 for therapeutic use. No potential conflicts of interest were disclosed by the other authors., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
132. Kinetic and structural roles for the surface in guiding SAS-6 self-assembly to direct centriole architecture.
- Author
-
Banterle N, Nievergelt AP, de Buhr S, Hatzopoulos GN, Brillard C, Andany S, Hübscher T, Sorgenfrei FA, Schwarz US, Gräter F, Fantner GE, and Gönczy P
- Subjects
- Chlamydomonas reinhardtii chemistry, Kinetics, Microscopy, Atomic Force, Models, Chemical, Molecular Dynamics Simulation, Organelle Biogenesis, Protein Conformation, Protein Multimerization, Cell Cycle Proteins chemistry, Centrioles chemistry
- Abstract
Discovering mechanisms governing organelle assembly is a fundamental pursuit in biology. The centriole is an evolutionarily conserved organelle with a signature 9-fold symmetrical chiral arrangement of microtubules imparted onto the cilium it templates. The first structure in nascent centrioles is a cartwheel, which comprises stacked 9-fold symmetrical SAS-6 ring polymers emerging orthogonal to a surface surrounding each resident centriole. The mechanisms through which SAS-6 polymerization ensures centriole organelle architecture remain elusive. We deploy photothermally-actuated off-resonance tapping high-speed atomic force microscopy to decipher surface SAS-6 self-assembly mechanisms. We show that the surface shifts the reaction equilibrium by ~10
4 compared to solution. Moreover, coarse-grained molecular dynamics and atomic force microscopy reveal that the surface converts the inherent helical propensity of SAS-6 polymers into 9-fold rings with residual asymmetry, which may guide ring stacking and impart chiral features to centrioles and cilia. Overall, our work reveals fundamental design principles governing centriole assembly., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
133. Discovery of a covalent inhibitor of heat shock protein 90 with antitumor activity that blocks the co-chaperone binding via C-terminal modification.
- Author
-
Li L, Chen N, Xia D, Xu S, Dai W, Tong Y, Wang L, Jiang Z, You Q, and Xu X
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Movement drug effects, Chaperonins chemistry, Chaperonins metabolism, HSP90 Heat-Shock Proteins metabolism, Humans, Isothiocyanates chemistry, Isothiocyanates metabolism, Male, Mice, Mice, Nude, Neoplasms drug therapy, Neoplasms pathology, Protein Binding, Structure-Activity Relationship, Sulfoxides chemistry, Sulfoxides metabolism, Transplantation, Heterologous, Antineoplastic Agents metabolism, Drug Discovery, HSP90 Heat-Shock Proteins antagonists & inhibitors
- Abstract
Heat shock protein (Hsp90), a critical molecular chaperone that regulates the maturation of a large number of oncogenic client proteins, plays an essential role in the growth of neoplastic cells. Herein, DDO-6600 is identified to covalent modification of Cys598 on Hsp90 from in silico study and is verified by a series of biological assays. We demonstrated that DDO-6600 covalently bound to Cys598 on the Hsp90 C terminus and exhibited antiproliferative activities against multiple tumor cells without inhibiting ATPase activity. Further studies showed that DDO-6600 disrupted the interaction between Hsp90 and Cdc37, which induced the degradation of kinase client proteins in multiple tumor cell lines, promoted apoptosis, and inhibited cell motility. Our findings offer mechanic insights into the covalent modification of Hsp90 and provide an alternative strategy for the development of Hsp90 covalent regulators or chemical probes to explore the therapeutical potential of Hsp90., Competing Interests: Declaration of interests Q.Y., X.X., L.L., N.C., W.D., Z.J., Xiaoke Guo, L.W., and Mengchen Lu are co-inventors on pending patent application CN 202110011243.5 that relates to pyrazole inhibitors of Hsp90 for medicinal applications. The authors declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
134. Native mass spectrometry and gas-phase fragmentation provide rapid and in-depth topological characterization of a PROTAC ternary complex.
- Author
-
Song JH, Wagner ND, Yan J, Li J, Huang RY, Balog AJ, Newitt JA, Chen G, and Gross ML
- Subjects
- Cell Cycle Proteins chemistry, Protein Interaction Maps, Proteolysis, Transcription Factors chemistry, Ubiquitin-Protein Ligases chemistry, Cell Cycle Proteins metabolism, Gases chemistry, Ion Mobility Spectrometry methods, Transcription Factors metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Proteolysis-targeting chimeras (PROTACs) represent a new direction in small-molecule therapeutics whereby a heterobifunctional linker to a protein of interest (POI) induces its ubiquitination-based proteolysis by recruiting an E3 ligase. Here, we show that charge reduction, native mass spectrometry, and gas-phase activation methods combine for an in-depth analysis of a PROTAC-linked ternary complex. Electron capture dissociation (ECD) of the intact POI-PROTAC-VCB complex (a trimeric subunit of an E3 ubiquitin ligase) promotes POI dissociation. Collision-induced dissociation (CID) causes elimination of the nonperipheral PROTAC, producing an intact VCB-POI complex not seen in solution but consistent with PROTAC-induced protein-protein interactions. In addition, we used ion mobility spectrometry (IMS) and collisional activation to identify the source of this unexpected dissociation. Together, the evidence shows that this integrated approach can be used to screen for ternary complex formation and PROTAC-protein contacts and may report on PROTAC-induced protein-protein interactions, a characteristic correlated with PROTAC selectivity and efficacy., Competing Interests: Declaration of interests Michael L. Gross is an unpaid member of the scientific advisory boards of Protein Metrics Inc. and Gen Next Technologies, two companies pursuing ideas in structural mass spectrometry., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
