Structure-Guided Functional Characterization of Enediyne Self-Sacrifice Resistance Proteins, CalU16 and CalU19
- Sherif I. Elshahawi ,
- Theresa A. Ramelot ,
- Jayaraman Seetharaman ,
- Jing Chen ,
- Shanteri Singh ,
- Yunhuang Yang ,
- Kari Pederson ,
- Madan K. Kharel ,
- Rong Xiao ,
- Scott Lew ,
- Ragothaman M. Yennamalli ,
- Mitchell D. Miller ,
- Fengbin Wang ,
- Liang Tong ,
- Gaetano T. Montelione ,
- Michael A. Kennedy ,
- Craig A. Bingman ,
- Haining Zhu ,
- George N. Phillips, Jr. , and
- Jon S. Thorson
Abstract

Calicheamicin γ1I (1) is an enediyne antitumor compound produced by Micromonospora echinospora spp. calichensis, and its biosynthetic gene cluster has been previously reported. Despite extensive analysis and biochemical study, several genes in the biosynthetic gene cluster of 1 remain functionally unassigned. Using a structural genomics approach and biochemical characterization, two proteins encoded by genes from the 1 biosynthetic gene cluster assigned as “unknowns”, CalU16 and CalU19, were characterized. Structure analysis revealed that they possess the STeroidogenic Acute Regulatory protein related lipid Transfer (START) domain known mainly to bind and transport lipids and previously identified as the structural signature of the enediyne self-resistance protein CalC. Subsequent study revealed calU16 and calU19 to confer resistance to 1, and reminiscent of the prototype CalC, both CalU16 and CalU19 were cleaved by 1in vitro. Through site-directed mutagenesis and mass spectrometry, we identified the site of cleavage in each protein and characterized their function in conferring resistance against 1. This report emphasizes the importance of structural genomics as a powerful tool for the functional annotation of unknown proteins.
Figure 1

Figure 1. (A) Selected structures of naturally occurring 10-membered enediynes. The “warhead” is highlighted in red. (B) Proposed mechanism of cycloaromatization of 1 and its effect on DNA scission and CalC self-sacrifice mechanism.
Results and Discussion
CalU16 Structure and CalU19 Homology Model
Figure 2

Figure 2. Structure of CalU16. (A) Overlay of NMR (green) and monomers of the crystal (brick- red) structures of CalU16. The N- and C-termini of the protein are labeled, while the dynamic loop is colored yellow. (B) Monomer with secondary structural elements labeled. The residues in the hydrophobic cavity are represented as stick models. (C) B-factors for the Cα atoms in the crystal structure (left) and Cα RMSD values from the NMR ensemble (right) are mapped to the color and tube diameter of “putty” traces showing the general agreement (correlation coefficient 0.559) between the structures.
property | value |
---|---|
Crystal Param. | |
space group | P61 |
molecules per asymmetric unit | 2 |
VM (Å3 Da–1) | 2.95 |
unit-cell param. (Å) | a = b = 51.85, c = 305.70 |
Data Collection Statistics | |
wavelength (Å) | 0.9790 |
resolution range (Å) | 50.00–2.50 (2.59–2.50) |
no. of reflections (measured/unique) | 236417/16051 |
completeness (%) | 99.9 (99.5) |
Rmergeb | 0.056 (0.169) |
redundancy | 14.8 (12.3) |
mean I/σ (I) | 27.4 (10.4) |
Refinement and Model Statistics | |
resolution range (Å) | 44.91–2.50 |
no. of reflections (work/test) | 15160/794 |
Rcrystc | 0.241 (0.325) |
Rfreed | 0.282 (0.371) |
twin operator | K, H, -L |
twin fraction | 0.50 |
RMSD bonds (Å) | 0.009 |
RMSD angles (deg) | 1.453 |
B factor (protein/solvent) (Å2) | 53.8/50.9 |
no. of protein atoms | 2497 |
no. of waters | 128 |
Ramachandran Plot (%)e | |
most favorable regions | 91.3 |
allowed regions | 5.8 |
disallowed regions | 2.9 |
Global Quality Scores (Raw/Z-Score) | |
Verify3D | 0.4/–1.0 |
ProsaII | 0.5/–0.6 |
Procheck G factor (phi-psi) | –0.6/–2.0 |
Procheck G factor (all) | –0.5/–2.7 |
Molprobity clash score | 9.6/–1.8 |
PDB ID | 4FPW |
Values in parentheses are for the highest resolution shell.
Rmerge = ∑h ∑i | Ii(h) – ⟨I(h)⟩|/∑h∑iIi(h), where Ii(h) is the intensity of an individual measurement of the reflection and ⟨I(h)⟩ is the mean intensity of the reflection.
Rcryst = ∑h ||Fobs| – |Fcalc||/∑h |Fobs|, where Fobs and Fcalc are the observed and calculated structure-factor amplitudes, respectively.
Rfree was calculated as Rcryst except that it uses 10% of the reflection data omitted from refinement.
Calculated using Molprobity. (71)
property | value |
---|---|
Completeness of Resonance Assignmentsb | |
backbone (%) | 98.3 |
side chain (%) | 97.9 |
aromatic (%) | 96.2 |
stereospecific methyl (%) | 85.8 |
Conformationally Restricting Constraintsc | |
distance constraints | |
total | 2522 |
intraresidue (i = j) | 471 |
sequential (|I – j| = 1) | 614 |
medium range (1 < |i – j| < 5) | 458 |
long-range (|i – j| ≥ 5) | 979 |
dihedral angle constraints | 222 |
hydrogen bond constraints | 192 |
NH RDC constraints (gel NH/NC′) | 75/59 |
no. of constraints per residue | 16.5 |
no. of long-range constraints per residue | 6.0 |
Residual Constraint Violationsc | |
avg. no. of distance violations per structure | |
0.1–0.2 Å | 24.8 |
0.2–0.5 Å | 6.2 |
> 0.5 Å | 0 |
avg no. of dihedral angle violations per structure | |
1–10° | 17.8 |
> 10° | 0.2 |
RDC Qrmsd (gel NH/NC′)d | 0.4/0.3 |
Model Qualityc | |
RMSD backbone atoms (Å)e | 0.7 |
RMSD heavy atoms (Å)e | 1.2 |
RMSD bond lengths (Å) | 0.018 |
RMSD bond angles (deg) | 1.3 |
MolProbity Ramachandran statisticsc,e | |
most favored regions (%) | 94.6 |
allowed regions (%) | 5.4 |
disallowed regions (%) | 0 |
global quality scores (Raw/Z-score)c | |
Verify3D | 0.4/–0.3 |
ProsaII | 0.5/–0.6 |
Procheck G factor (φ–ψ)e | –0.4/ −1.3 |
Procheck G factor (all)e | –0.3/–2.0 |
MolProbity clash score | 16.0/–1.2 |
RPF scoresf | |
recall/precision | 0.98 |
F-measure/DP-score | 0.94 |
Model Contents | |
ordered residue rangee | 7–65, 71–89, 95–140, 161–179 |
BMRB accession no. | 18547 |
PDB ID | 2LUZ |
Structural statistics computed for the ensemble of 20 deposited structures.
Computed using AVS software from the expected number of resonances, excluding: highly exchangeable protons (N-terminal, Lys, and Arg amino groups, hydroxyls of Ser, Thr, Tyr), carboxyls of Asp and Glu, nonprotonated aromatic carbons, for residues 1–182.
Calculated using PSVS 1.4. Average distance violations were calculated using the sum over r–6.
RDC goodness-of-fit quality factor Qrmsd determined using PALES.
Based on ordered residue ranges [S(φ) + S(ψ) > 1.8].
RPF scores reflecting the goodness-of-fit of the final ensemble of structures (including disordered residues) to the NOESY data and resonance assignments.
Structurally-Related Proteins
Figure 3

Figure 3. CalU16 structural homologues. (A) CalU16 (PDB: 4FPW); (B) CalC (PDB: 2L65), calicheamicin resistance protein; (C) TcmN Aro/Cyc (in complex with trans-dihydroquercetin; PDB: 3TVQ) involved in the biosynthesis of tetracenomycin; (D) Hyp-1 (in complex with ethylene glycol; PDB: 3IE5) involved in the biosynthesis of hypericin; (E) NCS (in complex with hydroxybenzaldehyde; PDB: 2VQ5) involved in the biosynthesis of norcoclaurine.
Heterologous Expression of calU16 or calU19 in E. coli Confers Resistance to Calicheamicin
Figure 4

Figure 4. CalU16 and CalU19 assays. Serial disc dilutions of 1 against (A) pSE28a-E. coli (control), (B) pSECalU16-E. coli (CalU16), (C) pSECalU19-E. coli (CalU19), (D) pJB2011-E. coli (CalC). Amount of 1 on discs 1–6 are 10 μg, 1 μg, 100 ng, 50 ng, 10 ng, and 1 ng, respectively. Coomassie-stained 8–12% SDS-PAGE gradient gel of (E) CalU16 and (F) CalU19 in the presence of DTT (lane 1), 1 (lane 2), and DTT and 1 (lane 3).
strain | MIC (μM) |
---|---|
pSE28a-E. coli | 0.036 |
pSECalU16-E. coli | 3 |
pSECalU19-E. coli | 12 |
pJB2011-E. coli | 12 |
pSEU16G128V-E. coli | 0.36 |
pSEU16G128R-E. coli | 0.18 |
pSEU16G142V-E. coli | 3 |
pSEU16G142R-E. coli | 3 |
pSEU19G177V-E. coli | 6 |
pSEU19G177R-E. coli | 0.36 |
pSEU19G181V-E. coli | 12 |
pSEU19G191V-E. coli | 12 |
pSEU19G196V-E. coli | 12 |
pSEU19G206V-E. coli | 12 |
CalU16 Biochemical Characterization
Figure 5

Figure 5. Site directed mutagenesis of CalU16 and CalU19. Docking models of (A) CalC (B) CalU16, and (C) CalU19 with mutated glycine residues represented as spheres where colored Gly residues indicate cleavage sites, wheat Gly residues indicate mutations that did not affect activity and calicheamicin (1) is represented as a stick model. Results of disc diffusion assay in CalU16 mutants (D) pSE28a-E. coli (control); (E) pSEU16G128V-E. coli; (F) pSEU16G128R-E. coli; (G) pSECalU16-E. coli; and CalU19 mutants (H) pSE28a-E. coli (control); (I) pSEU19G177V-E. coli; (J) pSEU19G177R-E. coli; (K) pSECalU19-E. coli. Amount of 1 on discs 1–6 are 10 μg, 1 μg, 100 ng, 50 ng, 10 ng, and 1 ng, respectively.
CalU19 Biochemical Characterization
Enediyne Specificity of CalU16 and CalU19
Conclusions
Methods
Strains, Materials, and General Methods
Parental Plasmids for Protein Production
Structure Determination of CalU16 by X-ray Crystallography
NMR Spectroscopy and Structure Determination of CalU16
Resistance Assays
Protein Cleavage Assay
Trypsin Digestion and Mass Analysis
Determination of Minimum Inhibitory Concentrations (MICs)
Supporting Information
Additional protocols and methods, supplemental figures, and supplemental tables. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
This work was supported by the National Institutes of Health (NIH) grants CA84374 (to J.S.T.), U01GM098248 (to G.N.P.), the National Center for Advancing Translational Sciences (UL1TR000117), a NIH PSI grant (U54-GM094597 to M.A.K. and G.T.M.) and the BioXFEL Science and Technology Center under National Science Foundation Grant No. 1231306. Enediynes for this study were generously provided by Pfizer (calicheamicin) and Bristol-Myers-Squibb (esperamicin and dynemicin). Proteomics analyses were conducted by the University of Kentucky Proteomics Core that is partially supported by grants from the National Institute of General Medical Sciences (P20GM103486), the National Cancer Institute (P30CA177558) and equipment acquired via a grant from National Center for Research Resources (1S10RR029127 to H.Z.). Protein NMR data collection was conducted at the Ohio Biomedicine Center of Excellence in Structural Biology and Metabolomics (Miami University). We thank Dr. J. Prestgard, Dr. T. Acton, and Dr. J. Everett at the Northeast Structural Genomics Consortium for technical assistance and J. Schwanof and R. Abramowitz for data collection assistance at beamline X4A, NSLS.
References
This article references 71 other publications.
- 1Maiese, W. M., Lechevalier, M. P., Lechevalier, H. A., Korshalla, J., Kuck, N., Fantini, A., Wildey, M. J., Thomas, J., and Greenstein, M. (1989) Calicheamicins, a novel family of antitumor antibiotics: taxonomy, fermentation and biological properties J. Antibiot. (Tokyo) 42, 558– 563[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXkt1SmurY%253D&md5=8944bd3a3bc333517c3ee816f01436bdCalicheamicins, a novel family of antitumor antibiotics: taxonomy, fermentation and biological propertiesMaiese, W. M.; Lechevalier, M. P.; Lechevalier, H. A.; Korshalla, J.; Kuck, N.; Fantini, A.; Wildey, M. J.; Thomas, J.; Greenstein, M.Journal of Antibiotics (1989), 42 (4), 558-63CODEN: JANTAJ; ISSN:0021-8820.A novel family of antitumor antibiotics, the calicheamicins, were isolated from the fermn. broth of Micromonospora echinospora subsp. calichensis. These antibiotics exhibited significant activity against gram-pos. and gram-neg. bacteria in vitro. Calicheamicin γ1I demonstrated antitumor activity against P388 leukemia and B16 melanoma in vivo.
- 2Lee, M. D., Manning, J. K., Williams, D. R., Kuck, N. A., Testa, R. T., and Borders, D. B. (1989) Calicheamicins, a novel family of antitumor antibiotics. 3. Isolation, purification and characterization of calicheamicins β 1Br, γ 1Br, α 2I, α 3I, β 1I, γ 1I, and δ 1I J. Antibiot. (Tokyo) 42, 1070– 1087[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlsFShsbg%253D&md5=2cb74746c1ed1b8f4ff887316d8b0e1fCalichemicins, a novel family of antitumor antibiotics. 3. Isolation, purification and characterization of calichemicins β1Br, γ1Br, α2I, α3I, β1I, γ1I, and δ1ILee, May D.; Manning, Joann K.; Williams, David R.; Kuck, Nydia A.; Testa, Raymond T.; Borders, Donald B.Journal of Antibiotics (1989), 42 (7), 1070-87CODEN: JANTAJ; ISSN:0021-8820.Novel antitumor antibiotics, calichemicins β1Br (I), γ1Br, α2I, α3I, β1I, γ1I, and δ1I, were recovered from the fermn. broth of Micromonospora echinospora calichensis by solvent extn., selective pptn., normal phase, reversed-phase, and partition chromatog. The individual components were characterized by their UV, IR, 1H, and 13C NMR spectra data.
- 3Lee, M. D., Dunne, T. S., Chang, C. C., Siegel, M. M., Morton, G. O., Ellestad, G. A., McGahren, W. J., and Borders, D. B. (1992) Calicheamicins, a novel family of antitumor antibiotics. 4. Structure elucidation of calicheamicins β 1Br, γ 1Br, α 2I, α 3I, β 1I, γ 1I, and δ 1I J. Am. Chem. Soc. 114, 985– 997
- 4Thorson, J. S., Sievers, E. L., Ahlert, J., Shepard, E., Whitwam, R. E., Onwueme, K. C., and Ruppen, M. (2000) Understanding and exploiting nature’s chemical arsenal: The past, present, and future of calicheamicin research Curr. Pharm. Des. 6, 1841– 1879[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXoslWntLo%253D&md5=8db2ae788ecfabec1e422181c508c689Understanding and exploiting nature's chemical arsenal: the past, present and future of calicheamicin researchThorson, Jon S.; Sievers, Eric L.; Ahlert, Joachim; Shepard, Erica; Whitwam, Ross E.; Onwueme, Kenolisa C.; Ruppen, MarkCurrent Pharmaceutical Design (2000), 6 (18), 1841-1879CODEN: CPDEFP; ISSN:1381-6128. (Bentham Science Publishers)A review with 234 refs. The enediyne antitumor antibiotics are appreciated for their novel mol. architecture, their remarkable biol. activity and their fascinating mode of action and many have spawned considerable interest as anticancer agents in the pharmaceutical industry. Of equal importance to these astonishing properties, the enediynes also offer a distinct opportunity to study the unparalleled biosyntheses of their unique mol. scaffolds and what promises to be unprecedented modes of self-resistance to highly reactive natural products. Elucidation of these aspects should unveil novel mechanistic enzymol., and may provide access to the rational biosynthetic modification of enediyne structure for new drug leads, the construction of enediyne overproducing strains and eventually lead to an enediyne combinatorial biosynthesis program. This article strives to compile and present the crit. research discoveries relevant to the clin. most promising enediyne, calicheamicin, from a historical perspective. Recent progress, particularly in the areas of biosynthesis, self-resistance, bio-engineering analogs and clin. studies are also highlighted.
- 5Galm, U., Hager, M. H., Van Lanen, S. G., Ju, J., Thorson, J. S., and Shen, B. (2005) Antitumor antibiotics: Bleomycin, enediynes, and mitomycin Chem. Rev. 105, 739– 758[ACS Full Text
], [CAS], Google Scholar
5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmslCrug%253D%253D&md5=7e21e51479e19b723d92113c80c58e51Antitumor Antibiotics: Bleomycin, Enediynes, and MitomycinGalm, Ute; Hager, Martin H.; Van Lanen, Steven G.; Ju, Jianhua; Thorson, Jon S.; Shen, BenChemical Reviews (Washington, DC, United States) (2005), 105 (2), 739-758CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. The review discusses discovery, biol. activities and resistance to bleomycin, enediynes, and mitomycin. - 6Liang, Z.-X. (2010) Complexity and simplicity in the biosynthesis of enediyne natural products Nat. Prod. Rep. 27, 499– 528[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjslGntrg%253D&md5=a235f078d92c01217637b8b8816f781cComplexity and simplicity in the biosynthesis of enediyne natural productsLiang, Zhao-XunNatural Product Reports (2010), 27 (4), 499-528CODEN: NPRRDF; ISSN:0265-0568. (Royal Society of Chemistry)A review. The review highlights the recent progress on the elucidation of the biosynthetic mechanism of enediyne natural products. A large body of knowledge has been gathered from the genetic, biochem. and structural studies since the sequencing of the first two enediyne gene clusters in 2002. Primary attention is devoted to the novel pathways and enzymes in the biosynthesis of the enediyne warhead as well as the peripheral moieties.
