1
|
Portugal J. Mithramycin and its analogs: Molecular features and antitumor action. Pharmacol Ther 2024; 260:108672. [PMID: 38838821 DOI: 10.1016/j.pharmthera.2024.108672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/09/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
The antitumor antibiotic mithramycin A (MTA) binds to G/C-rich DNA sequences in the presence of dications. MTA inhibits transcription regulated by the Sp1 transcription factor, often enhanced during tumor development. It shows antitumor activity, but its clinical use was discontinued due to toxic side effects. However, recent observations have led to its use being reconsidered. The MTA biosynthetic pathways have been modified to produce mithramycin analogs (mithralogs) that encompass lower toxicity and improved pharmacological activity. Some mithralogs reduce gene expression in human ovarian and prostate tumors, among other types of cancer. They down-regulate gene expression in various cellular processes, including Sp1-responsive genes that control tumor development. Moreover, MTA and several mithralogs, such as EC-8042 (DIG-MSK) and EC-8105, effectively treat Ewing sarcoma by inhibiting transcription controlled by the oncogenic EWS-FLI1 transcription factor.
Collapse
Affiliation(s)
- José Portugal
- Instituto de Diagnóstico Ambiental y Estudios del Agua, CSIC, E-08034 Barcelona, Spain.
| |
Collapse
|
2
|
Xu Z, Lee MC, Sheehan K, Fujii K, Rabl K, Rader G, Varney S, Sharma M, Eilers H, Kober K, Miaskowski C, Levine JD, Schumacher MA. Chemotherapy for pain: reversing inflammatory and neuropathic pain with the anticancer agent mithramycin A. Pain 2024; 165:54-74. [PMID: 37366593 PMCID: PMC10723648 DOI: 10.1097/j.pain.0000000000002972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/08/2023] [Accepted: 04/25/2023] [Indexed: 06/28/2023]
Abstract
ABSTRACT The persistence of inflammatory and neuropathic pain is poorly understood. We investigated a novel therapeutic paradigm by targeting gene networks that sustain or reverse persistent pain states. Our prior observations found that Sp1-like transcription factors drive the expression of TRPV1, a pain receptor, that is blocked in vitro by mithramycin A (MTM), an inhibitor of Sp1-like factors. Here, we investigate the ability of MTM to reverse in vivo models of inflammatory and chemotherapy-induced peripheral neuropathy (CIPN) pain and explore MTM's underlying mechanisms. Mithramycin reversed inflammatory heat hyperalgesia induced by complete Freund adjuvant and cisplatin-induced heat and mechanical hypersensitivity. In addition, MTM reversed both short-term and long-term (1 month) oxaliplatin-induced mechanical and cold hypersensitivity, without the rescue of intraepidermal nerve fiber loss. Mithramycin reversed oxaliplatin-induced cold hypersensitivity and oxaliplatin-induced TRPM8 overexpression in dorsal root ganglion (DRG). Evidence across multiple transcriptomic profiling approaches suggest that MTM reverses inflammatory and neuropathic pain through broad transcriptional and alternative splicing regulatory actions. Mithramycin-dependent changes in gene expression following oxaliplatin treatment were largely opposite to and rarely overlapped with changes in gene expression induced by oxaliplatin alone. Notably, RNAseq analysis revealed MTM rescue of oxaliplatin-induced dysregulation of mitochondrial electron transport chain genes that correlated with in vivo reversal of excess reactive oxygen species in DRG neurons. This finding suggests that the mechanism(s) driving persistent pain states such as CIPN are not fixed but are sustained by ongoing modifiable transcription-dependent processes.
Collapse
Affiliation(s)
- Zheyun Xu
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Man-Cheung Lee
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Kayla Sheehan
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Keisuke Fujii
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesiology, Wakayama Medical University, Wakayama, Japan
| | - Katalin Rabl
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Gabriella Rader
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Scarlett Varney
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Manohar Sharma
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Helge Eilers
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| | - Kord Kober
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, CA, United States
| | - Christine Miaskowski
- Department of Physiological Nursing, School of Nursing, University of California, San Francisco, CA, United States
| | - Jon D. Levine
- Division of Neuroscience, Departments of Medicine and Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Mark A. Schumacher
- Department of Anesthesia and Perioperative Care and the UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
3
|
Ye S, Molloy B, Pérez-Victoria I, Montero I, Braña AF, Olano C, Arca S, Martín J, Reyes F, Salas JA, Méndez C. Uncovering the Cryptic Gene Cluster ahb for 3-amino-4-hydroxybenzoate Derived Ahbamycins, by Searching SARP Regulator Encoding Genes in the Streptomyces argillaceus Genome. Int J Mol Sci 2023; 24:ijms24098197. [PMID: 37175904 PMCID: PMC10179220 DOI: 10.3390/ijms24098197] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/29/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Genome mining using standard bioinformatics tools has allowed for the uncovering of hidden biosynthesis gene clusters for specialized metabolites in Streptomyces genomes. In this work, we have used an alternative approach consisting in seeking "Streptomyces Antibiotic Regulatory Proteins" (SARP) encoding genes and analyzing their surrounding DNA region to unearth cryptic gene clusters that cannot be identified using standard bioinformatics tools. This strategy has allowed the unveiling of the new ahb cluster in Streptomyces argillaceus, which had not been retrieved before using antiSMASH. The ahb cluster is highly preserved in other Streptomyces strains, which suggests a role for their encoding compounds in specific environmental conditions. By combining overexpression of three regulatory genes and generation of different mutants, we were able to activate the ahb cluster, and to identify and chemically characterize the encoded compounds that we have named ahbamycins (AHBs). These constitute a new family of metabolites derived from 3-amino-4-hydroxybenzoate (3,4-AHBA) known for having antibiotic and antitumor activity. Additionally, by overexpressing three genes of the cluster (ahbH, ahbI, and ahbL2) for the synthesis and activation of 3,4-AHBA, a new hybrid compound, AHB18, was identified which had been produced from a metabolic crosstalk between the AHB and the argimycin P pathways. The identification of this new BGC opens the possibility to generate new compounds by combinatorial biosynthesis.
Collapse
Affiliation(s)
- Suhui Ye
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| | - Brian Molloy
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Ignacio Pérez-Victoria
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Armilla, 18016 Granada, Spain
| | - Ignacio Montero
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alfredo F Braña
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos Olano
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| | - Sonia Arca
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Jesús Martín
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Armilla, 18016 Granada, Spain
| | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Armilla, 18016 Granada, Spain
| | - José A Salas
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| | - Carmen Méndez
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
4
|
Jdanova S, Taylor MS. Mechanistic Study of the Copper(II)-Mediated Site-Selective O-Arylation of Glycosides with Arylboronic Acids. J Org Chem 2023; 88:3487-3498. [PMID: 36888595 DOI: 10.1021/acs.joc.2c02693] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Glycosides having multiple free OH groups have been shown to undergo site-selective O-arylations in the presence of arylboronic acids and copper(II) acetate. Herein, a mechanistic analysis of these Chan-Evans-Lam-type couplings is presented based on reaction kinetics, mass spectrometric analysis of reaction mixtures, and substituent effect studies. The results establish that the formation of a substrate-derived boronic ester accelerates the rate-determining transmetalation step. Intramolecular transfer of the aryl group from the boronic ester is ruled out in favor of a pathway in which the key pre-transmetalation assembly is generated from a boronic ester, a copper complex, and a second equivalent of arylboronic acid.