135. Structural basis and regulation of the reductive stress response.
- Author
-
Manford AG, Mena EL, Shih KY, Gee CL, McMinimy R, Martínez-González B, Sherriff R, Lew B, Zoltek M, Rodríguez-Pérez F, Woldesenbet M, Kuriyan J, and Rape M
- Subjects
- Amino Acids chemistry, Animals, Carrier Proteins chemistry, Carrier Proteins metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Female, Humans, Ions, Mice, Mutant Proteins metabolism, Mutation genetics, Protein Binding drug effects, Protein Stability drug effects, Reactive Oxygen Species metabolism, Structure-Activity Relationship, Substrate Specificity drug effects, Ubiquitin-Protein Ligase Complexes chemistry, Ubiquitin-Protein Ligase Complexes metabolism, Ubiquitination drug effects, Zinc pharmacology, Stress, Physiological drug effects
- Abstract
Although oxidative phosphorylation is best known for producing ATP, it also yields reactive oxygen species (ROS) as invariant byproducts. Depletion of ROS below their physiological levels, a phenomenon known as reductive stress, impedes cellular signaling and has been linked to cancer, diabetes, and cardiomyopathy. Cells alleviate reductive stress by ubiquitylating and degrading the mitochondrial gatekeeper FNIP1, yet it is unknown how the responsible E3 ligase CUL2
FEM1B can bind its target based on redox state and how this is adjusted to changing cellular environments. Here, we show that CUL2FEM1B relies on zinc as a molecular glue to selectively recruit reduced FNIP1 during reductive stress. FNIP1 ubiquitylation is gated by pseudosubstrate inhibitors of the BEX family, which prevent premature FNIP1 degradation to protect cells from unwarranted ROS accumulation. FEM1B gain-of-function mutation and BEX deletion elicit similar developmental syndromes, showing that the zinc-dependent reductive stress response must be tightly regulated to maintain cellular and organismal homeostasis., Competing Interests: Declaration of interests M.R. and J.K. are co-founders and members of the SAB of Nurix Tx. M.R. is on the SAB of Monte Rosa Tx and an iPartner at The Column Group. J.K. is on the SAB of Revolution Medicine and Carmot Tx., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
136. Cohesin mediates DNA loop extrusion by a "swing and clamp" mechanism.
- Author
-
Bauer BW, Davidson IF, Canena D, Wutz G, Tang W, Litos G, Horn S, Hinterdorfer P, and Peters JM
- Subjects
- Adenosine Triphosphatases metabolism, Adenosine Triphosphate metabolism, Binding Sites, Cell Cycle Proteins chemistry, DNA metabolism, Fluorescence Resonance Energy Transfer, HeLa Cells, Humans, Hydrolysis, Kinetics, Microscopy, Atomic Force, Models, Molecular, Nuclear Proteins metabolism, Protein Conformation, Cohesins, Cell Cycle Proteins metabolism, Chromosomal Proteins, Non-Histone metabolism, DNA chemistry, Nucleic Acid Conformation
- Abstract
Structural maintenance of chromosomes (SMC) complexes organize genome topology in all kingdoms of life and have been proposed to perform this function by DNA loop extrusion. How this process works is unknown. Here, we have analyzed how loop extrusion is mediated by human cohesin-NIPBL complexes, which enable chromatin folding in interphase cells. We have identified DNA binding sites and large-scale conformational changes that are required for loop extrusion and have determined how these are coordinated. Our results suggest that DNA is translocated by a spontaneous 50 nm-swing of cohesin's hinge, which hands DNA over to the ATPase head of SMC3, where upon binding of ATP, DNA is clamped by NIPBL. During this process, NIPBL "jumps ship" from the hinge toward the SMC3 head and might thereby couple the spontaneous hinge swing to ATP-dependent DNA clamping. These results reveal mechanistic principles of how cohesin-NIPBL and possibly other SMC complexes mediate loop extrusion., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
137. KCTD11 inhibits progression of lung cancer by binding to β-catenin to regulate the activity of the Wnt and Hippo pathways.
