Yi Tang

Find an error

Name: Tang, Yi
Organization: University of California , USA
Department: Department of Chemical and Biomolecular Engineering
Title: (PhD)

TOPICS

Co-reporter:Man-Cheng Tang, Xiaoqing Cui, Xueqian He, Zhuang Ding, Tianjiao Zhu, Yi Tang, and Dehai Li
Organic Letters October 6, 2017 Volume 19(Issue 19) pp:
Publication Date(Web):September 19, 2017
DOI:10.1021/acs.orglett.7b02653
Macrophorins are representative examples of isoprenoid epoxycyclohexenones containing cyclized drimane moieties. We located and characterized the biosynthetic gene cluster of macrophorin from Penicillium terrestris. MacJ encoded by this cluster was characterized to be the first example of a membrane-bound type-II terpene cyclase catalyzing the cyclization of meroterpenoids via direct protonation of the terminal olefinic bond in acyclic yanuthones. The late-stage functionalization and substrate promiscuity of MacJ make it a potential biocatalyst for the synthesis of macrophorin analogues.
Co-reporter:Wei Xu, Xiaolu Cai, Michael E. Jung, and Yi Tang
Journal of the American Chemical Society October 6, 2010 Volume 132(Issue 39) pp:13604-13607
Publication Date(Web):September 9, 2010
DOI:10.1021/ja107084d
The widely found fungal iterative PKS-NRPS hybrid megasynthetases are highly programmed biosynthetic machines involved in the synthesis of 3-acyltetramic acids and related natural products. In vitro analysis of iterative PKS-NRPS has been hampered by the difficulties associated with obtaining pure and functional forms of these large enzymes (>400 kDa). We successfully expressed Aspergillus nidulans aspyridone synthetase (ApdA) from an engineered Saccharomyces cerevisiae strain. The complete functions of ApdA and its enoylreductase partner ApdC are reconstituted in vitro and in S. cerevisiae with the production of preaspyridone 7. The programming rules of both the PKS and NRPS modules were then examined in vitro. The key interaction between the PKS and the NRPS was dissected and reconstituted in trans by using stand-alone modules. Analogs of 7 were synthesized through heterologous combinations of PKS and NRPS modules from different sources. Our results represent one of the largest, multidomain enzyme reconstituted to date and offer new opportunities for engineered biosynthesis of fungal natural products.
Co-reporter:Shu-Shan Gao, Marc Garcia-Borràs, Joyann S. Barber, Yang Hai, Abing Duan, Neil K. Garg, K. N. Houk, and Yi Tang
Journal of the American Chemical Society March 15, 2017 Volume 139(Issue 10) pp:3639-3639
Publication Date(Web):February 27, 2017
DOI:10.1021/jacs.7b01089
Hydroalkoxylation is a powerful and efficient method of forming C–O bonds and cyclic ethers in synthetic chemistry. In studying the biosynthesis of the fungal natural product herqueinone, we identified an enzyme that can perform an intramolecular enantioselective hydroalkoxylation reaction. PhnH catalyzes the addition of a phenol to the terminal olefin of a reverse prenyl group to give a dihydrobenzofuran product. The enzyme accelerates the reaction by 3 × 105-fold compared to the uncatalyzed reaction. PhnH belongs to a superfamily of proteins with a domain of unknown function (DUF3237), of which no member has a previously verified function. The discovery of PhnH demonstrates that enzymes can be used to promote the enantioselective hydroalkoxylation reaction and form cyclic ethers.
Co-reporter:Nicholas Liu, Yiu-Sun Hung, Shu-Shan Gao, Leibniz Hang, Yi Zou, Yit-Heng Chooi, and Yi Tang
Organic Letters July 7, 2017 Volume 19(Issue 13) pp:
Publication Date(Web):June 13, 2017
DOI:10.1021/acs.orglett.7b01534
Zaragozic acid A (1) is a potent cholesterol lowering, polyketide natural product made by various filamentous fungi. The reconstitution of enzymes responsible for the initial steps of the biosynthetic pathway of 1 is accomplished using an engineered fungal heterologous host. These initial steps feature the priming of a benzoic acid starter unit onto a highly reducing polyketide synthase (HRPKS), followed by oxaloacetate extension and product release to generate a tricarboxylic acid containing product 2. The reconstitution studies demonstrated that only three enzymes, HRPKS, citrate synthase, and hydrolase, are needed in A. nidulans to produce the structurally complex product.
Co-reporter:Michio Sato, Jacob E. Dander, Chizuru Sato, Yiu-Sun Hung, Shu-Shan Gao, Man-Cheng Tang, Leibniz Hang, Jaclyn M. Winter, Neil K. Garg, Kenji Watanabe, and Yi Tang
Journal of the American Chemical Society April 19, 2017 Volume 139(Issue 15) pp:5317-5317
Publication Date(Web):April 2, 2017
DOI:10.1021/jacs.7b02432
Fungal polyketide synthases (PKSs) can function collaboratively to synthesize natural products of significant structural diversity. Here we present a new mode of collaboration between a highly reducing PKS (HRPKS) and a PKS-nonribosomal peptide synthetase (PKS-NRPS) in the synthesis of oxaleimides from the Penicillium species. The HRPKS is recruited in the synthesis of an olefin-containing free amino acid, which is activated and incorporated by the adenylation domain of the PKS-NRPS. The precisely positioned olefin from the unnatural amino acid is proposed to facilitate a scaffold rearrangement of the PKS-NRPS product to forge the maleimide and succinimide cores of oxaleimides.
Co-reporter:John M. Billingsley, Anthony B. DeNicola, Joyann S. Barber, Man-Cheng Tang, ... Yi Tang
Metabolic Engineering 2017 Volume 44(Volume 44) pp:
Publication Date(Web):1 November 2017
DOI:10.1016/j.ymben.2017.09.006
Monoterpene indole alkaloids (MIAs) represent a structurally diverse, medicinally essential class of plant derived natural products. The universal MIA building block strictosidine was recently produced in the yeast Saccharomyces cerevisiae, setting the stage for optimization of microbial production. However, the irreversible reduction of pathway intermediates by yeast enzymes results in a non-recoverable loss of carbon, which has a strong negative impact on metabolic flux. In this study, we identified and engineered the determinants of biocatalytic selectivity which control flux towards the iridoid scaffold from which all MIAs are derived. Development of a bioconversion based production platform enabled analysis of the metabolic flux and interference around two critical steps in generating the iridoid scaffold: oxidation of 8-hydroxygeraniol to the dialdehyde 8-oxogeranial followed by reductive cyclization to form nepetalactol. In vitro reconstitution of previously uncharacterized shunt pathways enabled the identification of two distinct routes to a reduced shunt product including endogenous ‘ene’-reduction and non-productive reduction by iridoid synthase when interfaced with endogenous alcohol dehydrogenases. Deletion of five genes involved in α,β-unsaturated carbonyl metabolism resulted in a 5.2-fold increase in biocatalytic selectivity of the desired iridoid over reduced shunt product. We anticipate that our engineering strategies will play an important role in the development of S. cerevisiae for sustainable production of iridoids and MIAs.Download high-res image (178KB)Download full-size image
Co-reporter:Hsiao-Ching Lin; Travis C. McMahon; Ashay Patel; Michael Corsello; Adam Simon; Wei Xu; Muxun Zhao; K. N. Houk; Neil K. Garg
Journal of the American Chemical Society 2016 Volume 138(Issue 12) pp:4002-4005
Publication Date(Web):March 10, 2016
DOI:10.1021/jacs.6b01413
Dimeric indole alkaloids are structurally diverse natural products that have attracted significant attention from the synthetic and biosynthetic communities. Here, we describe the characterization of a P450 monooxygenase CnsC from Penicillium that catalyzes the heterodimeric coupling between two different indole moieties, tryptamine and aurantioclavine, to construct vicinal quaternary stereocenters and yield the heptacyclic communesin scaffold. We show, via biochemical characterization, substrate analogues, and computational methods that CnsC catalyzes the C3-C3′ carbon-carbon bond formation and controls the regioselectivities of the pair of subsequent aminal bond formations to yield the communesin core. Use of ω-N-methyltryptamine and tryptophol in place of tryptamine led to the enzymatic synthesis of isocommunesin compounds, which have not been isolated to date.
Co-reporter:Shu-Shan Gao; Abing Duan; Wei Xu; Peiyuan Yu; Leibniz Hang; K. N. Houk
Journal of the American Chemical Society 2016 Volume 138(Issue 12) pp:4249-4259
Publication Date(Web):March 15, 2016
DOI:10.1021/jacs.6b01528
Phenalenones are polyketide natural products that display diverse structures and biological activities. The core of phenalenones is a peri-fused tricyclic ring system cyclized from a linear polyketide precursor via an unresolved mechanism. Toward understanding the unusual cyclization steps, the phn biosynthetic gene cluster responsible for herqueinone biosynthesis was identified from the genome of Penicillium herquei. A nonreducing polyketide synthase (NR-PKS) PhnA was shown to synthesize the heptaketide backbone and cyclize it into the angular, hemiketal-containing naphtho-γ-pyrone prephenalenone. The product template (PT) domain of PhnA catalyzes only the C4–C9 aldol condensation, which is unprecedented among known PT domains. The transformation of prephenalenone to phenalenone requires an FAD-dependent monooxygenase (FMO) PhnB, which catalyzes the C2 aromatic hydroxylation of prephenalenone and ring opening of the γ-pyrone ring simultaneously. Density functional theory calculations provide insights into why the hydroxylated intermediate undergoes an aldol-like phenoxide–ketone cyclization to yield the phenalenone core. This study therefore unveiled new routes and biocatalysts for polyketide cyclization.
Co-reporter:Xia Yu, Fang Liu, Yi Zou, Man-Cheng Tang, Leibniz Hang, K. N. Houk, and Yi Tang
Journal of the American Chemical Society 2016 Volume 138(Issue 41) pp:13529-13532
Publication Date(Web):October 3, 2016
DOI:10.1021/jacs.6b09464
Nature synthesizes many strained natural products that have diverse biological activities. Uncovering these biosynthetic pathways may lead to biomimetic strategies for organic synthesis of such compounds. In this work, we elucidated the concise biosynthetic pathway of herquline A, a highly strained and reduced fungal piperazine alkaloid. The pathway builds on a nonribosomal peptide synthetase derived dityrosine piperazine intermediate. Following enzymatic reduction of the P450-cross-linked dicyclohexadienone, N-methylation of the piperazine serves as a trigger that leads to a cascade of stereoselective and nonenzymatic transformations. Computational analysis of key steps in the pathway rationalizes the observed reactivities.
