89 results on '"Jedrzejczak R"'
Search Results
2. Crystal Structure of CelD Cellulase from the Anaerobic Fungus Piromyces finnis
- Author
-
Dementieve, A., primary, Kim, Y., additional, Jedrzejczak, R., additional, Michalska, K., additional, and Joachimiak, A., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Methylation of RNA Cap in SARS-CoV-2 captured by serial crystallography
- Author
-
Wilamowski, M., primary, Sherrell, D.A., additional, Minasov, G., additional, Kim, Y., additional, Shuvalova, L., additional, Lavens, A., additional, Chard, R., additional, Maltseva, N., additional, Jedrzejczak, R., additional, Rosas-Lemus, M., additional, Saint, N., additional, Foster, I.T., additional, Michalska, K., additional, Satchell, K.J.F., additional, and Joachimiak, A, additional
- Published
- 2020
- Full Text
- View/download PDF
4. Crystal Structure of the Lipase Lip_vut5 from Goat Rumen metagenome.
- Author
-
Kim, Y., primary, Welk, L., additional, Mukendi, G., additional, Nkhi, G., additional, Motloi, T., additional, Jedrzejczak, R., additional, Feto, N., additional, and Joachimiak, A., additional
- Published
- 2020
- Full Text
- View/download PDF
5. Crystal structure of NDM-1 metallo-beta-lactamase in complex with Cd ions and a hydrolyzed beta-lactam ligand - new refinement
- Author
-
Kim, Y., primary, Raczynska, J.E., additional, Shabalin, I.G., additional, Jaskolski, M., additional, Minor, W., additional, Wlodawer, A., additional, Tesar, C., additional, Jedrzejczak, R., additional, Babnigg, J., additional, Mire, J., additional, Sacchettini, J., additional, and Joachimiak, A., additional
- Published
- 2017
- Full Text
- View/download PDF
6. Developments in the chemical analysis of environmental samples at the edge of the 21st century International Symposium held at Warsaw, Poland on 21–22 November, 1996
- Author
-
Dobkowski, Z. and Jedrzejczak, R.
- Published
- 1997
- Full Text
- View/download PDF
7. The Crystal structure of the N-terminal domain of a novel cellulases from Bacteroides coprocola
- Author
-
Tan, K., primary, Gu, M., additional, Jedrzejczak, R., additional, and Joachimiak, A., additional
- Published
- 2016
- Full Text
- View/download PDF
8. Crystal Structure of Metallothiol Transferase FosB 2 from Bacillus anthracis str. Ames
- Author
-
Maltseva, N., primary, Kim, Y., additional, Jedrzejczak, R., additional, Sharma, S.V., additional, Hamilton, C.J., additional, Anderson, W.F., additional, and Joachimiak, A., additional
- Published
- 2013
- Full Text
- View/download PDF
9. Crystal Structure of Histidine-containing Phosphotransfer Protein MtHPt1 from Medicago truncatula
- Author
-
Ruszkowski, M., primary, Brzezinski, K., additional, Jedrzejczak, R., additional, Dauter, M., additional, Dauter, Z., additional, Sikorski, M., additional, and Jaskolski, M., additional
- Published
- 2012
- Full Text
- View/download PDF
10. Human Mitochondrial Helicase Suv3
- Author
-
Dauter, Z., primary, Jedrzejczak, R., additional, Dauter, M., additional, Szczesny, R., additional, and Stepien, P., additional
- Published
- 2011
- Full Text
- View/download PDF
11. Human Mitochondrial Helicase Suv3 in Complex with Short RNA Fragment
- Author
-
Dauter, Z., primary, Jedrzejczak, R., additional, Dauter, M., additional, Wang, J., additional, Szczesny, R., additional, and Stepien, P., additional
- Published
- 2011
- Full Text
- View/download PDF
12. Structural Studies of ROK Fructokinase YdhR from Bacillus subtilis: Insights into Substrate Binding and Fructose Specificity
- Author
-
Nocek, B., primary, Stein, A.J., additional, Jedrzejczak, R., additional, Cuff, M.E., additional, Li, H., additional, Volkart, L., additional, and Joachimiak, A., additional
- Published
- 2011
- Full Text
- View/download PDF
13. Determination of total mercury in foods of plant origin in Poland by cold vapour atomic absorption spectrometry
- Author
-
Jedrzejczak, R., primary
- Published
- 2002
- Full Text
- View/download PDF
14. Developments in the chemical analysis of environmental samples at the edge of the 21 st century International Symposium held at Warsaw, Poland on 21-22 November, 1996
- Author
-
Dobkowski, Z., primary and Jedrzejczak, R., additional
- Published
- 1997
- Full Text
- View/download PDF
15. Oligomeric structure and regulation of Candida albicans glucosamine-6-phosphate synthase.
- Author
-
Milewski, S, Kuszczak, D, Jedrzejczak, R, Smith, R J, Brown, A J, and Gooday, G W
- Abstract
Candida albicans glucosamine-6-phosphate (GlcN-6-P) synthase was purified to apparent homogeneity with 52% yield from recombinant yeast YRSC-65 cells efficiently overexpressing the GFA1 gene. The pure enzyme exhibited Km(Gln) = 1.56 mM and Km(Fru-6-P) = 1.41 mM and catalyzed GlcN-6-P formation with kcat = 1150 min-1. The isoelectric point of 4.6 +/- 0.05 was estimated from isoelectric chromatofocusing. Gel filtration, native polyacrylamide gel electrophoresis, subunit cross-linking, and SDS-polyacrylamide gel electrophoresis showed that the native enzyme was a homotetramer of 79.5-kDa subunits, with an apparent molecular mass of 330-340 kDa. Results of chemical modification of the enzyme by group-specific reagents established an essential role of a cysteinyl residue at the glutamine-binding site and histidyl, lysyl, arginyl, and tyrosyl moieties at the Fru-6-P-binding site. GlcN-6-P synthase in crude extract was effectively inhibited by UDP-GlcNAc (IC50 = 0.67 mM). Purification of the enzyme markedly decreased the sensitivity to the inhibitor, but this could be restored by addition of another effector, glucose 6-phosphate. Binding of UDP-GlcNAc to the pure enzyme in the presence of Glc-6-P showed strong negative cooperativity, with nH = 0.54, whereas in the absence of this sugar phosphate no cooperative effect was observed. Pure enzyme was a substrate for cAMP-dependent protein kinase, the action of which led to the substantial increase of GlcN-6-P synthase activity, correlated with an extent of protein phosphorylation. The maximal level of activity was observed for the enzyme molecules containing 1. 21 +/- 0.08 mol of phosphate/mol of GlcN-6-P synthase. Monitoring of GlcN-6-P synthase activity and its sensitivity to UDP-GlcNAc during yeast --> mycelia transformation of C. albicans cells, under in situ conditions, revealed a marked increase of the former and a substantial fall of the latter.
- Published
- 1999
16. Evolution of a mine information system at the Endako mine.
- Author
-
Jedrzejczak R., McDowell M., Jedrzejczak R., and McDowell M.
- Abstract
Endako is an open pit molybdenum mine located in the north central region of British Columbia. The plant site includes crushing circuits, flotation circuits and roasting and refining plants. The existing and developing mine planning and engineering systems at Endako are discussed. The existing computer system and software operate well and give good results within individual departments. However, the system is not flexible enough for the whole operation. The new system allows data entry on an operation-wide basis and can be accessed by the operators from any level in the system. Information flow, record keeping and maintenance will be improved., Endako is an open pit molybdenum mine located in the north central region of British Columbia. The plant site includes crushing circuits, flotation circuits and roasting and refining plants. The existing and developing mine planning and engineering systems at Endako are discussed. The existing computer system and software operate well and give good results within individual departments. However, the system is not flexible enough for the whole operation. The new system allows data entry on an operation-wide basis and can be accessed by the operators from any level in the system. Information flow, record keeping and maintenance will be improved.
17. Developments in the chemical analysis of environmental samples at the edge of the 21st century International Symposium held at Warsaw, Poland on 21–22 November, 1996.
- Author
-
Dobkowski, Z. and Jedrzejczak, R.
- Published
- 1997
- Full Text
- View/download PDF
18. Total content of mercury, lead and cadmium in common edible fish and mussels from the Adriatic sea
- Author
-
Jureša, Dijana, Blanuša, Maja, Szteke, B., Kabata-Pendias, A., and Jedrzejczak, R.