Collapse
Affiliation(s)
- Sofia Jdanova
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6 Canada
| | - Mark S Taylor
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6 Canada
| |
Collapse
|
5
|
WANG H, WANG L, FAN K, PAN G. Tetracycline natural products: discovery, biosynthesis and engineering. Chin J Nat Med 2022; 20:773-794. [DOI: 10.1016/s1875-5364(22)60224-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Indexed: 11/03/2022]
|
6
|
Differential Impact of Random GC Tetrad Binding and Chromatin Events on Transcriptional Inhibition by Olivomycin A. Int J Mol Sci 2022; 23:ijms23168871. [PMID: 36012127 PMCID: PMC9408465 DOI: 10.3390/ijms23168871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022] Open
Abstract
Olivomycin A (OA), an antibiotic of the aureolic acid family, interferes with gene transcription upon forming complexes with GC-rich regions in the DNA minor groove. We demonstrate that the mechanism of transcriptional deregulation is not limited to OA interaction with GC-containing binding sites for transcription factors. Using electrophoretic mobility shift assays and DNAse I footprinting of cytomegalovirus (CMV) promoter fragments carrying OA-preferred GC tetrads (CMVwt), we showed OA binding specifically to GC islands. Replacement of G for A in these tetrads (CMVmut) abrogated OA binding. Furthermore, OA decreased RNA polymerase II (RNAPII) binding to the CMVwt promoter and inhibited the reporter gene expression. In line with the absence of OA binding sites in CMVmut DNA, the expression driven from this promoter was weakly sensitive to OA. In the endogenous genes OA decreased RNAPII on promoters and coding regions. In certain cases this phenomenon was concomitant with the increased histone 3 abundance. However, the sensitivity to OA did not correlate with GC patterns around transcription start sites, suggesting that certain GC stretches play unequal roles in OA-induced transcriptional perturbations. Thus, OA affects transcription via complex mechanisms in which GC tetranucleotide binding causes RNAPII/chromatin alterations differentially manifested in individual gene contexts.
Collapse
|
7
|
Pal R, Seleem MN. Discovery of a novel natural product inhibitor of Clostridioides difficile with potent activity in vitro and in vivo. PLoS One 2022; 17:e0267859. [PMID: 35939437 PMCID: PMC9359557 DOI: 10.1371/journal.pone.0267859] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/17/2022] [Indexed: 11/22/2022] Open
Abstract
Clostridioides difficile infection is a global health threat and remains the primary cause of hospital-acquired infections worldwide. The burgeoning incidence and severity of infections coupled with high rates of recurrence have created an urgent need for novel therapeutics. Here, we report a novel natural product scaffold as a potential anticlostridial lead with antivirulence properties and potent activity both in vitro and in vivo. A whole cell phenotypic screening of 1,000 purified natural products identified 6 compounds with potent activity against C. difficile (minimum inhibitory concentration (MIC) range from 0.03 to 2 μg/ml). All these 6 compounds were non-toxic to human colorectal cells. The natural product compounds also inhibited the production of key toxins, TcdA and TcdB, the key virulence determinants of C. difficile infection pathology. Additionally, the compounds exhibited rapid bactericidal activity and were superior to the standard-of-care antibiotic vancomycin, in reducing a high inoculum of C. difficile in vitro. Furthermore, a murine model of C. difficile infection revealed that compound NP-003875 conferred 100% protection to the infected mice from clinical manifestations of the disease. Collectively, the current study lays the foundation for further investigation of the natural product NP-003875 as a potential therapeutic choice for C. difficile infection.
Collapse
Affiliation(s)
- Rusha Pal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Mohamed N. Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
8
|
Becerril A, Pérez-Victoria I, Martín JM, Reyes F, Salas JA, Méndez C. Biosynthesis of Largimycins in Streptomyces argillaceus Involves Transient β-Alkylation and Cryptic Halogenation Steps Unprecedented in the Leinamycin Family. ACS Chem Biol 2022; 17:2320-2331. [PMID: 35830174 PMCID: PMC9396626 DOI: 10.1021/acschembio.2c00416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Largimycins A1 and A2 are key members of a recently identified
family of hybrid nonribosomal peptide polyketides belonging to the
scarcely represented group of antitumor leinamycins. They are encoded
by the gene cluster lrg of Streptomyces argillaceus. This cluster contains a halogenase gene and two sets of genes for
the biosynthesis and incorporation of β branches at C3 and C9.
Noticeably, largimycins A1 and A2 are nonhalogenated compounds and
only contain a β branch at C3. By generating mutants in those
genes and characterizing chemically their accumulated compounds, we
could confirm the existence of a chlorination step at C19, the introduction
of an acetyl-derived olefinic exomethylene group at C9, and a propionyl-derived
β branch at C3 in the biosynthesis pathway. Since the olefinic
exomethylene group and the chlorine atom are absent in the final products,
those biosynthetic steps can be considered cryptic in the overall
pathway but essential to generating keto and epoxide functionalities
at C9 and C18/C19, respectively. We propose that chlorination at C19
is utilized as an activation strategy that creates the precursor halohydrin
to finally yield the epoxy functionality at C18/C19. This represents
a novel strategy to create such functionalities and extends the small
number of natural product biosynthetic pathways that include a cryptic
chlorination step.
Collapse
Affiliation(s)
- Adriana Becerril
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain.,Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ignacio Pérez-Victoria
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Armilla, 18016 Granada, Spain
| | - Jesús M Martín
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Armilla, 18016 Granada, Spain
| | - Fernando Reyes
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Armilla, 18016 Granada, Spain
| | - Jose A Salas
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain.,Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| | - Carmen Méndez
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, 33006 Oviedo, Spain.,Instituto de Investigación Sanitaria de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
9
|
Carder HM, Wang Y, Wendlandt AE. Selective Axial-to-Equatorial Epimerization of Carbohydrates. J Am Chem Soc 2022; 144:11870-11877. [PMID: 35731921 DOI: 10.1021/jacs.2c04743] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Radical-mediated transformations have emerged as powerful methods for the synthesis of rare and unnatural branched, deoxygenated, and isomeric sugars. Here, we describe a radical-mediated axial-to-equatorial alcohol epimerization method to transform abundant glycans into rare isomers. The method delivers highly predictable and selective reaction outcomes that are complementary to other sugar isomerization methods. The synthetic utility of isomer interconversion is showcased through expedient glycan synthesis, including one-step glycodiversification. Mechanistic studies reveal that both site- and diastereoselectivities are achieved by highly selective H atom abstraction of equatorially disposed α-hydroxy C-H bonds.
Collapse
Affiliation(s)
- Hayden M Carder
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yong Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alison E Wendlandt
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
10
|
Li S, Mao Y, Zhang L, Wang M, Meng J, Liu X, Bai Y, Guo Y. Recent advances in microbial ε-poly-L-lysine fermentation and its diverse applications. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2022; 15:65. [PMID: 35710433 PMCID: PMC9205021 DOI: 10.1186/s13068-022-02166-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
The naturally occurring homo-polyamide biopolymer, ε-poly-L-lysine (ε-PL) consists of 25-35 L-lysine residues with amide linkages between α-carboxyl groups and ε-amino groups. ɛ-PL exhibits several useful properties because of its unusual structure, such as biodegradability, water solubility, no human toxicity, and broad-spectrum antibacterial activities; it is widely applied in the fields of food, medicine, clinical chemistry and electronics. However, current industrial production of ε-PL is only performed in a few countries. Based on an analysis of the physiological characteristics of ε-PL fermentation, current advances that enhance ε-PL fermentation, from strain improvement to product isolation are systematically reviewed, focusing on: (1) elucidating the metabolic pathway and regulatory mechanism of ε-PL synthesis; (2) enhancing biosynthetic performance through mutagenesis, fermentation optimization and metabolic engineering; and (3) understanding and improving the biological activity and functional properties of ε-PL. Finally, perspectives on engineering and exploiting ε-PL as a source material for the production of various advanced materials are also discussed, providing scientific guidelines for researchers to further improve the ε-PL fermentation process.