Co-reporter:Leibniz Hang, Nicholas Liu, and Yi Tang
ACS Catalysis 2016 Volume 6(Issue 9) pp:5935
Publication Date(Web):July 27, 2016
DOI:10.1021/acscatal.6b01559
Fungal polyketides are natural products with great chemical diversity that exhibit a wide range of biological activity. This chemical diversity often stems from specialized enzymes encoded in the biosynthetic gene cluster responsible for natural product biosynthesis. Fungal polyketide synthases (PKS) are megasynthases that produce the carbon scaffold for the molecules. Subsequent downstream tailoring enzymes such as oxygenases will then further modify the organic framework. In fungi, many of these enzymes have been found to work iteratively—catalyzing multiple similar reactions on different sites of the substrate. This Perspective will analyze several examples of fungal polyketides that are assembled from a scaffold-building iterative PKS and an accompanying, complexity-generating iterative tailoring oxygenase. In these examples, the PKS product is designed for downstream iterative oxygenations to elegantly generate additional complexity. Together, these iterative enzymes efficiently orchestrate the biosynthesis of elaborate natural products such as lovastatin, chaetoglobosin A, cytochalasin E, and aurovertin E.Keywords: biosynthesis; catalysis; monooxygenase; natural products; polyketide synthase
Co-reporter:Jessica M. Grandner, Ralph A. Cacho, Yi Tang, and K. N. Houk
ACS Catalysis 2016 Volume 6(Issue 7) pp:4506
Publication Date(Web):May 31, 2016
DOI:10.1021/acscatal.6b01068
Griseofulvin is an antifungal agent that has recently been determined to have potential antiviral and anticancer applications. The role of specific enzymes involved in the biosynthesis of this natural product has previously been determined, but the mechanism by which a p450 (GsfF), catalyzes the key oxidative cyclization of griseophenone B remains unknown. Using density functional theory (DFT), we have determined the mechanism of this oxidation that forms the oxa-spiro core of griseofulvin. Computations show GsfF preferentially performs phenolic O–H abstraction over epoxidation to catalyze the oxidation.Keywords: computations; DFT; griseofulvin; mechanism; O−H abstraction; p450
Co-reporter:Wei Xu, Lauren B. Raetz, Peng Wang, Yi Tang
Tetrahedron 2016 Volume 72(Issue 25) pp:3599-3604
Publication Date(Web):23 June 2016
DOI:10.1016/j.tet.2015.09.029
Cyclization steps in biosynthesis of aromatic polyketides are typically directed by specific enzymes known as cyclases. The fourth ring cyclization step that forms the fully aromatic pretetramid during tetracycline biosynthesis has not been resolved. Herein we report in vitro and in vivo studies on the fourth ring cyclization step in biosynthesis of SF2575 and related tetracyclic natural products. We demonstrate that an enzyme belonging to the ATP-dependent acyl-CoA ligase family catalyzes the C1–C18 Claisen condensation step that forms the A ring and yields the linear, tetracyclic aromatic compound that is the precursor to tetracyclic natural products. The enzyme, SsfL2, is well-conserved in all tetracycline biosynthetic gene clusters discovered to date. It is proposed that SsfL2 directly adenylates the tricyclic carboxylic acid to facilitate the final carbon-carbon bond formation, and coenzyme A is not required.
Co-reporter:Hideaki Oikawa and Yi Tang
The Journal of Antibiotics 2016 69(7) pp:471-472
Publication Date(Web):2016-07-01
DOI:10.1038/ja.2016.69
It is our great pleasure to be Guest Editors of this special issue of The Journal of Antibiotics dedicated to Professor David E Cane, upon his long-standing contribution as an editorial board member of this Journal. For over 45 years, Prof. Cane devoted much of his life to studying the biosynthesis of natural products.
Co-reporter:Ralph A. Cacho; Justin Thuss; Wei Xu; Randy Sanichar; Zhizeng Gao; Allison Nguyen; John C. Vederas
Journal of the American Chemical Society 2015 Volume 137(Issue 50) pp:15688-15691
Publication Date(Web):December 2, 2015
DOI:10.1021/jacs.5b11814
Highly reducing polyketide synthases (HR-PKSs) from fungi synthesize complex natural products using a single set of domains in a highly programmed, iterative fashion. The most enigmatic feature of HR-PKSs is how tailoring domains function selectively during different iterations of chain elongation to afford structural diversity. Using the lovastatin nonaketide synthase LovB as a model system and a variety of acyl substrates, we characterized the substrate specificity of the LovB methyltransferase (MT) domain. We showed that, while the MT domain displays methylation activity toward different β-ketoacyl groups, it is exceptionally selective toward its naturally programmed β-keto-dienyltetraketide substrate with respect to both chain length and functionalization. Accompanying characterization of the ketoreductase (KR) domain displays broader substrate specificity toward different β-ketoacyl groups. Our studies indicate that selective modifications by tailoring domains, such as the MTs, are achieved by higher kinetic efficiency on a particular substrate relative to the rate of transformation by other competing domains.
Co-reporter:Xu-Ming Mao; Zha-Jun Zhan; Matthew N. Grayson; Man-Cheng Tang; Wei Xu; Yong-Quan Li; Wen-Bing Yin; Hsiao-Ching Lin; Yit-Heng Chooi; K. N. Houk
Journal of the American Chemical Society 2015 Volume 137(Issue 37) pp:11904-11907
Publication Date(Web):September 4, 2015
DOI:10.1021/jacs.5b07816
Aurovertins are fungal polyketides that exhibit potent inhibition of adenosine triphosphate synthase. Aurovertins contain a 2,6-dioxabicyclo[3.2.1]octane ring that is proposed to be derived from a polyene precursor through regioselective oxidations and epoxide openings. In this study, we identified only four enzymes required to produce aurovertin E. The core polyketide synthase produces a polyene α-pyrone. Following pyrone O-methylation by a methyltransferase, a flavin-dependent mono-oxygenase and an epoxide hydrolase can iteratively transform the terminal triene portion of the precursor into the dioxabicyclo[3.2.1]octane scaffold. We demonstrate that a tetrahydrofuranyl polyene is the first stable intermediate in the transformation, which can undergo epoxidation and anti-Baldwin 6-endo-tet ring opening to yield the cyclic ether product. Our results further demonstrate the highly concise and efficient ways in which fungal biosynthetic pathways can generate complex natural product scaffolds.
Co-reporter:Yi Zou; Zhajun Zhan; Dehai Li; Mancheng Tang; Ralph A. Cacho; Kenji Watanabe
Journal of the American Chemical Society 2015 Volume 137(Issue 15) pp:4980-4983
Publication Date(Web):April 10, 2015
DOI:10.1021/jacs.5b03022
Modification of natural products with prenyl groups and the ensuing oxidative transformations are important for introducing structural complexity and biological activities. Penigequinolones (1) are potent insecticidal alkaloids that contain a highly modified 10-carbon prenyl group. Here we reveal an iterative prenylation mechanism for installing the 10-carbon unit using two aromatic prenyltransferases (PenI and PenG) present in the gene cluster of 1 from Penicillium thymicola. The initial Friedel–Crafts alkylation is catalyzed by PenI to yield dimethylallyl quinolone 6. The five-carbon side chain is then dehydrogenated by a flavin-dependent monooxygenase to give aryl diene 9, which serves as the electron-rich substrate for a second alkylation with dimethylallyl diphosphate to yield stryrenyl product 10. The completed, oxidized 10-carbon prenyl group then undergoes further structural morphing to yield yaequinolone C (12), the immediate precursor of 1. Our studies have therefore uncovered an unprecedented prenyl chain extension mechanism in natural product biosynthesis.
Co-reporter:Jaclyn M. Winter; Duilio Cascio; David Dietrich; Michio Sato; Kenji Watanabe; Michael R. Sawaya; John C. Vederas
Journal of the American Chemical Society 2015 Volume 137(Issue 31) pp:9885-9893
Publication Date(Web):July 14, 2015
DOI:10.1021/jacs.5b04520
Modular collaboration between iterative fungal polyketide synthases (IPKSs) is an important mechanism for generating structural diversity of polyketide natural products. Inter-PKS communication and substrate channeling are controlled in large by the starter unit acyl carrier protein transacylase (SAT) domain found in the accepting IPKS module. Here, we reconstituted the modular biosynthesis of the benzaldehyde core of the chaetoviridin and chaetomugilin azaphilone natural products using the IPKSs CazF and CazM. Our studies revealed a critical role of CazM’s SAT domain in selectively transferring a highly reduced triketide product from CazF. In contrast, a more oxidized triketide that is also produced by CazF and required in later stages of biosynthesis of the final product is not recognized by the SAT domain. The structural basis for the acyl unit selectivity was uncovered by the first X-ray structure of a fungal SAT domain, highlighted by a covalent hexanoyl thioester intermediate in the SAT active site. The crystal structure of SAT domain will enable protein engineering efforts aimed at mixing and matching different IPKS modules for the biosynthesis of new compounds.
Co-reporter:Man-Cheng Tang; Hsiao-Ching Lin; Dehai Li; Yi Zou; Jian Li; Wei Xu; Ralph A. Cacho; Maureen E. Hillenmeyer; Neil K. Garg
Journal of the American Chemical Society 2015 Volume 137(Issue 43) pp:13724-13727
Publication Date(Web):October 15, 2015
DOI:10.1021/jacs.5b06108
The structural diversity and biological activities of fungal indole diterpenes (IDTs) are generated in large part by the IDT cyclases (IDTCs). Identifying different IDTCs from IDT biosynthetic pathways is therefore important toward understanding how these enzymes introduce chemical diversity from a common linear precursor. However, IDTCs involved in the cyclization of the well-known aflavinine subgroup of IDTs have not been discovered. Here, using Saccharomyces cerevisiae as a heterologous host and a phylogenetically guided enzyme mining approach, we combinatorially assembled IDT biosynthetic pathways using IDTCs homologues identified from different fungal hosts. We identified the genetically standalone IDTCs involved in the cyclization of aflavinine and anominine and produced new IDTs not previously isolated. The cyclization mechanisms of the new IDTCs were proposed based on the yeast reconstitution results. Our studies demonstrate heterologous pathway assembly is a useful tool in the reconstitution of unclustered biosynthetic pathways.
Co-reporter:Dr. Xu-Ming Mao;Dr. Wei Xu;Dr. Dehai Li;Dr. Wen-Bing Yin;Dr. Yit-Heng Chooi;Dr. Yong-Quan Li;Dr. Yi Tang;Dr. Youcai Hu
Angewandte Chemie International Edition 2015 Volume 54( Issue 26) pp:7592-7596
Publication Date(Web):
DOI:10.1002/anie.201502452

Abstract

The small-molecule biosynthetic potential of most filamentous fungi has remained largely unexplored and represents an attractive source for the discovery of new compounds. Genome sequencing of Calcarisporium arbuscula, a mushroom-endophytic fungus, revealed 68 core genes that are involved in natural product biosynthesis. This is in sharp contrast to the predominant production of the ATPase inhibitors aurovertin B and D in the wild-type fungus. Inactivation of a histone H3 deacetylase led to pleiotropic activation and overexpression of more than 75 % of the biosynthetic genes. Sampling of the overproduced compounds led to the isolation of ten compounds of which four contained new structures, including the cyclic peptides arbumycin and arbumelin, the diterpenoid arbuscullic acid A, and the meroterpenoid arbuscullic acid B. Such epigenetic modifications therefore provide a rapid and global approach to mine the chemical diversity of endophytic fungi.

Co-reporter:Dr. Xu-Ming Mao;Dr. Wei Xu;Dr. Dehai Li;Dr. Wen-Bing Yin;Dr. Yit-Heng Chooi;Dr. Yong-Quan Li;Dr. Yi Tang;Dr. Youcai Hu
Angewandte Chemie 2015 Volume 127( Issue 26) pp:7702-7706
Publication Date(Web):
DOI:10.1002/ange.201502452

Abstract

The small-molecule biosynthetic potential of most filamentous fungi has remained largely unexplored and represents an attractive source for the discovery of new compounds. Genome sequencing of Calcarisporium arbuscula, a mushroom-endophytic fungus, revealed 68 core genes that are involved in natural product biosynthesis. This is in sharp contrast to the predominant production of the ATPase inhibitors aurovertin B and D in the wild-type fungus. Inactivation of a histone H3 deacetylase led to pleiotropic activation and overexpression of more than 75 % of the biosynthetic genes. Sampling of the overproduced compounds led to the isolation of ten compounds of which four contained new structures, including the cyclic peptides arbumycin and arbumelin, the diterpenoid arbuscullic acid A, and the meroterpenoid arbuscullic acid B. Such epigenetic modifications therefore provide a rapid and global approach to mine the chemical diversity of endophytic fungi.