- Subjects
human exposure ,mercury ,cadmium ,lead ,fish ,mussels ,atomic absorption spectrometry ,certified values ,provisional tolerable weekly intake - Abstract
Except for the occupationally exposed individuals consumption of fish is the main source of human exposure to mercury. Also exposure to lead and cadmium through fish is significant especially for the population living near the coast or on the islands. The aim of this study was to determine the total mercury, lead and cadmium concentrations of the common edible species of fresh fish and mussel from the different parts of the Adriatic Sea. Only muscular fish tissue, free of bones and total mussel tissue was taken for analysis. Total mercury concentration was measured by cold vapour atomic absorption spectrometry (Mercury Monitor LDC, UK) after digestion of sample by heating in concentrated nitric acid at 80 C, using closed glass tubes. Lead and cadmium were determined by electrothermal atomic absorption spectrometry (Varian model SpectrAA-300, Austria) after dry ashing of sample at 450 C. Deuterium lamp as background correction was used. Detection limits of analytical methods were 0.33 ng Hg ml-1, 0.47 ng Pb ml-1 and 0.07 ng Cd ml-1 for analyte solution or based on a sample wet weight: 3.3 ng Hg g-1, 0.94 ng Pb g-1 and 0.14 ng Cd g-1. Precision of the methods within a day, calculated as coefficients of variation of 10 analysis of the same fish sample were 17%, 38% and 23% for mercury, lead and cadmium, respectively. Methods accuracy was tested by the parallel analysis of a dogfish muscle reference material (DORM-2 from the National Research Council of Canada). The results were in good agreement with the certified values. The ranges of mercury, lead and cadmium concentrations in fish were 142-252 ng g-1, 9.6-44.1 ng g-1 and 5.1-49.1 ng g-1, respectively and mean metal concentrations in mussel were 134 ng Hg g-1, 121 ng Pb g-1 and 130 ng Cd g-1. The levels of analysed elements in fish and mussel did not exceed maximum permissible levels of mercury, lead and cadmium in fish and mussel set by the Croatian Ministry of Health. The moderate intake of fish and mussel does not exceed Provisional Tolerable Weekly Intake set by the Joint FAO/WHO Expert Committee on Food Additives.
- Published
- 2000
19. Epitopes recognition of SARS-CoV-2 nucleocapsid RNA binding domain by human monoclonal antibodies.
- Author
-
Kim Y, Maltseva N, Tesar C, Jedrzejczak R, Endres M, Ma H, Dugan HL, Stamper CT, Chang C, Li L, Changrob S, Zheng NY, Huang M, Ramanathan A, Wilson P, Michalska K, and Joachimiak A
- Abstract
Coronavirus nucleocapsid protein (NP) of SARS-CoV-2 plays a central role in many functions important for virus proliferation including packaging and protecting genomic RNA. The protein shares sequence, structure, and architecture with nucleocapsid proteins from betacoronaviruses. The N-terminal domain (NP
RBD ) binds RNA and the C-terminal domain is responsible for dimerization. After infection, NP is highly expressed and triggers robust host immune response. The anti-NP antibodies are not protective and not neutralizing but can effectively detect viral proliferation soon after infection. Two structures of SARS-CoV-2 NPRBD were determined providing a continuous model from residue 48 to 173, including RNA binding region and key epitopes. Five structures of NPRBD complexes with human mAbs were isolated using an antigen-bait sorting. Complexes revealed a distinct complement-determining regions and unique sets of epitope recognition. This may assist in the early detection of pathogens and designing peptide-based vaccines. Mutations that significantly increase viral load were mapped on developed, full length NP model, likely impacting interactions with host proteins and viral RNA., Competing Interests: The authors declare no competing interests., (© 2024 The Authors.)- Published
- 2024
- Full Text
- View/download PDF
20. Structure-based design of SARS-CoV-2 papain-like protease inhibitors.
- Author
-
Jadhav P, Huang B, Osipiuk J, Zhang X, Tan H, Tesar C, Endres M, Jedrzejczak R, Tan B, Deng X, Joachimiak A, Cai J, and Wang J
- Subjects
- Humans, SARS-CoV-2 metabolism, Pandemics, Antiviral Agents pharmacology, Antiviral Agents chemistry, Protease Inhibitors pharmacology, Protease Inhibitors chemistry, Papain chemistry, Papain genetics, Papain metabolism, COVID-19
- Abstract
The COVID-19 pandemic is caused by SARS-CoV-2, an RNA virus with high transmissibility and mutation rate. Given the paucity of orally bioavailable antiviral drugs to combat SARS-CoV-2 infection, there is a critical need for additional antivirals with alternative mechanisms of action. Papain-like protease (PL
pro ) is one of the two SARS-CoV-2 encoded viral cysteine proteases essential for viral replication. PLpro cleaves at three sites of the viral polyproteins. In addition, PLpro antagonizes the host immune response upon viral infection by cleaving ISG15 and ubiquitin from host proteins. Therefore, PLpro is a validated antiviral drug target. In this study, we report the X-ray crystal structures of papain-like protease (PLpro ) with two potent inhibitors, Jun9722 and Jun9843. Subsequently, we designed and synthesized several series of analogs to explore the structure-activity relationship, which led to the discovery of PLpro inhibitors with potent enzymatic inhibitory activity and antiviral activity against SARS-CoV-2. Together, the lead compounds are promising drug candidates for further development., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Jun Wang reports financial support was provided by National Institute of Allergy and Infectious Diseases. Jun Wang has patent #WO2022192665A1 pending to University of Arizona., (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
21. Structure and enzymatic characterization of CelD endoglucanase from the anaerobic fungus Piromyces finnis.
- Author
-
Dementiev A, Lillington SP, Jin S, Kim Y, Jedrzejczak R, Michalska K, Joachimiak A, and O'Malley MA
- Subjects
- Anaerobiosis, Glucans metabolism, Cellulase metabolism, Piromyces metabolism
- Abstract
Anaerobic fungi found in the guts of large herbivores are prolific biomass degraders whose genomes harbor a wealth of carbohydrate-active enzymes (CAZymes), of which only a handful are structurally or biochemically characterized. Here, we report the structure and kinetic rate parameters for a glycoside hydrolase (GH) family 5 subfamily 4 enzyme (CelD) from Piromyces finnis, a modular, cellulosome-incorporated endoglucanase that possesses three GH5 domains followed by two C-terminal fungal dockerin domains (double dockerin). We present the crystal structures of an apo wild-type CelD GH5 catalytic domain and its inactive E154A mutant in complex with cellotriose at 2.5 and 1.8 Å resolution, respectively, finding the CelD GH5 catalytic domain adopts the (β/α)
8 -barrel fold common to many GH5 enzymes. Structural superimposition of the apo wild-type structure with the E154A mutant-cellotriose complex supports a catalytic mechanism in which the E154 carboxylate side chain acts as an acid/base and E278 acts as a complementary nucleophile. Further analysis of the cellotriose binding pocket highlights a binding groove lined with conserved aromatic amino acids that when docked with larger cellulose oligomers is capable of binding seven glucose units and accommodating branched glucan substrates. Activity analyses confirm P. finnis CelD can hydrolyze mixed linkage glucan and xyloglucan, as well as carboxymethylcellulose (CMC). Measured kinetic parameters show the P. finnis CelD GH5 catalytic domain has CMC endoglucanase activity comparable to other fungal endoglucanases with kcat = 6.0 ± 0.6 s-1 and Km = 7.6 ± 2.1 g/L CMC. Enzyme kinetics were unperturbed by the addition or removal of the native C-terminal dockerin domains as well as the addition of a non-native N-terminal dockerin, suggesting strict modularity among the domains of CelD. KEY POINTS: • Anaerobic fungi host a wealth of industrially useful enzymes but are understudied. • P. finnis CelD has endoglucanase activity and structure common to GH5_4 enzymes. • CelD's kinetics do not change with domain fusion, exhibiting high modularity., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
22. Dual domain recognition determines SARS-CoV-2 PLpro selectivity for human ISG15 and K48-linked di-ubiquitin.
- Author
-
Wydorski PM, Osipiuk J, Lanham BT, Tesar C, Endres M, Engle E, Jedrzejczak R, Mullapudi V, Michalska K, Fidelis K, Fushman D, Joachimiak A, and Joachimiak LA
- Subjects
- Humans, Cytokines metabolism, Papain metabolism, Peptide Hydrolases metabolism, Ubiquitins metabolism, COVID-19, SARS-CoV-2 metabolism, Ubiquitin metabolism
- Abstract
The Papain-like protease (PLpro) is a domain of a multi-functional, non-structural protein 3 of coronaviruses. PLpro cleaves viral polyproteins and posttranslational conjugates with poly-ubiquitin and protective ISG15, composed of two ubiquitin-like (UBL) domains. Across coronaviruses, PLpro showed divergent selectivity for recognition and cleavage of posttranslational conjugates despite sequence conservation. We show that SARS-CoV-2 PLpro binds human ISG15 and K48-linked di-ubiquitin (K48-Ub
2 ) with nanomolar affinity and detect alternate weaker-binding modes. Crystal structures of untethered PLpro complexes with ISG15 and K48-Ub2 combined with solution NMR and cross-linking mass spectrometry revealed how the two domains of ISG15 or K48-Ub2 are differently utilized in interactions with PLpro. Analysis of protein interface energetics predicted differential binding stabilities of the two UBL/Ub domains that were validated experimentally. We emphasize how substrate recognition can be tuned to cleave specifically ISG15 or K48-Ub2 modifications while retaining capacity to cleave mono-Ub conjugates. These results highlight alternative druggable surfaces that would inhibit PLpro function., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
23. A Genomic Island of Vibrio cholerae Encodes a Three-Component Cytotoxin with Monomer and Protomer Forms Structurally Similar to Alpha-Pore-Forming Toxins.