- 7De Voss, J. J., Hangeland, J. J., and Townsend, C. A. (1990) Characterization of the in vitro cyclization chemistry of calicheamicin and its relation to DNA cleavage J. Am. Chem. Soc. 112, 4554– 4556[ACS Full Text
], [CAS], Google Scholar
7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXktFOmt7s%253D&md5=63fc400ddd02e5829adc9e80379cf355Characterization of the in vitro cyclization chemistry of calicheamicin and its relation to DNA cleavageDe Voss, James J.; Hangeland, Jon J.; Townsend, Craig A.Journal of the American Chemical Society (1990), 112 (11), 4554-6CODEN: JACSAT; ISSN:0002-7863.The proposed mechanism of thiol activation of calicheamicin γ1I (I), one of the diynene antitumor antibiotics, was examd. in variable-temp. NMR studies. Sep. reactions of the drug with Me thioglycolate and tri-n-butylphosphine led to the identification and characterization of a reactive intermediate resulting from cleavage of the allylic methyltrisulfide and intramol. β-addn. of the resulting thio(ate) to the enone system. This intermediate undergoes 1st-order decompn. at a convenient rate at about -10°. The end product of this process contains 2 H atoms abstracted from the medium consistent with the intervention of a 1,4-diyl formed in the proposed Bergman rearrangement. The relatively long soln. half-life of this intermediate (calcd. to be 4.5 s at 37° in conjunction with other findings led to the formulation of a kinetic scheme to evaluate thermodn. and kinetic factors in the sequence-selective cleavages of DNA characteristic of calicheamicin. - 8Chatterjee, M., Cramer, K. D., and Townsend, C. A. (1993) Kinetic nature of thiol activation in DNA cleavage by calicheamicin J. Am. Chem. Soc. 115, 3374– 3375
- 9Myers, A. G., Cohen, S. B., and Kwon, B. M. (1994) A study of the reaction of calicheamicin γ 1 with glutathione in the presence of double-stranded DNA J. Am. Chem. Soc. 116, 1255– 1271
- 10Zein, N., Sinha, A. M., McGahren, W. J., and Ellestad, G. A. (1988) Calicheamicin γ 1I: An antitumor antibiotic that cleaves double-stranded DNA site specifically Science 240, 1198– 1201
- 11Zein, N., Poncin, M., Nilakantan, R., and Ellestad, G. A. (1989) Calicheamicin γ 1I and DNA: Molecular recognition process responsible for site-specificity Science 244, 697– 699
- 12Mah, S. C., Price, M. A., Townsend, C. A., and Tullius, T. D. (1994) Features of DNA recognition for oriented binding and cleavage by calicheamicin Tetrahedron 50, 1361– 1378
- 13Biggins, J. B., Prudent, J. R., Marshall, D. J., and Thorson, J. S. (2006) A continuous assay for DNA cleavage using molecular break lights Methods Mol. Biol. 335, 83– 92[PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xjt1yrsLs%253D&md5=15a1230e3506d16698194bcaa77b65a4A continuous assay for DNA cleavage using molecular break lightsBiggins, John B.; Prudent, James R.; Marshall, David J.; Thorson, Jon S.Methods in Molecular Biology (Totowa, NJ, United States) (2006), 335 (Fluorescent Energy Transfer Nucleic Acid Probes), 83-92CODEN: MMBIED; ISSN:1064-3745. (Humana Press Inc.)Exploring the properties of mols. that cleave DNA (i.e., enzymic nucleases, chem. footprinting agents, and naturally occurring DNA cleaving antibiotics) has been an ongoing process with benefits extending toward both lab. and clin. applications. Despite the progress that has been made toward understanding the mechanics of DNA cleavage, a simple and continuous assay for detecting DNA cleavage has been lacking. Herein, the authors describe the mol. break light assay, wherein a single oligonucleotide modified by a 5'-fluorophore-3'-quencher pair adopting a stem-loop structure with an appropriate DNA recognition site, provides for the rapid assaying of DNA cleavage with high sensitivity. Furthermore, the described methodol. is highly convenient in that it is readily adaptable to common lab. fluorometers and multi-well plate/array systems, which may provide the basis for high-throughput screening of novel DNA cleaving agents. This assay may also be further extended to natural or "unnatural" transcription factor protection assay systems.
- 14Danishefsky, S. J. and Shair, M. D. (1996) Observations in the chemistry and biology of cyclic enediyne antibiotics: Total syntheses of calicheamicin γ1I and dynemicin A J. Org. Chem. 61, 16– 44
- 15Nicolaou, K. C., Hale, C. R. H., and Nilewski, C. (2012) A total synthesis trilogy: Calicheamicin γ1(I), Taxol, and brevetoxin A Chem. Rec. 12, 407– 441[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XoslKjsbc%253D&md5=d3605d104978d33d5ea7a81a7bf9e967A total synthesis trilogy: Calicheamicin γ1I, Taxol and brevetoxin ANicolaou, K. C.; Hale, Christopher R. H.; Nilewski, ChristianChemical Record (2012), 12 (4), 407-441CODEN: CRHEAK; ISSN:1527-8999. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Detailed behind-the-scenes accounts of the total syntheses of calicheamicin γ1I, Taxol and brevetoxin A are discussed with particular emphasis placed on strategies and tactics employed in these campaigns. DOI 10.1002/tcr.201200005.
- 16Ahlert, J., Shepard, E., Lomovskaya, N., Zazopoulos, E., Staffa, A., Bachmann, B. O., Huang, K., Fonstein, L., Czisny, A., Whitwam, R. E., Farnet, C. M., and Thorson, J. S. (2002) The calicheamicin gene cluster and its iterative type I enediyne PKS Science 297, 1173– 1176[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XmsVOht70%253D&md5=76ae8b38e6de9d1db318ca6538b09ac5The calicheamicin gene cluster and its iterative type I enediyne PKSAhlert, Joachim; Shepard, Erica; Lomovskaya, Natalia; Zazopoulos, Emmanuel; Staffa, Alfredo; Bachmann, Brian O.; Huang, Kexue; Fonstein, Leonid; Czisny, Anne; Whitwam, Ross E.; Farnet, Chris M.; Thorson, Jon S.Science (Washington, DC, United States) (2002), 297 (5584), 1173-1176CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The enediynes exemplify nature's ingenuity. We have cloned and characterized the biosynthetic locus coding for perhaps the most notorious member of the nonchromoprotein enediyne family, calicheamicin. This gene cluster contains an unusual polyketide synthase (PKS) that is demonstrated to be essential for enediyne biosynthesis. Comparison of the calicheamicin locus with the locus encoding the chromoprotein enediyne C-1027 reveals that the enediyne PKS is highly conserved among these distinct enediyne families. Contrary to previous hypotheses, this suggests that the chromoprotein and nonchromoprotein enediynes are generated by similar biosynthetic pathways.
- 17Liu, W., Ahlert, J., Gao, Q., Wendt-Pienkowski, E., Shen, B., and Thorson, J. S. (2003) Rapid PCR amplification of minimal enediyne polyketide synthase cassettes leads to a predictive familial classification model Proc. Natl. Acad. Sci. U.S.A. 100, 11959– 11963[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXotlGltL4%253D&md5=d50cfaa0360d5904aa9c7b0ea11b669dRapid PCR amplification of minimal enediyne polyketide synthase cassettes leads to a predictive familial classification modelLiu, Wen; Ahlert, Joachim; Gao, Qunjie; Wendt-Pienkowski, Evelyn; Shen, Ben; Thorson, Jon S.Proceedings of the National Academy of Sciences of the United States of America (2003), 100 (21), 11959-11963CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)A universal PCR method for the rapid amplification of minimal enediyne polyketide synthase (PKS) genes and the application of this methodol. to clone remaining prototypical genes from producers of structurally detd. enediynes in both family types are presented. A phylogenetic anal. of the new pool of bona fide enediyne PKS genes, consisting of three from 9-membered producers (neocarzinostatin, C1027, and maduropeptin) and three from 10-membered producers (calicheamicin, dynemicin, and esperamicin), reveals a clear genotypic distinction between the two structural families from which to form a predictive model. The results from this study support the postulation that the minimal enediyne PKS helps define the structural divergence of the enediyne core and provides the key tools for generating enediyne hybrid genes/mol. scaffolds; by using the model, a classification is also provided for the unknown enediyne PKS genes previously identified via genome scanning.
- 18Biggins, J. B., Onwueme, K. C., and Thorson, J. S. (2003) Resistance to enediyne antitumor antibiotics by CalC self-sacrifice Science 301, 1537– 1541
- 19Singh, S., Hager, M. H., Zhang, C., Griffith, B. R., Lee, M. S., Hallenga, K., Markley, J. L., and Thorson, J. S. (2006) Structural insight into the self-sacrifice mechanism of enediyne resistance ACS Chem. Biol. 1, 451– 460[ACS Full Text
], [CAS], Google Scholar
19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xot12mtbk%253D&md5=58226c7592f92d01dee192a28000ee15Structural insight into the self-sacrifice mechanism of enediyne resistanceSingh, Shanteri; Hager, Martin H.; Zhang, Changsheng; Griffith, Byron R.; Lee, Min S.; Hallenga, Klaas; Markley, John L.; Thorson, Jon S.ACS Chemical Biology (2006), 1 (7), 451-460CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The recent discovery of the first "self-sacrifice" mechanism for bacterial resistance to the enediyne antitumor antibiotics, where enediyne-induced proteolysis of the resistance protein CalC inactivates both the highly reactive metabolite and the resistance protein, revealed yet another ingenious bacterial mechanism for controlling reactive metabolites. As reported herein, the first 3D structures of CalC and CalC in complex with calicheamicin (CLM) divulge CalC to be a member of the steroidogenic acute regulatory protein (StAR)-related transfer (START) domain superfamily. In contrast to previous studies of proteins known to bind DNA-damaging natural products (e.g., bleomycins, mitomycins, and 9-membered chromoprotein enediynes), this is the 1st demonstrated involvement of a START domain fold. Consistent with the CalC self-sacrifice mechanism, CLM in complex with CalC is positioned for direct H abstraction from Gly113 to initiate the oxidative proteolysis-based resistance mechanism. These structural studies also illuminate, for the 1st time, a small DNA-binding region within CalC that may serve to localize CalC to the enediyne target (DNA). Given the role of START domains in nuclear/cytosolic transport and translocation, this structural study also may implicate START domains as post-endocytotic intracellular chaperones for enediyne-based therapeutics such as MyloTarg. - 20Zhang, C., Griffith, B. R., Fu, Q., Albermann, C., Fu, X., Lee, I.-K., Li, L., and Thorson, J. S. (2006) Exploiting the reversibility of natural product glycosyltransferase-catalyzed reactions Science 313, 1291– 1294[Crossref], [PubMed], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XoslOntb0%253D&md5=5f2b83f0499627c28f3ddfbf16880a97Exploiting the Reversibility of Natural Product Glycosyltransferase-Catalyzed ReactionsZhang, Changsheng; Griffith, Byron R.; Fu, Qiang; Albermann, Christoph; Fu, Xun; Lee, In-Kyoung; Li, Lingjun; Thorson, Jon S.Science (Washington, DC, United States) (2006), 313 (5791), 1291-1294CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Glycosyltransferases (GTs), an essential class of ubiquitous enzymes, are generally perceived as unidirectional catalysts. In contrast, we report that four glycosyltransferases from two distinct natural product biosynthetic pathways-calicheamicin and vancomycin-readily catalyze reversible reactions, allowing sugars and aglycons to be exchanged with ease. As proof of the broader applicability of these new reactions, more than 70 differentially glycosylated calicheamicin and vancomycin variants are reported. This study suggests the reversibility of GT-catalyzed reactions may be general and useful for generating exotic nucleotide sugars, establishing in vitro GT activity in complex systems, and enhancing natural product diversity.
- 21Johnson, H. D. and Thorson, J. S. (2008) Characterization of CalE10, the N-oxidase involved in calicheamicin hydroxyaminosugar formation J. Am. Chem. Soc. 130, 17662– 17663
- 22Belecki, K., Crawford, J. M., and Townsend, C. A. (2009) Production of octaketide polyenes by the calicheamicin polyketide synthase CalE8: Implications for the biosynthesis of enediyne core structures J. Am. Chem. Soc. 131, 12564– 12566[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVSisL7N&md5=ed8ce733ad40968e31b760f64e42465bProduction of Octaketide Polyenes by the Calicheamicin Polyketide Synthase CalE8: Implications for the Biosynthesis of Enediyne Core StructuresBelecki, Katherine; Crawford, Jason M.; Townsend, Craig A.Journal of the American Chemical Society (2009), 131 (35), 12564-12566CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Enediyne antibiotics are categorized according to the presence of either a 9- or 10-membered ring within their polyketide-derived core structures. Recent literature reports have favored the notion that biosynthetic divergence of the two structural families is detd. by the enediyne polyketide synthases (PKSs) alone. We now disclose the simultaneous in vitro prodn. of three octaketide polyenes by biosynthetic enzymes for the 10-membered enediyne calicheamicin γ1I, including the elusive β-keto acid precursor to a previously described C15 Me hexaenone. Alongside these two polyene products, we have addnl. detected a hydrocarbon heptaene previously isolated only from 9-membered enediyne systems. The discovery of the heptaene in the calicheamicin system promotes a more convergent model for the early steps of enediyne biosynthesis. Furthermore, the synthesis of this set of octaketides by the enediyne PKS CalE8 and thioesterase CalE7 suggests, in contrast to recent biosynthetic proposals, that accessory enzymes may be necessary to initiate differentiation to 9- or 10-membered enediyne precursors, either by modulation of enediyne PKS activity or by interception and modification of polyketide chain-extension intermediates. - 23Horsman, G. P., Chen, Y., Thorson, J. S., and Shen, B. (2010) Polyketide synthase chemistry does not direct biosynthetic divergence between 9- and 10-membered enediynes Proc. Natl. Acad. Sci. U.S.A. 107, 11331– 11335
- 24Gantt, R. W., Peltier-Pain, P., Singh, S., Zhou, M., and Thorson, J. S. (2013) Broadening the scope of glycosyltransferase-catalyzed sugar nucleotide synthesis Proc. Natl. Acad. Sci. U.S.A. 110, 7648– 7653[Crossref], [PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFGrtL8%253D&md5=bfd133c9e815a7c25d31b9b463e83e04Broadening the scope of glycosyltransferase-catalyzed sugar nucleotide synthesisGantt, Richard W.; Peltier-Pain, Pauline; Singh, Shanteri; Zhou, Maoquan; Thorson, Jon S.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (19), 7648-7653, S7648/1-S7648/72CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We described the integration of the general reversibility of glycosyltransferase-catalyzed reactions, artificial glycosyl donors, and a high throughput colorimetric screen to enable the engineering of glycosyltransferases for combinatorial sugar nucleotide synthesis. The best engineered catalyst from this study, the OleD Loki variant, contained the mutations P67T/I112P/T113M/S132F/A242I compared with the OleD wild-type sequence. Evaluated against the parental sequence OleD TDP16 variant used for screening, the OleD Loki variant displayed max. improvements in kcat/Km of >400-fold and >15-fold for formation of NDP-glucoses and UDP-sugars, resp. This OleD Loki variant also demonstrated efficient turnover with five variant NDP acceptors and six variant 2-chloro-4-nitrophenyl glycoside donors to produce 30 distinct NDP-sugars. This study highlights a convenient strategy to rapidly optimize glycosyltransferase catalysts for the synthesis of complex sugar nucleotides and the practical synthesis of a unique set of sugar nucleotides.
- 25Belecki, K. and Townsend, C. A. (2012) Environmental control of the calicheamicin polyketide synthase leads to detection of a programmed octaketide and a proposal for enediyne biosynthesis Angew. Chem., Int. Ed. 51, 11316– 11319
- 26Belecki, K. and Townsend, C. A. (2013) Biochemical determination of enzyme-bound metabolites: Preferential accumulation of a programmed octaketide on the enediyne polyketide synthase CalE8 J. Am. Chem. Soc. 135, 14339– 14348
- 27Ricart, A. D. (2011) Antibody-drug conjugates of calicheamicin derivative: Gemtuzumab Ozogamicin and Inotuzumab Ozogamicin Clin. Cancer Res. 17, 6417– 6427[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlSksrzF&md5=103b40cfe49632a86367414e0e97c011Antibody-Drug Conjugates of Calicheamicin Derivative: Gemtuzumab Ozogamicin and Inotuzumab OzogamicinRicart, Alejandro D.Clinical Cancer Research (2011), 17 (20), 6417-6427CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)A review. Antibody-drug conjugates (ADC) are an attractive approach for the treatment of acute myeloid leukemia and non-Hodgkin lymphomas, which in most cases, are inherently sensitive to cytotoxic agents. CD33 and CD22 are specific markers of myeloid leukemias and B-cell malignancies, resp. These endocytic receptors are ideal for an ADC strategy because they can effectively carry the cytotoxic payload into the cell. Gemtuzumab ozogamicin (GO, Mylotarg) and inotuzumab ozogamicin consist of a deriv. of calicheamicin (a potent DNA-binding cytotoxic antibiotic) linked to a humanized monoclonal IgG4 antibody directed against CD33 or CD22, resp. Both of these ADCs have a target-mediated pharmacokinetic disposition. GO was the first drug to prove the ADC concept in the clinic, specifically in phase II studies that included substantial proportions of older patients with relapsed acute myeloid leukemia. In contrast, in phase III studies, it has thus far failed to show clin. benefit in first-line treatment in combination with std. chemotherapy. Inotuzumab ozogamicin has shown remarkable clin. activity in relapsed/refractory B-cell non-Hodgkin lymphoma, and it has started phase III evaluation. The safety profile of these ADCs includes reversible myelosuppression (esp. neutropenia and thrombocytopenia), elevated hepatic transaminases, and hyperbilirubinemia. There have been postmarketing reports of hepatotoxicity, esp. veno-occlusive disease, assocd. with GO. The incidence is ∼2%, but patients who undergo hematopoietic stem cell transplantation have an increased risk. As we steadily move toward the goal of personalized medicine, these kinds of agents will provide a unique opportunity to treat selected patient subpopulations based on the expression of their specific tumor targets. Clin Cancer Res; 17(20); 6417-27.
- 28Trail, P. A. (2013) Antibody drug conjugates as cancer therapeutics Antibodies 2, 113– 129
- 29Krissinel, E. and Henrick, K. (2007) Inference of macromolecular assemblies from crystalline state J. Mol. Biol. 372, 774– 797[Crossref], [PubMed], [CAS], Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXpvFGktb8%253D&md5=a5c764cfc7dc129f53ddc31ef9d475faInference of Macromolecular Assemblies from Crystalline StateKrissinel, Evgeny; Henrick, KimJournal of Molecular Biology (2007), 372 (3), 774-797CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)The authors discuss basic phys.-chem. principles underlying the formation of stable macromol. complexes, which in many cases are likely to be the biol. units performing a certain physiol. function. The authors also consider available theor. approaches to the calcn. of macromol. affinity and entropy of complexation. The latter is shown to play an important role and make a major effect on complex size and symmetry. The authors develop a new method, based on chem. thermodn., for automatic detection of macromol. assemblies in the Protein Data Bank (PDB) entries that are the results of x-ray diffraction expts. As found, biol. units may be recovered at 80-90% success rate, which makes x-ray crystallog. an important source of exptl. data on macromol. complexes and protein-protein interactions. The method is implemented as a public WWW service (http://www.ebi.ac.uk/msd-srv/prot_int/pistart.html).
- 30Iyer, L. M., Koonin, E. V., and Aravind, L. (2001) Adaptations of the helix-grip fold for ligand binding and catalysis in the START domain superfamily Proteins 43, 134– 144
- 31Thorsell, A.-G., Lee, W. H., Persson, C., Siponen, M. I., Nilsson, M., Busam, R. D., Kotenyova, T., Schüler, H., and Lehtiö, L. (2011) Comparative structural analysis of lipid binding START domains PloS One 6, e19521
- 32Alpy, F. and Tomasetto, C. (2005) Give lipids a START: The StAR-related lipid transfer (START) domain in mammals J. Cell Sci. 118, 2791– 2801[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXnsVCjtrw%253D&md5=3b0a2eed630ac8e48b7fa6076a9d0266Give lipids a START: The StAR-related lipid transfer (START) domain in mammalsAlpy, Fabien; Tomasetto, CatherineJournal of Cell Science (2005), 118 (13), 2791-2801CODEN: JNCSAI; ISSN:0021-9533. (Company of Biologists Ltd.)A review. The steroidogenic acute regulatory protein (StAR)-related lipid transfer (START) domain is a protein module of ∼210 residues that binds lipids, including sterols. Fifteen mammalian proteins, STARD1-STARD15, possess a START domain and these can be grouped into 6 subfamilies. Cholesterol, 25-hydroxycholesterol, phosphatidylcholine, phosphatidylethanolamine, and ceramides are ligands for STARD1/STARD3/STARD5, STARD5, STARD2/STARD10, STARD10 and STARD11, resp. The lipids or sterols bound by the remaining 9 START proteins are unknown. Recent studies have shown that the C-terminal end of the domain plays a fundamental role, forming a lid over a deep lipid-binding pocket that shields the ligand from the external environment. The START domain can be regarded as a lipid-exchange and/or a lipid-sensing domain. Mammalian START proteins have diverse expression patterns and can be found free in the cytoplasm, attached to membranes or in the nucleus. They appear to function in a variety of distinct physiol. processes, such as lipid transfer between intracellular compartments, lipid metab., and modulation of signaling events. Mutation or misexpression of START proteins is linked to pathol. processes, including genetic disorders, autoimmune disease, and cancer.