Collapse
Affiliation(s)
- Shubo Li
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Yunren Mao
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Lifei Zhang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Miao Wang
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Jinhao Meng
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Xiaoling Liu
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Yunxia Bai
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| | - Yuan Guo
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, 530004, China.
| |
Collapse
|
11
|
Chen J, Li Y, Wang S, Zhang H, Du Y, Wu Q, Wang H. Targeting Clostridioides difficile: New uses for old drugs. Drug Discov Today 2022; 27:1862-1873. [PMID: 35390545 DOI: 10.1016/j.drudis.2022.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/03/2021] [Accepted: 03/30/2022] [Indexed: 12/30/2022]
Abstract
Clostridioides difficile bacteria can cause life-threatening diarrhea and colitis owing to limited treatment options and unacceptably high recurrence rates among infected patients. This necessitates the development of alternative routes for C. difficile treatment. Drug repurposing with new indications represents a proven shortcut. Here, we present a refined focus on 16 FDA-approved drugs that would be suitable for further development as potential anti-C. difficile drugs. Of these drugs, clinical trials have been conducted on five currently used drugs; however, ursodeoxycholic acid is the only drug to enter Phase IV clinical trials to date. Thus, drug repurposing promotes the study of mechanistic and therapeutic strategies, providing new options for the development of next-generation anti-C. difficile agents.
Collapse
Affiliation(s)
- Jianwei Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China; Macau University of Science and Technology, State Key Laboratory of Quality Research in Chinese Medicines, Macao
| | - Yasheng Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University & Anhui Center for Surveillance of Bacterial Resistance, Hefei, China
| | - Siqi Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Hongfang Zhang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Yujie Du
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Qiang Wu
- Macau University of Science and Technology, State Key Laboratory of Quality Research in Chinese Medicines, Macao.
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
12
|
Suh CE, Carder HM, Wendlandt AE. Selective Transformations of Carbohydrates Inspired by Radical-Based Enzymatic Mechanisms. ACS Chem Biol 2021; 16:1814-1828. [PMID: 33988380 DOI: 10.1021/acschembio.1c00190] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Enzymes are a longstanding source of inspiration for synthetic reaction development. However, enzymatic reactivity and selectivity are frequently untenable in a synthetic context, as the principles that govern control in an enzymatic setting often do not translate to small molecule catalysis. Recent synthetic methods have revealed the viability of using small molecule catalysts to promote highly selective radical-mediated transformations of minimally protected sugar substrates. These transformations share conceptual similarities with radical SAM enzymes found in microbial carbohydrate biosynthesis and present opportunities for synthetic chemists to access microbial and unnatural carbohydrate building blocks without the need for protecting groups or lengthy synthetic sequences. Here, we highlight strategies through which radical reaction pathways can enable the site-, regio-, and diastereoselective transformation of minimally protected carbohydrates in both synthetic and enzymatic systems.
Collapse
Affiliation(s)
- Carolyn E. Suh
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Hayden M. Carder
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alison E. Wendlandt
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
13
|
Carder HM, Suh CE, Wendlandt AE. A Unified Strategy to Access 2- and 4-Deoxygenated Sugars Enabled by Manganese-Promoted 1,2-Radical Migration. J Am Chem Soc 2021; 143:13798-13805. [PMID: 34406756 DOI: 10.1021/jacs.1c05993] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The selective manipulation of carbohydrate scaffolds is challenging due to the presence of multiple, nearly chemically indistinguishable O-H and C-H bonds. As a result, protecting-group-based synthetic strategies are typically necessary for carbohydrate modification. Here we report a concise semisynthetic strategy to access diverse 2- and 4-deoxygenated carbohydrates without relying on the exhaustive use of protecting groups to achieve site-selective reaction outcomes. Our approach leverages a Mn2+-promoted redox isomerization step, which proceeds via sugar radical intermediates accessed by neutral hydrogen atom abstraction under visible light-mediated photoredox conditions. The resulting deoxyketopyranosides feature chemically distinguishable functional groups and are readily transformed into diverse carbohydrate structures. To showcase the versatility of this method, we report expedient syntheses of the rare sugars l-ristosamine, l-olivose, l-mycarose, and l-digitoxose from commercial l-rhamnose. The findings presented here validate the potential for radical intermediates to facilitate the selective transformation of carbohydrates and showcase the step and efficiency advantages attendant to synthetic strategies that minimize a reliance upon protecting groups.
Collapse
Affiliation(s)
- Hayden M Carder
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Carolyn E Suh
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alison E Wendlandt
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
14
|
Isagulieva AK, Soshnikova NV, Shtil AA. Inhibition of the c-Myc Oncogene by the Aureolic Acid Group Antibiotics. DOKL BIOCHEM BIOPHYS 2021; 500:308-311. [PMID: 34697733 DOI: 10.1134/s1607672921050094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 11/23/2022]
Abstract
GC-rich stretches in the DNA minor groove are the established intracellular targets for the aureolic acid group of antibiotics such as olivomycin A and its semisynthetic analogue olivamide. We demonstrated here that both antibiotics at nanomolar concentrations inhibited transcription of the c-Myc oncogene in cultured human tumor cells. The mechanism of transcriptional inhibition did not require the full-length binding site for Sp1, a GC-dependent transcriptional factor. GC quartets with the nucleotide sequences optimal for drug binding are sufficient for c-Myc transcriptional block by the aureolic acid derivatives.
Collapse
Affiliation(s)
- A K Isagulieva
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.
- Gause Institute of New Antibiotics, Moscow, Russia.
| | - N V Soshnikova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A A Shtil
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
15
|
Meng S, Li X, Zhu J. Recent advances in direct synthesis of 2-deoxy glycosides and thioglycosides. Tetrahedron 2021. [DOI: 10.1016/j.tet.2021.132140] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Stevenson LJ, Bracegirdle J, Liu L, Sharrock AV, Ackerley DF, Keyzers RA, Owen JG. Metathramycin, a new bioactive aureolic acid discovered by heterologous expression of a metagenome derived biosynthetic pathway. RSC Chem Biol 2021; 2:556-567. [PMID: 34458799 PMCID: PMC8341913 DOI: 10.1039/d0cb00228c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Bacterial natural products have been a rich source of bioactive compounds for drug development, and advances in DNA sequencing, informatics and molecular biology have opened new avenues for their discovery. Here, we describe the isolation of an aureolic acid biosynthetic gene cluster from a metagenome library derived from a New Zealand soil sample. Heterologous expression of this pathway in Streptomyces albus resulted in the production and isolation of two new aureolic acid compounds, one of which (metathramycin, 6) possesses potent bioactivity against a human colon carcinoma cell line (HCT-116, IC50 = 14.6 nM). As metathramycin was a minor constituent of the fermentation extract, its discovery relied on a combination of approaches including bioactivity guided fractionation, MS/MS characterisation and pathway engineering. This study not only demonstrates the presence of previously uncharacterised aureolic acids in the environment, but also the value of an integrated natural product discovery approach which may be generally applicable to low abundance bioactive metabolites.