- Author
-
Herrera A, Kim Y, Chen J, Jedrzejczak R, Shukla S, Maltseva N, Joachimiak G, Welk L, Wiersum G, Jaroszewski L, Godzik A, Joachimiak A, and Satchell KJF
- Subjects
- Animals, Cytotoxins genetics, Cytotoxins metabolism, Escherichia coli genetics, Escherichia coli metabolism, Genomic Islands, Mice, Pore Forming Cytotoxic Proteins, Protein Subunits metabolism, Virulence Factors metabolism, Vibrio cholerae metabolism
- Abstract
Alpha-pore-forming toxins (α-PFTs) are secreted by many species of bacteria, including Escherichia coli, Aeromonas hydrophila, and Bacillus thuringiensis, as part of their arsenal of virulence factors, and are often cytotoxic. In particular, for α-PFTs, the membrane-spanning channel they form is composed of hydrophobic α-helices. These toxins oligomerize at the surface of target cells and transition from a soluble to a protomer state in which they expose their hydrophobic regions and insert into the membrane to form a pore. The pores may be composed of homooligomers of one component or heterooligomers with two or three components, resulting in bi- or tripartite toxins. The multicomponent α-PFTs are often expressed from a single operon. Recently, motility-associated killing factor A (MakA), an α-PFT, was discovered in Vibrio cholerae. We report that makA is found on the V. cholerae GI-10 genomic island within an operon containing genes for two other potential α-PFTs, MakB and MakE. We determined the X-ray crystal structures for MakA, MakB, and MakE and demonstrated that all three are structurally related to the α-PFT family in the soluble state, and we modeled their protomer state based on the α-PFT AhlB from A. hydrophila. We found that MakA alone is cytotoxic at micromolar concentrations. However, combining MakA with MakB and MakE is cytotoxic at nanomolar concentrations, with specificity for J774 macrophage cells. Our data suggest that MakA, -B, and -E are α-PFTs that potentially act as a tripartite pore-forming toxin with specificity for phagocytic cells. IMPORTANCE The bacterium Vibrio cholerae causes gastrointestinal, wound, and skin infections. The motility-associated killing factor A (MakA) was recently shown to be cytotoxic against colon, prostate, and other cancer cells. However, at the outset of this study, the capacity of MakA to damage cells in combination with other Mak proteins encoded in the same operon had not been elucidated. We determined the structures of three Mak proteins and established that they are structurally related to the α-PFTs. Compared to MakA alone, the combination of all three toxins was more potent specifically in mouse macrophages. This study highlights the idea that the Mak toxins are selectively cytotoxic and thus may function as a tripartite toxin with cell type specificity.
- Published
- 2022
- Full Text
- View/download PDF
24. Catalytically impaired TrpA subunit of tryptophan synthase from Chlamydia trachomatis is an allosteric regulator of TrpB.
- Author
-
Michalska K, Wellington S, Maltseva N, Jedrzejczak R, Selem-Mojica N, Rosas-Becerra LR, Barona-Gómez F, Hung DT, and Joachimiak A
- Subjects
- Allosteric Regulation, Amino Acid Sequence, Bacterial Proteins genetics, Bacterial Proteins metabolism, Binding Sites, Biocatalysis, Catalytic Domain, Chlamydia trachomatis chemistry, Cloning, Molecular, Crystallography, X-Ray, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, Kinetics, Models, Molecular, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Protein Subunits genetics, Protein Subunits metabolism, Pyridoxal Phosphate metabolism, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Alignment, Sequence Homology, Amino Acid, Substrate Specificity, Tryptophan biosynthesis, Tryptophan Synthase genetics, Tryptophan Synthase metabolism, Bacterial Proteins chemistry, Chlamydia trachomatis enzymology, Protein Subunits chemistry, Pyridoxal Phosphate chemistry, Tryptophan chemistry, Tryptophan Synthase chemistry
- Abstract
Intracellular growth and pathogenesis of Chlamydia species is controlled by the availability of tryptophan, yet the complete biosynthetic pathway for l-Trp is absent among members of the genus. Some representatives, however, preserve genes encoding tryptophan synthase, TrpAB - a bifunctional enzyme catalyzing the last two steps in l-Trp synthesis. TrpA (subunit α) converts indole-3-glycerol phosphate into indole and glyceraldehyde-3-phosphate (α reaction). The former compound is subsequently used by TrpB (subunit β) to produce l-Trp in the presence of l-Ser and a pyridoxal 5'-phosphate cofactor (β reaction). Previous studies have indicated that in Chlamydia, TrpA has lost its catalytic activity yet remains associated with TrpB to support the β reaction. Here, we provide detailed analysis of the TrpAB from C. trachomatis D/UW-3/CX, confirming that accumulation of mutations in the active site of TrpA renders it enzymatically inactive, despite the conservation of the catalytic residues. We also show that TrpA remains a functional component of the TrpAB complex, increasing the activity of TrpB by four-fold. The side chain of non-conserved βArg267 functions as cation effector, potentially rendering the enzyme less susceptible to the solvent ion composition. The observed structural and functional changes detected herein were placed in a broader evolutionary and genomic context, allowing identification of these mutations in relation to their trp gene contexts in which they occur. Moreover, in agreement with the in vitro data, partial relaxation of purifying selection for TrpA, but not for TrpB, was detected, reinforcing a partial loss of TrpA functions during the course of evolution., (© 2021 The Authors. Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)
- Published
- 2021
- Full Text
- View/download PDF
25. Masitinib is a broad coronavirus 3CL inhibitor that blocks replication of SARS-CoV-2.
- Author
-
Drayman N, DeMarco JK, Jones KA, Azizi SA, Froggatt HM, Tan K, Maltseva NI, Chen S, Nicolaescu V, Dvorkin S, Furlong K, Kathayat RS, Firpo MR, Mastrodomenico V, Bruce EA, Schmidt MM, Jedrzejczak R, Muñoz-Alía MÁ, Schuster B, Nair V, Han KY, O'Brien A, Tomatsidou A, Meyer B, Vignuzzi M, Missiakas D, Botten JW, Brooke CB, Lee H, Baker SC, Mounce BC, Heaton NS, Severson WE, Palmer KE, Dickinson BC, Joachimiak A, Randall G, and Tay S
- Subjects
- A549 Cells, Animals, Antiviral Agents chemistry, Antiviral Agents metabolism, Antiviral Agents therapeutic use, Benzamides, COVID-19 virology, Catalytic Domain, Coronavirus 3C Proteases chemistry, Coronavirus 3C Proteases metabolism, Coronavirus OC43, Human physiology, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors metabolism, HEK293 Cells, Humans, Inhibitory Concentration 50, Mice, Mice, Transgenic, Microbial Sensitivity Tests, Piperidines, Pyridines, SARS-CoV-2 enzymology, SARS-CoV-2 physiology, Thiazoles chemistry, Thiazoles metabolism, Thiazoles therapeutic use, Viral Load drug effects, Virus Replication drug effects, Antiviral Agents pharmacology, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus OC43, Human drug effects, Cysteine Proteinase Inhibitors pharmacology, SARS-CoV-2 drug effects, Thiazoles pharmacology, COVID-19 Drug Treatment
- Abstract
There is an urgent need for antiviral agents that treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We screened a library of 1900 clinically safe drugs against OC43, a human beta coronavirus that causes the common cold, and evaluated the top hits against SARS-CoV-2. Twenty drugs significantly inhibited replication of both viruses in cultured human cells. Eight of these drugs inhibited the activity of the SARS-CoV-2 main protease, 3CLpro, with the most potent being masitinib, an orally bioavailable tyrosine kinase inhibitor. X-ray crystallography and biochemistry show that masitinib acts as a competitive inhibitor of 3CLpro. Mice infected with SARS-CoV-2 and then treated with masitinib showed >200-fold reduction in viral titers in the lungs and nose, as well as reduced lung inflammation. Masitinib was also effective in vitro against all tested variants of concern (B.1.1.7, B.1.351, and P.1)., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
26. Transient and stabilized complexes of Nsp7, Nsp8, and Nsp12 in SARS-CoV-2 replication.
- Author
-
Wilamowski M, Hammel M, Leite W, Zhang Q, Kim Y, Weiss KL, Jedrzejczak R, Rosenberg DJ, Fan Y, Wower J, Bierma JC, Sarker AH, Tsutakawa SE, Pingali SV, O'Neill HM, Joachimiak A, and Hura GL
- Subjects
- Humans, Models, Molecular, RNA, Viral genetics, Scattering, Small Angle, Viral Nonstructural Proteins, Virus Replication, X-Ray Diffraction, COVID-19, SARS-CoV-2
- Abstract
The replication transcription complex (RTC) from the virus SARS-CoV-2 is responsible for recognizing and processing RNA for two principal purposes. The RTC copies viral RNA for propagation into new virus and for ribosomal transcription of viral proteins. To accomplish these activities, the RTC mechanism must also conform to a large number of imperatives, including RNA over DNA base recognition, basepairing, distinguishing viral and host RNA, production of mRNA that conforms to host ribosome conventions, interfacing with error checking machinery, and evading host immune responses. In addition, the RTC will discontinuously transcribe specific sections of viral RNA to amplify certain proteins over others. Central to SARS-CoV-2 viability, the RTC is therefore dynamic and sophisticated. We have conducted a systematic structural investigation of three components that make up the RTC: Nsp7, Nsp8, and Nsp12 (also known as RNA-dependent RNA polymerase). We have solved high-resolution crystal structures of the Nsp7/8 complex, providing insight into the interaction between the proteins. We have used small-angle x-ray and neutron solution scattering (SAXS and SANS) on each component individually as pairs and higher-order complexes and with and without RNA. Using size exclusion chromatography and multiangle light scattering-coupled SAXS, we defined which combination of components forms transient or stable complexes. We used contrast-matching to mask specific complex-forming components to test whether components change conformation upon complexation. Altogether, we find that individual Nsp7, Nsp8, and Nsp12 structures vary based on whether other proteins in their complex are present. Combining our crystal structure, atomic coordinates reported elsewhere, SAXS, SANS, and other biophysical techniques, we provide greater insight into the RTC assembly, mechanism, and potential avenues for disruption of the complex and its functions., (Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
27. 2'-O methylation of RNA cap in SARS-CoV-2 captured by serial crystallography.