Co-reporter:Elizabeth L. Noey;Nidhi Tibrewal;Gonzalo Jiménez-Osés;Sílvia Osuna;Jiyong Park;Carly M. Bond;Duilio Cascio;Jack Liang;Xiyun Zhang;Gjalt W. Huisman;Kendall N. Houk
PNAS 2015 112 (51 ) pp:E7065-E7072
Publication Date(Web):2015-12-22
DOI:10.1073/pnas.1507910112
Mutants of Lactobacillus kefir short-chain alcohol dehydrogenase, used here as ketoreductases (KREDs), enantioselectively reduce the pharmaceutically relevant substrates 3-thiacyclopentanone and 3-oxacyclopentanone. These substrates differ by only the heteroatom (S or O) in the ring, but the KRED mutants reduce them with different enantioselectivities. Kinetic studies show that these enzymes are more efficient with 3-thiacyclopentanone than with 3-oxacyclopentanone. X-ray crystal structures of apo- and NADP+-bound selected mutants show that the substrate-binding loop conformational preferences are modified by these mutations. Quantum mechanical calculations and molecular dynamics (MD) simulations are used to investigate the mechanism of reduction by the enzyme. We have developed an MD-based method for studying the diastereomeric transition state complexes and rationalize different enantiomeric ratios. This method, which probes the stability of the catalytic arrangement within the theozyme, shows a correlation between the relative fractions of catalytically competent poses for the enantiomeric reductions and the experimental enantiomeric ratio. Some mutations, such as A94F and Y190F, induce conformational changes in the active site that enlarge the small binding pocket, facilitating accommodation of the larger S atom in this region and enhancing S-selectivity with 3-thiacyclopentanone. In contrast, in the E145S mutant and the final variant evolved for large-scale production of the intermediate for the antibiotic sulopenem, R-selectivity is promoted by shrinking the small binding pocket, thereby destabilizing the pro-S orientation.
Co-reporter:Muxun Zhao ; Yarong Liu ; Renee S. Hsieh ; Nova Wang ; Wanyi Tai ; Kye-Il Joo ; Pin Wang ; Zhen Gu
Journal of the American Chemical Society 2014 Volume 136(Issue 43) pp:15319-15325
Publication Date(Web):October 7, 2014
DOI:10.1021/ja508083g
Encapsulating anticancer protein therapeutics in nanocarriers is an attractive option to minimize active drug destruction, increase local accumulation at the disease site, and decrease side effects to other tissues. Tumor-specific ligands can further facilitate targeting the nanocarriers to tumor cells and reduce nonspecific cellular internalization. Rationally designed non-covalent protein nanocapsules incorporating copper-free “click chemistry” moieties, polyethylene glycol (PEG) units, redox-sensitive cross-linker, and tumor-specific targeting ligands were synthesized to selectively deliver intracellular protein therapeutics into tumor cells via receptor-mediated endocytosis. These nanocapsules can be conjugated to different targeting ligands of choice, such as anti-Her2 antibody single-chain variable fragment (scFv) and luteinizing hormone releasing hormone (LHRH) peptide, resulting in specific and efficient accumulation within tumor cells overexpressing corresponding receptors. LHRH-conjugated nanocapsules selectively delivered recombinant human tumor suppressor protein p53 and its tumor-selective supervariant into targeted tumor cells, which led to reactivation of p53-mediated apoptosis. Our results validate a general approach for targeted protein delivery into tumor cells using cellular-responsive nanocarriers, opening up new opportunities for the development of intracellular protein-based anticancer treatment.
Co-reporter:Hsiao-Ching Lin ; Yuta Tsunematsu ; Sourabh Dhingra ; Wei Xu ; Manami Fukutomi ; Yit-Heng Chooi ; David E. Cane ; Ana M. Calvo ; Kenji Watanabe
Journal of the American Chemical Society 2014 Volume 136(Issue 11) pp:4426-4436
Publication Date(Web):February 26, 2014
DOI:10.1021/ja500881e
Fumagillin (1), a meroterpenoid from Aspergillus fumigatus, is known for its antiangiogenic activity due to binding to human methionine aminopeptidase 2. 1 has a highly oxygenated structure containing a penta-substituted cyclohexane that is generated by oxidative cleavage of the bicyclic sesquiterpene β-trans-bergamotene. The chemical nature, order, and biochemical mechanism of all the oxygenative tailoring reactions has remained enigmatic despite the identification of the biosynthetic gene cluster and the use of targeted-gene deletion experiments. Here, we report the identification and characterization of three oxygenases from the fumagillin biosynthetic pathway, including a multifunctional cytochrome P450 monooxygenase, a hydroxylating nonheme-iron-dependent dioxygenase, and an ABM family monooxygenase for oxidative cleavage of the polyketide moiety. Most significantly, the P450 monooxygenase is shown to catalyze successive hydroxylation, bicyclic ring-opening, and two epoxidations that generate the sesquiterpenoid core skeleton of 1. We also characterized a truncated polyketide synthase with a ketoreductase function that controls the configuration at C-5 of hydroxylated intermediates.
Co-reporter:Wei Xu, Diego J. Gavia and Yi Tang  
Natural Product Reports 2014 vol. 31(Issue 10) pp:1474-1487
Publication Date(Web):02 Sep 2014
DOI:10.1039/C4NP00073K
Covering: up to 2014 This review provides a summary of recent research advances in elucidating the biosynthesis of fungal indole alkaloids. The different strategies used to incorporate and derivatize the indole/indoline moieties in various families of fungal indole alkaloids will be discussed, including tryptophan-containing nonribosomal peptides, polyketide-nonribosomal peptide hybrids, and alkaloids derived from other indole building blocks. This review also includes a discussion regarding the downstream modifications that generate chemical and structural diversity among indole alkaloids.
Co-reporter:Yi Zou, Wei Xu, Yuta Tsunematsu, Mancheng Tang, Kenji Watanabe, and Yi Tang
Organic Letters 2014 Volume 16(Issue 24) pp:6390-6393
Publication Date(Web):December 10, 2014
DOI:10.1021/ol503179v
Biochemical studies of purified and dissected fungal polyketide synthase and nonribosomal peptide synthetase (PKS-NRPS) hybrid enzymes involved in biosynthesis of pseurotin and aspyridone indicate that one α-methylation step during polyketide synthesis is a prerequisite and a key checkpoint for chain transfer between PKS and NRPS modules. In the absence of the resulting γ-methyl feature, the completed polyketide intermediate is offloaded as an α-pyrone instead of being aminoacylated by the NRPS domain. These examples illustrate that precisely timed tailoring domain activities play critical roles in the overall programming of the iterative PKS (and NRPS) functions.
Co-reporter:Angelica O. Zabala, Yit-Heng Chooi, Moon Seok Choi, Hsiao-Ching Lin, and Yi Tang
ACS Chemical Biology 2014 Volume 9(Issue 7) pp:1576
Publication Date(Web):May 20, 2014
DOI:10.1021/cb500284t
Fungal highly reducing polyketide synthases (HRPKSs) are an enigmatic group of multidomain enzymes that catalyze the biosynthesis of structurally diverse compounds. This variety stems from their intrinsic programming rules, which permutate the use of tailoring domains and determine the overall number of iterative cycles. From genome sequencing and mining of the producing strain Eupenicillium brefeldianum ATCC 58665, we identified an HRPKS involved in the biosynthesis of an important protein transport-inhibitor Brefeldin A (BFA), followed by reconstitution of its activity in Saccharomyces cerevisiae and in vitro. Bref-PKS demonstrated an NADPH-dependent reductive tailoring specificity that led to the synthesis of four different octaketide products with varying degrees of reduction. Furthermore, contrary to what is expected from the structure of BFA, Bref-PKS is found to be a nonaketide synthase in the absence of an associated thiohydrolase Bref-TH. Such chain-length control by the partner thiohydrolase was found to be present in other HRPKS systems and highlights the importance of including tailoring enzyme activities in predicting fungal HRPKS functions and their products.
Co-reporter:Li Li, Peng Wang and Yi Tang
The Journal of Antibiotics 2014 67(1) pp:65-70
Publication Date(Web):September 11, 2013
DOI:10.1038/ja.2013.88
Glycosylation with deoxysugar is a common strategy used by nature to introduce structural diversity and biological activities among natural products. In this study, we biochemically confirmed the activities of SsfS6, a C-glycosyltransferase in the SF2575 biosynthetic pathway, as a regioselective D-olivose transferase that acts on the C-9 position of an anhydrotetracycline aglycon. To perform the glycosyl transfer reaction using Escherichia coli as a whole-cell biocatalyst, we reconstituted the biosynthesis of TDP-D-olivose using a heterologous pathway. Under in vivo conditions, SsfS6 transferred multiple endogenous sugar substrates, in addition to D-olivose, to the anhydrotetracycline substrate, demonstrating broad substrate tolerance and potential as a tetracycline-diversifying enzyme.
Co-reporter:Peng Wang ; Ghader Bashiri ; Xue Gao ; Michael R. Sawaya
Journal of the American Chemical Society 2013 Volume 135(Issue 19) pp:7138-7141
Publication Date(Web):April 26, 2013
DOI:10.1021/ja403516u
Tetracyclines are a group of natural products sharing a linearly fused four-ring scaffold, which is essential for their broad-spectrum antibiotic activities. Formation of the key precursor anhydrotetracycline 3 during oxytetracycline 1 biosynthesis has been previously characterized. However, the enzymatic steps that transform 3 into 1, including the additional hydroxylation at C5 and the final C5a–C11a reduction, have remained elusive. Here we report two redox enzymes, OxyS and OxyR, are sufficient to convert 3 to 1. OxyS catalyzes two sequential hydroxylations at C6 and C5 positions of 3 with opposite stereochemistry, while OxyR catalyzes the C5a–C11a reduction using F420 as a cofactor to produce 1. The crystal structure of OxyS was obtained to provide insights into the tandem C6- and C5-hydroxylation steps. The substrate specificities of OxyS and OxyR were shown to influence the relative ratio of 1 and tetracycline 2.
Co-reporter:Wei Jiang ; Ralph A. Cacho ; Grace Chiou ; Neil K. Garg ; Yi Tang ;Christopher T. Walsh
Journal of the American Chemical Society 2013 Volume 135(Issue 11) pp:4457-4466
Publication Date(Web):February 28, 2013
DOI:10.1021/ja312572v
The echinocandins are a small group of fungal N-acylated cyclic hexapeptides that are fungicidal for candida strains and fungistatic for aspergilli by targeting cell wall 1,3-β-glucan synthases. The side chains of all six amino acid building blocks have hydroxyl groups, including the nonproteinogenic 4R,5R-dihydroxy-Orn1, 4R-OH-Pro3, 3S,4S-dihydroxy-homoTyr4, and 3S-OH-4S-Me-Pro6. The echinocandin (ecd) gene cluster contains two predicted nonheme mononuclear iron oxygenase genes (ecdG,K) and one encoding a P450 type heme protein (ecdH). Deletion of the ecdH gene in the producing strain Emericella rugulosa generates an echinocandin scaffold (echinocandin D) lacking both hydroxyl groups on Orn1. Correspondingly, the ΔecdG strain failed to hydroxylate C3 of the homoTyr residue, and purified EcdG hydroxylated free L-homoTyr at C3. The ΔecdK strain failed to generate mature echinocandin unless supplemented with either 4R-Me-Pro or 3S-OH-4S-Me-Pro, indicating blockage of a step upstream of Me-Pro formation. Purified EcdK is a Leu 5-hydroxylase, acting iteratively at C5 to yield γ-Me-Glu-γ-semialdehyde in equilibrium with the cyclic imine product. Evaluation of deshydroxyechinocandin scaffolds in the in vitro anticandidal assays revealed up to a 3-fold loss of potency for the ΔecdG scaffolds, but a 3-fold gain of potency for the ΔecdH scaffold, in line with prior results on deoxyechinocandin homologues.