- 33Holm, L. and Rosenström, P. (2010) Dali server: Conservation mapping in 3D Nucleic Acids Res. 38, W545– 549[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXotVSqsr4%253D&md5=b7832262058d6a4f7d362f4ce565a666Dali server: conservation mapping in 3DHolm, Liisa; Rosenstrom, PaiviNucleic Acids Research (2010), 38 (Web Server), W545-W549CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Our web site (http://ekhidna.biocenter.helsinki.fi/daliserver) runs the Dali program for protein structure comparison. The web site consists of three parts: (i) the Dali server compares newly solved structures against structures in the Protein Data Bank (PDB), (ii) the Dali database allows browsing precomputed structural neighborhoods and (iii) the pairwise comparison generates suboptimal alignments for a pair of structures. Each part has its own query form and a common format for the results page. The inputs are either PDB identifiers or novel structures uploaded by the user. The results pages are hyperlinked to aid interactive anal. The web interface is simple and easy to use. The key purpose of interactive anal. is to check whether conserved residues line up in multiple structural alignments and how conserved residues and ligands cluster together in multiple structure superimpositions. In favorable cases, protein structure comparison can lead to evolutionary discoveries not detected by sequence anal.
- 34Krissinel, E. and Henrick, K. (2004) Secondary-structure matching (SSM), a new tool for fast protein structure alignment in three dimensions Acta Crystallogr., Sect. D: Biol. Crystallogr. 60, 2256– 2268[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVars7rL&md5=219f539e65e5fbc2cc631f937f544953Secondary-structure matching (SSM), a new tool for fast protein structure alignment in three dimensionsKrissinel, E.; Henrick, K.Acta Crystallographica, Section D: Biological Crystallography (2004), D60 (12, Pt. 1), 2256-2268CODEN: ABCRE6; ISSN:0907-4449. (Blackwell Publishing Ltd.)The present paper describes the SSM algorithm of protein structure comparison in three dimensions, which includes an original procedure of matching graphs built on the protein's secondary-structure elements, followed by an iterative three-dimensional alignment of protein backbone Cα atoms. The SSM results are compared with those obtained from other protein comparison servers, and the advantages and disadvantages of different scores that are used for structure recognition are discussed. A new score, balancing the r.m.s.d. and alignment length Nalign, is proposed. It is found that different servers agree reasonably well on the new score, while showing considerable differences in r.m.s.d. and Nalign.
- 35Ames, B. D., Korman, T. P., Zhang, W., Smith, P., Vu, T., Tang, Y., and Tsai, S.-C. (2008) Crystal structure and functional analysis of tetracenomycin ARO/CYC: Implications for cyclization specificity of aromatic polyketides Proc. Natl. Acad. Sci. U.S.A. 105, 5349– 5354
- 36Michalska, K., Fernandes, H., Sikorski, M., and Jaskolski, M. (2010) Crystal structure of Hyp-1, a St. John’s Wort protein implicated in the biosynthesis of hypericin J. Struct. Biol. 169, 161– 171
- 37Ilari, A., Franceschini, S., Bonamore, A., Arenghi, F., Botta, B., Macone, A., Pasquo, A., Bellucci, L., and Boffi, A. (2009) Structural basis of enzymatic (S)-norcoclaurine biosynthesis J. Biol. Chem. 284, 897– 904[Crossref], [PubMed], [CAS], Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsV2ntg%253D%253D&md5=01e0a2f7c5b59fa572b524d07938bd30Structural basis of enzymatic (S)-norcoclaurine biosynthesisIlari, Andrea; Franceschini, Stefano; Bonamore, Alessandra; Arenghi, Fabio; Botta, Bruno; Macone, Alberto; Pasquo, Alessandra; Bellucci, Luca; Boffi, AlbertoJournal of Biological Chemistry (2009), 284 (2), 897-904CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Norcoclaurine synthase (NCS) catalyzes the stereospecific Pictet-Spengler cyclization between dopamine and 4-hydroxyphenylacetaldehyde, the key step in the benzylisoquinoline alkaloid biosynthetic pathway. Here, the crystallog. structure of NCS from Thalictrum flavum in its complex with its substrate, dopamine, and the nonreactive substrate analog, 4-hydroxybenzaldehyde, was solved at 2.1 Å resoln. NCS shared no common features with the functionally correlated "Pictet-Spenglerases" that catalyze the 1st step of the indole alkaloid pathways and conformed to the overall fold of the Bet v1-like protein. The active site of NCS was located within a 20-Å-long catalytic tunnel and was shaped by the side-chains of the Tyr-108, Lys-122, Asp-141, and Glu-110 residues. The geometry of the amino acid side-chains with respect to the substrates revealed the structural determinants that govern the mechanism of the stereoselective Pictet-Spengler cyclization, thus establishing an excellent foundation for the understanding of the finer details of the catalytic process. Site-directed mutations of the relevant residues confirmed the assignment based on the crystallog. findings.
- 38Kofler, S., Asam, C., Eckhard, U., Wallner, M., Ferreira, F., and Brandstetter, H. (2012) Crystallographically mapped ligand binding differs in high and low IgE binding isoforms of birch pollen allergen bet v 1 J. Mol. Biol. 422, 109– 123
- 39Li, W., Wang, L., Sheng, X., Yan, C., Zhou, R., Hang, J., Yin, P., and Yan, N. (2013) Molecular basis for the selective and ABA-independent inhibition of PP2CA by PYL13 Cell Res. 23, 1369– 1379[Crossref], [PubMed], [CAS], Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWqu7nK&md5=06d90b8ae13d792940f0f7a15ff2efedMolecular basis for the selective and ABA-independent inhibition of PP2CA by PYL13Li, Wenqi; Wang, Li; Sheng, Xinlei; Yan, Chuangye; Zhou, Rui; Hang, Jing; Yin, Ping; Yan, NiengCell Research (2013), 23 (12), 1369-1379CODEN: CREEB6; ISSN:1001-0602. (NPG Nature Asia-Pacific)PYR1/PYL/RCAR family proteins (PYLs) are well-characterized abscisic acid (ABA) receptors. Among the 14 PYL members in Arabidopsis thaliana, PYL13 is ABA irresponsive and its function has remained elusive. Here, we show that PYL13 selectively inhibits the phosphatase activity of PP2CA independent of ABA. The crystal structure of PYL13-PP2CA complex, which was detd. at 2.4 Å resoln., elucidates the mol. basis for the specific recognition between PP2CA and PYL13. In addn. to the canonical interactions between PYLs and PP2Cs, an extra interface is identified involving an element in the vicinity of a previously uncharacterized CCCH zinc-finger (ZF) motif in PP2CA. Sequence blast identified another 56 ZF-contg. PP2Cs, all of which are from plants. The structure also reveals the mol. determinants for the ABA irresponsiveness of PYL13. Finally, biochem. anal. suggests that PYL13 may hetero-oligomerize with PYL10. These two PYLs antagonize each other in their resp. ABA-independent inhibitions of PP2Cs. The biochem. and structural studies provide important insights into the function of PYL13 in the stress response of plant and set up a foundation for future biotechnol. applications of PYL13.
- 40Pasternak, O., Bujacz, G. D., Fujimoto, Y., Hashimoto, Y., Jelen, F., Otlewski, J., Sikorski, M. M., and Jaskolski, M. (2006) Crystal structure of Vigna radiata cytokinin-specific binding protein in complex with zeatin Plant Cell 18, 2622– 2634
- 41Schirmer, T., Hoffimann-Sommergrube, K., Susani, M., Breiteneder, H., and Marković-Housley, Z. (2005) Crystal structure of the major celery allergen Api g 1: Molecular analysis of cross-reactivity J. Mol. Biol. 351, 1101– 1109
- 42Osipiuk, J., Wu, R., Moy, S., and Joachimiak, A. (2006) X-ray crystal structure of conserved hypothetical protein EF_2215 from . PDB ID: 2NN5. Unpublished.Google ScholarThere is no corresponding record for this reference.
- 43Singarapu, K., Eletsky, A., Sathyamoorthy, B., Sukumaran, D., Wang, D., Jiang, M., Ciccosanti, C., Xiao, R., Liu, J., Baran, M. C., Swapna, G., Acton, T. B., Rost, B., Montelione, G. T., and Szyperski, T. (2008) Solution NMR structure of protein encoded by gene BPP1335 from . PDB ID: 2K5G. Unpublished.Google ScholarThere is no corresponding record for this reference.
- 44Clark, B. J. (2012) The mammalian START domain protein family in lipid transport in health and disease J. Endocrinol. 212, 257– 275[Crossref], [PubMed], [CAS], Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XktlCrsb8%253D&md5=d3d9f925a2198cb3d6e53b15be5a3fecThe mammalian START domain protein family in lipid transport in health and diseaseClark, Barbara J.Journal of Endocrinology (2012), 212 (3), 257-275CODEN: JOENAK; ISSN:0022-0795. (BioScientifica Ltd.)A review. Lipid transfer proteins of the steroidogenic acute regulatory protein-related lipid transfer (START) domain family are defined by the presence of a conserved ∼210 amino acid sequence that folds into an α/β helix-grip structure forming a hydrophobic pocket for ligand binding. The mammalian START proteins bind diverse ligands, such as cholesterol, oxysterols, phospholipids, sphingolipids, and possibly fatty acids, and have putative roles in non-vesicular lipid transport, thioesterase enzymic activity, and tumor suppression. However, the biol. functions of many members of the START domain protein family are not well established. Recent research has focused on characterizing the cell-type distribution and regulation of the START proteins, examg. the specificity and directionality of lipid transport, and identifying disease states assocd. with dysregulation of START protein expression. This review summarizes the current concepts of the proposed physiol. and pathol. roles for the mammalian START domain proteins in cholesterol and lipid trafficking.
- 45Chang, A., Singh, S., Helmich, K. E., Goff, R. D., Bingman, C. A., Thorson, J. S., and Phillips, G. N., Jr. (2011) Complete set of glycosyltransferase structures in the calicheamicin biosynthetic pathway reveals the origin of regiospecificity Proc. Natl. Acad. Sci. U.S.A. 108, 17649– 17654
- 46Gerlt, J. A. and Babbitt, P. C. (2000) Can sequence determine function? Genome Biol. 1, 1– 10
- 47Whisstock, J. C. and Lesk, A. M. (2003) Prediction of protein function from protein sequence and structure Q. Rev. Biophys. 36, 307– 340[Crossref], [PubMed], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtlSjsw%253D%253D&md5=9540779034f73da3a6bf8fc32a130b32Prediction of protein function from protein sequence and structureWhisstock, James C.; Lesk, Arthur M.Quarterly Reviews of Biophysics (2003), 36 (3), 307-340CODEN: QURBAW; ISSN:0033-5835. (Cambridge University Press)A review. The sequence of a genome contains the plans of the possible life of an organism, but implementation of genetic information depends on the functions of the proteins and nucleic acids that it encodes. Many individual proteins of known sequence and structure present challenges to the understanding of their function. In particular, a no. of genes responsible for diseases have been identified but their specific functions are unknown. Whole-genome sequencing projects are a major source of proteins of unknown function. Annotation of a genome involves assignment of functions to gene products, in most cases on the basis of amino-acid sequence alone. 3D structure can aid the assignment of function, motivating the challenge of structural genomics projects to make structural information available for novel uncharacterized proteins. Structure-based identification of homologs often succeeds where sequence-alone-based methods fail, because in many cases evolution retains the folding pattern long after sequence similarity becomes undetectable. Nevertheless, prediction of protein function from sequence and structure is a difficult problem, because homologous proteins often have different functions. Many methods of function prediction rely on identifying similarity in sequence and/or structure between a protein of unknown function and one or more well-understood proteins. Alternative methods include inferring conservation patterns in members of a functionally uncharacterized family for which many sequences and structures are known. However, these inferences are tenuous. Such methods provide reasonable guesses at function, but are far from foolproof. It is therefore fortunate that the development of whole-organism approaches and comparative genomics permits other approaches to function prediction when the data are available. These include the use of protein-protein interaction patterns, and correlations between occurrences of related proteins in different organisms, as indicators of functional properties. Even if it is possible to ascribe a particular function to a gene product, the protein may have multiple functions. A fundamental problem is that function is in many cases an ill-defined concept. In this article we review the state of the art in function prediction and describe some of the underlying difficulties and successes.
- 48Schnoes, A. M., Brown, S. D., Dodevski, I., and Babbitt, P. C. (2009) Annotation error in public databases: Misannotation of molecular function in enzyme superfamilies PLoS Comput. Biol. 5, e1000605[Crossref], [PubMed], [CAS], Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1Mfis1SgtA%253D%253D&md5=1b018d0becdbf937c32bbfc10c611399Annotation error in public databases: misannotation of molecular function in enzyme superfamiliesSchnoes Alexandra M; Brown Shoshana D; Dodevski Igor; Babbitt Patricia CPLoS computational biology (2009), 5 (12), e1000605 ISSN:.Due to the rapid release of new data from genome sequencing projects, the majority of protein sequences in public databases have not been experimentally characterized; rather, sequences are annotated using computational analysis. The level of misannotation and the types of misannotation in large public databases are currently unknown and have not been analyzed in depth. We have investigated the misannotation levels for molecular function in four public protein sequence databases (UniProtKB/Swiss-Prot, GenBank NR, UniProtKB/TrEMBL, and KEGG) for a model set of 37 enzyme families for which extensive experimental information is available. The manually curated database Swiss-Prot shows the lowest annotation error levels (close to 0% for most families); the two other protein sequence databases (GenBank NR and TrEMBL) and the protein sequences in the KEGG pathways database exhibit similar and surprisingly high levels of misannotation that average 5%-63% across the six superfamilies studied. For 10 of the 37 families examined, the level of misannotation in one or more of these databases is >80%. Examination of the NR database over time shows that misannotation has increased from 1993 to 2005. The types of misannotation that were found fall into several categories, most associated with "overprediction" of molecular function. These results suggest that misannotation in enzyme superfamilies containing multiple families that catalyze different reactions is a larger problem than has been recognized. Strategies are suggested for addressing some of the systematic problems contributing to these high levels of misannotation.
- 49Baker, D. and Sali, A. (2001) Protein structure prediction and structural genomics Science 294, 93– 96[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXnsFyisbY%253D&md5=8b476f801ccb6223835f91e420edf57fProtein structure prediction and structural genomicsBaker, David; Sali, AndrejScience (Washington, DC, United States) (2001), 294 (5540), 93-96CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Genome sequencing projects are producing linear amino acid sequences, but full understanding of the biol. role of these proteins will require knowledge of their structure and function. Although exptl. structure detn. methods are providing high-resoln. structure information about a subset of the proteins, computational structure prediction methods will provide valuable information for the large fraction of sequences whose structures will not be detd. exptl. The first class of protein structure prediction methods, including threading and comparative modeling, rely on detectable similarity spanning most of the modeled sequence and at least one known structure. The second class of methods, de novo or ab initio methods, predict the structure from sequence alone, without relying on similarity at the fold level between the modeled sequence and any of the known structures. In this Viewpoint, we begin by describing the essential features of the methods, the accuracy of the models, and their application to the prediction and understanding of protein function, both for single proteins and on the scale of whole genomes. We then discuss the important role that protein structure prediction methods play in the growing worldwide effort in structural genomics.
- 50Hermann, J. C., Marti-Arbona, R., Fedorov, A. A., Fedorov, E., Almo, S. C., Shoichet, B. K., and Raushel, F. M. (2007) Structure-based activity prediction for an enzyme of unknown function Nature 448, 775– 779[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptVGrs7w%253D&md5=b0bd6612147fb848f49f8b9ccfd8384eStructure-based activity prediction for an enzyme of unknown functionHermann, Johannes C.; Marti-Arbona, Ricardo; Fedorov, Alexander A.; Fedorov, Elena; Almo, Steven C.; Shoichet, Brian K.; Raushel, Frank M.Nature (London, United Kingdom) (2007), 448 (7155), 775-779CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)With many genomes sequenced, a pressing challenge in biol. is predicting the function of the proteins that the genes encode. When proteins are unrelated to others of known activity, bioinformatics inference for function becomes problematic. It would thus be useful to interrogate protein structures for function directly. Here, we predict the function of an enzyme of unknown activity, Tm0936 from Thermotoga maritima, by docking high-energy intermediate forms of thousands of candidate metabolites. The docking hit list was dominated by adenine analogs, which appeared to undergo C6-deamination. Four of these, including 5-methylthioadenosine and S-adenosylhomocysteine (SAH), were tested as substrates, and three had substantial catalytic rate consts. (105 M-1 s-1). The x-ray crystal structure of the complex between Tm0936 and the product resulting from the deamination of SAH, S-inosylhomocysteine, was detd., and it corresponded closely to the predicted structure. The deaminated products can be further metabolized by T. maritima in a previously uncharacterized SAH degrdn. pathway. Structure-based docking with high-energy forms of potential substrates may be a useful tool to annotate enzymes for function.
- 51Guichou, J.-F. and Labesse, G. (2012) Fragment and conquer: From structure to complexes to function Structure 20, 1617– 1619
- 52Wright, G. D. (2007) The antibiotic resistome: The nexus of chemical and genetic diversity Nat. Rev. Microbiol. 5, 175– 186[Crossref], [PubMed], [CAS], Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhslOgur8%253D&md5=e98711cc6fee6a9713ad4fcfa9638d73The antibiotic resistome: the nexus of chemical and genetic diversityWright, Gerard D.Nature Reviews Microbiology (2007), 5 (3), 175-186CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)A review. Over the millennia, microorganisms have evolved evasion strategies to overcome a myriad of chem. and environmental challenges, including antimicrobial drugs. Even before the first clin. use of antibiotics more than 60 years ago, resistant organisms had been isolated. Moreover, the potential problem of the widespread distribution of antibiotic resistant bacteria was recognized by scientists and health-care specialists from the initial use of these drugs. Why is resistance inevitable and where does it come from. Understanding the mol. diversity that underlies resistance will inform our use of these drugs and guide efforts to develop new efficacious antibiotics.
- 53Cundliffe, E. and Demain, A. L. (2010) Avoidance of suicide in antibiotic-producing microbes J. Ind. Microbiol. Biotechnol. 37, 643– 672[Crossref], [PubMed], [CAS], Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsFGkt7c%253D&md5=714376ebb17316b0edd827dd2017fa39Avoidance of suicide in antibiotic-producing microbesCundliffe, Eric; Demain, Arnold L.Journal of Industrial Microbiology & Biotechnology (2010), 37 (7), 643-672CODEN: JIMBFL; ISSN:1367-5435. (Springer)A review. Many microbes synthesize potentially autotoxic antibiotics, mainly as secondary metabolites, against which they need to protect themselves. This is done in various ways, ranging from target-based strategies (i.e., modification of normal drug receptors or de novo synthesis of the latter in drug-resistant form) to the adoption of metabolic shielding and/or efflux strategies that prevent drug-target interactions. These self-defense mechanisms have been studied most intensively in antibiotic-producing prokaryotes, of which the most prolific are the actinomycetes. Only a few documented examples pertain to lower eukaryotes while higher organisms have hardly been addressed in this context. Thus, many plant alkaloids, variously described as herbivore repellents or nitrogen excretion devices, are truly antibiotics-even if toxic to humans. As just one example, bulbs of Narcissus spp. (including the King Alfred daffodil) accumulate narciclasine that binds to the larger subunit of the eukaryotic ribosome and inhibits peptide bond formation. However, ribosomes in the Amaryllidaceae have not been tested for possible resistance to narciclasine and other alkaloids. Clearly, the prevalence of suicide avoidance is likely to extend well beyond the remit of the present article.