- Author
-
Yang M, Han YM, Han Q, Rong XZ, Liu XF, and Ln XY
- Subjects
- Adult, Aged, Animals, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cell Line, Tumor, Cell Movement, Cell Proliferation, Disease Models, Animal, Epithelial-Mesenchymal Transition genetics, Female, Gene Expression, Heterografts, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Lung Neoplasms etiology, Lung Neoplasms pathology, Male, Mice, Middle Aged, Neoplasm Grading, Neoplasm Metastasis, Neoplasm Staging, Phosphorylation, Prognosis, Protein Binding, Protein Interaction Domains and Motifs, Protein Transport, Transcription Factors metabolism, Transferases chemistry, Transferases genetics, Cell Cycle Proteins metabolism, Hippo Signaling Pathway, Lung Neoplasms metabolism, Transferases metabolism, Wnt Signaling Pathway, beta Catenin metabolism
- Abstract
KCTD11 has been reported to be a potential tumour suppressor in several tumour types. However, the expression of KCTD11 and its role has not been reported in human non-small cell lung cancer (NSCLC). Whether its potential molecular mechanism is related to its BTB domain is also unknown. The expression of KCTD11 in 139 NSCLC tissue samples was detected by immunohistochemistry, and its correlation with clinicopathological factors was analysed. The effect of KCTD11 on the biological behaviour of lung cancer cells was verified in vitro and in vivo. Its effect on the epithelial-mesenchymal transition(EMT)process and the Wnt/β-catenin and Hippo/YAP pathways were observed by Western blot, dual-luciferase assay, RT-qPCR, immunofluorescence and immunoprecipitation. KCTD11 is under-expressed in lung cancer tissues and cells and was negatively correlated with the degree of differentiation, tumour-node-metastasis (TNM) stage and lymph node metastasis. Low KCTD11 expression was associated with poor prognosis. KCTD11 overexpression inhibited the proliferation and migration of lung cancer cells. Further studies indicated that KCTD11 inhibited the Wnt pathway, activated the Hippo pathway and inhibited EMT processes by inhibiting the nuclear translocation of β-catenin and YAP. KCTD11 lost its stimulatory effect on the Hippo pathway after knock down of β-catenin. These findings confirm that KCTD11 inhibits β-catenin and YAP nuclear translocation as well as the malignant phenotype of lung cancer cells by interacting with β-catenin. This provides an important experimental basis for the interaction between KCTD11, β-catenin and YAP, further revealing the link between the Wnt and Hippo pathways., (© 2021 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
138. Mass-sensitive particle tracking to elucidate the membrane-associated MinDE reaction cycle.
- Author
-
Heermann T, Steiert F, Ramm B, Hundt N, and Schwille P
- Subjects
- Adenosine Triphosphatases chemistry, Cell Cycle Proteins chemistry, Cell Membrane metabolism, Escherichia coli, Escherichia coli Proteins chemistry, Lipid Bilayers chemistry, Adenosine Triphosphatases metabolism, Biophysical Phenomena, Cell Cycle Proteins metabolism, Cell Membrane physiology, Escherichia coli Proteins metabolism, Lipid Bilayers metabolism
- Abstract
In spite of their great importance in biology, methods providing access to spontaneous molecular interactions with and on biological membranes have been sparse. The recent advent of mass photometry to quantify mass distributions of unlabeled biomolecules landing on surfaces raised hopes that this approach could be transferred to membranes. Here, by introducing a new interferometric scattering (iSCAT) image processing and analysis strategy adapted to diffusing particles, we enable mass-sensitive particle tracking (MSPT) of single unlabeled biomolecules on a supported lipid bilayer. We applied this approach to the highly nonlinear reaction cycles underlying MinDE protein self-organization. MSPT allowed us to determine the stoichiometry and turnover of individual membrane-bound MinD/MinDE protein complexes and to quantify their size-dependent diffusion. This study demonstrates the potential of MSPT to enhance our quantitative understanding of membrane-associated biological systems., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
139. Reversible amyloids of pyruvate kinase couple cell metabolism and stress granule disassembly.
- Author
-
Cereghetti G, Wilson-Zbinden C, Kissling VM, Diether M, Arm A, Yoo H, Piazza I, Saad S, Picotti P, Drummond DA, Sauer U, Dechant R, and Peter M
- Subjects
- Adenosine Triphosphate metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Fructosediphosphates metabolism, HSP70 Heat-Shock Proteins genetics, HSP70 Heat-Shock Proteins metabolism, Heat-Shock Proteins genetics, Heat-Shock Proteins metabolism, Pyruvate Kinase chemistry, Pyruvate Kinase genetics, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae growth & development, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Amyloid chemistry, Cell Cycle Proteins chemistry, Cytoplasmic Granules chemistry, Pyruvate Kinase metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Stress, Physiological
- Abstract
Cells respond to stress by blocking translation, rewiring metabolism and forming transient messenger ribonucleoprotein assemblies called stress granules (SGs). After stress release, re-establishing homeostasis and disassembling SGs requires ATP-consuming processes. However, the molecular mechanisms whereby cells restore ATP production and disassemble SGs after stress remain poorly understood. Here we show that upon stress, the ATP-producing enzyme Cdc19 forms inactive amyloids, and that their rapid re-solubilization is essential to restore ATP production and disassemble SGs in glucose-containing media. Cdc19 re-solubilization is initiated by the glycolytic metabolite fructose-1,6-bisphosphate, which directly binds Cdc19 amyloids, allowing Hsp104 and Ssa2 chaperone recruitment and aggregate re-solubilization. Fructose-1,6-bisphosphate then promotes Cdc19 tetramerization, which boosts its activity to further enhance ATP production and SG disassembly. Together, these results describe a molecular mechanism that is critical for stress recovery and directly couples cellular metabolism with SG dynamics via the regulation of reversible Cdc19 amyloids., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