- Author
-
Wilamowski M, Sherrell DA, Minasov G, Kim Y, Shuvalova L, Lavens A, Chard R, Maltseva N, Jedrzejczak R, Rosas-Lemus M, Saint N, Foster IT, Michalska K, Satchell KJF, and Joachimiak A
- Subjects
- Crystallography, Methylation, Methyltransferases chemistry, Methyltransferases metabolism, Multiprotein Complexes chemistry, Multiprotein Complexes metabolism, RNA Cap Analogs chemistry, RNA Cap Analogs metabolism, RNA Caps chemistry, RNA, Messenger chemistry, RNA, Viral chemistry, S-Adenosylhomocysteine chemistry, S-Adenosylhomocysteine metabolism, S-Adenosylmethionine chemistry, S-Adenosylmethionine metabolism, SARS-CoV-2 genetics, SARS-CoV-2 metabolism, Synchrotrons, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins metabolism, Viral Regulatory and Accessory Proteins chemistry, Viral Regulatory and Accessory Proteins metabolism, RNA Caps metabolism, RNA, Messenger metabolism, RNA, Viral metabolism, SARS-CoV-2 chemistry
- Abstract
The genome of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronavirus has a capping modification at the 5'-untranslated region (UTR) to prevent its degradation by host nucleases. These modifications are performed by the Nsp10/14 and Nsp10/16 heterodimers using S-adenosylmethionine as the methyl donor. Nsp10/16 heterodimer is responsible for the methylation at the ribose 2'-O position of the first nucleotide. To investigate the conformational changes of the complex during 2'-O methyltransferase activity, we used a fixed-target serial synchrotron crystallography method at room temperature. We determined crystal structures of Nsp10/16 with substrates and products that revealed the states before and after methylation, occurring within the crystals during the experiments. Here we report the crystal structure of Nsp10/16 in complex with Cap-1 analog (
m7 GpppAm2'-O ). Inhibition of Nsp16 activity may reduce viral proliferation, making this protein an attractive drug target., Competing Interests: The authors declare no competing interest., (Copyright © 2021 the Author(s). Published by PNAS.)- Published
- 2021
- Full Text
- View/download PDF
28. Mycobacterium tuberculosis Phe-tRNA synthetase: structural insights into tRNA recognition and aminoacylation.
- Author
-
Michalska K, Jedrzejczak R, Wower J, Chang C, Baragaña B, Gilbert IH, Forte B, and Joachimiak A
- Subjects
- Adenosine analogs & derivatives, Adenosine chemistry, Adenosine metabolism, Humans, Ligands, Models, Molecular, Mycobacterium tuberculosis genetics, Phenylalanine analogs & derivatives, Phenylalanine chemistry, Phenylalanine metabolism, Phenylalanine-tRNA Ligase metabolism, Protein Binding, RNA, Transfer, Phe metabolism, Thermus thermophilus enzymology, Mycobacterium tuberculosis enzymology, Phenylalanine-tRNA Ligase chemistry, RNA, Transfer, Phe chemistry, Transfer RNA Aminoacylation
- Abstract
Tuberculosis, caused by Mycobacterium tuberculosis, responsible for ∼1.5 million fatalities in 2018, is the deadliest infectious disease. Global spread of multidrug resistant strains is a public health threat, requiring new treatments. Aminoacyl-tRNA synthetases are plausible candidates as potential drug targets, because they play an essential role in translating the DNA code into protein sequence by attaching a specific amino acid to their cognate tRNAs. We report structures of M. tuberculosis Phe-tRNA synthetase complexed with an unmodified tRNAPhe transcript and either L-Phe or a nonhydrolyzable phenylalanine adenylate analog. High-resolution models reveal details of two modes of tRNA interaction with the enzyme: an initial recognition via indirect readout of anticodon stem-loop and aminoacylation ready state involving interactions of the 3' end of tRNAPhe with the adenylate site. For the first time, we observe the protein gate controlling access to the active site and detailed geometry of the acyl donor and tRNA acceptor consistent with accepted mechanism. We biochemically validated the inhibitory potency of the adenylate analog and provide the most complete view of the Phe-tRNA synthetase/tRNAPhe system to date. The presented topography of amino adenylate-binding and editing sites at different stages of tRNA binding to the enzyme provide insights for the rational design of anti-tuberculosis drugs., (© The Author(s) 2021. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
29. Tipiracil binds to uridine site and inhibits Nsp15 endoribonuclease NendoU from SARS-CoV-2.
- Author
-
Kim Y, Wower J, Maltseva N, Chang C, Jedrzejczak R, Wilamowski M, Kang S, Nicolaescu V, Randall G, Michalska K, and Joachimiak A
- Subjects
- A549 Cells, Antiviral Agents chemistry, Antiviral Agents pharmacokinetics, Catalytic Domain, Crystallography, X-Ray, Endoribonucleases chemistry, Endoribonucleases metabolism, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacokinetics, Humans, Ligands, Models, Molecular, Protein Conformation, Pyrrolidines chemistry, Pyrrolidines pharmacokinetics, Thymine chemistry, Thymine pharmacokinetics, Uridine metabolism, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins metabolism, Antiviral Agents pharmacology, COVID-19 virology, Endoribonucleases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Pyrrolidines pharmacology, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, Thymine pharmacology, Viral Nonstructural Proteins antagonists & inhibitors, COVID-19 Drug Treatment
- Abstract
SARS-CoV-2 Nsp15 is a uridine-specific endoribonuclease with C-terminal catalytic domain belonging to the EndoU family that is highly conserved in coronaviruses. As endoribonuclease activity seems to be responsible for the interference with the innate immune response, Nsp15 emerges as an attractive target for therapeutic intervention. Here we report the first structures with bound nucleotides and show how the enzyme specifically recognizes uridine moiety. In addition to a uridine site we present evidence for a second base binding site that can accommodate any base. The structure with a transition state analog, uridine vanadate, confirms interactions key to catalytic mechanisms. In the presence of manganese ions, the enzyme cleaves unpaired RNAs. This acquired knowledge was instrumental in identifying Tipiracil, an FDA approved drug that is used in the treatment of colorectal cancer, as a potential anti-COVID-19 drug. Using crystallography, biochemical, and whole-cell assays, we demonstrate that Tipiracil inhibits SARS-CoV-2 Nsp15 by interacting with the uridine binding pocket in the enzyme's active site. Our findings provide new insights for the development of uracil scaffold-based drugs.
- Published
- 2021
- Full Text
- View/download PDF
30. Structure of papain-like protease from SARS-CoV-2 and its complexes with non-covalent inhibitors.
- Author
-
Osipiuk J, Azizi SA, Dvorkin S, Endres M, Jedrzejczak R, Jones KA, Kang S, Kathayat RS, Kim Y, Lisnyak VG, Maki SL, Nicolaescu V, Taylor CA, Tesar C, Zhang YA, Zhou Z, Randall G, Michalska K, Snyder SA, Dickinson BC, and Joachimiak A
- Subjects
- Antiviral Agents pharmacology, Humans, Mutation, Polyproteins metabolism, Substrate Specificity, Virus Replication drug effects, Papain metabolism, Peptide Hydrolases metabolism, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology
- Abstract
The pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to expand. Papain-like protease (PLpro) is one of two SARS-CoV-2 proteases potentially targetable with antivirals. PLpro is an attractive target because it plays an essential role in cleavage and maturation of viral polyproteins, assembly of the replicase-transcriptase complex, and disruption of host responses. We report a substantive body of structural, biochemical, and virus replication studies that identify several inhibitors of the SARS-CoV-2 enzyme. We determined the high resolution structure of wild-type PLpro, the active site C111S mutant, and their complexes with inhibitors. This collection of structures details inhibitors recognition and interactions providing fundamental molecular and mechanistic insight into PLpro. All compounds inhibit the peptidase activity of PLpro in vitro, some block SARS-CoV-2 replication in cell culture assays. These findings will accelerate structure-based drug design efforts targeting PLpro to identify high-affinity inhibitors of clinical value.