- 54Allan, C. M., Hill, S., Morvaridi, S., Saiki, R., Johnson, J. S., Liau, W.-S., Hirano, K., Kawashima, T., Ji, Z., Loo, J. A., Shepherd, J. N., and Clarke, C. F. (2013) A conserved START domain coenzyme Q-binding polypeptide is required for efficient Q biosynthesis, respiratory electron transport, and antioxidant function in Saccharomyces cerevisiae Biochim. Biophys. Acta 1831, 776– 791
- 55Baker, J. R., Woolfson, D. N., Muskett, F. W., Stoneman, R. G., Urbaniak, M. D., and Caddick, S. (2007) Protein–small molecule interactions in neocarzinostatin, the prototypical enediyne chromoprotein antibiotic ChemBioChem 8, 704– 717
- 56Weigel, L. M., Clewell, D. B., Gill, S. R., Clark, N. C., McDougal, L. K., Flannagan, S. E., Kolonay, J. F., Shetty, J., Killgore, G. E., and Tenover, F. C. (2003) Genetic analysis of a high-level vancomycin-resistant isolate of Staphylococcus aureus Science 302, 1569– 1571[Crossref], [PubMed], [CAS], Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpt1SmsLk%253D&md5=62c5066b5d48e0249f84f8838eed8d61Genetic analysis of a high-level vancomycin-resistant isolate of Staphylococcus aureusWeigel, Linda M.; Clewell, Don B.; Gill, Steven R.; Clark, Nancye C.; McDougal, Linda K.; Flannagan, Susan E.; Kolonay, James F.; Shetty, Jyoti; Killgore, George E.; Tenover, Fred C.Science (Washington, DC, United States) (2003), 302 (5650), 1569-1571CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Vancomycin is usually reserved for treatment of serious infections, including those caused by multidrug-resistant Staphylococcus aureus. A clin. isolate of S. aureus with high-level resistance to vancomycin (minimal inhibitory concn. = 1024 μg/mL) was isolated in June 2002. This isolate harbored a 57.9-kilobase multiresistance conjugative plasmid within which Tn1546 (vanA) was integrated. Addnl. elements on the plasmid encoded resistance to trimethoprim (dfrA), β-lactams (blaZ), aminoglycosides (aacA-aphD), and disinfectants (qacC). Genetic analyses suggest that the long-anticipated transfer of vancomycin resistance to a methicillin-resistant S. aureus occurred in vivo by interspecies transfer of Tn1546 from a co-isolate of Enterococcus faecalis.
- 57Qureshi, N. K., Yin, S., and Boyle-Vavra, S. (2014) The role of the Staphylococcal VraTSR regulatory system on vancomycin resistance and vanA operon expression in vancomycin-resistant Staphylococcus aureus PloS One 9, e85873
- 58The PyMOL Molecular Graphics System, Version 1.3r1. (2010, August) Schrödinger, LLC., New York.Google ScholarThere is no corresponding record for this reference.
- 59Kearse, M., Moir, R., Wilson, A., Stones-Havas, S., Cheung, M., Sturrock, S., Buxton, S., Cooper, A., Markowitz, S., Duran, C., Thierer, T., Ashton, B., Meintjes, P., and Drummond, A. (2012) Geneious Basic: An integrated and extendable desktop software platform for the organization and analysis of sequence data Bioinformatics 28, 1647– 1649[Crossref], [PubMed], [CAS], Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38rovFKhtg%253D%253D&md5=284aaf2baa0b23d4f12aaa80316acfeeGeneious Basic: an integrated and extendable desktop software platform for the organization and analysis of sequence dataKearse Matthew; Moir Richard; Wilson Amy; Stones-Havas Steven; Cheung Matthew; Sturrock Shane; Buxton Simon; Cooper Alex; Markowitz Sidney; Duran Chris; Thierer Tobias; Ashton Bruce; Meintjes Peter; Drummond AlexeiBioinformatics (Oxford, England) (2012), 28 (12), 1647-9 ISSN:.UNLABELLED: The two main functions of bioinformatics are the organization and analysis of biological data using computational resources. Geneious Basic has been designed to be an easy-to-use and flexible desktop software application framework for the organization and analysis of biological data, with a focus on molecular sequences and related data types. It integrates numerous industry-standard discovery analysis tools, with interactive visualizations to generate publication-ready images. One key contribution to researchers in the life sciences is the Geneious public application programming interface (API) that affords the ability to leverage the existing framework of the Geneious Basic software platform for virtually unlimited extension and customization. The result is an increase in the speed and quality of development of computation tools for the life sciences, due to the functionality and graphical user interface available to the developer through the public API. Geneious Basic represents an ideal platform for the bioinformatics community to leverage existing components and to integrate their own specific requirements for the discovery, analysis and visualization of biological data. AVAILABILITY AND IMPLEMENTATION: Binaries and public API freely available for download at http://www.geneious.com/basic, implemented in Java and supported on Linux, Apple OSX and MS Windows. The software is also available from the Bio-Linux package repository at http://nebc.nerc.ac.uk/news/geneiousonbl.
- 60Acton, T. B., Xiao, R., Anderson, S., Aramini, J., Buchwald, W. A., Ciccosanti, C., Conover, K., Everett, J., Hamilton, K., Huang, Y. J., Janjua, H., Kornhaber, G., Lau, J., Lee, D. Y., Liu, G., Maglaqui, M., Ma, L., Mao, L., Patel, D., Rossi, P., Sahdev, S., Shastry, R., Swapna, G. V. T., Tang, Y., Tong, S., Wang, D., Wang, H., Zhao, L., and Montelione, G. T. (2011) Preparation of protein samples for NMR structure, function, and small-molecule screening studies Methods Enzymol. 493, 21– 60[Crossref], [PubMed], [CAS], Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXosleku7g%253D&md5=d46f532743d27d482799c9e51b3e0ed2Preparation of protein samples for NMR structure, function, and small-molecule screening studiesActon, Thomas B.; Xiao, Rong; Anderson, Stephen; Aramini, James; Buchwald, William A.; Ciccosanti, Colleen; Conover, Ken; Everett, John; Hamilton, Keith; Huang, Yuanpeng Janet; Janjua, Haleema; Kornhaber, Gregory; Lau, Jessica; Lee, Dong Yup; Liu, Gaohua; Maglaqui, Melissa; Ma, Lichung; Mao, Lei; Patel, Dayaban; Rossi, Paolo; Sahdev, Seema; Shastry, Ritu; Swapna, G. V. T.; Tang, Yeufeng; Tong, Saichiu; Wang, Dongyan; Wang, Huang; Zhao, Li; Montelione, Gaetano T.Methods in Enzymology (2011), 493 (Fragment-Based Drug Design), 21-60CODEN: MENZAU; ISSN:0076-6879. (Elsevier Inc.)A review. In this chapter, we conc. on the prodn. of high-quality protein samples for NMR studies, in particular, we provide an in-depth description of recent advances in the prodn. of NMR samples and their synergistic use with recent advancements in NMR hardware. We describe the protein prodn. platform of the Northeast Structural Genomics Consortium and outline our high-throughput strategies for producing high-quality protein samples for NMR studies. Our strategy is based on the cloning, expression, and purifn. of 6 × -His-tagged proteins using T7-based Escherichia coli systems and isotope enrichment in minimal media. We describe 96-well ligation-independent cloning and anal. expression systems, parallel preparative scale fermn., and high-throughput purifn. protocols. The 6 × -His affinity tag allows for a similar two-step purifn. procedure implemented in a parallel high-throughput fashion that routinely results in purity levels sufficient for NMR studies (>97% homogeneity). Using this platform, the protein open reading frames of over 17,500 different targeted proteins (or domains) have been cloned as over 28,000 constructs. Nearly 5000 of these proteins have been purified to homogeneity in tens of milligram quantities, resulting in more than 950 new protein structures, including more than 400 NMR structures, deposited in the Protein Data Bank. The Northeast Structural Genomics Consortium pipeline has been effective in producing protein samples of both prokaryotic and eukaryotic origin. Although this chapter describes our entire pipeline for producing isotope-enriched protein samples, it focuses on the major updates introduced during the last 5 years (Phase 2 of the National Institute of General Medical Sciences Protein Structure Initiative). Our advanced automated and/or parallel cloning, expression, purifn., and biophys. screening technologies are suitable for implementation in a large individual lab. or by a small group of collaborating investigators for structural biol., functional proteomics, ligand screening, and structural genomics research.
- 61Xiao, R., Anderson, S., Aramini, J., Belote, R., Buchwald, W. A., Ciccosanti, C., Conover, K., Everett, J. K., Hamilton, K., Huang, Y. J., Janjua, H., Jiang, M., Kornhaber, G. J., Lee, D. Y., Locke, J. Y., Ma, L.-C., Maglaqui, M., Mao, L., Mitra, S., Patel, D., Rossi, P., Sahdev, S., Sharma, S., Shastry, R., Swapna, G. V. T., Tong, S. N., Wang, D., Wang, H., Zhao, L., Montelione, G. T., and Acton, T. B. (2010) The high-throughput protein sample production platform of the Northeast Structural Genomics Consortium J. Struct. Biol. 172, 21– 33
- 62Luft, J. R., Wolfley, J. R., Said, M. I., Nagel, R. M., Lauricella, A. M., Smith, J. L., Thayer, M. H., Veatch, C. K., Snell, E. H., Malkowski, M. G., and Detitta, G. T. (2007) Efficient optimization of crystallization conditions by manipulation of drop volume ratio and temperature Protein Sci. Publ. Protein Soc. 16, 715– 722[Crossref], [PubMed], [CAS], Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXktVOmt7o%253D&md5=0be93bed89cf4b708f473d54f20a77ddEfficient optimization of crystallization conditions by manipulation of drop volume ratio and temperatureLuft, Joseph R.; Wolfley, Jennifer R.; Said, Meriem I.; Nagel, Raymond M.; Lauricella, Angela M.; Smith, Jennifer L.; Thayer, Max H.; Veatch, Christina K.; Snell, Edward H.; Malkowski, Michael G.; Detitta, George T.Protein Science (2007), 16 (4), 715-722CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)An efficient optimization method for the crystn. of biol. macromols. has been developed and tested. This builds on a successful high-throughput technique for the detn. of initial crystn. conditions. The optimization method takes an initial condition identified through screening and then varies the concn. of the macromol., precipitant, and the growth temp. in a systematic manner. The amt. of sample and no. of steps is minimized and no biochem. reformulation is required. In the current application a robotic liq. handling system enables high-throughput use, but the technique can easily be adapted in a nonautomated setting. This method has been applied successfully for the rapid optimization of crystn. conditions in nine representative cases.
- 63Otwinowski, Z. and Minor, W. (1997) Processing of X-ray diffraction data collected in oscillation mode Methods Enzymol. 276, 307– 326[Crossref], [PubMed], [CAS], Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXivFehsbw%253D&md5=c9536971d4e32cc35352c40fb9368131Processing of x-ray diffraction data collected in oscillation modeOtwinowski, Zbyszek; Minor, WladekMethods in Enzymology (1997), 276 (Macromolecular Crystallography, Part A), 307-326CODEN: MENZAU; ISSN:0076-6879. (Academic)Macromol. crystallog. is an iterative process. Rarely do the first crystals provide all the necessary data to solve the biol. problem being studied. Each step benefits from experience learned in previous steps. To monitor the progress, the HKL package provides 2 tools: (1) statistics, both weighted (χ2) and unweighted (R-merge), are provided, and the Bayesian reasoning and multicomponent error model facilitates obtaining the proper error ests. and (2) visualization of the process plays a double role by helping the operator to confirm that the process of data redn., including the resulting statistics, is correct, and allowing one to evaluate problems for which there are no good statistical criteria. Visualization also provides confidence that the point of diminishing returns in data collection and redn. has been reached. At that point, the effort should be directed to solving the structure. The methods presented here have been applied to solve a large variety of problems, from inorg. mols. with 5 Å unit cell to rotavirus of 700 Å diam. crystd. in 700 × 1000 × 1400 Å cell. Overall quality of the method was tested by many researchers by successful application of the programs to MAD structure detns.
- 64Sheldrick, G. M. (2010) Experimental phasing with SHELXC/D/E: Combining chain tracing with density modification Acta Crystallogr., Sect. D: Biol. Crystallogr. 66, 479– 485[Crossref], [PubMed], [CAS], Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFKisb4%253D&md5=0b73bdbc20100b89fe7de5aaf357aab6Experimental phasing with SHELXC/D/E: combining chain tracing with density modificationSheldrick, George M.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (4), 479-485CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)The programs SHELXC, SHELXD and SHELXE are designed to provide simple, robust and efficient exptl. phasing of macromols. by the SAD, MAD, SIR, SIRAS and RIP methods and are particularly suitable for use in automated structure-soln. pipelines. This paper gives a general account of exptl. phasing using these programs and describes the extension of iterative d. modification in SHELXE by the inclusion of automated protein main-chain tracing. This gives a good indication as to whether the structure has been solved and enables interpretable maps to be obtained from poorer starting phases. The autotracing algorithm starts with the location of possible seven-residue α-helixes and common tripeptides. After extension of these fragments in both directions, various criteria are used to decide whether to accept or reject the resulting poly-Ala traces. Noncrystallog. symmetry (NCS) is applied to the traced fragments, not to the d. Further features are the use of a 'no-go' map to prevent the traces from passing through heavy atoms or symmetry elements and a splicing technique to combine the best parts of traces (including those generated by NCS) that partly overlap.
- 65Terwilliger, T. C. and Berendzen, J. (1999) Automated MAD and MIR structure solution Acta Crystallogr., Sect. D: Biol. Crystallogr. 55, 849– 861
- 66Emsley, P., Lohkamp, B., Scott, W. G., and Cowtan, K. (2010) Features and development of Coot Acta Crystallogr., Sect. D: Biol. Crystallogr. 66, 486– 501[Crossref], [PubMed], [CAS], Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFKisb8%253D&md5=67262cbfc60004de5ef962d5c043c910Features and development of CootEmsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (4), 486-501CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)Coot is a mol.-graphics application for model building and validation of biol. macromols. The program displays electron-d. maps and at. models and allows model manipulations such as idealization, real-space refinement, manual rotation/translation, rigid-body fitting, ligand search, solvation, mutations, rotamers and Ramachandran idealization. Furthermore, tools are provided for model validation as well as interfaces to external programs for refinement, validation and graphics. The software is designed to be easy to learn for novice users, which is achieved by ensuring that tools for common tasks are 'discoverable' through familiar user-interface elements (menus and toolbars) or by intuitive behavior (mouse controls). Recent developments have focused on providing tools for expert users, with customisable key bindings, extensions and an extensive scripting interface. The software is under rapid development, but has already achieved very widespread use within the crystallog. community. The current state of the software is presented, with a description of the facilities available and of some of the underlying methods employed.
- 67Murshudov, G. N., Vagin, A. A., and Dodson, E. J. (1997) Refinement of macromolecular structures by the maximum-likelihood method Acta Crystallogr., Sect. D: Biol. Crystallogr. 53, 240– 255[Crossref], [PubMed], [CAS], Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXjs1Gnsb4%253D&md5=ec7f141ce1542f7ff458b98ecfe3f8afRefinement of macromolecular structures by the maximum-likelihood methodMurshudov, Garib N.; Vagin, Alexei A.; Dodson, Eleanor J.Acta Crystallographica, Section D: Biological Crystallography (1997), D53 (3), 240-255CODEN: ABCRE6; ISSN:0907-4449. (Munksgaard)A review with many refs. on the math. basis of max. likelihood. The likelihood function for macromol. structures is extended to include prior phase information and exptl. std. uncertainties. The assumption that different parts of a structure might have different errors is considered. A method for estg. σA using "free" reflections is described and its effects analyzed. The derived equations have been implemented in the program REFMAC. This has been tested on several proteins at different stages of refinement (bacterial α-amylase, cytochrome c', cross-linked insulin and oligopeptide binding protein). The results derived using the max.-likelihood residual are consistently better than those obtained from least-squares refinement.
- 68Laskowski, R., MacArthur, M., Moss, D., and Thornton, J. (1993) PROCHECK: A program to check the stereochemical quality of protein structures J. Appl. Crystallogr. 26, 283– 291[Crossref], [CAS], Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXit12lurY%253D&md5=fb69fc4410cd716aaaa7cc0db06b3ed2PROCHECK: a program to check the stereochemical quality of protein structuresLaskowski, Roman A.; MacArthur, Malcolm W.; Moss, David S.; Thornton, Janet M.Journal of Applied Crystallography (1993), 26 (2), 283-91CODEN: JACGAR; ISSN:0021-8898.The PROCHECK suite of programs provides a detailed check on the stereochem. of a protein structure. Its outputs comprise a no. of plots in PostScript format and a comprehensive residue-by-residue listing. These give an assessment of the overall quality of the structure as compared with well-refined structures of the same resoln. and also highlight regions that may need further investigation. The PROCHECK programs are useful for assessing the quality not only of protein structures in the process of being solved but also of existing structures and of those being modeled on known structures.
- 69Shi, J., Wang, Y., Zeng, L., Wu, Y., Deng, J., Zhang, Q., Lin, Y., Li, J., Kang, T., Tao, M., Rusinova, E., Zhang, G., Wang, C., Zhu, H., Yao, J., Zeng, Y.-X., Evers, B. M., Zhou, M.-M., and Zhou, B. P. (2014) Disrupting the interaction of BRD4 with diacetylated twist suppresses tumorigenesis in basal-like breast cancer Cancer Cell 25, 210– 225[Crossref], [PubMed], [CAS], Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitlCisrs%253D&md5=d207cbc1abb187a53d807452b4f10d8fDisrupting the Interaction of BRD4 with Diacetylated Twist Suppresses Tumorigenesis in Basal-like Breast CancerShi, Jian; Wang, Yifan; Zeng, Lei; Wu, Yadi; Deng, Jiong; Zhang, Qiang; Lin, Yiwei; Li, Junlin; Kang, Tiebang; Tao, Min; Rusinova, Elena; Zhang, Guangtao; Wang, Chi; Zhu, Haining; Yao, Jun; Zeng, Yi-Xin; Evers, B. Mark; Zhou, Ming-Ming; Zhou, Binhua P.Cancer Cell (2014), 25 (2), 210-225CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)Twist is a key transcription activator of epithelial-mesenchymal transition (EMT). It remains unclear how Twist induces gene expression. Here we report a mechanism by which Twist recruits BRD4 to direct WNT5A expression in basal-like breast cancer (BLBC). Twist contains a "histone H4-mimic" GK-X-GK motif that is diacetylated by Tip60. The diacetylated Twist binds the second bromodomain of BRD4, whose first bromodomain interacts with acetylated H4, thereby constructing an activated Twist/BRD4/P-TEFb/RNA-Pol II complex at the WNT5A promoter and enhancer. Pharmacol. inhibition of the Twist-BRD4 assocn. reduced WNT5A expression and suppressed invasion, cancer stem cell (CSC)-like properties, and tumorigenicity of BLBC cells. Our study indicates that the interaction with BRD4 is crit. for the oncogenic function of Twist in BLBC.
- 70Zhai, J., Ström, A.-L., Kilty, R., Venkatakrishnan, P., White, J., Everson, W. V., Smart, E. J., and Zhu, H. (2009) Proteomic characterization of lipid raft proteins in amyotrophic lateral sclerosis mouse spinal cord FEBS J. 276, 3308– 3323
- 71Chen, V. B., Arendall, W. B., 3rd, Headd, J. J., Keedy, D. A., Immormino, R. M., Kapral, G. J., Murray, L. W., Richardson, J. S., and Richardson, D. C. (2010) MolProbity: All-atom structure validation for macromolecular crystallography Acta Crystallogr., Sect. D: Biol. Crystallogr. 66, 12– 21[Crossref], [PubMed], [CAS], Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXit1Kktg%253D%253D&md5=b5fc7574f43f01dd6e43c3663ca4f779MolProbity: all-atom structure validation for macromolecular crystallographyChen, Vincent B.; Arendall, W. Bryan, III; Headd, Jeffrey J.; Keedy, Daniel A.; Immormino, Robert M.; Kapral, Gary J.; Murray, Laura W.; Richardson, Jane S.; Richardson, David C.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (1), 12-21CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)MolProbity is a structure-validation web service that provides broad-spectrum solidly based evaluation of model quality at both the global and local levels for both proteins and nucleic acids. It relies heavily on the power and sensitivity provided by optimized hydrogen placement and all-atom contact anal., complemented by updated versions of covalent-geometry and torsion-angle criteria. Some of the local corrections can be performed automatically in MolProbity and all of the diagnostics are presented in chart and graphical forms that help guide manual rebuilding. X-ray crystallog. provides a wealth of biol. important mol. data in the form of at. three-dimensional structures of proteins, nucleic acids and increasingly large complexes in multiple forms and states. Advances in automation, in everything from crystn. to data collection to phasing to model building to refinement, have made solving a structure using crystallog. easier than ever. However, despite these improvements, local errors that can affect biol. interpretation are widespread at low resoln. and even high-resoln. structures nearly all contain at least a few local errors such as Ramachandran outliers, flipped branched protein side chains and incorrect sugar puckers. It is crit. both for the crystallographer and for the end user that there are easy and reliable methods to diagnose and correct these sorts of errors in structures. MolProbity is the authors' contribution to helping solve this problem and this article reviews its general capabilities, reports on recent enhancements and usage, and presents evidence that the resulting improvements are now beneficially affecting the global database.