140. Loss of BubR1 acetylation provokes replication stress and leads to complex chromosomal rearrangements.
- Author
-
Park J, Yeu SY, Paik S, Kim H, Choi SY, Lee J, Jang J, Lee S, Koh Y, and Lee H
- Subjects
- Acetylation, Animals, Carcinogenesis genetics, Carcinogenesis metabolism, Cell Cycle Checkpoints, Cell Cycle Proteins chemistry, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Serine-Threonine Kinases chemistry, Aneuploidy, Carcinogenesis pathology, Cell Cycle Proteins physiology, Chromosomal Instability, Chromosome Segregation, Protein Serine-Threonine Kinases physiology, Telomere genetics, Tumor Suppressor Protein p53 physiology
- Abstract
Accurate chromosomal segregation during mitosis is regulated by the spindle assembly checkpoint (SAC). SAC failure results in aneuploidy, a hallmark of cancer. However, many studies have suggested that aneuploidy alone is not oncogenic. We have reported that BubR1 acetylation deficiency in mice (K243R/+) caused spontaneous tumorigenesis via weakened SAC signaling and unstable chromosome-spindle attachment, resulting in massive chromosomal mis-segregation. In addition to aneuploidy, cells derived from K243R/+ mice exhibited moderate genetic instability and chromosomal translocation. Here, we investigated how the loss of BubR1 acetylation led to genetic instability and chromosomal rearrangement. To rescue all chromosomal abnormalities generated by the loss of BubR1 acetylation during development, K243R/+ mice were crossed with p53-deficient mice. Genome-wide sequencing and spectral karyotyping of tumors derived from these double-mutant mice revealed that BubR1 acetylation deficiency was associated with complex chromosomal rearrangements, including Robertsonian-like whole-arm translocations. By analyzing the telomeres and centromeres in metaphase chromosome spreads, we found that BubR1 acetylation deficiency increased the collapse of stalled replication forks, commonly referred to as replication stress, and led to DNA damage and chromosomal rearrangements. BubR1 mutations that are critical in interacting with PCAF acetyltransferase and acetylating K250, L249F and A251P, were found from human cancers. Furthermore, a subset of human cancer cells exhibiting whole-arm translocation also displayed defects in BubR1 acetylation, supporting that defects in BubR1 acetylation in mitosis contributes to tumorigenesis. Collectively, loss of BubR1 acetylation provokes replication stress, particularly at the telomeres, leading to genetic instability and chromosomal rearrangement., (© 2021 Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
141. BRD4 orchestrates genome folding to promote neural crest differentiation.
- Author
-
Linares-Saldana R, Kim W, Bolar NA, Zhang H, Koch-Bojalad BA, Yoon S, Shah PP, Karnay A, Park DS, Luppino JM, Nguyen SC, Padmanabhan A, Smith CL, Poleshko A, Wang Q, Li L, Srivastava D, Vahedi G, Eom GH, Blobel GA, Joyce EF, and Jain R
- Subjects
- Animals, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Chromatin metabolism, Chromosomal Proteins, Non-Histone metabolism, Gene Expression Regulation, HEK293 Cells, Humans, Hydrophobic and Hydrophilic Interactions, Integrases metabolism, Mice, Models, Biological, Mouse Embryonic Stem Cells metabolism, Muscle Cells cytology, Neural Crest metabolism, Protein Binding, Protein Domains, Proteolysis, Transcription Factors chemistry, Transcription, Genetic, Cohesins, Cell Differentiation genetics, Genome, Neural Crest cytology, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
Higher-order chromatin structure regulates gene expression, and mutations in proteins mediating genome folding underlie developmental disorders known as cohesinopathies. However, the relationship between three-dimensional genome organization and embryonic development remains unclear. Here we define a role for bromodomain-containing protein 4 (BRD4) in genome folding, and leverage it to understand the importance of genome folding in neural crest progenitor differentiation. Brd4 deletion in neural crest results in cohesinopathy-like phenotypes. BRD4 interacts with NIPBL, a cohesin agonist, and BRD4 depletion or loss of the BRD4-NIPBL interaction reduces NIPBL occupancy, suggesting that BRD4 stabilizes NIPBL on chromatin. Chromatin interaction mapping and imaging experiments demonstrate that BRD4 depletion results in compromised genome folding and loop extrusion. Finally, mutation of individual BRD4 amino acids that mediate an interaction with NIPBL impedes neural crest differentiation into smooth muscle. Remarkably, loss of WAPL, a cohesin antagonist, rescues attenuated smooth muscle differentiation resulting from BRD4 loss. Collectively, our data reveal that BRD4 choreographs genome folding and illustrates the relevance of balancing cohesin activity for progenitor differentiation., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