Co-reporter:Zhizeng Gao ; Jingjing Wang ; Amy K. Norquay ; Kangjian Qiao ; Yi Tang ;John C. Vederas
Journal of the American Chemical Society 2013 Volume 135(Issue 5) pp:1735-1738
Publication Date(Web):January 28, 2013
DOI:10.1021/ja4001823
Iterative polyketide synthases (PKSs) are large, multifunctional enzymes that resemble eukaryotic fatty acid synthases but can make highly functionalized secondary metabolites using complex and unresolved programming rules. During biosynthesis of the kinase inhibitor hypothemycin by Hypomyces subiculosus, a highly reducing iterative PKS, Hpm8, cooperates with a nonreducing iterative PKS, Hpm3, to construct the advanced intermediate dehydrozearalenol (DHZ). The identity of putative intermediates in the formation of the highly reduced hexaketide portion of DHZ were confirmed by incorporation of 13C-labeled N-acetylcysteamine (SNAC) thioesters using the purified enzymes. The results show that Hpm8 can accept SNAC thioesters of intermediates that are ready for transfer from its acyl carrier protein domain to its ketosynthase domain and assemble them into DHZ in cooperation with Hpm3. Addition of certain structurally modified analogues of intermediates to Hpm8 and Hpm3 can produce DHZ derivatives.
Co-reporter:Yit-Heng Chooi ; Young J. Hong ; Ralph A. Cacho ; Dean J. Tantillo
Journal of the American Chemical Society 2013 Volume 135(Issue 45) pp:16805-16808
Publication Date(Web):October 25, 2013
DOI:10.1021/ja408966t
Viridicatumtoxin (1) is a tetracycline-like fungal meroterpenoid with a unique, fused spirobicyclic ring system. Puzzlingly, no dedicated terpene cyclase is found in the gene cluster identified in Penicillium aethiopicum. Cytochrome P450 enzymes VrtE and VrtK in the vrt gene cluster were shown to catalyze C5-hydroxylation and spirobicyclic ring formation, respectively. Feeding acyclic previridicatumtoxin to Saccharomyces cerevisiae expressing VrtK confirmed that VrtK is the sole enzyme required for cyclizing the geranyl moiety. Thus, VrtK is the first example of a P450 that can catalyze terpene cyclization, most likely via initial oxidation of C17 to an allylic carbocation. Quantum chemical modeling revealed a possible new tertiary carbocation intermediate E that forms after allylic carbocation formation. Intermediate E can readily undergo concerted 1,2-alkyl shift/1,3-hydride shift, either spontaneously or further aided by VrtK, followed by C7 Friedel–Crafts alkylation to afford 1. The most likely stereochemical course of the reaction was proposed on the basis of the results of our computations.
Co-reporter:Hsiao-Ching Lin ; Yit-Heng Chooi ; Sourabh Dhingra ; Wei Xu ; Ana M. Calvo
Journal of the American Chemical Society 2013 Volume 135(Issue 12) pp:4616-4619
Publication Date(Web):March 14, 2013
DOI:10.1021/ja312503y
Fumagillin 1 is a meroterpenoid from Aspergillus fumigatus that is known for its anti-angiogenic activity by binding to human methionine aminopeptidase 2. The genetic and molecular basis for biosynthesis of 1 had been an enigma despite the availability of the A. fumigatus genome sequence. Here, we report the identification and verification of the fma gene cluster, followed by characterization of the polyketide synthase and acyltransferase involved in biosynthesis of the dioic acid portion of 1. More significantly, we uncovered the elusive β-trans-bergamotene synthase in A. fumigatus as a membrane-bound terpene cyclase.
Co-reporter:Yit-Heng Chooi, Jinxu Fang, Hong Liu, Scott G. Filler, Pin Wang, and Yi Tang
Organic Letters 2013 Volume 15(Issue 4) pp:780-783
Publication Date(Web):January 31, 2013
DOI:10.1021/ol303435y
Activation of the polycyclic polyketide prenyltransferase (pcPTase)-containing silent clusters in Aspergillus fumigatus and Neosartorya fischeri led to isolation of a new metabolite neosartoricin (3). The structure of 3 was solved by X-ray crystallography and NMR to be a prenylated anthracenone. 3 exhibits T-cell antiproliferative activity with an IC50 of 3 μM, suggestive of a physiological role as an immunosuppressive agent.
Co-reporter:Jaclyn M. Winter, Grace Chiou, Ian R. Bothwell, Wei Xu, Neil K. Garg, Minkui Luo, and Yi Tang
Organic Letters 2013 Volume 15(Issue 14) pp:3774-3777
Publication Date(Web):July 9, 2013
DOI:10.1021/ol401723h
A strategy for introducing structural diversity into polyketides by exploiting the promiscuity of an in-line methyltransferase domain in a multidomain polyketide synthase is reported. In vitro investigations using the highly-reducing fungal polyketide synthase CazF revealed that its methyltransferase domain accepts the nonnatural cofactor propargylic Se-adenosyl-l-methionine and can transfer the propargyl moiety onto its growing polyketide chain. This propargylated polyketide product can then be further chain-extended and cyclized to form propargyl-α pyrone or be processed fully into the alkyne-containing 4′-propargyl-chaetoviridin A.
Co-reporter:Wen-Bing Yin, Yit Heng Chooi, Adam R. Smith, Ralph A. Cacho, Youcai Hu, Theodore C. White, and Yi Tang
ACS Synthetic Biology 2013 Volume 2(Issue 11) pp:629
Publication Date(Web):June 3, 2013
DOI:10.1021/sb400048b
Dermatophytes belonging to the Trichophyton and Arthroderma genera cause skin infections in humans and animals. From genome sequencing data, we mined a conserved gene cluster among dermatophytes that are homologous to one that produces an immunosuppressive polyketide in Aspergillus fumigatus. Using a recombination-based cloning strategy in yeast, we constructed fungal heterologous expression vectors that encode the cryptic clusters. When integrated into the model Aspergillus nidulans host, a structurally related compound neosartoricin B was formed, suggesting a possible role of this compound in the pathogenesis of these strains.Keywords: heterologous expression; natural products; polyketide; prenyltransferase;
Co-reporter:Ralph A. Cacho, Yi Tang
Chemistry & Biology 2013 Volume 20(Issue 1) pp:3-5
Publication Date(Web):24 January 2013
DOI:10.1016/j.chembiol.2013.01.005
In this issue of Chemistry & Biology, Zhang and colleagues developed a yeast cell surface display strategy to effectively evolve the substrate specificity of DhbE, one of the adenylation domains of the bacillibactin synthetase complex. The method yields DbhE variants that have dramatically altered substrate specifities toward unnatural aryl substrates.
Co-reporter:Xue Gao, Wei Jiang, Gonzalo Jiménez-Osés, Moon Seok Choi, Kendall N. Houk, Yi Tang, Christopher T. Walsh
Chemistry & Biology 2013 Volume 20(Issue 7) pp:870-878
Publication Date(Web):25 July 2013
DOI:10.1016/j.chembiol.2013.04.019
•NRPS AspA is reconstituted to synthesize multicyclic fungal alkaloids asperlicin C and D•The first module of AspA iteratively activates two molecules of anthranilate•CT cyclizes Ant-Ant-Trp into a macrolactam that undergoes transannular cyclization•Kinetic factors determine the regiochemical outcome of the transannular cyclizationsThe bimodular 276 kDa nonribosomal peptide synthetase AspA from Aspergillus alliaceus, heterologously expressed in Saccharomyces cerevisiae, converts tryptophan and two molecules of the aromatic β-amino acid anthranilate (Ant) into a pair of tetracyclic peptidyl alkaloids asperlicin C and D in a ratio of 10:1. The first module of AspA activates and processes two molecules of Ant iteratively to generate a tethered Ant-Ant-Trp-S-enzyme intermediate on module two. Release is postulated to involve tandem cyclizations, in which the first step is the macrocyclization of the linear tripeptidyl-S-enzyme, by the terminal condensation (CT) domain to generate the regioisomeric tetracyclic asperlicin scaffolds. Computational analysis of the transannular cyclization of the 11-membered macrocyclic intermediate shows that asperlicin C is the kinetically favored product due to the high stability of a conformation resembling the transition state for cyclization, while asperlicin D is thermodynamically more stable.
Co-reporter:Stuart W. Haynes, Xue Gao, Yi Tang, and Christopher T. Walsh
ACS Chemical Biology 2013 Volume 8(Issue 4) pp:741
Publication Date(Web):January 20, 2013
DOI:10.1021/cb3006787
Ardeemins are hexacyclic peptidyl alkaloids isolated from Aspergillus fischeri as agents that block efflux of anticancer drugs by MultiDrug Resistance (MDR) export pumps. To evaluate the biosynthetic logic and enzymatic machinery for ardeemin framework assembly, we sequenced the A. fischeri genome and identified the ardABC gene cluster. Through both genetic deletions and biochemical characterizations of purified ArdA and ArdB we show this ArdAB enzyme pair is sufficient to convert anthranilate (Ant), l-Ala, and l-Trp to ardeemin. ArdA is a 430 kDa trimodular nonribosomal peptide synthase (NRPS) that converts the three building blocks into a fumiquinazoline (FQ) regioisomer termed ardeemin FQ. ArdB is a prenyltransferase that takes tricyclic ardeemin FQ and dimethylallyl diphosphate to the hexacyclic ardeemin scaffold via prenylation at C2 of the Trp-derived indole moiety with intramolecular capture by an amide NH of the fumiquinazoline ring. The two-enzyme ArdAB pathway reveals remarkable efficiency in construction of the hexacyclic peptidyl alkaloid scaffold.
Co-reporter:Ralph A. Cacho, Yit-Heng Chooi, Hui Zhou, and Yi Tang
ACS Chemical Biology 2013 Volume 8(Issue 10) pp:2322
Publication Date(Web):August 25, 2013
DOI:10.1021/cb400541z
Griseofulvin (1) is a spirocyclic fungal natural product used in treatment of fungal dermatophytes. Formation of the spirocycle, or the grisan scaffold, from a benzophenone precursor is critical for the activity of 1. In this study, we have systematically characterized each of the biosynthetic enzymes related to the biogenesis of 1, including the characterization of a new polyketide synthase GsfA that synthesizes the benzophenone precursor and a cytochrome P450 GsfF that performs oxidative coupling between the orcinol and the phloroglucinol rings to yield the grisan structure. Notably, the finding of GsfF is in sharp contrast to the copper-dependent dihydrogeodin oxidase that performs a similar reaction in the geodin biosynthetic pathway. The biosynthetic knowledge enabled the in vitro total biosynthesis of 1 from malonyl-CoA using all purified enzyme components. This work therefore completely maps out the previously unresolved enzymology of the biosynthesis of a therapeutically relevant natural product
Co-reporter:Muxun Zhao, Biliang Hu, Zhen Gu, Kye-Il Joo, Pin Wang, Yi Tang
Nano Today 2013 Volume 8(Issue 1) pp:11-20
Publication Date(Web):February 2013
DOI:10.1016/j.nantod.2012.12.003
The development of stimuli-responsive, nano-scale therapeutics that selectively target and attack tumors is a major research focus in cancer nanotechnology. A potent therapeutic option is to directly arming the cancer cells with apoptotic-inducing proteins that are not affected by tumoral anti-apoptotic maneuvers. The avian virus-derived apoptin forms a high-molecular weight protein complex that selectively accumulates in the nucleus of cancer cell to induce apoptotic cell death. To achieve the efficient intracellular delivery of this tumor-selective protein in functional form, we synthesized degradable, sub-100 nm, core–shell protein nanocapsules containing the 2.4 MDa apoptin complexes. Recombinant apoptin is reversibly encapsulated in a positively charged, water soluble polymer shell and is released in native form in response to reducing conditions such as the cytoplasm. As characterized by confocal microscopy, the nanocapsules are efficiently internalized by mammalian cells lines, with accumulation of rhodamine-labeled apoptin in the nuclei of cancer cells only. Intracellularly released apoptin induced tumor-specific apoptosis in several cancer cell lines and inhibited tumor growth in vivo, demonstrating the potential of this polymer–protein combination as an anticancer therapeutic.Graphical abstractHighlights► We synthesized polymer nanocapsules for delivery of tumor-selective protein. ► The nanocapsule can be internalized by different tumor cells. ► The protein cargo is released intracellularly in the cytoplasm. ► Released complex induced apoptosis and inhibited tumor growth.