- Published
- 2021
- Full Text
- View/download PDF
31. Drug repurposing screen identifies masitinib as a 3CLpro inhibitor that blocks replication of SARS-CoV-2 in vitro .
- Author
-
Drayman N, Jones KA, Azizi SA, Froggatt HM, Tan K, Maltseva NI, Chen S, Nicolaescu V, Dvorkin S, Furlong K, Kathayat RS, Firpo MR, Mastrodomenico V, Bruce EA, Schmidt MM, Jedrzejczak R, Muñoz-Alía MÁ, Schuster B, Nair V, Botten JW, Brooke CB, Baker SC, Mounce BC, Heaton NS, Dickinson BC, Jaochimiak A, Randall G, and Tay S
- Abstract
There is an urgent need for anti-viral agents that treat SARS-CoV-2 infection. The shortest path to clinical use is repurposing of drugs that have an established safety profile in humans. Here, we first screened a library of 1,900 clinically safe drugs for inhibiting replication of OC43, a human beta-coronavirus that causes the common-cold and is a relative of SARS-CoV-2, and identified 108 effective drugs. We further evaluated the top 26 hits and determined their ability to inhibit SARS-CoV-2, as well as other pathogenic RNA viruses. 20 of the 26 drugs significantly inhibited SARS-CoV-2 replication in human lung cells (A549 epithelial cell line), with EC50 values ranging from 0.1 to 8 micromolar. We investigated the mechanism of action for these and found that masitinib, a drug originally developed as a tyrosine-kinase inhibitor for cancer treatment, strongly inhibited the activity of the SARS-CoV-2 main protease 3CLpro. X-ray crystallography revealed that masitinib directly binds to the active site of 3CLpro, thereby blocking its enzymatic activity. Mastinib also inhibited the related viral protease of picornaviruses and blocked picornaviruses replication. Thus, our results show that masitinib has broad anti-viral activity against two distinct beta-coronaviruses and multiple picornaviruses that cause human disease and is a strong candidate for clinical trials to treat SARS-CoV-2 infection.
- Published
- 2020
- Full Text
- View/download PDF
32. Crystal structures of SARS-CoV-2 ADP-ribose phosphatase: from the apo form to ligand complexes.
- Author
-
Michalska K, Kim Y, Jedrzejczak R, Maltseva NI, Stols L, Endres M, and Joachimiak A
- Abstract
Among 15 nonstructural proteins (Nsps), the newly emerging Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) encodes a large, multidomain Nsp3. One of its units is the ADP-ribose phosphatase domain (ADRP; also known as the macrodomain, MacroD), which is believed to interfere with the host immune response. Such a function appears to be linked to the ability of the protein to remove ADP-ribose from ADP-ribosylated proteins and RNA, yet the precise role and molecular targets of the enzyme remain unknown. Here, five high-resolution (1.07-2.01 Å) crystal structures corresponding to the apo form of the protein and its complexes with 2-( N -morpholino)ethanesulfonic acid (MES), AMP and ADP-ribose have been determined. The protein is shown to undergo conformational changes to adapt to the ligand in the manner previously observed in close homologues from other viruses. A conserved water molecule is also identified that may participate in hydrolysis. This work builds foundations for future structure-based research on ADRP, including the search for potential antiviral therapeutics., (© Karolina Michalska et al. 2020.)
- Published
- 2020
- Full Text
- View/download PDF
33. Crystal structure of Nsp15 endoribonuclease NendoU from SARS-CoV-2.
- Author
-
Kim Y, Jedrzejczak R, Maltseva NI, Wilamowski M, Endres M, Godzik A, Michalska K, and Joachimiak A
- Subjects
- Amino Acid Sequence, Betacoronavirus genetics, Betacoronavirus metabolism, Catalytic Domain, Cloning, Molecular, Crystallography, X-Ray, Endoribonucleases genetics, Endoribonucleases metabolism, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, Humans, Middle East Respiratory Syndrome Coronavirus genetics, Middle East Respiratory Syndrome Coronavirus metabolism, Models, Molecular, Oligonucleotides metabolism, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus metabolism, SARS-CoV-2, Sequence Alignment, Sequence Homology, Amino Acid, Substrate Specificity, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism, Betacoronavirus chemistry, Endoribonucleases chemistry, Middle East Respiratory Syndrome Coronavirus chemistry, Oligonucleotides chemistry, Severe acute respiratory syndrome-related coronavirus chemistry, Viral Nonstructural Proteins chemistry
- Abstract
Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) is rapidly spreading around the world. There is no existing vaccine or proven drug to prevent infections and stop virus proliferation. Although this virus is similar to human and animal SARS-CoVs and Middle East Respiratory Syndrome coronavirus (MERS-CoVs), the detailed information about SARS-CoV-2 proteins structures and functions is urgently needed to rapidly develop effective vaccines, antibodies, and antivirals. We applied high-throughput protein production and structure determination pipeline at the Center for Structural Genomics of Infectious Diseases to produce SARS-CoV-2 proteins and structures. Here we report two high-resolution crystal structures of endoribonuclease Nsp15/NendoU. We compare these structures with previously reported homologs from SARS and MERS coronaviruses., (© 2020 The Authors. Protein Science published by Wiley Periodicals, Inc. on behalf of The Protein Society.)
- Published
- 2020
- Full Text
- View/download PDF
34. Structural plasticity of SARS-CoV-2 3CL M pro active site cavity revealed by room temperature X-ray crystallography.
- Author
-
Kneller DW, Phillips G, O'Neill HM, Jedrzejczak R, Stols L, Langan P, Joachimiak A, Coates L, and Kovalevsky A
- Subjects
- Catalytic Domain, Coronavirus 3C Proteases, Crystallography, X-Ray, Cysteine Endopeptidases metabolism, Cysteine Proteinase Inhibitors metabolism, Ligands, Models, Molecular, Molecular Dynamics Simulation, Protein Binding, Protein Conformation, Protein Domains, Protein Structure, Secondary, SARS-CoV-2, Temperature, Viral Nonstructural Proteins antagonists & inhibitors, Viral Nonstructural Proteins metabolism, Betacoronavirus enzymology, Cysteine Endopeptidases chemistry, Viral Nonstructural Proteins chemistry
- Abstract
The COVID-19 disease caused by the SARS-CoV-2 coronavirus has become a pandemic health crisis. An attractive target for antiviral inhibitors is the main protease 3CL M
pro due to its essential role in processing the polyproteins translated from viral RNA. Here we report the room temperature X-ray structure of unliganded SARS-CoV-2 3CL Mpro , revealing the ligand-free structure of the active site and the conformation of the catalytic site cavity at near-physiological temperature. Comparison with previously reported low-temperature ligand-free and inhibitor-bound structures suggest that the room temperature structure may provide more relevant information at physiological temperatures for aiding in molecular docking studies.- Published
- 2020
- Full Text
- View/download PDF
35. The crystal structure of nsp10-nsp16 heterodimer from SARS-CoV-2 in complex with S-adenosylmethionine.
- Author
-
Rosas-Lemus M, Minasov G, Shuvalova L, Inniss NL, Kiryukhina O, Wiersum G, Kim Y, Jedrzejczak R, Maltseva NI, Endres M, Jaroszewski L, Godzik A, Joachimiak A, and Satchell KJF
- Abstract
SARS-CoV-2 is a member of the coronaviridae family and is the etiological agent of the respiratory Coronavirus Disease 2019. The virus has spread rapidly around the world resulting in over two million cases and nearly 150,000 deaths as of April 17, 2020. Since no treatments or vaccines are available to treat COVID-19 and SARS-CoV-2, respiratory complications derived from the infections have overwhelmed healthcare systems around the world. This virus is related to SARS-CoV-1, the virus that caused the 2002-2004 outbreak of Severe Acute Respiratory Syndrome. In January 2020, the Center for Structural Genomics of Infectious Diseases implemented a structural genomics pipeline to solve the structures of proteins essential for coronavirus replication-transcription. Here we show the first structure of the SARS-CoV-2 nsp10-nsp16 2'-O-methyltransferase complex with S-adenosylmethionine at a resolution of 1.80 Å. This heterodimer complex is essential for capping viral mRNA transcripts for efficient translation and to evade immune surveillance.
- Published
- 2020
- Full Text
- View/download PDF
36. Allosteric inhibitors of Mycobacterium tuberculosis tryptophan synthase.
- Author
-
Michalska K, Chang C, Maltseva NI, Jedrzejczak R, Robertson GT, Gusovsky F, McCarren P, Schreiber SL, Nag PP, and Joachimiak A
- Subjects
- Crystallography, X-Ray, Enzyme Inhibitors chemistry, Humans, Indoles chemistry, Ligands, Models, Molecular, Molecular Structure, Sulfonamides chemistry, Thiophenes chemistry, Tryptophan Synthase chemistry, Tryptophan Synthase metabolism, Allosteric Site drug effects, Enzyme Inhibitors pharmacology, Indoles pharmacology, Mycobacterium tuberculosis enzymology, Sulfonamides pharmacology, Thiophenes pharmacology, Tryptophan Synthase antagonists & inhibitors
- Abstract
Global dispersion of multidrug resistant bacteria is very common and evolution of antibiotic-resistance is occurring at an alarming rate, presenting a formidable challenge for humanity. The development of new therapeuthics with novel molecular targets is urgently needed. Current drugs primarily affect protein, nucleic acid, and cell wall synthesis. Metabolic pathways, including those involved in amino acid biosynthesis, have recently sparked interest in the drug discovery community as potential reservoirs of such novel targets. Tryptophan biosynthesis, utilized by bacteria but absent in humans, represents one of the currently studied processes with a therapeutic focus. It has been shown that tryptophan synthase (TrpAB) is required for survival of Mycobacterium tuberculosis in macrophages and for evading host defense, and therefore is a promising drug target. Here we present crystal structures of TrpAB with two allosteric inhibitors of M. tuberculosis tryptophan synthase that belong to sulfolane and indole-5-sulfonamide chemical scaffolds. We compare our results with previously reported structural and biochemical studies of another, azetidine-containing M. tuberculosis tryptophan synthase inhibitor. This work shows how structurally distinct ligands can occupy the same allosteric site and make specific interactions. It also highlights the potential benefit of targeting more variable allosteric sites of important metabolic enzymes., (© 2020 The Protein Society.)