142. The role of WDR76 protein in human diseases.
- Author
-
Yang J, Wang F, and Chen B
- Subjects
- Animals, Carcinogenesis, Computational Biology methods, Humans, Mice, Molecular Conformation, Neoplasm Metastasis, Peptides chemistry, Protein Domains, Proteomics methods, Ubiquitination, Cell Cycle Proteins chemistry, DNA-Binding Proteins chemistry, Ubiquitin chemistry, WD40 Repeats genetics
- Abstract
The WD40 repeat (WDR) domain is one of the most abundant protein interaction domains in the human proteome. More than 360 protein interaction domains have been annotated thus far. The WDR domains mediate interactions with peptide regions of important interaction partners in a variety of biological processes. Proteins with the WDR domain which typically contains a seven-bladed β propeller, are continuously being discovered. They represent a large class of proteins that are likely to play important roles. WD40 repeat domain-containing protein 76 (WDR76) is a member of WDR domain-containing proteins. Although it remains poorly understood, it is potentially involved in DNA damage repair, apoptosis, cell cycle progression, and gene expression regulation. Ongoing research on WDR76 is increasing the knowledge regarding its basic functions and role in different pathophysiological. The study of WDR76 is challenging due to the complexity of its interactions with its partners. In the present review, we summarized the current knowledge regarding WDR76, its physiological functions, the close relationship with human diseases, and potential opportunities for target therapy.
- Published
- 2021
- Full Text
- View/download PDF
143. Structural and mechanistic insights into the Artemis endonuclease and strategies for its inhibition.
- Author
-
Yosaatmadja Y, Baddock HT, Newman JA, Bielinski M, Gavard AE, Mukhopadhyay SMM, Dannerfjord AA, Schofield CJ, McHugh PJ, and Gileadi O
- Subjects
- B-Lymphocytes enzymology, Catalytic Domain genetics, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Crystallography, X-Ray, DNA End-Joining Repair genetics, DNA Repair genetics, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins chemistry, DNA-Binding Proteins genetics, Endonucleases antagonists & inhibitors, Endonucleases chemistry, Endonucleases genetics, Enzyme Inhibitors chemistry, Exodeoxyribonucleases chemistry, Exodeoxyribonucleases genetics, Humans, Phosphorylation genetics, Protein Folding, Severe Combined Immunodeficiency enzymology, Severe Combined Immunodeficiency pathology, T-Lymphocytes enzymology, Cell Cycle Proteins ultrastructure, DNA-Binding Proteins ultrastructure, Endonucleases ultrastructure, Exodeoxyribonucleases ultrastructure, Severe Combined Immunodeficiency genetics
- Abstract
Artemis (SNM1C/DCLRE1C) is an endonuclease that plays a key role in development of B- and T-lymphocytes and in dsDNA break repair by non-homologous end-joining (NHEJ). Artemis is phosphorylated by DNA-PKcs and acts to open DNA hairpin intermediates generated during V(D)J and class-switch recombination. Artemis deficiency leads to congenital radiosensitive severe acquired immune deficiency (RS-SCID). Artemis belongs to a superfamily of nucleases containing metallo-β-lactamase (MBL) and β-CASP (CPSF-Artemis-SNM1-Pso2) domains. We present crystal structures of the catalytic domain of wildtype and variant forms of Artemis, including one causing RS-SCID Omenn syndrome. The catalytic domain of the Artemis has similar endonuclease activity to the phosphorylated full-length protein. Our structures help explain the predominantly endonucleolytic activity of Artemis, which contrasts with the predominantly exonuclease activity of the closely related SNM1A and SNM1B MBL fold nucleases. The structures reveal a second metal binding site in its β-CASP domain unique to Artemis, which is amenable to inhibition by compounds including ebselen. By combining our structural data with that from a recently reported Artemis structure, we were able model the interaction of Artemis with DNA substrates. The structures, including one of Artemis with the cephalosporin ceftriaxone, will help enable the rational development of selective SNM1 nuclease inhibitors., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