Cited By
This article is cited by 22 publications.
- Paramita Pal, Jamie R. Alley, Craig A. Townsend. Examining Heterodimerization by Aryl C–N Coupling in Dynemicin Biosynthesis. ACS Chemical Biology 2023, Article ASAP.
- Nagaraju Mupparapu, Lauren Brewster, Katrina F. Ostrom, Sherif I. Elshahawi. Late‐Stage Chemoenzymatic Installation of Hydroxy‐Bearing Allyl Moiety on the Indole Ring of Tryptophan‐Containing Peptides. Chemistry – A European Journal 2022, 28 (20) https://doi.org/10.1002/chem.202104614
- Xiaohui Yan. Anthraquinone-fused enediynes: discovery, biosynthesis and development. Natural Product Reports 2022, 39 (3) , 703-728. https://doi.org/10.1039/D1NP00054C
- Dulce Ramírez-Rendon, Ajit Kumar Passari, Beatriz Ruiz-Villafán, Romina Rodríguez-Sanoja, Sergio Sánchez, Arnold L. Demain. Impact of novel microbial secondary metabolites on the pharma industry. Applied Microbiology and Biotechnology 2022, 106 (5-6) , 1855-1878. https://doi.org/10.1007/s00253-022-11821-5
- Eshita Khera, Shujun Dong, Haolong Huang, Laureen de Bever, Floris L. van Delft, Greg M. Thurber. Cellular-Resolution Imaging of Bystander Payload Tissue Penetration from Antibody-Drug Conjugates. Molecular Cancer Therapeutics 2022, 21 (2) , 310-321. https://doi.org/10.1158/1535-7163.MCT-21-0580
- Abigael J. Kosgei, Mitchell D. Miller, Minakshi Bhardwaj, Weijun Xu, Jon S. Thorson, Steven G. Van Lanen, George N. Phillips. The crystal structure of DynF from the dynemicin-biosynthesis pathway of Micromonospora chersina. Acta Crystallographica Section F Structural Biology Communications 2022, 78 (1) , 1-7. https://doi.org/10.1107/S2053230X21012322
- Sarah K. Alvarado, Mitchell D. Miller, Minakshi Bhardwaj, Jon S. Thorson, Steven G. Van Lanen, George N. Phillips. Structural characterization of DynU16, a START/Bet v1-like protein involved in dynemicin biosynthesis. Acta Crystallographica Section F Structural Biology Communications 2021, 77 (10) , 328-333. https://doi.org/10.1107/S2053230X21008943
- Nagaraju Mupparapu, Yu‐Hsin Cindy Lin, Tae Ho Kim, Sherif I. Elshahawi. Regiospecific Synthesis of Calcium‐Independent Daptomycin Antibiotics using a Chemoenzymatic Method. Chemistry – A European Journal 2021, 27 (12) , 4176-4182. https://doi.org/10.1002/chem.202005100
- Ajeeth Adhikari, Christiana N. Teijaro, Craig A. Townsend, Ben Shen. Biosynthesis of Enediyne Natural Products. 2020, 365-414. https://doi.org/10.1016/B978-0-12-409547-2.14651-7
- Hiroshi Ogawara. Comparison of Antibiotic Resistance Mechanisms in Antibiotic-Producing and Pathogenic Bacteria. Molecules 2019, 24 (19) , 3430. https://doi.org/10.3390/molecules24193430
- Timothy A. Wencewicz. Crossroads of Antibiotic Resistance and Biosynthesis. Journal of Molecular Biology 2019, 431 (18) , 3370-3399. https://doi.org/10.1016/j.jmb.2019.06.033
- Ellis C. O’Neill, Michelle Schorn, Charles B. Larson, Natalie Millán-Aguiñaga. Targeted antibiotic discovery through biosynthesis-associated resistance determinants: target directed genome mining. Critical Reviews in Microbiology 2019, 45 (3) , 255-277. https://doi.org/10.1080/1040841X.2019.1590307
- Xian-Feng Hou, Yu-Jiao Song, Mei Zhang, Wenxian Lan, Song Meng, Chunxi Wang, Hai-Xue Pan, Chunyang Cao, Gong-Li Tang. Enzymology of Anthraquinone-γ-Pyrone Ring Formation in Complex Aromatic Polyketide Biosynthesis. Angewandte Chemie 2018, 130 (41) , 13663-13667. https://doi.org/10.1002/ange.201806729
- Xian-Feng Hou, Yu-Jiao Song, Mei Zhang, Wenxian Lan, Song Meng, Chunxi Wang, Hai-Xue Pan, Chunyang Cao, Gong-Li Tang. Enzymology of Anthraquinone-γ-Pyrone Ring Formation in Complex Aromatic Polyketide Biosynthesis. Angewandte Chemie International Edition 2018, 57 (41) , 13475-13479. https://doi.org/10.1002/anie.201806729
- Elodie Tenconi, Sébastien Rigali. Self-resistance mechanisms to DNA-damaging antitumor antibiotics in actinobacteria. Current Opinion in Microbiology 2018, 45 , 100-108. https://doi.org/10.1016/j.mib.2018.03.003
- Chin-Yuan Chang, Xiaohui Yan, Ivana Crnovcic, Thibault Annaval, Changsoo Chang, Boguslaw Nocek, Jeffrey D. Rudolf, Dong Yang, Hindra, Gyorgy Babnigg, Andrzej Joachimiak, George N. Phillips, Ben Shen. Resistance to Enediyne Antitumor Antibiotics by Sequestration. Cell Chemical Biology 2018, 25 (9) , 1075-1085.e4. https://doi.org/10.1016/j.chembiol.2018.05.012
- Yinyin Cai, Zhijun Liao, Ying Ju, Juan Liu, Yong Mao, Xiangrong Liu. Resistance gene identification from Larimichthys crocea with machine learning techniques. Scientific Reports 2016, 6 (1) https://doi.org/10.1038/srep38367
- Jeffrey D Rudolf, Xiaohui Yan, Ben Shen. Genome neighborhood network reveals insights into enediyne biosynthesis and facilitates prediction and prioritization for discovery. Journal of Industrial Microbiology and Biotechnology 2016, 43 (2-3) , 261-276. https://doi.org/10.1007/s10295-015-1671-0
- John K. Everett, Roberto Tejero, Sarath B. K. Murthy, Thomas B. Acton, James M. Aramini, Michael C. Baran, Jordi Benach, John R. Cort, Alexander Eletsky, Farhad Forouhar, Rongjin Guan, Alexandre P. Kuzin, Hsiau-Wei Lee, Gaohua Liu, Rajeswari Mani, Binchen Mao, Jeffrey L. Mills, Alexander F. Montelione, Kari Pederson, Robert Powers, Theresa Ramelot, Paolo Rossi, Jayaraman Seetharaman, David Snyder, G. V. T. Swapna, Sergey M. Vorobiev, Yibing Wu, Rong Xiao, Yunhuang Yang, Cheryl H. Arrowsmith, John F. Hunt, Michael A. Kennedy, James H. Prestegard, Thomas Szyperski, Liang Tong, Gaetano T. Montelione. A community resource of experimental data for NMR / X-ray crystal structure pairs. Protein Science 2016, 25 (1) , 30-45. https://doi.org/10.1002/pro.2774
- M.S. Vela Gurovic. Isolation and Identification of Enediynes and Cycloaromatized Derivatives. 2016, 29-62. https://doi.org/10.1016/B978-0-444-63603-4.00002-4
- Lu Han, Shanteri Singh, Jon S. Thorson, George N. Phillips. Loop dynamics of thymidine diphosphate-rhamnose 3′-O-methyltransferase (CalS11), an enzyme in calicheamicin biosynthesis. Structural Dynamics 2016, 3 (1) , 012004. https://doi.org/10.1063/1.4941368
- Sherif I. Elshahawi, Khaled A. Shaaban, Madan K. Kharel, Jon S. Thorson. A comprehensive review of glycosylated bacterial natural products. Chemical Society Reviews 2015, 44 (21) , 7591-7697. https://doi.org/10.1039/C4CS00426D
Abstract
Figure 1
Figure 1. (A) Selected structures of naturally occurring 10-membered enediynes. The “warhead” is highlighted in red. (B) Proposed mechanism of cycloaromatization of 1 and its effect on DNA scission and CalC self-sacrifice mechanism.
Figure 2
Figure 2. Structure of CalU16. (A) Overlay of NMR (green) and monomers of the crystal (brick- red) structures of CalU16. The N- and C-termini of the protein are labeled, while the dynamic loop is colored yellow. (B) Monomer with secondary structural elements labeled. The residues in the hydrophobic cavity are represented as stick models. (C) B-factors for the Cα atoms in the crystal structure (left) and Cα RMSD values from the NMR ensemble (right) are mapped to the color and tube diameter of “putty” traces showing the general agreement (correlation coefficient 0.559) between the structures.
Figure 3
Figure 3. CalU16 structural homologues. (A) CalU16 (PDB: 4FPW); (B) CalC (PDB: 2L65), calicheamicin resistance protein; (C) TcmN Aro/Cyc (in complex with trans-dihydroquercetin; PDB: 3TVQ) involved in the biosynthesis of tetracenomycin; (D) Hyp-1 (in complex with ethylene glycol; PDB: 3IE5) involved in the biosynthesis of hypericin; (E) NCS (in complex with hydroxybenzaldehyde; PDB: 2VQ5) involved in the biosynthesis of norcoclaurine.
Figure 4
Figure 4. CalU16 and CalU19 assays. Serial disc dilutions of 1 against (A) pSE28a-E. coli (control), (B) pSECalU16-E. coli (CalU16), (C) pSECalU19-E. coli (CalU19), (D) pJB2011-E. coli (CalC). Amount of 1 on discs 1–6 are 10 μg, 1 μg, 100 ng, 50 ng, 10 ng, and 1 ng, respectively. Coomassie-stained 8–12% SDS-PAGE gradient gel of (E) CalU16 and (F) CalU19 in the presence of DTT (lane 1), 1 (lane 2), and DTT and 1 (lane 3).
Figure 5
Figure 5. Site directed mutagenesis of CalU16 and CalU19. Docking models of (A) CalC (B) CalU16, and (C) CalU19 with mutated glycine residues represented as spheres where colored Gly residues indicate cleavage sites, wheat Gly residues indicate mutations that did not affect activity and calicheamicin (1) is represented as a stick model. Results of disc diffusion assay in CalU16 mutants (D) pSE28a-E. coli (control); (E) pSEU16G128V-E. coli; (F) pSEU16G128R-E. coli; (G) pSECalU16-E. coli; and CalU19 mutants (H) pSE28a-E. coli (control); (I) pSEU19G177V-E. coli; (J) pSEU19G177R-E. coli; (K) pSECalU19-E. coli. Amount of 1 on discs 1–6 are 10 μg, 1 μg, 100 ng, 50 ng, 10 ng, and 1 ng, respectively.
References
ARTICLE SECTIONSThis article references 71 other publications.
- 1Maiese, W. M., Lechevalier, M. P., Lechevalier, H. A., Korshalla, J., Kuck, N., Fantini, A., Wildey, M. J., Thomas, J., and Greenstein, M. (1989) Calicheamicins, a novel family of antitumor antibiotics: taxonomy, fermentation and biological properties J. Antibiot. (Tokyo) 42, 558– 563[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXkt1SmurY%253D&md5=8944bd3a3bc333517c3ee816f01436bdCalicheamicins, a novel family of antitumor antibiotics: taxonomy, fermentation and biological propertiesMaiese, W. M.; Lechevalier, M. P.; Lechevalier, H. A.; Korshalla, J.; Kuck, N.; Fantini, A.; Wildey, M. J.; Thomas, J.; Greenstein, M.Journal of Antibiotics (1989), 42 (4), 558-63CODEN: JANTAJ; ISSN:0021-8820.A novel family of antitumor antibiotics, the calicheamicins, were isolated from the fermn. broth of Micromonospora echinospora subsp. calichensis. These antibiotics exhibited significant activity against gram-pos. and gram-neg. bacteria in vitro. Calicheamicin γ1I demonstrated antitumor activity against P388 leukemia and B16 melanoma in vivo.
- 2Lee, M. D., Manning, J. K., Williams, D. R., Kuck, N. A., Testa, R. T., and Borders, D. B. (1989) Calicheamicins, a novel family of antitumor antibiotics. 3. Isolation, purification and characterization of calicheamicins β 1Br, γ 1Br, α 2I, α 3I, β 1I, γ 1I, and δ 1I J. Antibiot. (Tokyo) 42, 1070– 1087[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL1MXlsFShsbg%253D&md5=2cb74746c1ed1b8f4ff887316d8b0e1fCalichemicins, a novel family of antitumor antibiotics. 3. Isolation, purification and characterization of calichemicins β1Br, γ1Br, α2I, α3I, β1I, γ1I, and δ1ILee, May D.; Manning, Joann K.; Williams, David R.; Kuck, Nydia A.; Testa, Raymond T.; Borders, Donald B.Journal of Antibiotics (1989), 42 (7), 1070-87CODEN: JANTAJ; ISSN:0021-8820.Novel antitumor antibiotics, calichemicins β1Br (I), γ1Br, α2I, α3I, β1I, γ1I, and δ1I, were recovered from the fermn. broth of Micromonospora echinospora calichensis by solvent extn., selective pptn., normal phase, reversed-phase, and partition chromatog. The individual components were characterized by their UV, IR, 1H, and 13C NMR spectra data.
- 3Lee, M. D., Dunne, T. S., Chang, C. C., Siegel, M. M., Morton, G. O., Ellestad, G. A., McGahren, W. J., and Borders, D. B. (1992) Calicheamicins, a novel family of antitumor antibiotics. 4. Structure elucidation of calicheamicins β 1Br, γ 1Br, α 2I, α 3I, β 1I, γ 1I, and δ 1I J. Am. Chem. Soc. 114, 985– 997
- 4Thorson, J. S., Sievers, E. L., Ahlert, J., Shepard, E., Whitwam, R. E., Onwueme, K. C., and Ruppen, M. (2000) Understanding and exploiting nature’s chemical arsenal: The past, present, and future of calicheamicin research Curr. Pharm. Des. 6, 1841– 1879[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXoslWntLo%253D&md5=8db2ae788ecfabec1e422181c508c689Understanding and exploiting nature's chemical arsenal: the past, present and future of calicheamicin researchThorson, Jon S.; Sievers, Eric L.; Ahlert, Joachim; Shepard, Erica; Whitwam, Ross E.; Onwueme, Kenolisa C.; Ruppen, MarkCurrent Pharmaceutical Design (2000), 6 (18), 1841-1879CODEN: CPDEFP; ISSN:1381-6128. (Bentham Science Publishers)A review with 234 refs. The enediyne antitumor antibiotics are appreciated for their novel mol. architecture, their remarkable biol. activity and their fascinating mode of action and many have spawned considerable interest as anticancer agents in the pharmaceutical industry. Of equal importance to these astonishing properties, the enediynes also offer a distinct opportunity to study the unparalleled biosyntheses of their unique mol. scaffolds and what promises to be unprecedented modes of self-resistance to highly reactive natural products. Elucidation of these aspects should unveil novel mechanistic enzymol., and may provide access to the rational biosynthetic modification of enediyne structure for new drug leads, the construction of enediyne overproducing strains and eventually lead to an enediyne combinatorial biosynthesis program. This article strives to compile and present the crit. research discoveries relevant to the clin. most promising enediyne, calicheamicin, from a historical perspective. Recent progress, particularly in the areas of biosynthesis, self-resistance, bio-engineering analogs and clin. studies are also highlighted.
- 5Galm, U., Hager, M. H., Van Lanen, S. G., Ju, J., Thorson, J. S., and Shen, B. (2005) Antitumor antibiotics: Bleomycin, enediynes, and mitomycin Chem. Rev. 105, 739– 758[ACS Full Text
], [CAS], Google Scholar
5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXmslCrug%253D%253D&md5=7e21e51479e19b723d92113c80c58e51Antitumor Antibiotics: Bleomycin, Enediynes, and MitomycinGalm, Ute; Hager, Martin H.; Van Lanen, Steven G.; Ju, Jianhua; Thorson, Jon S.; Shen, BenChemical Reviews (Washington, DC, United States) (2005), 105 (2), 739-758CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. The review discusses discovery, biol. activities and resistance to bleomycin, enediynes, and mitomycin. - 6Liang, Z.-X. (2010) Complexity and simplicity in the biosynthesis of enediyne natural products Nat. Prod. Rep. 27, 499– 528[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXjslGntrg%253D&md5=a235f078d92c01217637b8b8816f781cComplexity and simplicity in the biosynthesis of enediyne natural productsLiang, Zhao-XunNatural Product Reports (2010), 27 (4), 499-528CODEN: NPRRDF; ISSN:0265-0568. (Royal Society of Chemistry)A review. The review highlights the recent progress on the elucidation of the biosynthetic mechanism of enediyne natural products. A large body of knowledge has been gathered from the genetic, biochem. and structural studies since the sequencing of the first two enediyne gene clusters in 2002. Primary attention is devoted to the novel pathways and enzymes in the biosynthesis of the enediyne warhead as well as the peripheral moieties.
- 7De Voss, J. J., Hangeland, J. J., and Townsend, C. A. (1990) Characterization of the in vitro cyclization chemistry of calicheamicin and its relation to DNA cleavage J. Am. Chem. Soc. 112, 4554– 4556[ACS Full Text
], [CAS], Google Scholar
7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXktFOmt7s%253D&md5=63fc400ddd02e5829adc9e80379cf355Characterization of the in vitro cyclization chemistry of calicheamicin and its relation to DNA cleavageDe Voss, James J.; Hangeland, Jon J.; Townsend, Craig A.Journal of the American Chemical Society (1990), 112 (11), 4554-6CODEN: JACSAT; ISSN:0002-7863.The proposed mechanism of thiol activation of calicheamicin γ1I (I), one of the diynene antitumor antibiotics, was examd. in variable-temp. NMR studies. Sep. reactions of the drug with Me thioglycolate and tri-n-butylphosphine led to the identification and characterization of a reactive intermediate resulting from cleavage of the allylic methyltrisulfide and intramol. β-addn. of the resulting thio(ate) to the enone system. This intermediate undergoes 1st-order decompn. at a convenient rate at about -10°. The end product of this process contains 2 H atoms abstracted from the medium consistent with the intervention of a 1,4-diyl formed in the proposed Bergman rearrangement. The relatively long soln. half-life of this intermediate (calcd. to be 4.5 s at 37° in conjunction with other findings led to the formulation of a kinetic scheme to evaluate thermodn. and kinetic factors in the sequence-selective cleavages of DNA characteristic of calicheamicin. - 8Chatterjee, M., Cramer, K. D., and Townsend, C. A. (1993) Kinetic nature of thiol activation in DNA cleavage by calicheamicin J. Am. Chem. Soc. 115, 3374– 3375
- 9Myers, A. G., Cohen, S. B., and Kwon, B. M. (1994) A study of the reaction of calicheamicin γ 1 with glutathione in the presence of double-stranded DNA J. Am. Chem. Soc. 116, 1255– 1271
- 10Zein, N., Sinha, A. M., McGahren, W. J., and Ellestad, G. A. (1988) Calicheamicin γ 1I: An antitumor antibiotic that cleaves double-stranded DNA site specifically Science 240, 1198– 1201
- 11Zein, N., Poncin, M., Nilakantan, R., and Ellestad, G. A. (1989) Calicheamicin γ 1I and DNA: Molecular recognition process responsible for site-specificity Science 244, 697– 699
- 12Mah, S. C., Price, M. A., Townsend, C. A., and Tullius, T. D. (1994) Features of DNA recognition for oriented binding and cleavage by calicheamicin Tetrahedron 50, 1361– 1378
- 13Biggins, J. B., Prudent, J. R., Marshall, D. J., and Thorson, J. S. (2006) A continuous assay for DNA cleavage using molecular break lights Methods Mol. Biol. 335, 83– 92[PubMed], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xjt1yrsLs%253D&md5=15a1230e3506d16698194bcaa77b65a4A continuous assay for DNA cleavage using molecular break lightsBiggins, John B.; Prudent, James R.; Marshall, David J.; Thorson, Jon S.Methods in Molecular Biology (Totowa, NJ, United States) (2006), 335 (Fluorescent Energy Transfer Nucleic Acid Probes), 83-92CODEN: MMBIED; ISSN:1064-3745. (Humana Press Inc.)Exploring the properties of mols. that cleave DNA (i.e., enzymic nucleases, chem. footprinting agents, and naturally occurring DNA cleaving antibiotics) has been an ongoing process with benefits extending toward both lab. and clin. applications. Despite the progress that has been made toward understanding the mechanics of DNA cleavage, a simple and continuous assay for detecting DNA cleavage has been lacking. Herein, the authors describe the mol. break light assay, wherein a single oligonucleotide modified by a 5'-fluorophore-3'-quencher pair adopting a stem-loop structure with an appropriate DNA recognition site, provides for the rapid assaying of DNA cleavage with high sensitivity. Furthermore, the described methodol. is highly convenient in that it is readily adaptable to common lab. fluorometers and multi-well plate/array systems, which may provide the basis for high-throughput screening of novel DNA cleaving agents. This assay may also be further extended to natural or "unnatural" transcription factor protection assay systems.