- Published
- 2020
- Full Text
- View/download PDF
37. Structure of Calcarisporiella thermophila Hsp104 Disaggregase that Antagonizes Diverse Proteotoxic Misfolding Events.
- Author
-
Michalska K, Zhang K, March ZM, Hatzos-Skintges C, Pintilie G, Bigelow L, Castellano LM, Miles LJ, Jackrel ME, Chuang E, Jedrzejczak R, Shorter J, Chiu W, and Joachimiak A
- Subjects
- Cryoelectron Microscopy, Crystallography, X-Ray, DNA-Binding Proteins antagonists & inhibitors, Fungal Proteins chemistry, Fungal Proteins pharmacology, Humans, Models, Molecular, Peptides antagonists & inhibitors, Protein Conformation, alpha-Helical, Proteostasis Deficiencies prevention & control, alpha-Synuclein antagonists & inhibitors, Adenosine Triphosphatases chemistry, Adenosine Triphosphatases pharmacology, Mucorales enzymology
- Abstract
Hsp104 is an AAA+ protein disaggregase with powerful amyloid-remodeling activity. All nonmetazoan eukaryotes express Hsp104 while eubacteria express an Hsp104 ortholog, ClpB. However, most studies have focused on Hsp104 from Saccharomyces cerevisiae and ClpB orthologs from two eubacterial species. Thus, the natural spectrum of Hsp104/ClpB molecular architectures and protein-remodeling activities remains largely unexplored. Here, we report two structures of Hsp104 from the thermophilic fungus Calcarisporiella thermophila (CtHsp104), a 2.70Å crystal structure and 4.0Å cryo-electron microscopy structure. Both structures reveal left-handed, helical assemblies with all domains clearly resolved. We thus provide the highest resolution and most complete view of Hsp104 hexamers to date. We also establish that CtHsp104 antagonizes several toxic protein-misfolding events in vivo where S. cerevisiae Hsp104 is ineffective, including rescue of TDP-43, polyglutamine, and α-synuclein toxicity. We suggest that natural Hsp104 variation is an invaluable, untapped resource for illuminating therapeutic disaggregases for fatal neurodegenerative diseases., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
38. Crystal structure of the ligand-binding domain of a LysR-type transcriptional regulator: transcriptional activation via a rotary switch.
- Author
-
Kim Y, Chhor G, Tsai CS, Winans JB, Jedrzejczak R, Joachimiak A, and Winans SC
- Subjects
- Arginine pharmacology, Bacterial Proteins genetics, Binding Sites physiology, Carrier Proteins genetics, Carrier Proteins metabolism, Crystallography, X-Ray, DNA, Bacterial genetics, DNA-Binding Proteins genetics, Membrane Proteins genetics, Membrane Proteins metabolism, Promoter Regions, Genetic drug effects, Promoter Regions, Genetic genetics, Protein Binding physiology, Transcription Factors genetics, Agrobacterium tumefaciens genetics, Arginine analogs & derivatives, Bacterial Proteins metabolism, DNA-Binding Proteins metabolism, Protein Structure, Quaternary drug effects, Transcription Factors metabolism, Transcriptional Activation genetics
- Abstract
LysR-type transcriptional regulators (LTTRs) generally bind to target promoters in two conformations, depending on the availability of inducing ligands. OccR is an LTTR that regulates the octopine catabolism operon of Agrobacterium tumefaciens. OccR binds to a site located between the divergent occQ and occR promoters. Octopine triggers a conformational change that activates the occQ promoter, and does not affect autorepression. This change shortens the length of bound DNA and relaxes a high-angle DNA bend. Here, we describe the crystal structure of the ligand-binding domain (LBD) of OccR apoprotein and holoprotein. Pairs of LBDs form dimers with extensive hydrogen bonding, while pairs of dimers interact via a single helix, creating a tetramer interface. Octopine causes a 70° rotation of each dimer with respect to the opposite dimer, precisely at the tetramer interface. We modeled the DNA binding domain (DBD), linker helix and bound DNA onto the apoprotein and holoprotein. The two DBDs of the modeled apoprotein lie far apart and the bound DNA between them has a high-angle DNA bend. In contrast, the two DBDs of the holoprotein lie closer to each other, with a low DNA bend angle. This inter-dimer pivot fully explains earlier studies of this LTTR., (© 2018 John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
39. Structural Insights into the Free-Standing Condensation Enzyme SgcC5 Catalyzing Ester-Bond Formation in the Biosynthesis of the Enediyne Antitumor Antibiotic C-1027.
- Author
-
Chang CY, Lohman JR, Huang T, Michalska K, Bigelow L, Rudolf JD, Jedrzejczak R, Yan X, Ma M, Babnigg G, Joachimiak A, Phillips GN Jr, and Shen B
- Subjects
- Catalysis, Antibiotics, Antineoplastic chemistry, Antibiotics, Antineoplastic metabolism, Bacterial Proteins chemistry, Bacterial Proteins genetics, Bacterial Proteins metabolism, Enediynes chemistry, Enediynes metabolism, Genes, Bacterial, Peptide Synthases chemistry, Peptide Synthases genetics, Peptide Synthases metabolism, Streptomyces enzymology, Streptomyces genetics
- Abstract
C-1027 is a chromoprotein enediyne antitumor antibiotic, consisting of the CagA apoprotein and the C-1027 chromophore. The C-1027 chromophore features a nine-membered enediyne core appended with three peripheral moieties, including an ( S)-3-chloro-5-hydroxy-β-tyrosine. In a convergent biosynthesis of the C-1027 chromophore, the ( S)-3-chloro-5-hydroxy-β-tyrosine moiety is appended to the enediyne core by the free-standing condensation enzyme SgcC5. Unlike canonical condensation domains from the modular nonribosomal peptide synthetases that catalyze amide-bond formation, SgcC5 catalyzes ester-bond formation, as demonstrated in vitro, between SgcC2-tethered ( S)-3-chloro-5-hydroxy-β-tyrosine and ( R)-1-phenyl-1,2-ethanediol, a mimic of the enediyne core as an acceptor substrate. Here, we report that (i) genes encoding SgcC5 homologues are widespread among both experimentally confirmed and bioinformatically predicted enediyne biosynthetic gene clusters, forming a new clade of condensation enzymes, (ii) SgcC5 shares a similar overall structure with the canonical condensation domains but forms a homodimer in solution, the active site of which is located in a cavity rather than a tunnel typically seen in condensation domains, and (iii) the catalytic histidine of SgcC5 activates the 2-hydroxyl group, while a hydrogen-bond network in SgcC5 prefers the R-enantiomer of the acceptor substrate, accounting for the regio- and stereospecific ester-bond formation between SgcC2-tethered ( S)-3-chloro-5-hydroxy-β-tyrosine and ( R)-1-phenyl-1,2-ethanediol upon acid-base catalysis. These findings expand the catalytic repertoire and reveal new insights into the structure and mechanism of condensation enzymes.
- Published
- 2018
- Full Text
- View/download PDF
40. Structural Evidence of a Major Conformational Change Triggered by Substrate Binding in DapE Enzymes: Impact on the Catalytic Mechanism.
- Author
-
Nocek B, Reidl C, Starus A, Heath T, Bienvenue D, Osipiuk J, Jedrzejczak R, Joachimiak A, Becker DP, and Holz RC
- Subjects
- Amidohydrolases genetics, Amidohydrolases metabolism, Amino Acid Substitution, Bacterial Proteins genetics, Bacterial Proteins metabolism, Catalysis, Catalytic Domain, Crystallography, X-Ray, Diaminopimelic Acid metabolism, Dimerization, Haemophilus influenzae genetics, Hydrogen Bonding, Models, Molecular, Mutagenesis, Site-Directed, Neisseria meningitidis enzymology, Neisseria meningitidis genetics, Protein Binding, Protein Conformation, Recombinant Proteins chemistry, Rotation, Substrate Specificity, Succinic Acid metabolism, Zinc chemistry, Amidohydrolases chemistry, Bacterial Proteins chemistry, Haemophilus influenzae enzymology
- Abstract
The X-ray crystal structure of the dapE-encoded N-succinyl-l,l-diaminopimelic acid desuccinylase from Haemophilus influenzae (HiDapE) bound by the products of hydrolysis, succinic acid and l,l-DAP, was determined at 1.95 Å. Surprisingly, the structure bound to the products revealed that HiDapE undergoes a significant conformational change in which the catalytic domain rotates ∼50° and shifts ∼10.1 Å (as measured at the position of the Zn atoms) relative to the dimerization domain. This heretofore unobserved closed conformation revealed significant movements within the catalytic domain compared to that of wild-type HiDapE, which results in effectively closing off access to the dinuclear Zn(II) active site with the succinate carboxylate moiety bridging the dinculear Zn(II) cluster in a μ-1,3 fashion forming a bis(μ-carboxylato)dizinc(II) core with a Zn-Zn distance of 3.8 Å. Surprisingly, His194.B, which is located on the dimerization domain of the opposing chain ∼10.1 Å from the dinuclear Zn(II) active site, forms a hydrogen bond (2.9 Å) with the oxygen atom of succinic acid bound to Zn2, forming an oxyanion hole. As the closed structure forms upon substrate binding, the movement of His194.B by more than ∼10 Å is critical, based on site-directed mutagenesis data, for activation of the scissile carbonyl carbon of the substrate for nucleophilic attack by a hydroxide nucleophile. Employing the HiDapE product-bound structure as the starting point, a reverse engineering approach called product-based transition-state modeling provided structural models for each major catalytic step. These data provide insight into the catalytic reaction mechanism and also the future design of new, potent inhibitors of DapE enzymes.