Collapse
Affiliation(s)
- Luke J Stevenson
- School of Biological Sciences, Victoria University of Wellington Wellington New Zealand .,Maurice Wilkins Centre for Molecular Biodiscovery New Zealand.,Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington Wellington New Zealand
| | - Joe Bracegirdle
- Maurice Wilkins Centre for Molecular Biodiscovery New Zealand.,Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington Wellington New Zealand.,School of Chemical and Physical Sciences, Victoria University of Wellington Wellington New Zealand
| | - Liwei Liu
- School of Biological Sciences, Victoria University of Wellington Wellington New Zealand
| | - Abigail V Sharrock
- School of Biological Sciences, Victoria University of Wellington Wellington New Zealand .,Maurice Wilkins Centre for Molecular Biodiscovery New Zealand.,Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington Wellington New Zealand
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington Wellington New Zealand .,Maurice Wilkins Centre for Molecular Biodiscovery New Zealand.,Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington Wellington New Zealand
| | - Robert A Keyzers
- Maurice Wilkins Centre for Molecular Biodiscovery New Zealand.,Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington Wellington New Zealand.,School of Chemical and Physical Sciences, Victoria University of Wellington Wellington New Zealand
| | - Jeremy G Owen
- School of Biological Sciences, Victoria University of Wellington Wellington New Zealand .,Maurice Wilkins Centre for Molecular Biodiscovery New Zealand.,Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington Wellington New Zealand
| |
Collapse
|
17
|
Zhu HJ, Zhang B, Wang L, Wang W, Liu SH, Igarashi Y, Bashiri G, Tan RX, Ge HM. Redox Modifications in the Biosynthesis of Alchivemycin A Enable the Formation of Its Key Pharmacophore. J Am Chem Soc 2021; 143:4751-4757. [PMID: 33736434 DOI: 10.1021/jacs.1c00516] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Redox enzymes play a critical role in transforming nascent scaffolds into structurally complex and biologically active natural products. Alchivemycin A (AVM, 1) is a highly oxidized polycyclic compound with potent antimicrobial activity and features a rare 2H-tetrahydro-4,6-dioxo-1,2-oxazine (TDO) ring system. The scaffold of AVM has previously been shown to be biosynthesized by a hybrid polyketide synthase-nonribosomal peptide synthetase (PKS-NRPS) pathway. In this study, we present a postassembly secondary metabolic network involving six redox enzymes that leads to AVM formation. We characterize this complex redox network using in vivo gene deletions, in vitro biochemical assays, and one-pot enzymatic total synthesis. Importantly, we show that an FAD-dependent monooxygenase catalyzes oxygen insertion into an amide bond to form the key TDO ring in AVM, an unprecedented function of flavoenzymes. We also show that the TDO ring is essential to the antimicrobial activity of AVM, likely through targeting the β-subunit of RNA polymerase. As further evidence, we show that AvmK, a β-subunit of RNA synthase, can confer self-resistance to AVM via target modification. Our findings expand the repertoire of functions of flavoenzymes and provide insight into antimicrobial and biocatalyst development based on AVM.
Collapse
Affiliation(s)
- Hong Jie Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Bo Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Lan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Wen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Shuang He Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University, Toyama 939-0398, Japan
| | - Ghader Bashiri
- Laboratory of Molecular and Microbial Biochemistry, School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Ren Xiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Hui Ming Ge
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| |
Collapse
|
18
|
Estupiñán Ó, Rendueles C, Suárez P, Rey V, Murillo D, Morís F, Gutiérrez G, Blanco-López MDC, Matos M, Rodríguez R. Nano-Encapsulation of Mithramycin in Transfersomes and Polymeric Micelles for the Treatment of Sarcomas. J Clin Med 2021; 10:jcm10071358. [PMID: 33806182 PMCID: PMC8037461 DOI: 10.3390/jcm10071358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are aggressive tumors which often show a poor response to current treatments. As a promising therapeutic alternative, we focused on mithramycin (MTM), a natural antibiotic with a promising anti-tumor activity but also a relevant systemic toxicity. Therefore, the encapsulation of MTM in nano-delivery systems may represent a way to increase its therapeutic window. Here, we designed novel transfersomes and PLGA polymeric micelles by combining different membrane components (phosphatidylcholine, Span 60, Tween 20 and cholesterol) to optimize the nanoparticle size, polydispersity index (PDI) and encapsulation efficiency (EE). Using both thin film hydration and the ethanol injection methods we obtained MTM-loaded transferosomes displaying an optimal hydrodynamic diameter of 100–130 nm and EE values higher than 50%. Additionally, we used the emulsion/solvent evaporation method to synthesize polymeric micelles with a mean size of 228 nm and a narrow PDI, capable of encapsulating MTM with EE values up to 87%. These MTM nano-delivery systems mimicked the potent anti-tumor activity of free MTM, both in adherent and cancer stem cell-enriched tumorsphere cultures of myxoid liposarcoma and chondrosarcoma models. Similarly to free MTM, nanocarrier-delivered MTM efficiently inhibits the signaling mediated by the pro-oncogenic factor SP1. In summary, we provide new formulations for the efficient encapsulation of MTM which may constitute a safer delivering alternative to be explored in future clinical uses.
Collapse
Affiliation(s)
- Óscar Estupiñán
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
| | - Claudia Rendueles
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
| | - Paula Suárez
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
| | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Dzohara Murillo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
| | | | - Gemma Gutiérrez
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
- Asturias University Institute of Biotechnology, University of Oviedo, 33006 Oviedo, Spain;
| | - María del Carmen Blanco-López
- Asturias University Institute of Biotechnology, University of Oviedo, 33006 Oviedo, Spain;
- Department of Physical and Analytical Chemistry, University of Oviedo, 33006 Oviedo, Spain
| | - María Matos
- Department of Chemical and Environmental Engineering, University of Oviedo, 33006 Oviedo, Spain; (C.R.); (P.S.); (G.G.)
- Asturias University Institute of Biotechnology, University of Oviedo, 33006 Oviedo, Spain;
- Correspondence: (M.M.); (R.R.)
| | - René Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)—Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Ó.E.); (V.R.); (D.M.)
- Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
- CIBER en Oncología (CIBERONC), 28029 Madrid, Spain
- Correspondence: (M.M.); (R.R.)
| |
Collapse
|
19
|
Dimakos V, Taylor MS. Recent advances in the direct O-arylation of carbohydrates. Org Biomol Chem 2021; 19:514-524. [PMID: 33331387 DOI: 10.1039/d0ob02009e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Methods for the O-arylation of hydroxyl and hemiacetal groups in carbohydrates via C(sp2)-O bond formation are discussed. Such methods provide an alternative disconnection to the traditional approach of nucleophilic substitution between a sugar-derived electrophile and a phenol or phenoxide nucleophile. They have led to new opportunities for stereoselectivity, site-selectivity and chemoselectivity in the preparation of O-aryl glycosides and carbohydrate-derived aryl ethers, compounds that are useful for a broad range of applications in medicinal chemistry, glycobiology and organic synthesis.
Collapse
Affiliation(s)
- Victoria Dimakos
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON M5S 3H6, Canada.
| | - Mark S Taylor
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON M5S 3H6, Canada.
| |
Collapse
|
20
|
Zheng J, Hoffman KL, Chen JS, Shivappa N, Sood A, Browman GJ, Dirba DD, Hanash S, Wei P, Hebert JR, Petrosino JF, Schembre SM, Daniel CR. Dietary inflammatory potential in relation to the gut microbiome: results from a cross-sectional study. Br J Nutr 2020; 124:931-942. [PMID: 32475373 PMCID: PMC7554089 DOI: 10.1017/s0007114520001853] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diet has direct and indirect effects on health through inflammation and the gut microbiome. We investigated total dietary inflammatory potential via the literature-derived index (Dietary Inflammatory Index (DII®)) with gut microbiota diversity, composition and function. In cancer-free patient volunteers initially approached at colonoscopy and healthy volunteers recruited from the medical centre community, we assessed 16S ribosomal DNA in all subjects who provided dietary assessments and stool samples (n 101) and the gut metagenome in a subset of patients with residual fasting blood samples (n 34). Associations of energy-adjusted DII scores with microbial diversity and composition were examined using linear regression, permutational multivariate ANOVA and linear discriminant analysis. Spearman correlation was used to evaluate associations of species and pathways with DII and circulating inflammatory markers. Across DII levels, α- and β-diversity did not significantly differ; however, Ruminococcus torques, Eubacterium nodatum, Acidaminococcus intestini and Clostridium leptum were more abundant in the most pro-inflammatory diet group, while Akkermansia muciniphila was enriched in the most anti-inflammatory diet group. With adjustment for age and BMI, R. torques, E. nodatum and A. intestini remained significantly associated with a more pro-inflammatory diet. In the metagenomic and fasting blood subset, A. intestini was correlated with circulating plasminogen activator inhibitor-1, a pro-inflammatory marker (rho = 0·40), but no associations remained significant upon correction for multiple testing. An index reflecting overall inflammatory potential of the diet was associated with specific microbes, but not overall diversity of the gut microbiome in our study. Findings from this preliminary study warrant further research in larger samples and prospective cohorts.