- 14Danishefsky, S. J. and Shair, M. D. (1996) Observations in the chemistry and biology of cyclic enediyne antibiotics: Total syntheses of calicheamicin γ1I and dynemicin A J. Org. Chem. 61, 16– 44
- 15Nicolaou, K. C., Hale, C. R. H., and Nilewski, C. (2012) A total synthesis trilogy: Calicheamicin γ1(I), Taxol, and brevetoxin A Chem. Rec. 12, 407– 441[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XoslKjsbc%253D&md5=d3605d104978d33d5ea7a81a7bf9e967A total synthesis trilogy: Calicheamicin γ1I, Taxol and brevetoxin ANicolaou, K. C.; Hale, Christopher R. H.; Nilewski, ChristianChemical Record (2012), 12 (4), 407-441CODEN: CRHEAK; ISSN:1527-8999. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Detailed behind-the-scenes accounts of the total syntheses of calicheamicin γ1I, Taxol and brevetoxin A are discussed with particular emphasis placed on strategies and tactics employed in these campaigns. DOI 10.1002/tcr.201200005.
- 16Ahlert, J., Shepard, E., Lomovskaya, N., Zazopoulos, E., Staffa, A., Bachmann, B. O., Huang, K., Fonstein, L., Czisny, A., Whitwam, R. E., Farnet, C. M., and Thorson, J. S. (2002) The calicheamicin gene cluster and its iterative type I enediyne PKS Science 297, 1173– 1176[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XmsVOht70%253D&md5=76ae8b38e6de9d1db318ca6538b09ac5The calicheamicin gene cluster and its iterative type I enediyne PKSAhlert, Joachim; Shepard, Erica; Lomovskaya, Natalia; Zazopoulos, Emmanuel; Staffa, Alfredo; Bachmann, Brian O.; Huang, Kexue; Fonstein, Leonid; Czisny, Anne; Whitwam, Ross E.; Farnet, Chris M.; Thorson, Jon S.Science (Washington, DC, United States) (2002), 297 (5584), 1173-1176CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The enediynes exemplify nature's ingenuity. We have cloned and characterized the biosynthetic locus coding for perhaps the most notorious member of the nonchromoprotein enediyne family, calicheamicin. This gene cluster contains an unusual polyketide synthase (PKS) that is demonstrated to be essential for enediyne biosynthesis. Comparison of the calicheamicin locus with the locus encoding the chromoprotein enediyne C-1027 reveals that the enediyne PKS is highly conserved among these distinct enediyne families. Contrary to previous hypotheses, this suggests that the chromoprotein and nonchromoprotein enediynes are generated by similar biosynthetic pathways.
- 17Liu, W., Ahlert, J., Gao, Q., Wendt-Pienkowski, E., Shen, B., and Thorson, J. S. (2003) Rapid PCR amplification of minimal enediyne polyketide synthase cassettes leads to a predictive familial classification model Proc. Natl. Acad. Sci. U.S.A. 100, 11959– 11963[Crossref], [PubMed], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXotlGltL4%253D&md5=d50cfaa0360d5904aa9c7b0ea11b669dRapid PCR amplification of minimal enediyne polyketide synthase cassettes leads to a predictive familial classification modelLiu, Wen; Ahlert, Joachim; Gao, Qunjie; Wendt-Pienkowski, Evelyn; Shen, Ben; Thorson, Jon S.Proceedings of the National Academy of Sciences of the United States of America (2003), 100 (21), 11959-11963CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)A universal PCR method for the rapid amplification of minimal enediyne polyketide synthase (PKS) genes and the application of this methodol. to clone remaining prototypical genes from producers of structurally detd. enediynes in both family types are presented. A phylogenetic anal. of the new pool of bona fide enediyne PKS genes, consisting of three from 9-membered producers (neocarzinostatin, C1027, and maduropeptin) and three from 10-membered producers (calicheamicin, dynemicin, and esperamicin), reveals a clear genotypic distinction between the two structural families from which to form a predictive model. The results from this study support the postulation that the minimal enediyne PKS helps define the structural divergence of the enediyne core and provides the key tools for generating enediyne hybrid genes/mol. scaffolds; by using the model, a classification is also provided for the unknown enediyne PKS genes previously identified via genome scanning.
- 18Biggins, J. B., Onwueme, K. C., and Thorson, J. S. (2003) Resistance to enediyne antitumor antibiotics by CalC self-sacrifice Science 301, 1537– 1541
- 19Singh, S., Hager, M. H., Zhang, C., Griffith, B. R., Lee, M. S., Hallenga, K., Markley, J. L., and Thorson, J. S. (2006) Structural insight into the self-sacrifice mechanism of enediyne resistance ACS Chem. Biol. 1, 451– 460[ACS Full Text
], [CAS], Google Scholar
19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xot12mtbk%253D&md5=58226c7592f92d01dee192a28000ee15Structural insight into the self-sacrifice mechanism of enediyne resistanceSingh, Shanteri; Hager, Martin H.; Zhang, Changsheng; Griffith, Byron R.; Lee, Min S.; Hallenga, Klaas; Markley, John L.; Thorson, Jon S.ACS Chemical Biology (2006), 1 (7), 451-460CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)The recent discovery of the first "self-sacrifice" mechanism for bacterial resistance to the enediyne antitumor antibiotics, where enediyne-induced proteolysis of the resistance protein CalC inactivates both the highly reactive metabolite and the resistance protein, revealed yet another ingenious bacterial mechanism for controlling reactive metabolites. As reported herein, the first 3D structures of CalC and CalC in complex with calicheamicin (CLM) divulge CalC to be a member of the steroidogenic acute regulatory protein (StAR)-related transfer (START) domain superfamily. In contrast to previous studies of proteins known to bind DNA-damaging natural products (e.g., bleomycins, mitomycins, and 9-membered chromoprotein enediynes), this is the 1st demonstrated involvement of a START domain fold. Consistent with the CalC self-sacrifice mechanism, CLM in complex with CalC is positioned for direct H abstraction from Gly113 to initiate the oxidative proteolysis-based resistance mechanism. These structural studies also illuminate, for the 1st time, a small DNA-binding region within CalC that may serve to localize CalC to the enediyne target (DNA). Given the role of START domains in nuclear/cytosolic transport and translocation, this structural study also may implicate START domains as post-endocytotic intracellular chaperones for enediyne-based therapeutics such as MyloTarg. - 20Zhang, C., Griffith, B. R., Fu, Q., Albermann, C., Fu, X., Lee, I.-K., Li, L., and Thorson, J. S. (2006) Exploiting the reversibility of natural product glycosyltransferase-catalyzed reactions Science 313, 1291– 1294[Crossref], [PubMed], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XoslOntb0%253D&md5=5f2b83f0499627c28f3ddfbf16880a97Exploiting the Reversibility of Natural Product Glycosyltransferase-Catalyzed ReactionsZhang, Changsheng; Griffith, Byron R.; Fu, Qiang; Albermann, Christoph; Fu, Xun; Lee, In-Kyoung; Li, Lingjun; Thorson, Jon S.Science (Washington, DC, United States) (2006), 313 (5791), 1291-1294CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Glycosyltransferases (GTs), an essential class of ubiquitous enzymes, are generally perceived as unidirectional catalysts. In contrast, we report that four glycosyltransferases from two distinct natural product biosynthetic pathways-calicheamicin and vancomycin-readily catalyze reversible reactions, allowing sugars and aglycons to be exchanged with ease. As proof of the broader applicability of these new reactions, more than 70 differentially glycosylated calicheamicin and vancomycin variants are reported. This study suggests the reversibility of GT-catalyzed reactions may be general and useful for generating exotic nucleotide sugars, establishing in vitro GT activity in complex systems, and enhancing natural product diversity.
- 21Johnson, H. D. and Thorson, J. S. (2008) Characterization of CalE10, the N-oxidase involved in calicheamicin hydroxyaminosugar formation J. Am. Chem. Soc. 130, 17662– 17663
- 22Belecki, K., Crawford, J. M., and Townsend, C. A. (2009) Production of octaketide polyenes by the calicheamicin polyketide synthase CalE8: Implications for the biosynthesis of enediyne core structures J. Am. Chem. Soc. 131, 12564– 12566[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVSisL7N&md5=ed8ce733ad40968e31b760f64e42465bProduction of Octaketide Polyenes by the Calicheamicin Polyketide Synthase CalE8: Implications for the Biosynthesis of Enediyne Core StructuresBelecki, Katherine; Crawford, Jason M.; Townsend, Craig A.Journal of the American Chemical Society (2009), 131 (35), 12564-12566CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Enediyne antibiotics are categorized according to the presence of either a 9- or 10-membered ring within their polyketide-derived core structures. Recent literature reports have favored the notion that biosynthetic divergence of the two structural families is detd. by the enediyne polyketide synthases (PKSs) alone. We now disclose the simultaneous in vitro prodn. of three octaketide polyenes by biosynthetic enzymes for the 10-membered enediyne calicheamicin γ1I, including the elusive β-keto acid precursor to a previously described C15 Me hexaenone. Alongside these two polyene products, we have addnl. detected a hydrocarbon heptaene previously isolated only from 9-membered enediyne systems. The discovery of the heptaene in the calicheamicin system promotes a more convergent model for the early steps of enediyne biosynthesis. Furthermore, the synthesis of this set of octaketides by the enediyne PKS CalE8 and thioesterase CalE7 suggests, in contrast to recent biosynthetic proposals, that accessory enzymes may be necessary to initiate differentiation to 9- or 10-membered enediyne precursors, either by modulation of enediyne PKS activity or by interception and modification of polyketide chain-extension intermediates. - 23Horsman, G. P., Chen, Y., Thorson, J. S., and Shen, B. (2010) Polyketide synthase chemistry does not direct biosynthetic divergence between 9- and 10-membered enediynes Proc. Natl. Acad. Sci. U.S.A. 107, 11331– 11335
- 24Gantt, R. W., Peltier-Pain, P., Singh, S., Zhou, M., and Thorson, J. S. (2013) Broadening the scope of glycosyltransferase-catalyzed sugar nucleotide synthesis Proc. Natl. Acad. Sci. U.S.A. 110, 7648– 7653[Crossref], [PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFGrtL8%253D&md5=bfd133c9e815a7c25d31b9b463e83e04Broadening the scope of glycosyltransferase-catalyzed sugar nucleotide synthesisGantt, Richard W.; Peltier-Pain, Pauline; Singh, Shanteri; Zhou, Maoquan; Thorson, Jon S.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (19), 7648-7653, S7648/1-S7648/72CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We described the integration of the general reversibility of glycosyltransferase-catalyzed reactions, artificial glycosyl donors, and a high throughput colorimetric screen to enable the engineering of glycosyltransferases for combinatorial sugar nucleotide synthesis. The best engineered catalyst from this study, the OleD Loki variant, contained the mutations P67T/I112P/T113M/S132F/A242I compared with the OleD wild-type sequence. Evaluated against the parental sequence OleD TDP16 variant used for screening, the OleD Loki variant displayed max. improvements in kcat/Km of >400-fold and >15-fold for formation of NDP-glucoses and UDP-sugars, resp. This OleD Loki variant also demonstrated efficient turnover with five variant NDP acceptors and six variant 2-chloro-4-nitrophenyl glycoside donors to produce 30 distinct NDP-sugars. This study highlights a convenient strategy to rapidly optimize glycosyltransferase catalysts for the synthesis of complex sugar nucleotides and the practical synthesis of a unique set of sugar nucleotides.
- 25Belecki, K. and Townsend, C. A. (2012) Environmental control of the calicheamicin polyketide synthase leads to detection of a programmed octaketide and a proposal for enediyne biosynthesis Angew. Chem., Int. Ed. 51, 11316– 11319
- 26Belecki, K. and Townsend, C. A. (2013) Biochemical determination of enzyme-bound metabolites: Preferential accumulation of a programmed octaketide on the enediyne polyketide synthase CalE8 J. Am. Chem. Soc. 135, 14339– 14348
- 27Ricart, A. D. (2011) Antibody-drug conjugates of calicheamicin derivative: Gemtuzumab Ozogamicin and Inotuzumab Ozogamicin Clin. Cancer Res. 17, 6417– 6427[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlSksrzF&md5=103b40cfe49632a86367414e0e97c011Antibody-Drug Conjugates of Calicheamicin Derivative: Gemtuzumab Ozogamicin and Inotuzumab OzogamicinRicart, Alejandro D.Clinical Cancer Research (2011), 17 (20), 6417-6427CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)A review. Antibody-drug conjugates (ADC) are an attractive approach for the treatment of acute myeloid leukemia and non-Hodgkin lymphomas, which in most cases, are inherently sensitive to cytotoxic agents. CD33 and CD22 are specific markers of myeloid leukemias and B-cell malignancies, resp. These endocytic receptors are ideal for an ADC strategy because they can effectively carry the cytotoxic payload into the cell. Gemtuzumab ozogamicin (GO, Mylotarg) and inotuzumab ozogamicin consist of a deriv. of calicheamicin (a potent DNA-binding cytotoxic antibiotic) linked to a humanized monoclonal IgG4 antibody directed against CD33 or CD22, resp. Both of these ADCs have a target-mediated pharmacokinetic disposition. GO was the first drug to prove the ADC concept in the clinic, specifically in phase II studies that included substantial proportions of older patients with relapsed acute myeloid leukemia. In contrast, in phase III studies, it has thus far failed to show clin. benefit in first-line treatment in combination with std. chemotherapy. Inotuzumab ozogamicin has shown remarkable clin. activity in relapsed/refractory B-cell non-Hodgkin lymphoma, and it has started phase III evaluation. The safety profile of these ADCs includes reversible myelosuppression (esp. neutropenia and thrombocytopenia), elevated hepatic transaminases, and hyperbilirubinemia. There have been postmarketing reports of hepatotoxicity, esp. veno-occlusive disease, assocd. with GO. The incidence is ∼2%, but patients who undergo hematopoietic stem cell transplantation have an increased risk. As we steadily move toward the goal of personalized medicine, these kinds of agents will provide a unique opportunity to treat selected patient subpopulations based on the expression of their specific tumor targets. Clin Cancer Res; 17(20); 6417-27.
- 28Trail, P. A. (2013) Antibody drug conjugates as cancer therapeutics Antibodies 2, 113– 129
- 29Krissinel, E. and Henrick, K. (2007) Inference of macromolecular assemblies from crystalline state J. Mol. Biol. 372, 774– 797[Crossref], [PubMed], [CAS], Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXpvFGktb8%253D&md5=a5c764cfc7dc129f53ddc31ef9d475faInference of Macromolecular Assemblies from Crystalline StateKrissinel, Evgeny; Henrick, KimJournal of Molecular Biology (2007), 372 (3), 774-797CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)The authors discuss basic phys.-chem. principles underlying the formation of stable macromol. complexes, which in many cases are likely to be the biol. units performing a certain physiol. function. The authors also consider available theor. approaches to the calcn. of macromol. affinity and entropy of complexation. The latter is shown to play an important role and make a major effect on complex size and symmetry. The authors develop a new method, based on chem. thermodn., for automatic detection of macromol. assemblies in the Protein Data Bank (PDB) entries that are the results of x-ray diffraction expts. As found, biol. units may be recovered at 80-90% success rate, which makes x-ray crystallog. an important source of exptl. data on macromol. complexes and protein-protein interactions. The method is implemented as a public WWW service (http://www.ebi.ac.uk/msd-srv/prot_int/pistart.html).
- 30Iyer, L. M., Koonin, E. V., and Aravind, L. (2001) Adaptations of the helix-grip fold for ligand binding and catalysis in the START domain superfamily Proteins 43, 134– 144
- 31Thorsell, A.-G., Lee, W. H., Persson, C., Siponen, M. I., Nilsson, M., Busam, R. D., Kotenyova, T., Schüler, H., and Lehtiö, L. (2011) Comparative structural analysis of lipid binding START domains PloS One 6, e19521
- 32Alpy, F. and Tomasetto, C. (2005) Give lipids a START: The StAR-related lipid transfer (START) domain in mammals J. Cell Sci. 118, 2791– 2801[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXnsVCjtrw%253D&md5=3b0a2eed630ac8e48b7fa6076a9d0266Give lipids a START: The StAR-related lipid transfer (START) domain in mammalsAlpy, Fabien; Tomasetto, CatherineJournal of Cell Science (2005), 118 (13), 2791-2801CODEN: JNCSAI; ISSN:0021-9533. (Company of Biologists Ltd.)A review. The steroidogenic acute regulatory protein (StAR)-related lipid transfer (START) domain is a protein module of ∼210 residues that binds lipids, including sterols. Fifteen mammalian proteins, STARD1-STARD15, possess a START domain and these can be grouped into 6 subfamilies. Cholesterol, 25-hydroxycholesterol, phosphatidylcholine, phosphatidylethanolamine, and ceramides are ligands for STARD1/STARD3/STARD5, STARD5, STARD2/STARD10, STARD10 and STARD11, resp. The lipids or sterols bound by the remaining 9 START proteins are unknown. Recent studies have shown that the C-terminal end of the domain plays a fundamental role, forming a lid over a deep lipid-binding pocket that shields the ligand from the external environment. The START domain can be regarded as a lipid-exchange and/or a lipid-sensing domain. Mammalian START proteins have diverse expression patterns and can be found free in the cytoplasm, attached to membranes or in the nucleus. They appear to function in a variety of distinct physiol. processes, such as lipid transfer between intracellular compartments, lipid metab., and modulation of signaling events. Mutation or misexpression of START proteins is linked to pathol. processes, including genetic disorders, autoimmune disease, and cancer.
- 33Holm, L. and Rosenström, P. (2010) Dali server: Conservation mapping in 3D Nucleic Acids Res. 38, W545– 549[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXotVSqsr4%253D&md5=b7832262058d6a4f7d362f4ce565a666Dali server: conservation mapping in 3DHolm, Liisa; Rosenstrom, PaiviNucleic Acids Research (2010), 38 (Web Server), W545-W549CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Our web site (http://ekhidna.biocenter.helsinki.fi/daliserver) runs the Dali program for protein structure comparison. The web site consists of three parts: (i) the Dali server compares newly solved structures against structures in the Protein Data Bank (PDB), (ii) the Dali database allows browsing precomputed structural neighborhoods and (iii) the pairwise comparison generates suboptimal alignments for a pair of structures. Each part has its own query form and a common format for the results page. The inputs are either PDB identifiers or novel structures uploaded by the user. The results pages are hyperlinked to aid interactive anal. The web interface is simple and easy to use. The key purpose of interactive anal. is to check whether conserved residues line up in multiple structural alignments and how conserved residues and ligands cluster together in multiple structure superimpositions. In favorable cases, protein structure comparison can lead to evolutionary discoveries not detected by sequence anal.