144. Conformational scanning of individual EF-hand motifs of calcium sensor protein centrin-1.
- Author
-
Phanindranath R, Sudhakar DVS, Thangaraj K, and Sharma Y
- Subjects
- Genes, Reporter, Humans, Magnesium metabolism, Protein Unfolding, Tryptophan metabolism, Calcium metabolism, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, EF Hand Motifs
- Abstract
Centrin-1, a Ca
2+ sensor protein of the centrin family is a crucial player for cell division in eukaryotes and plays a key role in the microtubule organising centre. Despite being regarded as a calcium sensor with a matched structure to calmodulin/troponin C, the protein undergoes mild changes in conformation and binds Ca2+ with moderate affinity. We present an in-depth analysis of the Ca2+ sensing by individual EF-hand motifs of centrin-1 and address unsolved questions of the rationales for moderate affinity and conformational transitions of the protein. Employing the more sensitive approach of Trp scanning of individual EF-hand motif, we have undertaken an exhaustive investigation of Ca2+ binding to individual EF-hand motifs, named EF1 to EF4. All four EF-hand motifs of centrin-1 are structural as all of them bind both Ca2+ and Mg2+ . EF1 and EF4 are the most flexible sites as they undergo drastic conformational changes following Ca2+ binding, whereas EF3 responds to Ca2+ minimally. On the other hand, EF2 moves towards the protein surface upon binding Ca2+ . The independent filling mode of Ca2+ to EF-hand motifs and lack of intermotif communication explain the lack of cooperativity of binding, thus constraining centrin-1 to a moderate affinity binding protein. Thus, centrin-1 is distinct from other calcium sensors such as calmodulin., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
145. A Peptidomimetic Ligand Targeting the Chromodomain of MPP8 Reveals HRP2's Association with the HUSH Complex.
- Author
-
Waybright JM, Clinkscales SE, Barnash KD, Budziszewski GR, Rectenwald JM, Chiarella AM, Norris-Drouin JL, Cholensky SH, Pearce KH, Herring LE, McGinty RK, Hathaway NA, and James LI
- Subjects
- Cell Cycle Proteins metabolism, Fluorescence Resonance Energy Transfer, Histones chemistry, Hydrophobic and Hydrophilic Interactions, Intercellular Signaling Peptides and Proteins metabolism, Ligands, Lysine chemistry, Mass Spectrometry, Methylation, Models, Molecular, Peptidomimetics metabolism, Phosphoproteins metabolism, Protein Binding, Protein Domains, Protein Processing, Post-Translational, Proteomics, Structure-Activity Relationship, Cell Cycle Proteins chemistry, Peptidomimetics chemistry, Phosphoproteins chemistry
- Abstract
The interpretation of histone post-translational modifications (PTMs), specifically lysine methylation, by specific classes of "reader" proteins marks an important aspect of epigenetic control of gene expression. Methyl-lysine (Kme) readers often regulate gene expression patterns through the recognition of a specific Kme PTM while participating in or recruiting large protein complexes that contain enzymatic or chromatin remodeling activity. Understanding the composition of these Kme-reader-containing protein complexes can serve to further our understanding of the biological roles of Kme readers, while small molecule chemical tools can be valuable reagents in interrogating novel protein-protein interactions. Here, we describe our efforts to target the chromodomain of M-phase phosphoprotein 8 (MPP8), a member of the human silencing hub (HUSH) complex and a histone 3 lysine 9 trimethyl (H3K9me3) reader that is vital for heterochromatin formation and has specific roles in cancer metastasis. Utilizing a one-bead, one-compound (OBOC) combinatorial screening approach, we identified UNC5246, a peptidomimetic ligand capable of interacting with the MPP8 chromodomain in the context of the HUSH complex. Additionally, a biotinylated derivative of UNC5246 facilitated chemoproteomics studies which revealed hepatoma-derived growth factor-related protein 2 (HRP2) as a novel protein associated with MPP8. HRP2 was further shown to colocalize with MPP8 at the E-cadherin gene locus, suggesting a possible role in cancer cell plasticity.
- Published
- 2021
- Full Text
- View/download PDF
146. Solid-Phase Assembly of Multienzyme Systems into Artificial Cellulosomes.
- Author
-
Zeballos N, Diamanti E, Benítez-Mateos AI, Schmidt-Dannert C, and López-Gallego F
- Subjects
- Transaminases metabolism, Transaminases chemistry, Cohesins, Porosity, Chromosomal Proteins, Non-Histone metabolism, Chromosomal Proteins, Non-Histone chemistry, Cell Cycle Proteins metabolism, Cell Cycle Proteins chemistry, Sepharose chemistry, Amines chemistry, Amines metabolism, Cellulosomes metabolism, Cellulosomes chemistry, Enzymes, Immobilized chemistry, Enzymes, Immobilized metabolism, Alcohol Dehydrogenase metabolism, Alcohol Dehydrogenase chemistry
- Abstract
We herein describe a bioinspired solid-phase assembly of a multienzyme system scaffolded on an artificial cellulosome. An alcohol dehydrogenase and an ω-transaminase were fused to cohesin and dockerin domains to drive their sequential and ordered coimmobilization on agarose porous microbeads. The resulting immobilized scaffolded enzymatic cellulosome was characterized through quartz crystal microbalance with dissipation and confocal laser scanning microscopy to demonstrate that both enzymes interact with each other and physically colocalize within the microbeads. Finally, the assembled multifunctional heterogeneous biocatalyst was tested for the one-pot conversion of alcohols into amines. By using the physically colocalized enzymatic system confined into porous microbeads, the yield of the corresponding amine was 1.3 and 10 times higher than the spatially segregated immobilized system and the free enzymes, respectively. This work establishes the basis of a new concept to organize multienzyme systems at the nanoscale within solid and porous immobilization carriers.