Co-reporter:Wei Xu;Dr. Yit-Heng Chooi;Jin W. Choi; Shuang Li; John C. Vederas; Nancy A. DaSilva; Yi Tang
Angewandte Chemie 2013 Volume 125( Issue 25) pp:6600-6603
Publication Date(Web):
DOI:10.1002/ange.201302406
Co-reporter:Wei Xu;Dr. Yit-Heng Chooi;Jin W. Choi; Shuang Li; John C. Vederas; Nancy A. DaSilva; Yi Tang
Angewandte Chemie International Edition 2013 Volume 52( Issue 25) pp:6472-6475
Publication Date(Web):
DOI:10.1002/anie.201302406
Co-reporter:Jaclyn M. Winter ; Michio Sato ; Satoru Sugimoto ; Grace Chiou ; Neil K. Garg ; Yi Tang ;Kenji Watanabe
Journal of the American Chemical Society 2012 Volume 134(Issue 43) pp:17900-17903
Publication Date(Web):October 16, 2012
DOI:10.1021/ja3090498
We report the identification and characterization of the caz biosynthetic cluster from Chaetomium globosum and the characterization of a highly reducing polyketide synthase (PKS) that acts in both a sequential and convergent manner with a nonreducing PKS to form the chaetomugilin and chaetoviridin azaphilones. Genetic inactivation studies verified the involvement of individual caz genes in the biosynthesis of the azaphilones. Through in vitro reconstitution, we demonstrated the in vitro synthesis of chaetoviridin A from the pyranoquinone intermediate cazisochromene using the highly reducing PKS and an acyltransferase.
Co-reporter:Stuart W. Haynes ; Xue Gao ; Yi Tang ;Christopher T. Walsh
Journal of the American Chemical Society 2012 Volume 134(Issue 42) pp:17444-17447
Publication Date(Web):October 2, 2012
DOI:10.1021/ja308371z
Members of the asperlicin family of fungal metabolites produced by Aspergillus alliaceus are known potent CCKA antagonists. Herein, we report the identification of the gene cluster responsible for directing their biosynthesis. We validate and probe the pathway by genetic manipulation, and provide the first biochemical characterization of the oxidative cyclization en route to the heptacyclic asperlicin E by reconstituting the activity of the FAD depend monooxygenase AspB. This report provides the first genetic characterization of a NRPS assembly line that efficiently activates two anthranilate building blocks and illustrates the remarkably efficient biosynthesis of the complex heptacyclic asperlicin E.
Co-reporter:Ralph A. Cacho ; Wei Jiang ; Yit-Heng Chooi ; Christopher T. Walsh
Journal of the American Chemical Society 2012 Volume 134(Issue 40) pp:16781-16790
Publication Date(Web):September 21, 2012
DOI:10.1021/ja307220z
Echinocandins are a family of fungal lipidated cyclic hexapeptide natural products. Due to their effectiveness as antifungal agents, three semisynthetic derivatives have been developed and approved for treatment of human invasive candidiasis. All six of the amino acid residues are hydroxylated, including 4R,5R-dihydroxy-l-ornithine, 4R-hydroxyl-l-proline, 3S,4S-dihydroxy-l-homotyrosine, and 3S-hydroxyl-4S-methyl-l-proline. We report here the biosynthetic gene cluster of echinocandin B 1 from Emericella rugulosa NRRL 11440 containing genes encoding for a six-module nonribosomal peptide synthetase EcdA, an acyl-AMP ligase EcdI, and oxygenases EcdG, EcdH, and EcdK. We showed EcdI activates linoleate as linoleyl-AMP and installs it on the first thiolation domain of EcdA. We have also established through ATP–PPi exchange assay that EcdA loads l-ornithine in the first module. A separate hty gene cluster encodes four enzymes for de novo generation of l-homotyrosine from acetyl-CoA and 4-hydroxyphenyl-pyruvate is found from the sequenced genome. Deletions in the ecdA, and htyA genes validate their essential roles in echinocandin B production. Five predicted iron-centered oxygenase genes, ecdG, ecdH, ecdK, htyE, and htyF, in the two separate ecd and hty clusters are likely to be the tailoring oxygenases for maturation of the nascent NRPS lipohexapeptidolactam product.
Co-reporter:Yit-Heng Chooi ; Peng Wang ; Jinxu Fang ; Yanran Li ; Katherine Wu ; Pin Wang
Journal of the American Chemical Society 2012 Volume 134(Issue 22) pp:9428-9437
Publication Date(Web):May 16, 2012
DOI:10.1021/ja3028636
The prenyltransferase (PTase) gene vrtC was proposed to be involved in viridicatumtoxin (1) biosynthesis in Penicillium aethiopicum. Targeted gene deletion and reconstitution of recombinant VrtC activity in vitro established that VrtC is a geranyl transferase that catalyzes a regiospecific Friedel–Crafts alkylation of the naphthacenedione carboxamide intermediate 2 at carbon 6 with geranyl diphosphate. VrtC can function in the absence of divalent ions and can utilize similar naphthacenedione substrates, such as the acetyl-primed TAN-1612 (4). Genome mining using the VrtC protein sequence leads to the identification of a homologous group of PTase genes in the genomes of human and animal-associated fungi. Three enzymes encoded by this new subgroup of PTase genes from Neosartorya fischeri, Microsporum canis, and Trichophyton tonsurans were shown to be able to catalyze transfer of dimethylallyl to several tetracyclic naphthacenedione substrates in vitro. In total, seven C5- or C10-prenylated naphthacenedione compounds were generated. The regioselectivity of these new polycyclic PTases (pcPTases) was confirmed by characterization of product 9 obtained from biotransformation of 4 in Escherichia coli expressing the N. fischeri pcPTase gene. The discovery of this new subgroup of PTases extends our enzymatic tools for modifying polycyclic compounds and enables genome mining of new prenylated polyketides.
Co-reporter:Peng Wang, Xue Gao, Yi Tang
Current Opinion in Chemical Biology 2012 Volume 16(3–4) pp:362-369
Publication Date(Web):August 2012
DOI:10.1016/j.cbpa.2012.04.008
Redox enzymes such as FAD-dependent and cytochrome P450 oxygenases play indispensible roles in generating structural complexity during natural product biosynthesis. In the pre-assembly steps, redox enzymes can convert garden variety primary metabolites into unique starter and extender building blocks. In the post-assembly tailoring steps, redox cascades can transform nascent scaffolds into structurally complex final products. In this review, we will discuss several recently characterized redox enzymes in the biosynthesis of polyketides and nonribosomal peptides.Graphical abstractHighlights► Redox enzymes play important roles in pre-assembly and post-assembly modifications. ► Both PKS and NRPS pathways use redox enzymes to introduce structural complexity. ► Recent discoveries of novel redox enzymes are discussed.
Co-reporter:Qian Liu, Fen Yao, Yit Heng Chooi, Qianjin Kang, Wei Xu, Yanran Li, Yucheng Shao, Yuefeng Shi, Zixin Deng, Yi Tang, Delin You
Chemistry & Biology 2012 Volume 19(Issue 2) pp:243-253
Publication Date(Web):24 February 2012
DOI:10.1016/j.chembiol.2011.12.018
Piericidins are a class of α-pyridone antibiotics that inhibit mitochondrial respiratory chain and exhibit antimicrobial, antifungal, and antitumor activities. Sequential analysis of Streptomyces piomogeues var. Hangzhouwanensis genome revealed six modular polyketide synthases, an amidotransferase, two methyltransferases, and a monooxygenase for piericidin A1 production. Gene functional analysis and deletion results provide overview of the biosynthesis pathway. Furthermore, in vitro characterization of the terminal polyketide synthase module with the thioesterase domain using β-ketoacyl substrates was performed. That revealed a pathway where the α-pyridone ring formation is dependent on hydrolysis of the product β, δ-diketo carboxylic acid by the C-terminal thioesterase followed by amidation and cyclization. These findings set the stage to investigate unusual enzymatic mechanisms in α-pyridone antibiotics biosynthesis, provide a foundation for genome mining of α-pyridone antibiotics, and produce analogs by molecular engineering.Graphical AbstractFigure optionsDownload full-size imageDownload high-quality image (190 K)Download as PowerPoint slideHighlights► Genome scanning revealed a pathway for α-pyridone antibiotics in bacteria ► Gene replacements verified pieA, C, D are involved in piericidin A1 biosynthesis ► In vitro characterization showed that PieA-TE mediates polyketide hydrolysis ► Biosynthesis of α-pyridone via hydrolysis followed by amidation and cyclization
Co-reporter:Angelica O. Zabala, Wei Xu, Yit-Heng Chooi, Yi Tang
Chemistry & Biology 2012 Volume 19(Issue 8) pp:1049-1059
Publication Date(Web):24 August 2012
DOI:10.1016/j.chembiol.2012.07.004
Azaphilones are a class of fungal metabolites characterized by a highly oxygenated pyrano-quinone bicyclic core and exhibiting a broad range of bioactivities. Although widespread among various fungi, their biosynthesis has not been thoroughly elucidated. By activation of a silent (aza) gene cluster in Aspergillus niger ATCC 1015, we discovered six azaphilone compounds, azanigerones A–F (1, 3–7). Transcriptional analysis and deletion of a key polyketide synthase (PKS) gene further confirmed the involvement of the aza gene cluster. The biosynthetic pathway was shown to involve the convergent actions of a highly reducing PKS and a non-reducing PKS. Most significantly, in vitro reaction of a key flavin-dependent monooxygenase encoded in the cluster with an early benzaldehyde intermediate revealed its roles in hydroxylation and pyran-ring formation to afford the characteristic bicylic core shared by azaphilones.Graphical AbstractFigure optionsDownload full-size imageDownload high-quality image (244 K)Download as PowerPoint slideHighlights► Discovery of an azaphilone pathway in Aspergillus niger ► Six azaphilone compounds were isolated and characterized ► The biosynthesis involves convergent actions of an HR-PKS and an NR-PKS ► Hydroxylation by a monooxygenase promotes pyran-ring formation
Co-reporter:Angelica O. Zabala;Ralph A. Cacho
Journal of Industrial Microbiology & Biotechnology 2012 Volume 39( Issue 2) pp:227-241
Publication Date(Web):2012 February
DOI:10.1007/s10295-011-1044-2
A dazzling array of enzymes is used by nature in making structurally complex natural products. These enzymes constitute a molecular toolbox that may be used in the construction and fine-tuning of pharmaceutically active molecules. Aided by technological advancements in protein engineering, it is now possible to tailor the activities and specificities of these enzymes as biocatalysts in the production of both natural products and their unnatural derivatives. These efforts are crucial in drug discovery and development, where there is a continuous quest for more potent agents. Both rational and random evolution techniques have been utilized in engineering these enzymes. This review will highlight some examples from several large families of natural products.
Co-reporter:Peng Wang;Dr. Woncheol Kim;Dr. Lauren B. Pickens;Xue Gao ; Yi Tang
Angewandte Chemie 2012 Volume 124( Issue 44) pp:11298-11302
Publication Date(Web):
DOI:10.1002/ange.201205426
Co-reporter:Anuradha Biswas, Ying Liu, Tianfei Liu, Guoping Fan, Yi Tang
Biomaterials 2012 33(21) pp: 5459-5467
Publication Date(Web):
DOI:10.1016/j.biomaterials.2012.04.012
Co-reporter:Peng Wang;Dr. Woncheol Kim;Dr. Lauren B. Pickens;Xue Gao ; Yi Tang
Angewandte Chemie International Edition 2012 Volume 51( Issue 44) pp:11136-11140
Publication Date(Web):
DOI:10.1002/anie.201205426
Co-reporter:Yit-Heng Chooi and Yi Tang
The Journal of Organic Chemistry 2012 Volume 77(Issue 22) pp:9933-9953
Publication Date(Web):August 31, 2012
DOI:10.1021/jo301592k
The iterative type I polyketide synthases (IPKSs) are central to the biosynthesis of an enormously diverse array of natural products in fungi. These natural products, known as polyketides, exhibit a wide range of biological activities and include clinically important drugs as well as undesirable toxins. The PKSs synthesize these structurally diverse polyketides via a series of decarboxylative condensations of malonyl-CoA extender units and β-keto modifications in a highly programmed manner. Significant progress has been made over the past few years in understanding the biosynthetic mechanism and programming of fungal PKSs. The continuously expanding fungal genome sequence data have sparked genome-directed discoveries of new fungal PKSs and associated products. The increasing number of fungal PKSs that have been linked to their products along with in-depth biochemical and structural characterizations of these large enzymes have remarkably improved our knowledge on the molecular basis for polyketide structural diversity in fungi. This Perspective highlights the recent advances and examines how the newly expanded paradigm has contributed to our ability to link fungal PKS genes to chemical structures and vice versa. The knowledge will help us navigate through the logarithmically expanding seas of genomic information for polyketide compound discovery and provided opportunities to reprogram these megasynthases to generate new chemical entities.