Collapse
Affiliation(s)
- Jiali Zheng
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Kristi L Hoffman
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030, USA
| | - Jiun-Sheng Chen
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Quantitative Sciences Program, The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX77030, USA
| | - Nitin Shivappa
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC29208, USA
| | - Akhil Sood
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Internal Medicine, University of Texas Medical Branch, Galveston, TX77555, USA
| | - Gladys J Browman
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Danika D Dirba
- Department of Behavioral Science, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Peng Wei
- Quantitative Sciences Program, The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX77030, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - James R Hebert
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC29208, USA
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030, USA
| | - Susan M Schembre
- Department of Behavioral Science, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Department of Family and Community Medicine, University of Arizona, Tucson, AZ85721, USA
| | - Carrie R Daniel
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
- Quantitative Sciences Program, The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX77030, USA
| |
Collapse
|
21
|
Antibacterial Activity of Chromomycins from a Marine-Derived Streptomyces microflavus. Mar Drugs 2020; 18:md18100522. [PMID: 33096696 PMCID: PMC7588889 DOI: 10.3390/md18100522] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
A marine-derived actinomycete (Streptomyces sp. MBTI36) exhibiting antibacterial activities was investigated in the present study. The strain was identified using genetic techniques. The 16S rDNA sequence of the isolate indicated that it was most closely related to Streptomyces microflavus. Furthermore, a new chromomycin A9 (1), along with chromomycin Ap (2), chromomycin A2 (3), and chromomycin A3 (4), were isolated from the ethyl acetate extract. Their structures were determined using extensive spectroscopic methods including 1D and 2D NMR, and HRMS, as well as comparisons with previously reported data. Compounds 1–4 showed potent antibacterial activities against Gram-positive bacteria including methicillin-resistant Staphylococcus aureus (MRSA). During a passage experiment, minimum inhibitory concentration (MIC) values for compounds 1–4 showed no more than a 4-fold increase from the starting MIC value, indicating that no resistance was detected over the 21 passages.
Collapse
|
22
|
Protection of all cleavable sites of DNA with the multiple CGCG or continuous CGG sites from the restriction enzyme, indicative of simultaneous binding of small ligands. Bioorg Med Chem 2020; 28:115730. [PMID: 33069073 DOI: 10.1016/j.bmc.2020.115730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 11/21/2022]
Abstract
The anthracenone ligands (1-12) with a keto-phenol and a hydroxamic acid unit were synthesized and evaluated by a restriction enzyme inhibition assay. DNA substrates composed of multiple CGCG or CGG sites are fully hydrolyzed by a restriction enzyme that is selective for each sequence. Under such conditions, the full-length DNA substrate remains only when the ligand binds to all binding sites and protects it from hydrolysis by the restriction enzymes. In the assay using AccII and the 50-mer DNA substrates containing a different number of CGCG sites at different non-binding AT base pair intervals, the more the CGCG sites, the more the full-length DNA increased. Namely, simultaneous binding of the ligand (5) to the CGCG sites increased in the order of (CGCG)5>(CGCG)2>(CGCG)1. Furthermore, the length of the spacer of the hydroxamic acid to the anthracenone skeleton played an important role in the preference for the number of the d(A/T) base pairs between the CGCG sites. The long spacer-ligand (5) showed a preference to the CGCG sites with five AT pairs, and the short spacer-ligand (10) to that with two AT pairs. The ligand (12) with the shortest spacer showed a preference in simultaneous binding to the 54-mer DNA composed of 16 continuous CGG sites in the assay using the restriction enzyme Fnu4HI that hydrolyzes the d(GCGGC)/d(CGCCG) site. Application of these ligands to biological systems including the repeat DNA sequence should be of significant interest.
Collapse
|
23
|
Discrimination between G/C Binding Sites by Olivomycin A Is Determined by Kinetics of the Drug-DNA Interaction. Int J Mol Sci 2020; 21:ijms21155299. [PMID: 32722584 PMCID: PMC7432603 DOI: 10.3390/ijms21155299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/25/2022] Open
Abstract
Olivomycin A (OA) exerts its cytotoxic potency due to binding to the minor groove of the G/C-rich DNA and interfering with replication and transcription. Screening of the complete set of tetranucleotide G/C sites by electrophoretic mobility gel shift assay (EMSA) revealed that the sites containing central GC or GG dinucleotides were able to bind OA, whereas the sites with the central CG dinucleotide were not. However, studies of equilibrium OA binding in solution by fluorescence, circular dichroism and isothermal titration calorimetry failed to confirm the sequence preference of OA, indicating instead a similar type of complex and comparable affinity of OA to all G/C binding sites. This discrepancy was resolved by kinetics analysis of the drug–DNA interaction: the dissociation rate significantly differed between SGCS, SGGS and SCGS sites (S stands for G or C), thereby explaining the disintegration of the complexes during EMSA. The functional relevance of the revealed differential kinetics of OA–DNA interaction was demonstrated in an in vitro transcription assay. These findings emphasize the crucial role of kinetics in the mechanism of OA action and provide an important approach to the screening of new drug candidates.
Collapse
|
24
|
Kormanec J, Novakova R, Csolleiova D, Feckova L, Rezuchova B, Sevcikova B, Homerova D. The antitumor antibiotic mithramycin: new advanced approaches in modification and production. Appl Microbiol Biotechnol 2020; 104:7701-7721. [DOI: 10.1007/s00253-020-10782-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
|
25
|
Zabala D, Song L, Dashti Y, Challis GL, Salas JA, Méndez C. Heterologous reconstitution of the biosynthesis pathway for 4-demethyl-premithramycinone, the aglycon of antitumor polyketide mithramycin. Microb Cell Fact 2020; 19:111. [PMID: 32448325 PMCID: PMC7247220 DOI: 10.1186/s12934-020-01368-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mithramycin is an anti-tumor compound of the aureolic acid family produced by Streptomyces argillaceus. Its biosynthesis gene cluster has been cloned and characterized, and several new analogs with improved pharmacological properties have been generated through combinatorial biosynthesis. To further study these compounds as potential new anticancer drugs requires their production yields to be improved significantly. The biosynthesis of mithramycin proceeds through the formation of the key intermediate 4-demethyl-premithramycinone. Extensive studies have characterized the biosynthesis pathway from this intermediate to mithramycin. However, the biosynthesis pathway for 4-demethyl-premithramycinone remains unclear. RESULTS Expression of cosmid cosAR7, containing a set of mithramycin biosynthesis genes, in Streptomyces albus resulted in the production of 4-demethyl-premithramycinone, delimiting genes required for its biosynthesis. Inactivation of mtmL, encoding an ATP-dependent acyl-CoA ligase, led to the accumulation of the tricyclic intermediate 2-hydroxy-nogalonic acid, proving its essential role in the formation of the fourth ring of 4-demethyl-premithramycinone. Expression of different sets of mithramycin biosynthesis genes as cassettes in S. albus and analysis of the resulting metabolites, allowed the reconstitution of the biosynthesis pathway for 4-demethyl-premithramycinone, assigning gene functions and establishing the order of biosynthetic steps. CONCLUSIONS We established the biosynthesis pathway for 4-demethyl-premithramycinone, and identified the minimal set of genes required for its assembly. We propose that the biosynthesis starts with the formation of a linear decaketide by the minimal polyketide synthase MtmPKS. Then, the cyclase/aromatase MtmQ catalyzes the cyclization of the first ring (C7-C12), followed by formation of the second and third rings (C5-C14; C3-C16) catalyzed by the cyclase MtmY. Formation of the fourth ring (C1-C18) requires MtmL and MtmX. Finally, further oxygenation and reduction is catalyzed by MtmOII and MtmTI/MtmTII respectively, to generate the final stable tetracyclic intermediate 4-demethyl-premithramycinone. Understanding the biosynthesis of this compound affords enhanced possibilities to generate new mithramycin analogs and improve their production titers for bioactivity investigation.