- Published
- 2021
- Full Text
- View/download PDF
147. MAD2L2 dimerization and TRIP13 control shieldin activity in DNA repair.
- Author
-
de Krijger I, Föhr B, Pérez SH, Vincendeau E, Serrat J, Thouin AM, Susvirkar V, Lescale C, Paniagua I, Hoekman L, Kaur S, Altelaar M, Deriano L, Faesen AC, and Jacobs JJL
- Subjects
- ATPases Associated with Diverse Cellular Activities chemistry, ATPases Associated with Diverse Cellular Activities metabolism, Animals, Binding Sites, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Cell Line, Tumor, Cisplatin pharmacology, DNA chemistry, DNA metabolism, DNA Breaks, Double-Stranded, DNA-Binding Proteins chemistry, DNA-Binding Proteins metabolism, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Gene Expression, HEK293 Cells, HeLa Cells, Humans, Mad2 Proteins chemistry, Mad2 Proteins metabolism, Mice, Phthalazines pharmacology, Piperazines pharmacology, Protein Binding, Protein Interaction Domains and Motifs, Protein Multimerization, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, ATPases Associated with Diverse Cellular Activities genetics, Cell Cycle Proteins genetics, DNA genetics, DNA Repair, DNA-Binding Proteins genetics, Mad2 Proteins genetics
- Abstract
MAD2L2 (REV7) plays an important role in DNA double-strand break repair. As a member of the shieldin complex, consisting of MAD2L2, SHLD1, SHLD2 and SHLD3, it controls DNA repair pathway choice by counteracting DNA end-resection. Here we investigated the requirements for shieldin complex assembly and activity. Besides a dimerization-surface, HORMA-domain protein MAD2L2 has the extraordinary ability to wrap its C-terminus around SHLD3, likely creating a very stable complex. We show that appropriate function of MAD2L2 within shieldin requires its dimerization, mediated by SHLD2 and accelerating MAD2L2-SHLD3 interaction. Dimerization-defective MAD2L2 impairs shieldin assembly and fails to promote NHEJ. Moreover, MAD2L2 dimerization, along with the presence of SHLD3, allows shieldin to interact with the TRIP13 ATPase, known to drive topological switches in HORMA-domain proteins. We find that appropriate levels of TRIP13 are important for proper shieldin (dis)assembly and activity in DNA repair. Together our data provide important insights in the dependencies for shieldin activity., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
148. Structural Insight into the Interaction of Sendai Virus C Protein with Alix To Stimulate Viral Budding.
- Author
-
Oda K, Matoba Y, Sugiyama M, and Sakaguchi T
- Subjects
- Amino Acid Sequence, Animals, Binding, Competitive, Cell Line, Crystallography, X-Ray, Humans, Interferon-alpha genetics, Interferon-alpha metabolism, Interferon-beta genetics, Interferon-beta metabolism, Models, Molecular, Protein Binding, Protein Conformation, Protein Domains, Sendai virus chemistry, Sendai virus genetics, Sendai virus metabolism, Signal Transduction, Virion physiology, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins metabolism, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Endosomal Sorting Complexes Required for Transport chemistry, Endosomal Sorting Complexes Required for Transport metabolism, Sendai virus physiology, Viral Proteins chemistry, Viral Proteins metabolism, Virus Release
- Abstract
Sendai virus (SeV), belonging to the Respirovirus genus of the family Paramyxoviridae , harbors an accessory protein, named C protein, which facilitates viral pathogenicity in mice. In addition, the C protein is known to stimulate the budding of virus-like particles by binding to the host ALG-2 interacting protein X (Alix), a component of the endosomal sorting complexes required for transport (ESCRT) machinery. However, small interfering RNA (siRNA)-mediated gene knockdown studies suggested that neither Alix nor C protein is related to SeV budding. In the present study, we determined the crystal structure of a complex comprising the C-terminal half of the C protein (Y3) and the Bro1 domain of Alix at a resolution of 2.2 Å to investigate the role of the complex in SeV budding. The structure revealed that a novel consensus sequence, LXXW, which is conserved among Respirovirus C proteins, is important for Alix binding. SeV possessing a mutated C protein with reduced Alix-binding affinity showed impaired virus production, which correlated with the binding affinity. Infectivity analysis showed a 160-fold reduction at 12 h postinfection compared with nonmutated virus, while C protein competes with CHMP4, one subunit of the ESCRT-III complex, for binding to Alix. All together, these results highlight the critical role of C protein in SeV budding. IMPORTANCE Human parainfluenza virus type I (hPIV1) is a respiratory pathogen affecting young children, immunocompromised patients, and the elderly, with no available vaccines or antiviral drugs. Sendai virus (SeV), a murine counterpart of hPIV1, has been studied extensively to determine the molecular and biological properties of hPIV1. These viruses possess a multifunctional accessory protein, C protein, which is essential for stimulating viral reproduction, but its role in budding remains controversial. In the present study, the crystal structure of the C-terminal half of the SeV C protein associated with the Bro1 domain of Alix, a component of cell membrane modulating machinery ESCRT, was elucidated. Based on the structure, we designed mutant C proteins with different binding affinities to Alix and showed that the interaction between C and Alix is vital for viral budding. These findings provide new insights into the development of new antiviral drugs against hPIV1.