Co-reporter:Yanran Li ; Yit-Heng Chooi ; Yuewei Sheng ; Joan S. Valentine
Journal of the American Chemical Society 2011 Volume 133(Issue 39) pp:15773-15785
Publication Date(Web):August 25, 2011
DOI:10.1021/ja206906d
The linear tetracyclic TAN-1612 (1) and BMS-192548 (2) were isolated from different filamentous fungal strains and have been examined as potential neuropeptide Y and neurokinin-1 receptor antagonists, respectively. Although the biosynthesis of fungal aromatic polyketides has attracted much interest in recent years, the biosynthetic mechanism for such naphthacenedione-containing products has not been established. Using a targeted genome mining approach, we first located the ada gene cluster responsible for the biosynthesis of 1 in Aspergillus niger ATCC 1015. The connection between 1 and the ada pathway was verified through overexpression of the Zn2Cys6-type pathway-specific transcriptional regulator AdaR and subsequent gene expression analysis. The enzymes encoded in the ada gene cluster share high sequence similarities to the known apt pathway linked to the biosynthesis of anthraquinone asperthecin 3. Subsequent comparative investigation of these two highly homologous gene clusters by heterologous pathway reconstitution in Saccharomyces cerevisiae revealed a novel α-hydroxylation-dependent Claisen cyclization cascade, which involves a flavin-dependent monooxygenase that hydroxylates the α-carbon of an acyl carrier protein-bound polyketide and a bifunctional metallo-β-lactamase-type thioesterase (MβL-TE). The bifunctional MβL-TE catalyzes the fourth ring cyclization to afford the naphthacenedione scaffold upon α-hydroxylation, whereas it performs hydrolytic release of an anthracenone product in the absence of α-hydroxylation. Through in vitro biochemical characterizations and metal analyses, we verified that the apt MβL-TE is a dimanganese enzyme and requires both Mn2+ cations for the observed activities. The MβL-TE is the first example of a thioesterase in polyketide biosynthesis that catalyzes the Claisen-like condensation without an α/β hydrolase fold and forms no covalent bond with the substrate. These mechanistic features should be general to the biosynthesis of tetracyclic naphthacenedione compounds in fungi.
Co-reporter:Xue Gao ; Yit-Heng Chooi ; Brian D. Ames ; Peng Wang ; Christopher T. Walsh
Journal of the American Chemical Society 2011 Volume 133(Issue 8) pp:2729-2741
Publication Date(Web):February 7, 2011
DOI:10.1021/ja1101085
Tremorgenic mycotoxins are a group of indole alkaloids which include the quinazoline-containing tryptoquivaline (2) that are capable of eliciting intermittent or sustained tremors in vertebrate animals. The biosynthesis of this group of bioactive compounds, which are characterized by an acetylated quinazoline ring connected to a 6−5−5 imidazoindolone ring system via a 5-membered spirolactone, has remained uncharacterized. Here, we report the identification of a gene cluster (tqa) from P. aethiopicum that is involved in the biosynthesis of tryptoquialanine (1), which is structurally similar to 2. The pathway has been confirmed to go through an intermediate common to the fumiquinazoline pathway, fumiquinazoline F, which originates from a fungal trimodular nonribosomal peptide synthetase (NRPS). By systematically inactivating every biosynthetic gene in the cluster, followed by isolation and characterization of the intermediates, we were able to establish the biosynthetic sequence of the pathway. An unusual oxidative opening of the pyrazinone ring by an FAD-dependent berberine bridge enzyme-like oxidoreductase has been proposed based on genetic knockout studies. Notably, a 2-aminoisobutyric acid (AIB)-utilizing NRPS module has been identified and reconstituted in vitro, along with two putative enzymes of unknown functions that are involved in the synthesis of the unnatural amino acid by genetic analysis. This work provides new genetic and biochemical insights into the biosynthesis of this group of fungal alkaloids, including the tremorgens related to 2.
Co-reporter:Zhen Gu, Muxun Zhao, Yuewei Sheng, Laurent A. Bentolila, and Yi Tang
Analytical Chemistry 2011 Volume 83(Issue 6) pp:2324
Publication Date(Web):February 16, 2011
DOI:10.1021/ac103236g
Mercury is a highly hazardous and widespread pollutant with bioaccumulative properties. Novel approaches that meet the criteria of desired selectivity, high sensitivity, good biocompatibility, and low background interference in natural settings are continuously being explored. We herein describe a new strategy utilizing the combination of infrared fluorescent protein (IFP) and its chromophore as an infrared fluorescence probe for mercury ion (Hg(II)) detection. Hg(II) has been validated to have specific binding affinity to a cysteine residue (C24) of IFP, thereby inhibiting the conjugation of IFP chromophore biliverdin (BV) to C24 and “turning off” the infrared emission of IFP. The IFP/BV sensor has high selectivity toward Hg(II) among other metal ions over a broad pH range. The in vitro detection limit was determined to be less than 50 nM. As a genetically encoded probe, we demonstrate the IFP/BV sensor can serve as a tool to detect Hg(II) in living organisms or tissues. Moreover, we have exploited a protein-agarose hydrogel-based paper assay to immobilize IFP for detection of Hg(II) in a portable and robust fashion.
Co-reporter:Michael J. Meehan, Xinkai Xie, Xiling Zhao, Wei Xu, Yi Tang, and Pieter C. Dorrestein
Biochemistry 2011 Volume 50(Issue 2) pp:
Publication Date(Web):November 11, 2010
DOI:10.1021/bi1014776
There are very few fungal polyketide synthases that have been characterized by mass spectrometry. In this paper we describe the in vitro reconstitution and FT-ICR-MS verification of the full activity of an intact 277 kDa fungal polyketide synthase LovF of the lovastatin biosynthetic pathway. We report here both the verification of the reconstitution of fully functional holo-LovF by using 13C-labeled malonyl-CoA to form α-methylbutyrate functionality and also detection of five predicted intermediates covalently bound to the 4′-phosphopantetheine at the acyl carrier protein (ACP) active site utilizing the phosphopantetheine ejection assay and high-resolution mass spectrometry. Under in vitro conditions, the diketide acetoacetyl intermediate did not accumulate on the ACP active site of holo-LovF following incubation with malonyl-CoA substrate. We found that incubation of holo-LovF with acetoacetyl-CoA served as an effective means of loading the diketide intermediate onto the ACP active site of LovF. Our results demonstrate that subsequent α-methylation of the acetoacetyl intermediate stabilizes the intermediate onto the ACP active site and facilitates the formation and mass spectrometric detection of additional intermediates en route to the formation of α-methylbutyrate.
Co-reporter:Muxun Zhao, Anuradha Biswas, Biliang Hu, Kye-Il Joo, Pin Wang, Zhen Gu, Yi Tang
Biomaterials 2011 32(22) pp: 5223-5230
Publication Date(Web):
DOI:10.1016/j.biomaterials.2011.03.060
Co-reporter:Anuradha Biswas, Kye-Il Joo, Jing Liu, Muxun Zhao, Guoping Fan, Pin Wang, Zhen Gu, and Yi Tang
ACS Nano 2011 Volume 5(Issue 2) pp:1385
Publication Date(Web):January 26, 2011
DOI:10.1021/nn1031005
Proteins possess distinct intracellular roles allowing them to have vast therapeutic applications. However, due to poor cellular permeability and fragility of most proteins, intracellular delivery of native, active proteins is challenging. We describe a biomimetic protein delivery vehicle which is degradable upon the digestion by furin, a ubiquitous intracellular protease, to release encapsulated cargos. Proteins were encapsulated in a nanosized matrix prepared with monomers and a bisacrylated peptide cross-linker which can be specifically recognized and cleaved by furin. Release of encapsulated protein was confirmed in a cell-free system upon proteolytic degradation of nanocapsules. In vitro cell culture studies demonstrated successful intracellular delivery of both nuclear and cytosolic proteins and confirmed the importance of furin-degradable construction for native protein release. This endoprotease-mediated intracellular delivery system may be extended to effectively deliver various biological therapeutics.Keywords: endoprotease; intracellular; nanocapsule; polymer; protein delivery
Co-reporter:Hui Zhou ; Kangjian Qiao ; Zhizeng Gao ; Michael J. Meehan ; Jesse W.-H. Li ; Xiling Zhao ; Pieter C. Dorrestein ; John C. Vederas
Journal of the American Chemical Society 2010 Volume 132(Issue 13) pp:4530-4531
Publication Date(Web):March 11, 2010
DOI:10.1021/ja100060k
Hypothemycin is a macrolide protein kinase inhibitor from the fungus Hypomyces subiculosus. During biosynthesis, its carbon framework is assembled by two iterative polyketide synthases (PKSs), Hpm8 (highly reducing) and Hpm3 (nonreducing). These were heterologously expressed in Saccharomyces cerevisiae BJ5464-NpgA, purified to near homogeneity, and reconstituted in vitro to produce (6′S,10′S)-trans-7′,8′-dehydrozearalenol (1) from malonyl-CoA and NADPH. The structure of 1 was determined by X-ray crystallographic analysis. In the absence of functional Hpm3, the reducing PKS Hpm8 produces and offloads truncated pyrone products instead of the expected hexaketide. The nonreducing Hpm3 is able to accept an N-acetylcysteamine thioester of a correctly functionalized hexaketide to form 1, but it is unable to initiate polyketide formation from malonyl-CoA. We show that the starter-unit:ACP transacylase (SAT) of Hpm3 is critical for crosstalk between the two enzymes and that the rate of biosynthesis of 1 is determined by the rate of hexaketide formation by Hpm8.
Co-reporter:Hui Zhou, Yanran Li and Yi Tang  
Natural Product Reports 2010 vol. 27(Issue 6) pp:839-868
Publication Date(Web):31 Mar 2010
DOI:10.1039/B911518H
Covering: up to the end of 2009
Co-reporter:Zhen Gu, Anuradha Biswas, Kye-Il Joo, Biliang Hu, Pin Wang and Yi Tang  
Chemical Communications 2010 vol. 46(Issue 35) pp:6467-6469
Publication Date(Web):26 Jul 2010
DOI:10.1039/C0CC01439G
We describe a FRET-based protease detection strategy, using a single-fluorescent-protein nanogel as donor and a dark quencher as acceptor linked by a photolabile caged-peptide. This design enables probing of protease activity in a UV-responsive fashion.