- 34Krissinel, E. and Henrick, K. (2004) Secondary-structure matching (SSM), a new tool for fast protein structure alignment in three dimensions Acta Crystallogr., Sect. D: Biol. Crystallogr. 60, 2256– 2268[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVars7rL&md5=219f539e65e5fbc2cc631f937f544953Secondary-structure matching (SSM), a new tool for fast protein structure alignment in three dimensionsKrissinel, E.; Henrick, K.Acta Crystallographica, Section D: Biological Crystallography (2004), D60 (12, Pt. 1), 2256-2268CODEN: ABCRE6; ISSN:0907-4449. (Blackwell Publishing Ltd.)The present paper describes the SSM algorithm of protein structure comparison in three dimensions, which includes an original procedure of matching graphs built on the protein's secondary-structure elements, followed by an iterative three-dimensional alignment of protein backbone Cα atoms. The SSM results are compared with those obtained from other protein comparison servers, and the advantages and disadvantages of different scores that are used for structure recognition are discussed. A new score, balancing the r.m.s.d. and alignment length Nalign, is proposed. It is found that different servers agree reasonably well on the new score, while showing considerable differences in r.m.s.d. and Nalign.
- 35Ames, B. D., Korman, T. P., Zhang, W., Smith, P., Vu, T., Tang, Y., and Tsai, S.-C. (2008) Crystal structure and functional analysis of tetracenomycin ARO/CYC: Implications for cyclization specificity of aromatic polyketides Proc. Natl. Acad. Sci. U.S.A. 105, 5349– 5354
- 36Michalska, K., Fernandes, H., Sikorski, M., and Jaskolski, M. (2010) Crystal structure of Hyp-1, a St. John’s Wort protein implicated in the biosynthesis of hypericin J. Struct. Biol. 169, 161– 171
- 37Ilari, A., Franceschini, S., Bonamore, A., Arenghi, F., Botta, B., Macone, A., Pasquo, A., Bellucci, L., and Boffi, A. (2009) Structural basis of enzymatic (S)-norcoclaurine biosynthesis J. Biol. Chem. 284, 897– 904[Crossref], [PubMed], [CAS], Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsV2ntg%253D%253D&md5=01e0a2f7c5b59fa572b524d07938bd30Structural basis of enzymatic (S)-norcoclaurine biosynthesisIlari, Andrea; Franceschini, Stefano; Bonamore, Alessandra; Arenghi, Fabio; Botta, Bruno; Macone, Alberto; Pasquo, Alessandra; Bellucci, Luca; Boffi, AlbertoJournal of Biological Chemistry (2009), 284 (2), 897-904CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Norcoclaurine synthase (NCS) catalyzes the stereospecific Pictet-Spengler cyclization between dopamine and 4-hydroxyphenylacetaldehyde, the key step in the benzylisoquinoline alkaloid biosynthetic pathway. Here, the crystallog. structure of NCS from Thalictrum flavum in its complex with its substrate, dopamine, and the nonreactive substrate analog, 4-hydroxybenzaldehyde, was solved at 2.1 Å resoln. NCS shared no common features with the functionally correlated "Pictet-Spenglerases" that catalyze the 1st step of the indole alkaloid pathways and conformed to the overall fold of the Bet v1-like protein. The active site of NCS was located within a 20-Å-long catalytic tunnel and was shaped by the side-chains of the Tyr-108, Lys-122, Asp-141, and Glu-110 residues. The geometry of the amino acid side-chains with respect to the substrates revealed the structural determinants that govern the mechanism of the stereoselective Pictet-Spengler cyclization, thus establishing an excellent foundation for the understanding of the finer details of the catalytic process. Site-directed mutations of the relevant residues confirmed the assignment based on the crystallog. findings.
- 38Kofler, S., Asam, C., Eckhard, U., Wallner, M., Ferreira, F., and Brandstetter, H. (2012) Crystallographically mapped ligand binding differs in high and low IgE binding isoforms of birch pollen allergen bet v 1 J. Mol. Biol. 422, 109– 123
- 39Li, W., Wang, L., Sheng, X., Yan, C., Zhou, R., Hang, J., Yin, P., and Yan, N. (2013) Molecular basis for the selective and ABA-independent inhibition of PP2CA by PYL13 Cell Res. 23, 1369– 1379[Crossref], [PubMed], [CAS], Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslWqu7nK&md5=06d90b8ae13d792940f0f7a15ff2efedMolecular basis for the selective and ABA-independent inhibition of PP2CA by PYL13Li, Wenqi; Wang, Li; Sheng, Xinlei; Yan, Chuangye; Zhou, Rui; Hang, Jing; Yin, Ping; Yan, NiengCell Research (2013), 23 (12), 1369-1379CODEN: CREEB6; ISSN:1001-0602. (NPG Nature Asia-Pacific)PYR1/PYL/RCAR family proteins (PYLs) are well-characterized abscisic acid (ABA) receptors. Among the 14 PYL members in Arabidopsis thaliana, PYL13 is ABA irresponsive and its function has remained elusive. Here, we show that PYL13 selectively inhibits the phosphatase activity of PP2CA independent of ABA. The crystal structure of PYL13-PP2CA complex, which was detd. at 2.4 Å resoln., elucidates the mol. basis for the specific recognition between PP2CA and PYL13. In addn. to the canonical interactions between PYLs and PP2Cs, an extra interface is identified involving an element in the vicinity of a previously uncharacterized CCCH zinc-finger (ZF) motif in PP2CA. Sequence blast identified another 56 ZF-contg. PP2Cs, all of which are from plants. The structure also reveals the mol. determinants for the ABA irresponsiveness of PYL13. Finally, biochem. anal. suggests that PYL13 may hetero-oligomerize with PYL10. These two PYLs antagonize each other in their resp. ABA-independent inhibitions of PP2Cs. The biochem. and structural studies provide important insights into the function of PYL13 in the stress response of plant and set up a foundation for future biotechnol. applications of PYL13.
- 40Pasternak, O., Bujacz, G. D., Fujimoto, Y., Hashimoto, Y., Jelen, F., Otlewski, J., Sikorski, M. M., and Jaskolski, M. (2006) Crystal structure of Vigna radiata cytokinin-specific binding protein in complex with zeatin Plant Cell 18, 2622– 2634
- 41Schirmer, T., Hoffimann-Sommergrube, K., Susani, M., Breiteneder, H., and Marković-Housley, Z. (2005) Crystal structure of the major celery allergen Api g 1: Molecular analysis of cross-reactivity J. Mol. Biol. 351, 1101– 1109
- 42Osipiuk, J., Wu, R., Moy, S., and Joachimiak, A. (2006) X-ray crystal structure of conserved hypothetical protein EF_2215 from . PDB ID: 2NN5. Unpublished.Google ScholarThere is no corresponding record for this reference.
- 43Singarapu, K., Eletsky, A., Sathyamoorthy, B., Sukumaran, D., Wang, D., Jiang, M., Ciccosanti, C., Xiao, R., Liu, J., Baran, M. C., Swapna, G., Acton, T. B., Rost, B., Montelione, G. T., and Szyperski, T. (2008) Solution NMR structure of protein encoded by gene BPP1335 from . PDB ID: 2K5G. Unpublished.Google ScholarThere is no corresponding record for this reference.
- 44Clark, B. J. (2012) The mammalian START domain protein family in lipid transport in health and disease J. Endocrinol. 212, 257– 275[Crossref], [PubMed], [CAS], Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XktlCrsb8%253D&md5=d3d9f925a2198cb3d6e53b15be5a3fecThe mammalian START domain protein family in lipid transport in health and diseaseClark, Barbara J.Journal of Endocrinology (2012), 212 (3), 257-275CODEN: JOENAK; ISSN:0022-0795. (BioScientifica Ltd.)A review. Lipid transfer proteins of the steroidogenic acute regulatory protein-related lipid transfer (START) domain family are defined by the presence of a conserved ∼210 amino acid sequence that folds into an α/β helix-grip structure forming a hydrophobic pocket for ligand binding. The mammalian START proteins bind diverse ligands, such as cholesterol, oxysterols, phospholipids, sphingolipids, and possibly fatty acids, and have putative roles in non-vesicular lipid transport, thioesterase enzymic activity, and tumor suppression. However, the biol. functions of many members of the START domain protein family are not well established. Recent research has focused on characterizing the cell-type distribution and regulation of the START proteins, examg. the specificity and directionality of lipid transport, and identifying disease states assocd. with dysregulation of START protein expression. This review summarizes the current concepts of the proposed physiol. and pathol. roles for the mammalian START domain proteins in cholesterol and lipid trafficking.
- 45Chang, A., Singh, S., Helmich, K. E., Goff, R. D., Bingman, C. A., Thorson, J. S., and Phillips, G. N., Jr. (2011) Complete set of glycosyltransferase structures in the calicheamicin biosynthetic pathway reveals the origin of regiospecificity Proc. Natl. Acad. Sci. U.S.A. 108, 17649– 17654
- 46Gerlt, J. A. and Babbitt, P. C. (2000) Can sequence determine function? Genome Biol. 1, 1– 10
- 47Whisstock, J. C. and Lesk, A. M. (2003) Prediction of protein function from protein sequence and structure Q. Rev. Biophys. 36, 307– 340[Crossref], [PubMed], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXmtlSjsw%253D%253D&md5=9540779034f73da3a6bf8fc32a130b32Prediction of protein function from protein sequence and structureWhisstock, James C.; Lesk, Arthur M.Quarterly Reviews of Biophysics (2003), 36 (3), 307-340CODEN: QURBAW; ISSN:0033-5835. (Cambridge University Press)A review. The sequence of a genome contains the plans of the possible life of an organism, but implementation of genetic information depends on the functions of the proteins and nucleic acids that it encodes. Many individual proteins of known sequence and structure present challenges to the understanding of their function. In particular, a no. of genes responsible for diseases have been identified but their specific functions are unknown. Whole-genome sequencing projects are a major source of proteins of unknown function. Annotation of a genome involves assignment of functions to gene products, in most cases on the basis of amino-acid sequence alone. 3D structure can aid the assignment of function, motivating the challenge of structural genomics projects to make structural information available for novel uncharacterized proteins. Structure-based identification of homologs often succeeds where sequence-alone-based methods fail, because in many cases evolution retains the folding pattern long after sequence similarity becomes undetectable. Nevertheless, prediction of protein function from sequence and structure is a difficult problem, because homologous proteins often have different functions. Many methods of function prediction rely on identifying similarity in sequence and/or structure between a protein of unknown function and one or more well-understood proteins. Alternative methods include inferring conservation patterns in members of a functionally uncharacterized family for which many sequences and structures are known. However, these inferences are tenuous. Such methods provide reasonable guesses at function, but are far from foolproof. It is therefore fortunate that the development of whole-organism approaches and comparative genomics permits other approaches to function prediction when the data are available. These include the use of protein-protein interaction patterns, and correlations between occurrences of related proteins in different organisms, as indicators of functional properties. Even if it is possible to ascribe a particular function to a gene product, the protein may have multiple functions. A fundamental problem is that function is in many cases an ill-defined concept. In this article we review the state of the art in function prediction and describe some of the underlying difficulties and successes.
- 48Schnoes, A. M., Brown, S. D., Dodevski, I., and Babbitt, P. C. (2009) Annotation error in public databases: Misannotation of molecular function in enzyme superfamilies PLoS Comput. Biol. 5, e1000605[Crossref], [PubMed], [CAS], Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1Mfis1SgtA%253D%253D&md5=1b018d0becdbf937c32bbfc10c611399Annotation error in public databases: misannotation of molecular function in enzyme superfamiliesSchnoes Alexandra M; Brown Shoshana D; Dodevski Igor; Babbitt Patricia CPLoS computational biology (2009), 5 (12), e1000605 ISSN:.Due to the rapid release of new data from genome sequencing projects, the majority of protein sequences in public databases have not been experimentally characterized; rather, sequences are annotated using computational analysis. The level of misannotation and the types of misannotation in large public databases are currently unknown and have not been analyzed in depth. We have investigated the misannotation levels for molecular function in four public protein sequence databases (UniProtKB/Swiss-Prot, GenBank NR, UniProtKB/TrEMBL, and KEGG) for a model set of 37 enzyme families for which extensive experimental information is available. The manually curated database Swiss-Prot shows the lowest annotation error levels (close to 0% for most families); the two other protein sequence databases (GenBank NR and TrEMBL) and the protein sequences in the KEGG pathways database exhibit similar and surprisingly high levels of misannotation that average 5%-63% across the six superfamilies studied. For 10 of the 37 families examined, the level of misannotation in one or more of these databases is >80%. Examination of the NR database over time shows that misannotation has increased from 1993 to 2005. The types of misannotation that were found fall into several categories, most associated with "overprediction" of molecular function. These results suggest that misannotation in enzyme superfamilies containing multiple families that catalyze different reactions is a larger problem than has been recognized. Strategies are suggested for addressing some of the systematic problems contributing to these high levels of misannotation.
- 49Baker, D. and Sali, A. (2001) Protein structure prediction and structural genomics Science 294, 93– 96[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXnsFyisbY%253D&md5=8b476f801ccb6223835f91e420edf57fProtein structure prediction and structural genomicsBaker, David; Sali, AndrejScience (Washington, DC, United States) (2001), 294 (5540), 93-96CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Genome sequencing projects are producing linear amino acid sequences, but full understanding of the biol. role of these proteins will require knowledge of their structure and function. Although exptl. structure detn. methods are providing high-resoln. structure information about a subset of the proteins, computational structure prediction methods will provide valuable information for the large fraction of sequences whose structures will not be detd. exptl. The first class of protein structure prediction methods, including threading and comparative modeling, rely on detectable similarity spanning most of the modeled sequence and at least one known structure. The second class of methods, de novo or ab initio methods, predict the structure from sequence alone, without relying on similarity at the fold level between the modeled sequence and any of the known structures. In this Viewpoint, we begin by describing the essential features of the methods, the accuracy of the models, and their application to the prediction and understanding of protein function, both for single proteins and on the scale of whole genomes. We then discuss the important role that protein structure prediction methods play in the growing worldwide effort in structural genomics.
- 50Hermann, J. C., Marti-Arbona, R., Fedorov, A. A., Fedorov, E., Almo, S. C., Shoichet, B. K., and Raushel, F. M. (2007) Structure-based activity prediction for an enzyme of unknown function Nature 448, 775– 779[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptVGrs7w%253D&md5=b0bd6612147fb848f49f8b9ccfd8384eStructure-based activity prediction for an enzyme of unknown functionHermann, Johannes C.; Marti-Arbona, Ricardo; Fedorov, Alexander A.; Fedorov, Elena; Almo, Steven C.; Shoichet, Brian K.; Raushel, Frank M.Nature (London, United Kingdom) (2007), 448 (7155), 775-779CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)With many genomes sequenced, a pressing challenge in biol. is predicting the function of the proteins that the genes encode. When proteins are unrelated to others of known activity, bioinformatics inference for function becomes problematic. It would thus be useful to interrogate protein structures for function directly. Here, we predict the function of an enzyme of unknown activity, Tm0936 from Thermotoga maritima, by docking high-energy intermediate forms of thousands of candidate metabolites. The docking hit list was dominated by adenine analogs, which appeared to undergo C6-deamination. Four of these, including 5-methylthioadenosine and S-adenosylhomocysteine (SAH), were tested as substrates, and three had substantial catalytic rate consts. (105 M-1 s-1). The x-ray crystal structure of the complex between Tm0936 and the product resulting from the deamination of SAH, S-inosylhomocysteine, was detd., and it corresponded closely to the predicted structure. The deaminated products can be further metabolized by T. maritima in a previously uncharacterized SAH degrdn. pathway. Structure-based docking with high-energy forms of potential substrates may be a useful tool to annotate enzymes for function.
- 51Guichou, J.-F. and Labesse, G. (2012) Fragment and conquer: From structure to complexes to function Structure 20, 1617– 1619
- 52Wright, G. D. (2007) The antibiotic resistome: The nexus of chemical and genetic diversity Nat. Rev. Microbiol. 5, 175– 186[Crossref], [PubMed], [CAS], Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhslOgur8%253D&md5=e98711cc6fee6a9713ad4fcfa9638d73The antibiotic resistome: the nexus of chemical and genetic diversityWright, Gerard D.Nature Reviews Microbiology (2007), 5 (3), 175-186CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)A review. Over the millennia, microorganisms have evolved evasion strategies to overcome a myriad of chem. and environmental challenges, including antimicrobial drugs. Even before the first clin. use of antibiotics more than 60 years ago, resistant organisms had been isolated. Moreover, the potential problem of the widespread distribution of antibiotic resistant bacteria was recognized by scientists and health-care specialists from the initial use of these drugs. Why is resistance inevitable and where does it come from. Understanding the mol. diversity that underlies resistance will inform our use of these drugs and guide efforts to develop new efficacious antibiotics.
- 53Cundliffe, E. and Demain, A. L. (2010) Avoidance of suicide in antibiotic-producing microbes J. Ind. Microbiol. Biotechnol. 37, 643– 672[Crossref], [PubMed], [CAS], Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsFGkt7c%253D&md5=714376ebb17316b0edd827dd2017fa39Avoidance of suicide in antibiotic-producing microbesCundliffe, Eric; Demain, Arnold L.Journal of Industrial Microbiology & Biotechnology (2010), 37 (7), 643-672CODEN: JIMBFL; ISSN:1367-5435. (Springer)A review. Many microbes synthesize potentially autotoxic antibiotics, mainly as secondary metabolites, against which they need to protect themselves. This is done in various ways, ranging from target-based strategies (i.e., modification of normal drug receptors or de novo synthesis of the latter in drug-resistant form) to the adoption of metabolic shielding and/or efflux strategies that prevent drug-target interactions. These self-defense mechanisms have been studied most intensively in antibiotic-producing prokaryotes, of which the most prolific are the actinomycetes. Only a few documented examples pertain to lower eukaryotes while higher organisms have hardly been addressed in this context. Thus, many plant alkaloids, variously described as herbivore repellents or nitrogen excretion devices, are truly antibiotics-even if toxic to humans. As just one example, bulbs of Narcissus spp. (including the King Alfred daffodil) accumulate narciclasine that binds to the larger subunit of the eukaryotic ribosome and inhibits peptide bond formation. However, ribosomes in the Amaryllidaceae have not been tested for possible resistance to narciclasine and other alkaloids. Clearly, the prevalence of suicide avoidance is likely to extend well beyond the remit of the present article.
- 54Allan, C. M., Hill, S., Morvaridi, S., Saiki, R., Johnson, J. S., Liau, W.-S., Hirano, K., Kawashima, T., Ji, Z., Loo, J. A., Shepherd, J. N., and Clarke, C. F. (2013) A conserved START domain coenzyme Q-binding polypeptide is required for efficient Q biosynthesis, respiratory electron transport, and antioxidant function in Saccharomyces cerevisiae Biochim. Biophys. Acta 1831, 776– 791
- 55Baker, J. R., Woolfson, D. N., Muskett, F. W., Stoneman, R. G., Urbaniak, M. D., and Caddick, S. (2007) Protein–small molecule interactions in neocarzinostatin, the prototypical enediyne chromoprotein antibiotic ChemBioChem 8, 704– 717
- 56Weigel, L. M., Clewell, D. B., Gill, S. R., Clark, N. C., McDougal, L. K., Flannagan, S. E., Kolonay, J. F., Shetty, J., Killgore, G. E., and Tenover, F. C. (2003) Genetic analysis of a high-level vancomycin-resistant isolate of Staphylococcus aureus Science 302, 1569– 1571[Crossref], [PubMed], [CAS], Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXpt1SmsLk%253D&md5=62c5066b5d48e0249f84f8838eed8d61Genetic analysis of a high-level vancomycin-resistant isolate of Staphylococcus aureusWeigel, Linda M.; Clewell, Don B.; Gill, Steven R.; Clark, Nancye C.; McDougal, Linda K.; Flannagan, Susan E.; Kolonay, James F.; Shetty, Jyoti; Killgore, George E.; Tenover, Fred C.Science (Washington, DC, United States) (2003), 302 (5650), 1569-1571CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Vancomycin is usually reserved for treatment of serious infections, including those caused by multidrug-resistant Staphylococcus aureus. A clin. isolate of S. aureus with high-level resistance to vancomycin (minimal inhibitory concn. = 1024 μg/mL) was isolated in June 2002. This isolate harbored a 57.9-kilobase multiresistance conjugative plasmid within which Tn1546 (vanA) was integrated. Addnl. elements on the plasmid encoded resistance to trimethoprim (dfrA), β-lactams (blaZ), aminoglycosides (aacA-aphD), and disinfectants (qacC). Genetic analyses suggest that the long-anticipated transfer of vancomycin resistance to a methicillin-resistant S. aureus occurred in vivo by interspecies transfer of Tn1546 from a co-isolate of Enterococcus faecalis.