- Published
- 2021
- Full Text
- View/download PDF
149. Development of a Novel Cell Surface Attachment System to Display Multi-Protein Complex Using the Cohesin-Dockerin Binding Pair.
- Author
-
Ko HJ, Song H, and Choi IG
- Subjects
- Amino Acid Sequence, Archaeal Proteins chemistry, Archaeal Proteins genetics, Archaeoglobus fulgidus, Cell Cycle Proteins chemistry, Cell Cycle Proteins genetics, Cell Membrane genetics, Cell Membrane metabolism, Chromosomal Proteins, Non-Histone chemistry, Chromosomal Proteins, Non-Histone genetics, Disaccharidases chemistry, Disaccharidases genetics, Disaccharidases metabolism, Escherichia coli genetics, Escherichia coli metabolism, Luminescent Proteins chemistry, Luminescent Proteins genetics, Luminescent Proteins metabolism, Protein Binding, Protein Folding, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Red Fluorescent Protein, Cohesins, Archaeal Proteins metabolism, Cell Cycle Proteins metabolism, Cell Surface Display Techniques methods, Chromosomal Proteins, Non-Histone metabolism
- Abstract
Autodisplay of a multimeric protein complex on a cell surface is limited by intrinsic factors such as the types and orientations of anchor modules. Moreover, improper folding of proteins to be displayed often hinders functional cell surface display. While overcoming these drawbacks, we ultimately extended the applicability of the autodisplay platform to the display of a protein complex. We designed and constructed a cell surface attachment (CSA) system that uses a noncovalent protein-protein interaction. We employed the high-affinity interaction mediated by an orthogonal cohesin-dockerin (Coh-Doc) pair from Archaeoglobus fulgidus to build the CSA system. Then, we validated the orthogonal Coh-Doc binding by attaching a monomeric red fluorescent protein to the cell surface. In addition, we evaluated the functional anchoring of proteins fused with the Doc module to the autodisplayed Coh module on the surface of Escherichia coli . The designed CSA system was applied to create a functional attachment of dimeric α-neoagarobiose hydrolase to the surface of E. coli cells.
- Published
- 2021
- Full Text
- View/download PDF
150. Studies of Interaction Mechanism between Pyrido [3,4- d ] Pyrimidine Inhibitors and Mps1.
- Author
-
Xing C, Zhou X, Chen C, Sun W, Zheng Q, and Liang D
- Subjects
- Catalytic Domain, Cell Cycle Proteins antagonists & inhibitors, Crystallography, X-Ray, Humans, Molecular Docking Simulation, Molecular Dynamics Simulation, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases chemistry, Protein-Tyrosine Kinases chemistry, Pyrimidines pharmacology, Cell Cycle Proteins chemistry, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases antagonists & inhibitors, Pyrimidines chemistry
- Abstract
Monopolar spindle 1 (Mps1), a dual-specific kinase, is related to the proper execution of chromosome biorientation and mitotic checkpoint signaling. The overexpression of Mps1 promotes the occurrence of cancer or the survival of aneuploid cancer cells, in other words, the reduction of Mps1 will severely reduce the viability of human cancer cells. Therefore, Mps1 is a potential target for cancer treatment. Recently, a series of novel pyrido [3,4- d ] pyrimidine derivatives targeting Mps1 with high biological activity were synthesized. The crystal structure of Mps1 in complex with pyrido [3,4- d ] pyrimidine derivatives was also reported, but there were no specific mechanism studies for this series of small molecule inhibitors. In this study, complexes binding modes were probed by molecular docking and further validated by molecular dynamics simulations and the molecular mechanics/generalized Born surface area (MM/GBSA) method. The results indicated that the van der Waals interactions and the nonpolar solvation energies were responsible to the basis for favorable binding free energies, all inhibitors interacted with residues I531, V539, M602, C604, N606, I607, L654, I663, and P673 of Mps1. By analyzing the hydrogen bonds, we found the residues G605 and K529 in Mps1 formed stable hydrogen bonds with compounds, it was more conducive to activities of Mps1 inhibitors. According to the above analysis, we further designed five new compounds. We found that compounds IV and V were better potential Mps1 inhibitors through docking and ADMET prediction. The obtained new insights not only were helpful in understanding the binding mode of inhibitors in Mps1, but also provided important references for further rational design of Mps1 inhibitors.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.