Collapse
Affiliation(s)
- Daniel Zabala
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), University of Oviedo, Oviedo, Spain
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Lijiang Song
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Yousef Dashti
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
| | - Gregory L Challis
- Department of Chemistry, University of Warwick, Coventry, CV4 7AL, UK
- Warwick Integrative Synthetic Biology Centre, University of Warwick, Coventry, CV4 7AL, UK
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - José A Salas
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), University of Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria de Asturias (ISPA), Oviedo, Spain
| | - Carmen Méndez
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), University of Oviedo, Oviedo, Spain.
- Instituto de Investigación Sanitaria de Asturias (ISPA), Oviedo, Spain.
| |
Collapse
|
26
|
Mrudulakumari Vasudevan U, Lee EY. Flavonoids, terpenoids, and polyketide antibiotics: Role of glycosylation and biocatalytic tactics in engineering glycosylation. Biotechnol Adv 2020; 41:107550. [PMID: 32360984 DOI: 10.1016/j.biotechadv.2020.107550] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/19/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
Flavonoids, terpenoids, and polyketides are structurally diverse secondary metabolites used widely as pharmaceuticals and nutraceuticals. Most of these molecules exist in nature as glycosides, in which sugar residues act as a decisive factor in their architectural complexity and bioactivity. Engineering glycosylation through selective trimming or extension of the sugar residues in these molecules is a prerequisite to their commercial production as well to creating novel derivatives with specialized functions. Traditional chemical glycosylation methods are tedious and can offer only limited end-product diversity. New in vitro and in vivo biocatalytic tools have emerged as outstanding platforms for engineering glycosylation in these three classes of secondary metabolites to create a large repertoire of versatile glycoprofiles. As knowledge has increased about secondary metabolite-associated promiscuous glycosyltransferases and sugar biosynthetic machinery, along with phenomenal progress in combinatorial biosynthesis, reliable industrial production of unnatural secondary metabolites has gained momentum in recent years. This review highlights the significant role of sugar residues in naturally occurring flavonoids, terpenoids, and polyketide antibiotics. General biocatalytic tools used to alter the identity and pattern of sugar molecules are described, followed by a detailed illustration of diverse strategies used in the past decade to engineer glycosylation of these valuable metabolites, exemplified with commercialized products and patents. By addressing the challenges involved in current bio catalytic methods and considering the perspectives portrayed in this review, exceptional drugs, flavors, and aromas from these small molecules could come to dominate the natural-product industry.
Collapse
Affiliation(s)
| | - Eun Yeol Lee
- Department of Chemical Engineering, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
27
|
Pal R, Seleem MN. Screening of Natural Products and Approved Oncology Drug Libraries for Activity against Clostridioides difficile. Sci Rep 2020; 10:5966. [PMID: 32249833 PMCID: PMC7136261 DOI: 10.1038/s41598-020-63029-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/24/2020] [Indexed: 12/17/2022] Open
Abstract
Clostridioides difficile is the most common cause of healthcare-associated diarrhea. Infection of the gastrointestinal tract with this Gram-positive, obligate anaerobe can lead to potentially life-threatening conditions in the antibiotic-treated populace. New therapeutics are urgently needed to treat this infection and prevent its recurrence. Here, we screened two libraries from the National Cancer Institute, namely, the natural product set III library (117 compounds) and the approved oncology drugs set V library (114 compounds), against C. difficile. In the two libraries screened, 17 compounds from the natural product set III library and 7 compounds from the approved oncology drugs set V library were found to exhibit anticlostridial activity. The most potent FDA-approved drugs (mitomycin C and mithramycin A) and a promising natural product (aureomycin) were further screened against 20 clinical isolates of C. difficile. The anticancer drugs, mitomycin C (MIC50 = 0.25 μg/ml) and mithramycin A (MIC50 = 0.015 μg/ml), and the naturally derived tetracycline derivative, aureomycin (MIC50 = 0.06 μg/ml), exhibited potent activity against C. difficile strains. Mithramycin A and aureomycin were further found to inhibit toxin production by this pathogen. Given their efficacy, these compounds can provide a quick supplement to current treatment to address the unmet needs in treating C. difficile infection and preventing its recurrence.
Collapse
Affiliation(s)
- Rusha Pal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA. .,Purdue Institute of Inflammation, Immunology, and Infectious Disease, West Lafayette, IN, 47907, USA.
| |
Collapse
|
28
|
Main trends in the design of semi-synthetic antibiotics of a new generation. RUSSIAN CHEMICAL REVIEWS 2020. [PMCID: PMC7149660 DOI: 10.1070/rcr4892] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
This review summarizes main advances achieved by Russian researchers in the synthesis and characterization of semi-synthetic antibiotics of a new generation in the period from 2004 to 2019. The following classes of compounds are considered as the basis for modification: polycyclic antibacterial glycopeptides of the vancomycin group, classical macrolides, antifungal polyene macrolides, the antitumour antibiotic olivomycin A, antitumour anthracyclines and broad-spectrum antibiotics, in particular, oligomycin A, heliomycin and some other. Main trends in the design of modern anti-infective and antitumour agents over this period are considered in relation to original natural antibiotics, which have been independently discovered by Russian researchers. It is shown that a new type of hybrid structures can, in principle, be synthesized based on glycopeptides, macrolides and other antibiotics, including heterodimers containing a new benzoxaborole pharmacophore. The review addresses the influence of the length of the spacer between two antibiotic molecules on the biological activity of hybrid structures. A combination of genetic engineering techniques and methods of organic synthesis is shown to be useful for the design of new potent antifungal antibiotics based on polyenes of the amphotericin B group. Many new semi-synthetic analogues exhibit important biological properties, such as a broad spectrum of activity and low toxicity. Emphasis is given to certain aspects related to investigation of a broad range of biological activity and mechanisms of action of new derivatives. The bibliography includes 101 references.
Collapse
|
29
|
Vellingiri B, Iyer M, Devi Subramaniam M, Jayaramayya K, Siama Z, Giridharan B, Narayanasamy A, Abdal Dayem A, Cho SG. Understanding the Role of the Transcription Factor Sp1 in Ovarian Cancer: from Theory to Practice. Int J Mol Sci 2020; 21:E1153. [PMID: 32050495 PMCID: PMC7038193 DOI: 10.3390/ijms21031153] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 12/23/2022] Open
Abstract
Ovarian cancer (OC) is one of the deadliest cancers among women contributing to high risk of mortality, mainly owing to delayed detection. There is no specific biomarker for its detection in early stages. However, recent findings show that over-expression of specificity protein 1 (Sp1) is involved in many OC cases. The ubiquitous transcription of Sp1 apparently mediates the maintenance of normal and cancerous biological processes such as cell growth, differentiation, angiogenesis, apoptosis, cellular reprogramming and tumorigenesis. Sp1 exerts its effects on cellular genes containing putative GC-rich Sp1-binding site in their promoters. A better understanding of the mechanisms underlying Sp1 transcription factor (TF) regulation and functions in OC tumorigenesis could help identify novel prognostic markers, to target cancer stem cells (CSCs) by following cellular reprogramming and enable the development of novel therapies for future generations. In this review, we address the structure, function, and biology of Sp1 in normal and cancer cells, underpinning the involvement of Sp1 in OC tumorigenesis. In addition, we have highlighted the influence of Sp1 TF in cellular reprogramming of iPSCs and how it plays a role in controlling CSCs. This review highlights the drugs targeting Sp1 and their action on cancer cells. In conclusion, we predict that research in this direction will be highly beneficial for OC treatment, and chemotherapeutic drugs targeting Sp1 will emerge as a promising therapy for OC.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, India
| | - Mahalaxmi Iyer
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, India; (M.I.); (K.J.)