- Published
- 2018
- Full Text
- View/download PDF
41. X-ray crystal structures of the pheromone-binding domains of two quorum-hindered transcription factors, YenR of Yersinia enterocolitica and CepR2 of Burkholderia cenocepacia.
- Author
-
Kim Y, Chhor G, Tsai CS, Fox G, Chen CS, Winans NJ, Jedrzejczak R, Joachimiak A, and Winans SC
- Subjects
- Bacterial Proteins genetics, Burkholderia cenocepacia chemistry, Crystallography, X-Ray, DNA-Binding Proteins chemistry, Gene Expression Regulation, Bacterial, Homoserine chemistry, Pheromones chemistry, Protein Conformation, Protein Domains genetics, Protein Folding, Trans-Activators genetics, Transcription Factors chemistry, Yersinia enterocolitica chemistry, Bacterial Proteins chemistry, Homoserine analogs & derivatives, Lactones chemistry, Trans-Activators chemistry
- Abstract
The ability of LuxR-type proteins to regulate transcription is controlled by bacterial pheromones, N-acylhomoserine lactones (AHLs). Most LuxR-family proteins require their cognate AHLs for activity, and some of them require AHLs for folding and stability, and for protease-resistance. However, a few members of this family are able to fold, dimerize, bind DNA, and regulate transcription in the absence of AHLs; moreover, these proteins are antagonized by their cognate AHLs. One such protein is YenR of Yersinia enterocolitica, which is antagonized by N-3-oxohexanoyl-l-homoserine lactone (OHHL). This pheromone is produced by the OHHL synthase, a product of the adjacent yenI gene. Another example is CepR2 of Burkholderia cenocepacia, which is antagonized by N-octanoyl-l-homoserine lactone (OHL), whose synthesis is directed by the cepI gene of the same bacterium. Here, we describe the high-resolution crystal structures of the AHL binding domains of YenR and CepR2. YenR was crystallized in the presence and absence of OHHL. While this ligand does not cause large scale changes in the YenR structure, it does alter the orientation of several highly conserved YenR residues within and near the pheromone-binding pocket, which in turn caused a significant movement of a surface-exposed loop., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
42. Crystal Structure of Thioesterase SgcE10 Supporting Common Polyene Intermediates in 9- and 10-Membered Enediyne Core Biosynthesis.
- Author
-
Annaval T, Rudolf JD, Chang CY, Lohman JR, Kim Y, Bigelow L, Jedrzejczak R, Babnigg G, Joachimiak A, Phillips GN Jr, and Shen B
- Abstract
Enediynes are potent natural product anticancer antibiotics, and are classified as 9- or 10-membered according to the size of their enediyne core carbon skeleton. Both 9- and 10-membered enediyne cores are biosynthesized by the enediyne polyketide synthase (PKSE), thioesterase (TE), and PKSE-associated enzymes. Although the divergence between 9- and 10-membered enediyne core biosynthesis remains unclear, it has been observed that nascent polyketide intermediates, tethered to the acyl carrier protein (ACP) domain of PKSE, could be released by TE in the absence of the PKSE-associated enzymes. In this study, we determined the crystal structure of SgcE10, the TE that participates in the biosynthesis of the 9-membered enediyne C-1027. Structural comparison of SgcE10 with CalE7 and DynE7, two TEs that participate in the biosynthesis of the 10-membered enediynes calicheamicin and dynemicin, respectively, revealed that they share a common α/β hot-dog fold. The amino acids involved in both substrate binding and catalysis are conserved among SgcE10, CalE7, and DynE7. The volume and the shape of the substrate-binding channel and active site in SgcE10, CalE7, and DynE7 confirm that TEs from both 9- and 10-membered enediyne biosynthetic machineries bind the linear form of similar ACP-tethered polyene intermediates. Taken together, these findings further support the proposal that the divergence between 9- and 10-membered enediyne core biosynthesis occurs beyond PKSE and TE catalysis.
- Published
- 2017
- Full Text
- View/download PDF
43. The CDI toxin of Yersinia kristensenii is a novel bacterial member of the RNase A superfamily.
- Author
-
Batot G, Michalska K, Ekberg G, Irimpan EM, Joachimiak G, Jedrzejczak R, Babnigg G, Hayes CS, Joachimiak A, and Goulding CW
- Subjects
- Bacterial Toxins metabolism, Crystallography, X-Ray, Models, Molecular, Protein Conformation, RNA metabolism, Ribonuclease, Pancreatic metabolism, Bacterial Toxins chemistry, Endoribonucleases chemistry, Ribonuclease, Pancreatic chemistry, Yersinia enzymology
- Abstract
Contact-dependent growth inhibition (CDI) is an important mechanism of inter-bacterial competition found in many Gram-negative pathogens. CDI+ cells express cell-surface CdiA proteins that bind neighboring bacteria and deliver C-terminal toxin domains (CdiA-CT) to inhibit target-cell growth. CDI+ bacteria also produce CdiI immunity proteins, which specifically neutralize cognate CdiA-CT toxins to prevent self-inhibition. Here, we present the crystal structure of the CdiA-CT/CdiIYkris complex from Yersinia kristensenii ATCC 33638. CdiA-CTYkris adopts the same fold as angiogenin and other RNase A paralogs, but the toxin does not share sequence similarity with these nucleases and lacks the characteristic disulfide bonds of the superfamily. Consistent with the structural homology, CdiA-CTYkris has potent RNase activity in vitro and in vivo. Structure-guided mutagenesis reveals that His175, Arg186, Thr276 and Tyr278 contribute to CdiA-CTYkris activity, suggesting that these residues participate in substrate binding and/or catalysis. CdiIYkris binds directly over the putative active site and likely neutralizes toxicity by blocking access to RNA substrates. Significantly, CdiA-CTYkris is the first non-vertebrate protein found to possess the RNase A superfamily fold, and homologs of this toxin are associated with secretion systems in many Gram-negative and Gram-positive bacteria. These observations suggest that RNase A-like toxins are commonly deployed in inter-bacterial competition., (© The Author(s) 2017. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2017
- Full Text
- View/download PDF
44. A microbial sensor for organophosphate hydrolysis exploiting an engineered specificity switch in a transcription factor.
- Author
-
Jha RK, Kern TL, Kim Y, Tesar C, Jedrzejczak R, Joachimiak A, and Strauss CE
- Subjects
- Crystallography, X-Ray, Flow Cytometry, Hydrolysis, Ligands, Molecular Docking Simulation, Nitrophenols metabolism, Organophosphates chemistry, Paraoxon metabolism, Plasmids metabolism, Structural Homology, Protein, Transcription Factors chemistry, Bacterial Proteins metabolism, Biosensing Techniques, Organophosphates metabolism, Protein Engineering, Transcription Factors metabolism
- Abstract
A whole-cell biosensor utilizing a transcription factor (TF) is an effective tool for sensitive and selective detection of specialty chemicals or anthropogenic molecules, but requires access to an expanded repertoire of TFs. Using homology modeling and ligand docking for binding pocket identification, assisted by conservative mutations in the pocket, we engineered a novel specificity in an Acinetobacter TF, PobR, to 'sense' a chemical p-nitrophenol (pNP) and measured the response via a fluorescent protein reporter expressed from a PobR promoter. Out of 10(7) variants of PobR, four were active when dosed with pNP, with two mutants showing a specificity switch from the native effector 4-hydroxybenzoate (4HB). One of the mutants, pNPmut1 was then used to create a smart microbial cell responding to pNP production from hydrolysis of an insecticide, paraoxon, in a coupled assay involving phosphotriesterase (PTE) enzyme expressed from a separate promoter. We show the fluorescence of the cells correlated with the catalytic efficiency of the PTE variant expressed in each cell. High selectivity between similar molecules (4HB versus pNP), high sensitivity for pNP detection (∼2 μM) and agreement of apo- and holo-structures of PobR scaffold with predetermined computational models are other significant results presented in this work., (Published by Oxford University Press on behalf of Nucleic Acids Research 2016. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2016
- Full Text
- View/download PDF
45. Structural dynamics of a methionine γ-lyase for calicheamicin biosynthesis: Rotation of the conserved tyrosine stacking with pyridoxal phosphate.