Co-reporter:Yit-Heng Chooi, Ralph Cacho, Yi Tang
Chemistry & Biology 2010 Volume 17(Issue 5) pp:483-494
Publication Date(Web):28 May 2010
DOI:10.1016/j.chembiol.2010.03.015
Penicillium aethiopicum produces two structurally interesting and biologically active polyketides: the tetracycline-like viridicatumtoxin 1 and the classic antifungal agent griseofulvin 2. Here, we report the concurrent discovery of the two corresponding biosynthetic gene clusters (vrt and gsf) by 454 shotgun sequencing. Gene deletions confirmed that two nonreducing PKSs (NRPKSs), vrtA and gsfA, are required for the biosynthesis of 1 and 2, respectively. Both PKSs share similar domain architectures and lack a C-terminal thioesterase domain. We identified gsfI as the chlorinase involved in the biosynthesis of 2, because deletion of gsfI resulted in the accumulation of decholorogriseofulvin 3. Comparative analysis with the P. chrysogenum genome revealed that both clusters are embedded within conserved syntenic regions of P. aethiopicum chromosomes. Discovery of the vrt and gsf clusters provided the basis for genetic and biochemical studies of the pathways.Graphical AbstractFigure optionsDownload full-size imageDownload high-quality image (522 K)Download as PowerPoint slideHighlights► 454 sequencing unveiled the gene clusters for viridicatumtoxin 1 and griseofulvin 2 ► vrt and gsf clusters lie within conserved syntenic regions of P. aethiopicum genome ► VrtA and GsfA are nonreducing PKSs required for biosynthesis of 1 and 2 ► gsfI encodes chlorinase involved in the biosynthesis of 2
Co-reporter:Zhen Gu and Yi Tang  
Lab on a Chip 2010 vol. 10(Issue 15) pp:1946-1951
Publication Date(Web):30 Apr 2010
DOI:10.1039/C001335H
The ability to form functional polymeric patterning structures has important implications for the studies of cell biology, tissue engineering, and medical diagnostics. We have developed a novel enzyme-assisted photolithography (EAPL) method for spatial functionalization of hydrogels via a high throughput fashion. A bisacrylated peptide crosslinker, containing a protease cleavable amino acid sequence and caged by a photolabile moiety, is used during hydrogel polymerization. A facile two-step process is employed, including UV exposure to decage the peptide crosslinker at a desired area and protease development to specifically digest gels at UV treated regions only. Importantly, proteolysis of the peptide bonds generates free nucleophilic amine groups at the patterned area that can be further functionalized. Using this strategy and caspase-3 as the enzyme developer, we demonstrate the simultaneous generation of topographical and functional patterns into poly(ethylene glycol) (PEG) hydrogels. We show that 20 μm-wide line arrays functionalized with arginine–glycine–aspartic acid (RGD)-containing peptides can be used to generate cell patterns with individual cell resolution. We also fabricated arrays 20 μm diameter cavities decorated with B lymphocyte specific anti-CD19, which was used to achieve a 600-fold enrichment of B-cells from a 0.1% starting B-cell mixture. The simple fabrication process, straightforward chemistry and an all-aqueous based biocompatible and environmentally friendly approach render EAPL a versatile platform to construct biologically responsive 2D patterns or 3D scaffolds for lab-on-a-chip systems and tissue engineering.
Co-reporter:Yit-Heng Chooi, Yi Tang
Chemistry & Biology 2010 Volume 17(Issue 8) pp:791-793
Publication Date(Web):27 August 2010
DOI:10.1016/j.chembiol.2010.08.001
Surfactin is a member of the lipopeptide family of antibiotics, which includes the clinical drug daptomycin (Cubicin). The potency of these antibiotics is affected by the attached lipid chain, which is incorporated into the nonribosomally assembled peptidyl backbone via a process known as lipoinitiation. Kraas et al. (2010) have provided valuable insights into the lipoinitiation mechanism, which will be useful for future biosynthetic modifications of lipopeptide antibiotics.
Co-reporter:Xue Gao;Peng Wang
Applied Microbiology and Biotechnology 2010 Volume 88( Issue 6) pp:1233-1242
Publication Date(Web):2010 December
DOI:10.1007/s00253-010-2860-4
Polyketides are important bioactive natural products biosynthesized by bacteria, fungi, and plants. The enzymes that synthesize polyketides are collectively referred to as polyketide synthases (PKSs). Because many of the natural hosts that produce polyketides are difficult to culture or manipulate, establishing a universal heterologous host that is genetically tractable has become an important goal toward the engineered biosynthesis of polyketides and analogues. Here, we summarize the recent progresses in engineering Escherichia coli as a heterologous host for reconstituting PKSs of different types. Our increased understanding of PKS enzymology and structural biology, combined with new tools in protein engineering, metabolic engineering, and synthetic biology, has firmly established E. coli as a powerful host for producing polyketides.
Co-reporter:Zhen Gu, Ming Yan, Biliang Hu, Kye-Il Joo, Anuradha Biswas, Yu Huang, Yunfeng Lu, Pin Wang and Yi Tang
Nano Letters 2009 Volume 9(Issue 12) pp:4533-4538
Publication Date(Web):2017-2-22
DOI:10.1021/nl902935b
Target proteins can be functionally encapsulated using a cocoon-like polymeric nanocapsule formed by interfacial polymerization. The nanocapsule is cross-linked by peptides that can be proteolyzed by proteases upon which the protein cargo is released. The protease-mediated degradation process can be controlled in a spatiotemporal fashion through modification of the peptide cross-linker with photolabile moieties. We demonstrate the utility of this approach through the cytoplasmic delivery of the apoptosis inducing caspase-3 to cancer cells.
Co-reporter:Xinkai Xie ; Michael J. Meehan ; Wei Xu ; Pieter C. Dorrestein
Journal of the American Chemical Society 2009 Volume 131(Issue 24) pp:8388-8389
Publication Date(Web):May 28, 2009
DOI:10.1021/ja903203g
LovF is a highly reducing polyketide synthase (HR-PKS) from the filamentous fungus Aspergillus terreus. LovF synthesizes the α-S-methylbutyrate side chain that is subsequently transferred to monacolin J to yield the cholesterol-lowering natural product lovastatin. In the report, we expressed the full length LovF and reconstituted the megasynthase activities in vitro. We confirmed the diketide product of LovF is offloaded from the LovF ACP domain by the dissociated acyltransferase LovD. This represents the first example of acyltransferase-mediated release of polyketide products from fungal PKSs. We determined LovD primarily interacts with the ACP domain of LovF and the protein−protein interactions lead to highly efficient transfer of the diketide product. The catalytic efficiency is enhanced nearly 1 × 106-fold when LovF was used as the acyl carrier instead of N-acetylcysteamine. Reconstitution and characterization of the LovF offloading mechanism provide new insights into the functions of fungal HR-PKS.
Co-reporter:Lauren B. Pickens ; Woncheol Kim ; Peng Wang ; Hui Zhou ; Kenji Watanabe ; Shuichi Gomi
Journal of the American Chemical Society 2009 Volume 131(Issue 48) pp:17677-17689
Publication Date(Web):November 12, 2009
DOI:10.1021/ja907852c
SF2575 1 is a tetracycline polyketide produced by Streptomyces sp. SF2575 and displays exceptionally potent anticancer activity toward a broad range of cancer cell lines. The structure of SF2575 is characterized by a highly substituted tetracycline aglycon. The modifications include methylation of the C-6 and C-12a hydroxyl groups, acylation of the 4-(S)-hydroxyl with salicylic acid, C-glycosylation of the C-9 of the D-ring with d-olivose and further acylation of the C4′-hydroxyl of d-olivose with the unusual angelic acid. Understanding the biosynthesis of SF2575 can therefore expand the repertoire of enzymes that can modify tetracyclines, and facilitate engineered biosynthesis of SF2575 analogues. In this study, we identified, sequenced, and functionally analyzed the ssf biosynthetic gene cluster which contains 40 putative open reading frames. Genes encoding enzymes that can assemble the tetracycline aglycon, as well as installing these unique structural features, are found in the gene cluster. Biosynthetic intermediates were isolated from the SF2575 culture extract to suggest the order of pendant-group addition is C-9 glycosylation, C-4 salicylation, and O-4′ angelylcylation. Using in vitro assays, two enzymes that are responsible for C-4 acylation of salicylic acid were identified. These enzymes include an ATP-dependent salicylyl-CoA ligase SsfL1 and a putative GDSL family acyltransferase SsfX3, both of which were shown to have relaxed substrate specificity toward substituted benzoic acids. Since the salicylic acid moiety is critically important for the anticancer properties of SF2575, verification of the activities of SsfL1 and SsfX3 sets the stage for biosynthetic modification of the C-4 group toward structure−activity relationship studies of SF2575. Using heterologous biosynthesis in Streptomyces lividans, we also determined that biosynthesis of the SF2575 tetracycline aglycon 8 parallels that of oxytetracycline 4 and diverges after the assembly of 4-keto-anhydrotetracycline 51. The minimal ssf polyketide synthase together with the amidotransferase SsfD produced the amidated decaketide backbone that is required for the formation of 2-naphthacenecarboxamide skeleton. Additional enzymes, such as cyclases C-6 methyltransferase and C-4/C-12a dihydroxylase, were functionally reconstituted.
Co-reporter:Xue Gao, Xinkai Xie, Inna Pashkov, Michael R. Sawaya, Janel Laidman, Wenjun Zhang, Ralph Cacho, Todd O. Yeates, Yi Tang
Chemistry & Biology 2009 Volume 16(Issue 10) pp:1064-1074
Publication Date(Web):30 October 2009
DOI:10.1016/j.chembiol.2009.09.017
Enzymes from natural product biosynthetic pathways are attractive candidates for creating tailored biocatalysts to produce semisynthetic pharmaceutical compounds. LovD is an acyltransferase that converts the inactive monacolin J acid (MJA) into the cholesterol-lowering lovastatin. LovD can also synthesize the blockbuster drug simvastatin using MJA and a synthetic α-dimethylbutyryl thioester, albeit with suboptimal properties as a biocatalyst. Here we used directed evolution to improve the properties of LovD toward semisynthesis of simvastatin. Mutants with improved catalytic efficiency, solubility, and thermal stability were obtained, with the best mutant displaying an ∼11-fold increase in an Escherichia coli-based biocatalytic platform. To understand the structural basis of LovD enzymology, seven X-ray crystal structures were determined, including the parent LovD, an improved mutant G5, and G5 cocrystallized with ligands. Comparisons between the structures reveal that beneficial mutations stabilize the structure of G5 in a more compact conformation that is favorable for catalysis.
Co-reporter:Peng Wang;Wenjun Zhang;Jixun Zhan Dr.
ChemBioChem 2009 Volume 10( Issue 9) pp:1544-1550
Publication Date(Web):
DOI:10.1002/cbic.200900122
Co-reporter:Jixun Zhan Dr.;Kangjian Qiao
ChemBioChem 2009 Volume 10( Issue 9) pp:1447-1452
Publication Date(Web):
DOI:10.1002/cbic.200900082
Co-reporter:Wenjun Zhang
Journal of Medicinal Chemistry 2008 Volume 51(Issue 9) pp:2629-2633
Publication Date(Web):April 5, 2008
DOI:10.1021/jm701269v
Co-reporter:Jixun Zhan Dr.;Kenji Watanabe
ChemBioChem 2008 Volume 9( Issue 11) pp:1710-1715
Publication Date(Web):
DOI:10.1002/cbic.200800178
Co-reporter:Wenjun Zhang;Yanran Li
PNAS 2008 Volume 105 (Issue 52 ) pp:20683-20688
Publication Date(Web):2008-12-30
DOI:10.1073/pnas.0809084105
Bacterial aromatic polyketides are important therapeutic compounds including front line antibiotics and anticancer drugs. It is one of the last remaining major classes of natural products of which the biosynthesis has not been reconstituted in the genetically superior host Escherichia coli. Here, we demonstrate the engineered biosynthesis of bacterial aromatic polyketides in E. coli by using a dissected and reassembled fungal polyketide synthase (PKS). The minimal PKS of the megasynthase PKS4 from Gibberella fujikuroi was extracted by using two approaches. The first approach yielded a stand-alone Ketosynthase (KS)_malonyl-CoA:ACP transferase (MAT) didomain and an acyl-carrier protein (ACP) domain, whereas the second approach yielded a compact PKS (PKS_WJ) that consists of KS, MAT, and ACP on a single polypeptide. Both minimal PKSs produced nonfungal polyketides cyclized via different regioselectivity, whereas the fungal-specific C2-C7 cyclization mode was not observed. The kinetic properties of the two minimal PKSs were characterized to confirm both PKSs can synthesize polyketides with similar efficiency as the parent PKS4 megasynthase. Both minimal PKSs interacted effectively with exogenous polyketide cyclases as demonstrated by the synthesis of predominantly PK8 3 or NonaSEK4 6 in the presence of a C9-C14 or a C7-C12 cyclase, respectively. When PKS_WJ and downstream tailoring enzymes were expressed in E. coli, the expected nonaketide anthraquinone SEK26 was recovered in good titer. High-cell density fermentation was performed to demonstrate the scale-up potential of the in vivo platform for the biosynthesis of bacterial polyketides. Using engineered fungal PKSs can therefore be a general approach toward the heterologous biosynthesis of bacterial aromatic polyketides in E. coli.