| | - Mohana Devi Subramaniam
- Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai 600006, India;
| | - Kaavya Jayaramayya
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, India; (M.I.); (K.J.)
| | - Zothan Siama
- Department of Zoology, School of Life-science, Mizoram University, Aizawl 796004, Mizoram, India;
| | - Bupesh Giridharan
- R&D Wing, Sree Balaji Medical College and Hospital (SBMCH), BIHER, Chromepet, Chennai 600044, Tamil Nadu, India;
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India;
| | - Ahmed Abdal Dayem
- Molecular & Cellular Reprogramming Center, Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea;
| | - Ssang-Goo Cho
- Molecular & Cellular Reprogramming Center, Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea;
| |
Collapse
|
30
|
An "olivomycin A" derivative from a sponge-associated Streptomyces sp. strain SP 85. 3 Biotech 2019; 9:439. [PMID: 31750037 DOI: 10.1007/s13205-019-1964-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/22/2019] [Indexed: 12/25/2022] Open
Abstract
We isolated an actinobacterium, Streptomyces sp. strain SP 85 from the marine sponge Dysidea avara. Polyphasic identification of the microorganism showed that the strain SP 85 had high 16S rRNA gene similarity (99%) with Streptomyces olivaceus strain NBRC 12805, while some physiological and biochemical differences were observed. A cytotoxic compound, SP 85 was isolated from the active culture extract of the strain SP 85 by bioassay-guided purification over silica gel column chromatography, preparative TLC, and HPLC. The structure elucidation based on the spectroscopic analysis, including UV, ESI-MS, and 13C NMR data revealed that SP 85 compound is an analog of anti-tumor drug, "olivomycin A". The SP 85 compound showed high cytotoxic activity against three human cancer cell lines, including SW480, HepG2, and MCF7 with IC50 values of 16, 93, and 78 nM, respectively. SP 85 exhibited significantly (2-10 times) higher cytotoxicity against the tumor cell lines in comparison with HUVECs as the normal cell line, which also induced apoptosis in the tested cancerous cell line. This is the first report on the production of an "olivomycin A" derivative by a sponge-associated Streptomyces, showing the great potential of sponge-associated actinobacteria in producing cytotoxic natural products.
Collapse
|
31
|
Ogawara H. Comparison of Antibiotic Resistance Mechanisms in Antibiotic-Producing and Pathogenic Bacteria. Molecules 2019; 24:E3430. [PMID: 31546630 PMCID: PMC6804068 DOI: 10.3390/molecules24193430] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/18/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antibiotic resistance poses a tremendous threat to human health. To overcome this problem, it is essential to know the mechanism of antibiotic resistance in antibiotic-producing and pathogenic bacteria. This paper deals with this problem from four points of view. First, the antibiotic resistance genes in producers are discussed related to their biosynthesis. Most resistance genes are present within the biosynthetic gene clusters, but some genes such as paromomycin acetyltransferases are located far outside the gene cluster. Second, when the antibiotic resistance genes in pathogens are compared with those in the producers, resistance mechanisms have dependency on antibiotic classes, and, in addition, new types of resistance mechanisms such as Eis aminoglycoside acetyltransferase and self-sacrifice proteins in enediyne antibiotics emerge in pathogens. Third, the relationships of the resistance genes between producers and pathogens are reevaluated at their amino acid sequence as well as nucleotide sequence levels. Pathogenic bacteria possess other resistance mechanisms than those in antibiotic producers. In addition, resistance mechanisms are little different between early stage of antibiotic use and the present time, e.g., β-lactam resistance in Staphylococcus aureus. Lastly, guanine + cytosine (GC) barrier in gene transfer to pathogenic bacteria is considered. Now, the resistance genes constitute resistome composed of complicated mixture from divergent environments.
Collapse
Affiliation(s)
- Hiroshi Ogawara
- HO Bio Institute, 33-9, Yushima-2, Bunkyo-ku, Tokyo 113-0034, Japan.
- Department of Biochemistry, Meiji Pharmaceutical University, 522-1, Noshio-2, Kiyose, Tokyo 204-8588, Japan.
| |
Collapse
|
32
|
Shaw M, Kumar A. Additive‐Free Gold(III)‐Catalyzed Stereoselective Synthesis of 2‐Deoxyglycosides Using Phenylpropiolate Glycosides as Donors. Chem Asian J 2019; 14:4651-4658. [DOI: 10.1002/asia.201900888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/05/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Mukta Shaw
- Department of ChemistryIndian Institute of Technology Patna, Bihta 801106 Bihar India
| | - Amit Kumar
- Department of ChemistryIndian Institute of Technology Patna, Bihta 801106 Bihar India
| |
Collapse
|
33
|
Dimakos V, Liu JJW, Ge Z, Taylor MS. Copper-mediated anomeric O-arylation with organoboron reagents. Org Biomol Chem 2019; 17:5671-5674. [PMID: 31123748 DOI: 10.1039/c9ob01022j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Copper-mediated couplings of arylboroxines with glycosyl hemiacetals furnish O-aryl glycosides via Csp2-O bond formation. The method enables the anomeric O-arylation of protected pyranose and furanose derivatives, and is tolerant of functionalized arylboroxine partners. Whereas mixtures of anomers are formed from glucopyranose, galactopyranose and arabinofuranose hemiacetals, the α-anomer is generated selectively from mannopyranose and mannofuranose-derived substrates.
Collapse
Affiliation(s)
- Victoria Dimakos
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON, M5S 3H6 Canada.
| | | | | | | |
Collapse
|
34
|
Sissung TM, Huang PA, Hauke RJ, McCrea EM, Peer CJ, Barbier RH, Strope JD, Ley AM, Zhang M, Hong JA, Venzon D, Jackson JP, Brouwer KR, Grohar P, Glod J, Widemann BC, Heller T, Schrump DS, Figg WD. Severe Hepatotoxicity of Mithramycin Therapy Caused by Altered Expression of Hepatocellular Bile Transporters. Mol Pharmacol 2019; 96:158-167. [PMID: 31175181 DOI: 10.1124/mol.118.114827] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Mithramycin demonstrates preclinical anticancer activity, but its therapeutic dose is limited by the development of hepatotoxicity that remains poorly characterized. A pharmacogenomics characterization of mithramycin-induced transaminitis revealed that hepatotoxicity is associated with germline variants in genes involved in bile disposition: ABCB4 (multidrug resistance 3) rs2302387 and ABCB11 [bile salt export pump (BSEP)] rs4668115 reduce transporter expression (P < 0.05) and were associated with ≥grade 3 transaminitis developing 24 hours after the third infusion of mithramycin (25 mcg/kg, 6 hours/infusion, every day ×7, every 28 days; P < 0.0040). A similar relationship was observed in a pediatric cohort. We therefore undertook to characterize the mechanism of mithramycin-induced acute transaminitis. As mithramycin affects cellular response to bile acid treatment by altering the expression of multiple bile transporters (e.g., ABCB4, ABCB11, sodium/taurocholate cotransporting polypeptide, organic solute transporter α/β) in several cell lines [Huh7, HepaRG, HepaRG BSEP (-/-)] and primary human hepatocytes, we hypothesized that mithramycin inhibited bile-mediated activation of the farnesoid X receptor (FXR). FXR was downregulated in all hepatocyte cell lines and primary human hepatocytes (P < 0.0001), and mithramycin inhibited chenodeoxycholic acid- and GW4046-induced FXR-galactose-induced gene 4 luciferase reporter activity (P < 0.001). Mithramycin promoted glycochenodeoxycholic acid-induced cytotoxicity in ABCB11 (-/-) cells and increased the overall intracellular concentration of bile acids in primary human hepatocytes grown in sandwich culture (P < 0.01). Mithramycin is a FXR expression and FXR transactivation inhibitor that inhibits bile flow and potentiates bile-induced cellular toxicity, particularly in cells with low ABCB11 function. These results suggest that mithramycin causes hepatotoxicity through derangement of bile acid disposition; results also suggest that pharmacogenomic markers may be useful to identify patients who may tolerate higher mithramycin doses. SIGNIFICANCE STATEMENT: The present study characterizes a novel mechanism of drug-induced hepatotoxicity in which mithramycin not only alters farnesoid X receptor (FXR) and small heterodimer partner gene expression but also inhibits bile acid binding to FXR, resulting in deregulation of cellular bile homeostasis. Two novel single-nucleotide polymorphisms in bile flow transporters are associated with mithramycin-induced liver function test elevations, and the present results are the rationale for a genotype-directed clinical trial using mithramycin in patients with thoracic malignancies.