- Author
-
Cao H, Tan K, Wang F, Bigelow L, Yennamalli RM, Jedrzejczak R, Babnigg G, Bingman CA, Joachimiak A, Kharel MK, Singh S, Thorson JS, and Phillips GN Jr
- Abstract
CalE6 from Micromonospora echinospora is a (pyridoxal 5' phosphate) PLP-dependent methionine γ-lyase involved in the biosynthesis of calicheamicins. We report the crystal structure of a CalE6 2-(N-morpholino)ethanesulfonic acid complex showing ligand-induced rotation of Tyr100, which stacks with PLP, resembling the corresponding tyrosine rotation of true catalytic intermediates of CalE6 homologs. Elastic network modeling and crystallographic ensemble refinement reveal mobility of the N-terminal loop, which involves both tetrameric assembly and PLP binding. Modeling and comparative structural analysis of PLP-dependent enzymes involved in Cys/Met metabolism shine light on the functional implications of the intrinsic dynamic properties of CalE6 in catalysis and holoenzyme maturation.
- Published
- 2016
- Full Text
- View/download PDF
46. Gene selection and cloning approaches for co-expression and production of recombinant protein-protein complexes.
- Author
-
Babnigg G, Jedrzejczak R, Nocek B, Stein A, Eschenfeldt W, Stols L, Marshall N, Weger A, Wu R, Donnelly M, and Joachimiak A
- Subjects
- Computational Biology methods, Escherichia coli genetics, Escherichia coli metabolism, Gene Fusion, Gene Order, Genetic Vectors, Models, Molecular, Multiprotein Complexes metabolism, Operon, Protein Binding, Protein Conformation, Recombinant Fusion Proteins isolation & purification, Recombinant Fusion Proteins metabolism, Solubility, Cloning, Molecular methods, Gene Expression, Multiprotein Complexes chemistry, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics
- Abstract
Multiprotein complexes play essential roles in all cells and X-ray crystallography can provide unparalleled insight into their structure and function. Many of these complexes are believed to be sufficiently stable for structural biology studies, but the production of protein-protein complexes using recombinant technologies is still labor-intensive. We have explored several strategies for the identification and cloning of heterodimers and heterotrimers that are compatible with the high-throughput (HTP) structural biology pipeline developed for single proteins. Two approaches are presented and compared which resulted in co-expression of paired genes from a single expression vector. Native operons encoding predicted interacting proteins were selected from a repertoire of genomes, and cloned directly to expression vector. In an alternative approach, Helicobacter pylori proteins predicted to interact strongly were cloned, each associated with translational control elements, then linked into an artificial operon. Proteins were then expressed and purified by standard HTP protocols, resulting to date in the structure determination of two H. pylori complexes.
- Published
- 2015
- Full Text
- View/download PDF
47. Structure of a cupin protein Plu4264 from Photorhabdus luminescens subsp. laumondii TTO1 at 1.35 Å resolution.
- Author
-
Weerth RS, Michalska K, Bingman CA, Yennamalli RM, Li H, Jedrzejczak R, Wang F, Babnigg G, Joachimiak A, Thomas MG, and Phillips GN Jr
- Subjects
- Binding Sites, Crystallography, X-Ray, Manganese chemistry, Models, Molecular, Photorhabdus chemistry, Protein Structure, Secondary, Bacterial Proteins chemistry
- Abstract
Proteins belonging to the cupin superfamily have a wide range of catalytic and noncatalytic functions. Cupin proteins commonly have the capacity to bind a metal ion with the metal frequently determining the function of the protein. We have been investigating the function of homologous cupin proteins that are conserved in more than 40 species of bacteria. To gain insights into the potential function of these proteins we have solved the structure of Plu4264 from Photorhabdus luminescens TTO1 at a resolution of 1.35 Å and identified manganese as the likely natural metal ligand of the protein., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
48. RsaM: a transcriptional regulator of Burkholderia spp. with novel fold.
- Author
-
Michalska K, Chhor G, Clancy S, Jedrzejczak R, Babnigg G, Winans SC, and Joachimiak A
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Conserved Sequence, Crystallography, X-Ray, Models, Molecular, Protein Structure, Quaternary, Protein Structure, Tertiary, Quorum Sensing, Structural Homology, Protein, Bacterial Proteins chemistry, Burkholderia, Transcription Factors chemistry
- Abstract
Burkholderia cepacia complex is a set of closely related bacterial species that are notorious pathogens of cystic fibrosis patients, responsible for life-threatening lung infections. Expression of several virulence factors of Burkholderia cepacia complex is controlled by a mechanism known as quorum sensing (QS). QS is a means of bacterial communication used to coordinate gene expression in a cell-density-dependent manner. The system involves the production of diffusible signaling molecules (N-acyl-l-homoserine lactones, AHLs), that bind to cognate transcriptional regulators and influence their ability to regulate gene expression. One such system that is highly conserved in Burkholderia cepacia complex consists of CepI and CepR. CepI is AHL synthase, whereas CepR is an AHL-dependent transcription factor. In most members of the Burkholderia cepacia complex group, the cepI and cepR genes are divergently transcribed and separated by additional genes. One of them, bcam1869, encodes the BcRsaM protein, which was recently postulated to modulate the abundance or activity of CepI or CepR. Here, we show the crystal structure of BcRsaM from B. cenocepacia J2315. It is a single-domain protein with unique topology and presents a novel fold. The protein is a dimer in the crystal and in solution. This regulator has no known DNA-binding motifs and direct binding of BcRsaM to the cepI promoter could not be detected in in vitro assays. Therefore, we propose that the modulatory action of RsaM might result from interactions with other components of the QS machinery rather than from direct association with the DNA promoter., Database: The atomic coordinates and structure factors have been deposited in the Protein Data Bank under entry 4O2H., Structured Digital Abstract: BcRsaM and BcRsaM bind by x-ray crystallography (View interaction) BcRsaM and BcRsaM bind by molecular sieving (View interaction)., (© 2014 FEBS.)
- Published
- 2014
- Full Text
- View/download PDF
49. The dimerization domain in DapE enzymes is required for catalysis.
- Author
-
Nocek B, Starus A, Makowska-Grzyska M, Gutierrez B, Sanchez S, Jedrzejczak R, Mack JC, Olsen KW, Joachimiak A, and Holz RC
- Subjects
- Amidohydrolases genetics, Bacterial Proteins genetics, Catalysis, Catalytic Domain, Crystallography, X-Ray, Kinetics, Molecular Dynamics Simulation, Mutagenesis, Site-Directed, Mutation, Vibrio cholerae enzymology, Amidohydrolases chemistry, Amidohydrolases metabolism, Bacterial Proteins chemistry, Bacterial Proteins metabolism
- Abstract
The emergence of antibiotic-resistant bacterial strains underscores the importance of identifying new drug targets and developing new antimicrobial compounds. Lysine and meso-diaminopimelic acid are essential for protein production and bacterial peptidoglycan cell wall remodeling and are synthesized in bacteria by enzymes encoded within dap operon. Therefore dap enzymes may serve as excellent targets for developing a new class of antimicrobial agents. The dapE-encoded N-succinyl-L,L-diaminopimelic acid desuccinylase (DapE) converts N-succinyl-L,L-diaminopimelic acid to L,L-diaminopimelic acid and succinate. The enzyme is composed of catalytic and dimerization domains, and belongs to the M20 peptidase family. To understand the specific role of each domain of the enzyme we engineered dimerization domain deletion mutants of DapEs from Haemophilus influenzae and Vibrio cholerae, and characterized these proteins structurally and biochemically. No activity was observed for all deletion mutants. Structural comparisons of wild-type, inactive monomeric DapE enzymes with other M20 peptidases suggest that the dimerization domain is essential for DapE enzymatic activity. Structural analysis and molecular dynamics simulations indicate that removal of the dimerization domain increased the flexibility of a conserved active site loop that may provide critical interactions with the substrate.
- Published
- 2014
- Full Text
- View/download PDF
50. New LIC vectors for production of proteins from genes containing rare codons.
- Author
-
Eschenfeldt WH, Makowska-Grzyska M, Stols L, Donnelly MI, Jedrzejczak R, and Joachimiak A
- Subjects
- Biotinylation, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Gene Order, Kinetics, Ligands, Plasmids genetics, Protein Binding, Proteins isolation & purification, Proteins metabolism, RNA, Transfer chemistry, RNA, Transfer genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins isolation & purification, Recombinant Fusion Proteins metabolism, Codon, Genetic Vectors genetics, Proteins genetics
- Abstract
In the effort to produce proteins coded by diverse genomes, structural genomics projects often must express genes containing codons that are rare in the production strain. To address this problem, genes expressing tRNAs corresponding to those codons are typically coexpressed from a second plasmid in the host strain, or from genes incorporated into production plasmids. Here we describe the modification of a series of LIC pMCSG vectors currently used in the high-throughput (HTP) production of proteins to include crucial tRNA genes covering rare codons for Arg (AGG/AGA) and Ile (AUA). We also present variants of these new vectors that allow analysis of ligand binding or co-expression of multiple proteins introduced through two independent LIC steps. Additionally, to accommodate the cloning of multiple large proteins, the size of the plasmids was reduced by approximately one kilobase through the removal of non-essential DNA from the base vector. Production of proteins from core vectors of this series validated the desired enhanced capabilities: higher yields of proteins expressed from genes with rare codons occurred in most cases, biotinylated derivatives enabled detailed automated ligand binding analysis, and multiple proteins introduced by dual LIC cloning were expressed successfully and in near balanced stoichiometry, allowing tandem purification of interacting proteins.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.