Co-reporter:Hui Zhou;Jixun Zhan;Kenji Watanabe;Xinkai Xie
PNAS 2008 Volume 105 (Issue 17 ) pp:6249-6254
Publication Date(Web):2008-04-29
DOI:10.1073/pnas.0800657105
Resorcylic acid lactones represent a unique class of fungal polyketides and display a wide range of biological activities, such as nanomolar inhibitors of Hsp90 and MAP kinase. The biosynthesis of these compounds is proposed to involve two fungal polyketide synthases (PKS) that function collaboratively to yield a 14-membered macrolactone with a resorcylate core. We report here the reconstitution of Gibberella zeae PKS13, which is the nonreducing PKS associated with zearalenone biosynthesis. Using a small molecule mimic of the natural hexaketide starter unit, we reconstituted the entire repertoire of PKS13 activities in vitro, including starter-unit selection, iterative condensation, regioselective C2–C7 cyclization, and macrolactone formation. PKS13 synthesized both natural 14-membered and previously uncharacterized 16-membered resorcylic acid lactones, indicating relaxed control in both iterative elongation and macrocyclization. PKS13 exhibited broad starter-unit specificities toward fatty acyl-CoAs ranging in sizes between C6 and C16 and displayed the highest activity toward decanoyl-CoA. PKS13 was shown to be active in Escherichia coli and synthesized numerous alkyl pyrones and alkyl resorcylic esters without exogenously supplied precursors. We demonstrated that PKS13 can interact with E. coli fatty acid biosynthetic machinery and can be primed with fatty-acyl ACPp at low-micromolar concentrations. PKS13 synthesized new polyketides in E. coli when the culture was supplemented with synthetic precursors, showcasing its utility in precursor-directed biosynthesis. PKS13 is therefore a highly versatile polyketide macrolactone synthase that is useful in the engineered biosynthesis of polyketides, including resorcylic acid lactones that are not found in nature.
Co-reporter:Xinkai Xie, Kenji Watanabe, Wladyslaw A. Wojcicki, Clay C.C. Wang, Yi Tang
Chemistry & Biology 2006 Volume 13(Issue 11) pp:1161-1169
Publication Date(Web):November 2006
DOI:10.1016/j.chembiol.2006.09.008
The natural product lovastatin and its semisynthetic, more effective derivative, simvastatin, are important drugs for the treatment of hypercholesterolemia. Here, we report the biochemical characterization of a dedicated acyltransferase, LovD, encoded in the lovastatin biosynthetic pathway. We demonstrate that LovD has broad substrate specificity towards the acyl carrier, the acyl substrate, and the decalin acyl acceptor. LovD can efficiently catalyze the acyl transfer from coenzyme A thioesters or N-acetylcysteamine (SNAC) thioesters to monacolin J. When α-dimethylbutyryl-SNAC was used as the acyl donor, LovD was able to convert monacolin J and 6-hydroxyl-6-desmethylmonacolin J into simvastatin and huvastatin, respectively. Using the Escherichia coli LovD overexpression strain as a whole-cell biocatalyst, preparative amounts of simvastatin were synthesized in a single fermentation step. Our results demonstrate LovD is an attractive enzyme for engineered biosynthesis of pharmaceutically important cholesterol-lowering drugs.
Co-reporter:Jaclyn M Winter, Yi Tang
Current Opinion in Biotechnology (October 2012) Volume 23(Issue 5) pp:736-743
Publication Date(Web):1 October 2012
DOI:10.1016/j.copbio.2011.12.016
Small molecules produced in Nature possess exquisite chemical diversity and continue to be an inspiration for the development of new therapeutic agents. In their host organisms, natural products are assembled and modified using dedicated biosynthetic pathways. By rationally reprogramming and manipulating these pathways, unnatural metabolites containing enhanced structural features that were otherwise inaccessible can be obtained. Additionally, new chemical entities can be synthesized by developing the enzymes that carry out these complicated chemical reactions into biocatalysts. In this review, we will discuss a variety of combinatorial biosynthetic strategies, their technical challenges, and highlight some recent (since 2007) examples of rationally designed metabolites, as well as platforms that have been established for the production and modification of clinically important pharmaceutical compounds.Graphical abstractDownload high-res image (149KB)Download full-size imageHighlights► Reprogramming natural product assembly lines have yielded new compounds. ► Substrate specificity of tailoring enzymes can be exploited to afford derivatives. ► Expression and manipulation of pathways have been performed in surrogate hosts.
Co-reporter:Xinkai Xie, Wilson W. Wong, Yi Tang
Metabolic Engineering (July 2007) Volume 9(Issue 4) pp:379-386
Publication Date(Web):1 July 2007
DOI:10.1016/j.ymben.2007.05.006
Simvastatin is an important cholesterol lowering compound and is currently synthesized from the natural product lovastatin via multistep chemical synthesis. We have previously reported the use of an Escherichia coli strain BL21(DE3)/pAW31 as the host for whole-cell biocatalytic conversion of monacolin J acid to simvastatin acid. During fermentation and bioconversion, unknown E. coli enzyme(s) hydrolyzed the membrane permeable thioester substrate dimethylbutyryl-S-methyl mercaptopropionate (DMB-S-MMP) to the free acid, significantly decreased the efficiencies of the whole-cell bioconversion and the downstream purification steps. Using the Keio K-12 Singe-Gene Knockout collection, we identified BioH as the sole enzyme responsible for the observed substrate hydrolysis. Purification and reconstitution of E. coli BioH activity in vitro confirmed its function. BioH catalyzed the rapid hydrolysis of DMB-S-MMP with kcat and Km values of 260±45 s−1 and 229±26 μM, respectively. This is in agreement with previous reports that BioH can function as a carboxylesterase towards fatty acid esters. YT2, which is a ΔbioH mutant of BL21(DE3), did not hydrolyze DMB-S-MMP during prolonged fermentation and was used as an alternative host for whole-cell biocatalysis. The rate of simvastatin acid synthesis in YT2 was significantly faster than in BL21(DE3) and 99% conversion of 15 mM simvastatin acid in less than 12 h was achieved. Furthermore, the engineered host required significantly less DMB-S-MMP to be added to accomplish complete conversion. Finally, simvastatin acid synthesized using YT2 can be readily purified from fermentation broth and no additional steps to remove the hydrolyzed dimethylbutyryl-S-mercaptopropionic acid is required. Together, the proteomic and metabolic engineering approaches render the whole-cell biocatalytic process more robust and economically attractive.
Co-reporter:Blaine A Pfeifer, Yi Tang
Current Opinion in Biotechnology (December 2016) Volume 42() pp:vi-vii
Publication Date(Web):1 December 2016
DOI:10.1016/j.copbio.2016.11.002
Co-reporter:John M Billingsley, Anthony B DeNicola, Yi Tang
Current Opinion in Biotechnology (December 2016) Volume 42() pp:74-83
Publication Date(Web):1 December 2016
DOI:10.1016/j.copbio.2016.02.033
•New tools allow rapid refactoring and assembly of gene clusters for optimized expression in yeast.•DNA double-strand break technologies enable efficient integration of biosynthetic pathways.•Rewiring of yeast metabolic network generates versatile platform strains.•Novel strategies facilitated by synthetic biology increase natural product titers.The explosion of genomic sequence data and the significant advancements in synthetic biology have led to the development of new technologies for natural products discovery and production. Using powerful genetic tools, the yeast Saccharomyces cerevisiae has been engineered as a production host for natural product pathways from bacterial, fungal, and plant species. With an expanding library of characterized genetic parts, biosynthetic pathways can be refactored for optimized expression in yeast. New engineering strategies have enabled the increased production of valuable secondary metabolites by tuning metabolic pathways. Improvements in high-throughput screening methods have facilitated the rapid identification of variants with improved biosynthetic capabilities. In this review, we focus on the molecular tools and engineering strategies that have recently empowered heterologous natural product biosynthesis.Current Opinion in Biotechnology 2016, 42:74–83This review comes from a themed issue on Pharmaceutical biotechnologyEdited by Blaine Pfeifer and Yi TangFor a complete overview see the Issue and the EditorialAvailable online 16th March 2016http://dx.doi.org/10.1016/j.copbio.2016.02.0330958-1669/© 2016 Elsevier Ltd. All rights reserved.Download high-res image (86KB)Download full-size image
Co-reporter:Hui Zhou, Xinkai Xie, Yi Tang
Current Opinion in Biotechnology (December 2008) Volume 19(Issue 6) pp:590-596
Publication Date(Web):1 December 2008
DOI:10.1016/j.copbio.2008.10.012
Many biologically active natural products are produced by the host organisms using dedicated biosynthetic pathways. The programming rules of these pathways may be rationally manipulated through combinatorial biosynthesis to produce natural products that contain structural variations or enhanced pharmacological properties. Furthermore, these pathways contain enzymes that can be harvested as powerful biocatalysts for the synthesis of both new drugs and existing blockbuster therapeutics. This review will highlight recent advances in exploring natural product biosynthetic pathways for new compounds, novel enzymes and useful biocatalysts.
Co-reporter:Wei Xu ; Xiaolu Cai ; Michael E. Jung
Journal of the American Chemical Society () pp:
Publication Date(Web):September 9, 2010
DOI:10.1021/ja107084d
The widely found fungal iterative PKS-NRPS hybrid megasynthetases are highly programmed biosynthetic machines involved in the synthesis of 3-acyltetramic acids and related natural products. In vitro analysis of iterative PKS-NRPS has been hampered by the difficulties associated with obtaining pure and functional forms of these large enzymes (>400 kDa). We successfully expressed Aspergillus nidulans aspyridone synthetase (ApdA) from an engineered Saccharomyces cerevisiae strain. The complete functions of ApdA and its enoylreductase partner ApdC are reconstituted in vitro and in S. cerevisiae with the production of preaspyridone 7. The programming rules of both the PKS and NRPS modules were then examined in vitro. The key interaction between the PKS and the NRPS was dissected and reconstituted in trans by using stand-alone modules. Analogs of 7 were synthesized through heterologous combinations of PKS and NRPS modules from different sources. Our results represent one of the largest, multidomain enzyme reconstituted to date and offer new opportunities for engineered biosynthesis of fungal natural products.
Co-reporter:Kangjian Qiao ; Hui Zhou ; Wei Xu ; Wenjun Zhang ; Neil Garg
Organic Letters () pp:
Publication Date(Web):March 8, 2011
DOI:10.1021/ol200288w
A nonribosomal peptide synthetase-like enzyme (NRPS325) from Aspergillus terreus was reconstituted in vitro and was shown to synthesize thiopyrazines using an unprecedented mechanism. Substrate promiscuity of NRPS325 toward different amino acids and free thiols was explored to produce >60 different thiopyrazine compounds.
Co-reporter:Zhen Gu, Anuradha Biswas, Kye-Il Joo, Biliang Hu, Pin Wang and Yi Tang
Chemical Communications 2010 - vol. 46(Issue 35) pp:NaN6469-6469
Publication Date(Web):2010/07/26
DOI:10.1039/C0CC01439G
We describe a FRET-based protease detection strategy, using a single-fluorescent-protein nanogel as donor and a dark quencher as acceptor linked by a photolabile caged-peptide. This design enables probing of protease activity in a UV-responsive fashion.
2,5-bis(4-hydroxybenzyl)pyrazine
Fosamprenavir