- 57Qureshi, N. K., Yin, S., and Boyle-Vavra, S. (2014) The role of the Staphylococcal VraTSR regulatory system on vancomycin resistance and vanA operon expression in vancomycin-resistant Staphylococcus aureus PloS One 9, e85873
- 58The PyMOL Molecular Graphics System, Version 1.3r1. (2010, August) Schrödinger, LLC., New York.Google ScholarThere is no corresponding record for this reference.
- 59Kearse, M., Moir, R., Wilson, A., Stones-Havas, S., Cheung, M., Sturrock, S., Buxton, S., Cooper, A., Markowitz, S., Duran, C., Thierer, T., Ashton, B., Meintjes, P., and Drummond, A. (2012) Geneious Basic: An integrated and extendable desktop software platform for the organization and analysis of sequence data Bioinformatics 28, 1647– 1649[Crossref], [PubMed], [CAS], Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38rovFKhtg%253D%253D&md5=284aaf2baa0b23d4f12aaa80316acfeeGeneious Basic: an integrated and extendable desktop software platform for the organization and analysis of sequence dataKearse Matthew; Moir Richard; Wilson Amy; Stones-Havas Steven; Cheung Matthew; Sturrock Shane; Buxton Simon; Cooper Alex; Markowitz Sidney; Duran Chris; Thierer Tobias; Ashton Bruce; Meintjes Peter; Drummond AlexeiBioinformatics (Oxford, England) (2012), 28 (12), 1647-9 ISSN:.UNLABELLED: The two main functions of bioinformatics are the organization and analysis of biological data using computational resources. Geneious Basic has been designed to be an easy-to-use and flexible desktop software application framework for the organization and analysis of biological data, with a focus on molecular sequences and related data types. It integrates numerous industry-standard discovery analysis tools, with interactive visualizations to generate publication-ready images. One key contribution to researchers in the life sciences is the Geneious public application programming interface (API) that affords the ability to leverage the existing framework of the Geneious Basic software platform for virtually unlimited extension and customization. The result is an increase in the speed and quality of development of computation tools for the life sciences, due to the functionality and graphical user interface available to the developer through the public API. Geneious Basic represents an ideal platform for the bioinformatics community to leverage existing components and to integrate their own specific requirements for the discovery, analysis and visualization of biological data. AVAILABILITY AND IMPLEMENTATION: Binaries and public API freely available for download at http://www.geneious.com/basic, implemented in Java and supported on Linux, Apple OSX and MS Windows. The software is also available from the Bio-Linux package repository at http://nebc.nerc.ac.uk/news/geneiousonbl.
- 60Acton, T. B., Xiao, R., Anderson, S., Aramini, J., Buchwald, W. A., Ciccosanti, C., Conover, K., Everett, J., Hamilton, K., Huang, Y. J., Janjua, H., Kornhaber, G., Lau, J., Lee, D. Y., Liu, G., Maglaqui, M., Ma, L., Mao, L., Patel, D., Rossi, P., Sahdev, S., Shastry, R., Swapna, G. V. T., Tang, Y., Tong, S., Wang, D., Wang, H., Zhao, L., and Montelione, G. T. (2011) Preparation of protein samples for NMR structure, function, and small-molecule screening studies Methods Enzymol. 493, 21– 60[Crossref], [PubMed], [CAS], Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXosleku7g%253D&md5=d46f532743d27d482799c9e51b3e0ed2Preparation of protein samples for NMR structure, function, and small-molecule screening studiesActon, Thomas B.; Xiao, Rong; Anderson, Stephen; Aramini, James; Buchwald, William A.; Ciccosanti, Colleen; Conover, Ken; Everett, John; Hamilton, Keith; Huang, Yuanpeng Janet; Janjua, Haleema; Kornhaber, Gregory; Lau, Jessica; Lee, Dong Yup; Liu, Gaohua; Maglaqui, Melissa; Ma, Lichung; Mao, Lei; Patel, Dayaban; Rossi, Paolo; Sahdev, Seema; Shastry, Ritu; Swapna, G. V. T.; Tang, Yeufeng; Tong, Saichiu; Wang, Dongyan; Wang, Huang; Zhao, Li; Montelione, Gaetano T.Methods in Enzymology (2011), 493 (Fragment-Based Drug Design), 21-60CODEN: MENZAU; ISSN:0076-6879. (Elsevier Inc.)A review. In this chapter, we conc. on the prodn. of high-quality protein samples for NMR studies, in particular, we provide an in-depth description of recent advances in the prodn. of NMR samples and their synergistic use with recent advancements in NMR hardware. We describe the protein prodn. platform of the Northeast Structural Genomics Consortium and outline our high-throughput strategies for producing high-quality protein samples for NMR studies. Our strategy is based on the cloning, expression, and purifn. of 6 × -His-tagged proteins using T7-based Escherichia coli systems and isotope enrichment in minimal media. We describe 96-well ligation-independent cloning and anal. expression systems, parallel preparative scale fermn., and high-throughput purifn. protocols. The 6 × -His affinity tag allows for a similar two-step purifn. procedure implemented in a parallel high-throughput fashion that routinely results in purity levels sufficient for NMR studies (>97% homogeneity). Using this platform, the protein open reading frames of over 17,500 different targeted proteins (or domains) have been cloned as over 28,000 constructs. Nearly 5000 of these proteins have been purified to homogeneity in tens of milligram quantities, resulting in more than 950 new protein structures, including more than 400 NMR structures, deposited in the Protein Data Bank. The Northeast Structural Genomics Consortium pipeline has been effective in producing protein samples of both prokaryotic and eukaryotic origin. Although this chapter describes our entire pipeline for producing isotope-enriched protein samples, it focuses on the major updates introduced during the last 5 years (Phase 2 of the National Institute of General Medical Sciences Protein Structure Initiative). Our advanced automated and/or parallel cloning, expression, purifn., and biophys. screening technologies are suitable for implementation in a large individual lab. or by a small group of collaborating investigators for structural biol., functional proteomics, ligand screening, and structural genomics research.
- 61Xiao, R., Anderson, S., Aramini, J., Belote, R., Buchwald, W. A., Ciccosanti, C., Conover, K., Everett, J. K., Hamilton, K., Huang, Y. J., Janjua, H., Jiang, M., Kornhaber, G. J., Lee, D. Y., Locke, J. Y., Ma, L.-C., Maglaqui, M., Mao, L., Mitra, S., Patel, D., Rossi, P., Sahdev, S., Sharma, S., Shastry, R., Swapna, G. V. T., Tong, S. N., Wang, D., Wang, H., Zhao, L., Montelione, G. T., and Acton, T. B. (2010) The high-throughput protein sample production platform of the Northeast Structural Genomics Consortium J. Struct. Biol. 172, 21– 33
- 62Luft, J. R., Wolfley, J. R., Said, M. I., Nagel, R. M., Lauricella, A. M., Smith, J. L., Thayer, M. H., Veatch, C. K., Snell, E. H., Malkowski, M. G., and Detitta, G. T. (2007) Efficient optimization of crystallization conditions by manipulation of drop volume ratio and temperature Protein Sci. Publ. Protein Soc. 16, 715– 722[Crossref], [PubMed], [CAS], Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXktVOmt7o%253D&md5=0be93bed89cf4b708f473d54f20a77ddEfficient optimization of crystallization conditions by manipulation of drop volume ratio and temperatureLuft, Joseph R.; Wolfley, Jennifer R.; Said, Meriem I.; Nagel, Raymond M.; Lauricella, Angela M.; Smith, Jennifer L.; Thayer, Max H.; Veatch, Christina K.; Snell, Edward H.; Malkowski, Michael G.; Detitta, George T.Protein Science (2007), 16 (4), 715-722CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)An efficient optimization method for the crystn. of biol. macromols. has been developed and tested. This builds on a successful high-throughput technique for the detn. of initial crystn. conditions. The optimization method takes an initial condition identified through screening and then varies the concn. of the macromol., precipitant, and the growth temp. in a systematic manner. The amt. of sample and no. of steps is minimized and no biochem. reformulation is required. In the current application a robotic liq. handling system enables high-throughput use, but the technique can easily be adapted in a nonautomated setting. This method has been applied successfully for the rapid optimization of crystn. conditions in nine representative cases.
- 63Otwinowski, Z. and Minor, W. (1997) Processing of X-ray diffraction data collected in oscillation mode Methods Enzymol. 276, 307– 326[Crossref], [PubMed], [CAS], Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXivFehsbw%253D&md5=c9536971d4e32cc35352c40fb9368131Processing of x-ray diffraction data collected in oscillation modeOtwinowski, Zbyszek; Minor, WladekMethods in Enzymology (1997), 276 (Macromolecular Crystallography, Part A), 307-326CODEN: MENZAU; ISSN:0076-6879. (Academic)Macromol. crystallog. is an iterative process. Rarely do the first crystals provide all the necessary data to solve the biol. problem being studied. Each step benefits from experience learned in previous steps. To monitor the progress, the HKL package provides 2 tools: (1) statistics, both weighted (χ2) and unweighted (R-merge), are provided, and the Bayesian reasoning and multicomponent error model facilitates obtaining the proper error ests. and (2) visualization of the process plays a double role by helping the operator to confirm that the process of data redn., including the resulting statistics, is correct, and allowing one to evaluate problems for which there are no good statistical criteria. Visualization also provides confidence that the point of diminishing returns in data collection and redn. has been reached. At that point, the effort should be directed to solving the structure. The methods presented here have been applied to solve a large variety of problems, from inorg. mols. with 5 Å unit cell to rotavirus of 700 Å diam. crystd. in 700 × 1000 × 1400 Å cell. Overall quality of the method was tested by many researchers by successful application of the programs to MAD structure detns.
- 64Sheldrick, G. M. (2010) Experimental phasing with SHELXC/D/E: Combining chain tracing with density modification Acta Crystallogr., Sect. D: Biol. Crystallogr. 66, 479– 485[Crossref], [PubMed], [CAS], Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFKisb4%253D&md5=0b73bdbc20100b89fe7de5aaf357aab6Experimental phasing with SHELXC/D/E: combining chain tracing with density modificationSheldrick, George M.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (4), 479-485CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)The programs SHELXC, SHELXD and SHELXE are designed to provide simple, robust and efficient exptl. phasing of macromols. by the SAD, MAD, SIR, SIRAS and RIP methods and are particularly suitable for use in automated structure-soln. pipelines. This paper gives a general account of exptl. phasing using these programs and describes the extension of iterative d. modification in SHELXE by the inclusion of automated protein main-chain tracing. This gives a good indication as to whether the structure has been solved and enables interpretable maps to be obtained from poorer starting phases. The autotracing algorithm starts with the location of possible seven-residue α-helixes and common tripeptides. After extension of these fragments in both directions, various criteria are used to decide whether to accept or reject the resulting poly-Ala traces. Noncrystallog. symmetry (NCS) is applied to the traced fragments, not to the d. Further features are the use of a 'no-go' map to prevent the traces from passing through heavy atoms or symmetry elements and a splicing technique to combine the best parts of traces (including those generated by NCS) that partly overlap.
- 65Terwilliger, T. C. and Berendzen, J. (1999) Automated MAD and MIR structure solution Acta Crystallogr., Sect. D: Biol. Crystallogr. 55, 849– 861
- 66Emsley, P., Lohkamp, B., Scott, W. G., and Cowtan, K. (2010) Features and development of Coot Acta Crystallogr., Sect. D: Biol. Crystallogr. 66, 486– 501[Crossref], [PubMed], [CAS], Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXksFKisb8%253D&md5=67262cbfc60004de5ef962d5c043c910Features and development of CootEmsley, P.; Lohkamp, B.; Scott, W. G.; Cowtan, K.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (4), 486-501CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)Coot is a mol.-graphics application for model building and validation of biol. macromols. The program displays electron-d. maps and at. models and allows model manipulations such as idealization, real-space refinement, manual rotation/translation, rigid-body fitting, ligand search, solvation, mutations, rotamers and Ramachandran idealization. Furthermore, tools are provided for model validation as well as interfaces to external programs for refinement, validation and graphics. The software is designed to be easy to learn for novice users, which is achieved by ensuring that tools for common tasks are 'discoverable' through familiar user-interface elements (menus and toolbars) or by intuitive behavior (mouse controls). Recent developments have focused on providing tools for expert users, with customisable key bindings, extensions and an extensive scripting interface. The software is under rapid development, but has already achieved very widespread use within the crystallog. community. The current state of the software is presented, with a description of the facilities available and of some of the underlying methods employed.
- 67Murshudov, G. N., Vagin, A. A., and Dodson, E. J. (1997) Refinement of macromolecular structures by the maximum-likelihood method Acta Crystallogr., Sect. D: Biol. Crystallogr. 53, 240– 255[Crossref], [PubMed], [CAS], Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXjs1Gnsb4%253D&md5=ec7f141ce1542f7ff458b98ecfe3f8afRefinement of macromolecular structures by the maximum-likelihood methodMurshudov, Garib N.; Vagin, Alexei A.; Dodson, Eleanor J.Acta Crystallographica, Section D: Biological Crystallography (1997), D53 (3), 240-255CODEN: ABCRE6; ISSN:0907-4449. (Munksgaard)A review with many refs. on the math. basis of max. likelihood. The likelihood function for macromol. structures is extended to include prior phase information and exptl. std. uncertainties. The assumption that different parts of a structure might have different errors is considered. A method for estg. σA using "free" reflections is described and its effects analyzed. The derived equations have been implemented in the program REFMAC. This has been tested on several proteins at different stages of refinement (bacterial α-amylase, cytochrome c', cross-linked insulin and oligopeptide binding protein). The results derived using the max.-likelihood residual are consistently better than those obtained from least-squares refinement.
- 68Laskowski, R., MacArthur, M., Moss, D., and Thornton, J. (1993) PROCHECK: A program to check the stereochemical quality of protein structures J. Appl. Crystallogr. 26, 283– 291[Crossref], [CAS], Google Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3sXit12lurY%253D&md5=fb69fc4410cd716aaaa7cc0db06b3ed2PROCHECK: a program to check the stereochemical quality of protein structuresLaskowski, Roman A.; MacArthur, Malcolm W.; Moss, David S.; Thornton, Janet M.Journal of Applied Crystallography (1993), 26 (2), 283-91CODEN: JACGAR; ISSN:0021-8898.The PROCHECK suite of programs provides a detailed check on the stereochem. of a protein structure. Its outputs comprise a no. of plots in PostScript format and a comprehensive residue-by-residue listing. These give an assessment of the overall quality of the structure as compared with well-refined structures of the same resoln. and also highlight regions that may need further investigation. The PROCHECK programs are useful for assessing the quality not only of protein structures in the process of being solved but also of existing structures and of those being modeled on known structures.
- 69Shi, J., Wang, Y., Zeng, L., Wu, Y., Deng, J., Zhang, Q., Lin, Y., Li, J., Kang, T., Tao, M., Rusinova, E., Zhang, G., Wang, C., Zhu, H., Yao, J., Zeng, Y.-X., Evers, B. M., Zhou, M.-M., and Zhou, B. P. (2014) Disrupting the interaction of BRD4 with diacetylated twist suppresses tumorigenesis in basal-like breast cancer Cancer Cell 25, 210– 225[Crossref], [PubMed], [CAS], Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitlCisrs%253D&md5=d207cbc1abb187a53d807452b4f10d8fDisrupting the Interaction of BRD4 with Diacetylated Twist Suppresses Tumorigenesis in Basal-like Breast CancerShi, Jian; Wang, Yifan; Zeng, Lei; Wu, Yadi; Deng, Jiong; Zhang, Qiang; Lin, Yiwei; Li, Junlin; Kang, Tiebang; Tao, Min; Rusinova, Elena; Zhang, Guangtao; Wang, Chi; Zhu, Haining; Yao, Jun; Zeng, Yi-Xin; Evers, B. Mark; Zhou, Ming-Ming; Zhou, Binhua P.Cancer Cell (2014), 25 (2), 210-225CODEN: CCAECI; ISSN:1535-6108. (Elsevier Inc.)Twist is a key transcription activator of epithelial-mesenchymal transition (EMT). It remains unclear how Twist induces gene expression. Here we report a mechanism by which Twist recruits BRD4 to direct WNT5A expression in basal-like breast cancer (BLBC). Twist contains a "histone H4-mimic" GK-X-GK motif that is diacetylated by Tip60. The diacetylated Twist binds the second bromodomain of BRD4, whose first bromodomain interacts with acetylated H4, thereby constructing an activated Twist/BRD4/P-TEFb/RNA-Pol II complex at the WNT5A promoter and enhancer. Pharmacol. inhibition of the Twist-BRD4 assocn. reduced WNT5A expression and suppressed invasion, cancer stem cell (CSC)-like properties, and tumorigenicity of BLBC cells. Our study indicates that the interaction with BRD4 is crit. for the oncogenic function of Twist in BLBC.
- 70Zhai, J., Ström, A.-L., Kilty, R., Venkatakrishnan, P., White, J., Everson, W. V., Smart, E. J., and Zhu, H. (2009) Proteomic characterization of lipid raft proteins in amyotrophic lateral sclerosis mouse spinal cord FEBS J. 276, 3308– 3323
- 71Chen, V. B., Arendall, W. B., 3rd, Headd, J. J., Keedy, D. A., Immormino, R. M., Kapral, G. J., Murray, L. W., Richardson, J. S., and Richardson, D. C. (2010) MolProbity: All-atom structure validation for macromolecular crystallography Acta Crystallogr., Sect. D: Biol. Crystallogr. 66, 12– 21[Crossref], [PubMed], [CAS], Google Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXit1Kktg%253D%253D&md5=b5fc7574f43f01dd6e43c3663ca4f779MolProbity: all-atom structure validation for macromolecular crystallographyChen, Vincent B.; Arendall, W. Bryan, III; Headd, Jeffrey J.; Keedy, Daniel A.; Immormino, Robert M.; Kapral, Gary J.; Murray, Laura W.; Richardson, Jane S.; Richardson, David C.Acta Crystallographica, Section D: Biological Crystallography (2010), 66 (1), 12-21CODEN: ABCRE6; ISSN:0907-4449. (International Union of Crystallography)MolProbity is a structure-validation web service that provides broad-spectrum solidly based evaluation of model quality at both the global and local levels for both proteins and nucleic acids. It relies heavily on the power and sensitivity provided by optimized hydrogen placement and all-atom contact anal., complemented by updated versions of covalent-geometry and torsion-angle criteria. Some of the local corrections can be performed automatically in MolProbity and all of the diagnostics are presented in chart and graphical forms that help guide manual rebuilding. X-ray crystallog. provides a wealth of biol. important mol. data in the form of at. three-dimensional structures of proteins, nucleic acids and increasingly large complexes in multiple forms and states. Advances in automation, in everything from crystn. to data collection to phasing to model building to refinement, have made solving a structure using crystallog. easier than ever. However, despite these improvements, local errors that can affect biol. interpretation are widespread at low resoln. and even high-resoln. structures nearly all contain at least a few local errors such as Ramachandran outliers, flipped branched protein side chains and incorrect sugar puckers. It is crit. both for the crystallographer and for the end user that there are easy and reliable methods to diagnose and correct these sorts of errors in structures. MolProbity is the authors' contribution to helping solve this problem and this article reviews its general capabilities, reports on recent enhancements and usage, and presents evidence that the resulting improvements are now beneficially affecting the global database.
Supporting Information
Supporting Information
ARTICLE SECTIONSAdditional protocols and methods, supplemental figures, and supplemental tables. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.