Collapse
Affiliation(s)
- Tristan M Sissung
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Phoebe A Huang
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Ralph J Hauke
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Edel M McCrea
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Cody J Peer
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Roberto H Barbier
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Jonathan D Strope
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Ariel M Ley
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Mary Zhang
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Julie A Hong
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - David Venzon
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Jonathan P Jackson
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Kenneth R Brouwer
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Patrick Grohar
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Jon Glod
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Brigitte C Widemann
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Theo Heller
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - David S Schrump
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - William D Figg
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| |
Collapse
|
35
|
Biosynthesis of Polyketides in Streptomyces. Microorganisms 2019; 7:microorganisms7050124. [PMID: 31064143 PMCID: PMC6560455 DOI: 10.3390/microorganisms7050124] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/24/2019] [Accepted: 04/27/2019] [Indexed: 12/12/2022] Open
Abstract
Polyketides are a large group of secondary metabolites that have notable variety in their structure and function. Polyketides exhibit a wide range of bioactivities such as antibacterial, antifungal, anticancer, antiviral, immune-suppressing, anti-cholesterol, and anti-inflammatory activity. Naturally, they are found in bacteria, fungi, plants, protists, insects, mollusks, and sponges. Streptomyces is a genus of Gram-positive bacteria that has a filamentous form like fungi. This genus is best known as one of the polyketides producers. Some examples of polyketides produced by Streptomyces are rapamycin, oleandomycin, actinorhodin, daunorubicin, and caprazamycin. Biosynthesis of polyketides involves a group of enzyme activities called polyketide synthases (PKSs). There are three types of PKSs (type I, type II, and type III) in Streptomyces responsible for producing polyketides. This paper focuses on the biosynthesis of polyketides in Streptomyces with three structurally-different types of PKSs.
Collapse
|
36
|
Furuta N, Tsukagoshi S, Hirayanagi K, Ikeda Y. Suppression of the yeast elongation factor Spt4 ortholog reduces expanded SCA36 GGCCUG repeat aggregation and cytotoxicity. Brain Res 2019; 1711:29-40. [DOI: 10.1016/j.brainres.2018.12.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/28/2018] [Accepted: 12/31/2018] [Indexed: 12/18/2022]
|
37
|
Sergeev AV, Tevyashova AN, Vorobyov AP, Gromova ES. The Effect of Antitumor Antibiotic Olivomycin A and Its New Semi-synthetic Derivative Olivamide on the Activity of Murine DNA Methyltransferase Dnmt3a. BIOCHEMISTRY (MOSCOW) 2019; 84:62-70. [PMID: 30927527 DOI: 10.1134/s0006297919010085] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Olivomycin A is a highly active antitumor drug that belongs to the family of aureolic acid antibiotics. The antitumor effect of olivomycin A is related to its ability to bind to the DNA minor groove in GC-rich regions as Mg2+-coordinated complexes. Characterization of cellular targets of olivomycin A and its mechanism of action is crucial for the successful application of this antibiotic in clinical practice and development of semi-synthetic derivatives with improved pharmacological properties. Previously, we have shown that minor groove ligands are able to disrupt the key epigenetic process of DNA methylation. In this paper, we have studied the impact of olivomycin A and its improved semi-synthetic analogue N,N-dimethylaminoethylamide of 1'-des-(2,3-dihydroxy-n-butyroyl)-1'-carboxy-olivomycin A (olivamide) on the functioning of de novo DNA methyltransferase Dnmt3a (enzyme that carries out methylation of cytosine residues in the DNA CG-sites in eukaryotic cells) using an in vitro system consisting of the murine Dnmt3a catalytic domain and a 30-mer DNA duplex containing four consecutive GC pairs. We have shown that olivomycin A and olivamide inhibit Dnmt3a with IC50 of 6 ± 1 and 7.1 ± 0.7 μM, respectively. Neither olivomycin A nor olivamide interfered with the formation of the specific enzyme-substrate complex; however, olivomycin A prevented formation of the covalent DNA-Dnmt3a intermediate that is necessary for the methylation reaction to proceed. The inhibitory effects of olivomycin A and olivamide can be explained by the disruption of the enzyme catalytic loop movement through the DNA minor groove (the reaction stage that precedes the covalent bond formation between DNA and the enzyme). The results of this work indicate the epigenetic contribution to the antitumor effect of aureolic acid group antibiotics.
Collapse
Affiliation(s)
- A V Sergeev
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia.
| | - A N Tevyashova
- Gause Institute of New Antibiotics, Moscow, 119021, Russia.,D. Mendeleev University of Chemical Technology of Russia, Moscow, 125047, Russia
| | - A P Vorobyov
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia
| | - E S Gromova
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia
| |
Collapse
|
38
|
Hou C, Rohr J, Parkin S, Tsodikov OV. How mithramycin stereochemistry dictates its structure and DNA binding function. MEDCHEMCOMM 2019; 10:735-741. [PMID: 31191864 DOI: 10.1039/c9md00100j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/28/2019] [Indexed: 12/16/2022]
Abstract
An aureolic acid natural product mithramycin (MTM) has been known for its potent antineoplastic properties. MTM inhibits cell growth by binding in the minor groove of double-stranded DNA as a dimer, in which the two molecules of MTM are coordinated to each other through a divalent metal ion. A crystal structure of an MTM analogue, MTM SA-Phe, in the active metal ion-coordinated dimeric form demonstrates how the stereochemical features of MTM define the helicity of the dimeric scaffold for its binding to a right-handed DNA double helix. We also show crystallographically and biochemically that MTM, but not MTM SA-Phe, can be inactivated by boric acid through formation of a large macrocyclic species, in which two molecules of MTM are crosslinked to each other through 3-side chain-boron-sugar intermolecular bonds. We discuss these structural and biochemical properties in the context of MTM biosynthesis and the design of MTM analogues as anticancer therapeutics.
Collapse
Affiliation(s)
- Caixia Hou
- University of Kentucky , Department of Pharmaceutical Sciences , College of Pharmacy , Lexington , KY 40536-0596 , USA . ;
| | - Jürgen Rohr
- University of Kentucky , Department of Pharmaceutical Sciences , College of Pharmacy , Lexington , KY 40536-0596 , USA . ;
| | - Sean Parkin
- University of Kentucky , Department of Chemistry , Lexington , KY 40506-0055 , USA .
| | - Oleg V Tsodikov
- University of Kentucky , Department of Pharmaceutical Sciences , College of Pharmacy , Lexington , KY 40536-0596 , USA . ;
| |
Collapse
|
39
|
Prajapati D, Kumari N, Dave K, Chatupale V, Pohnerkar J. Chromomycin, an antibiotic produced by Streptomyces flaviscleroticus might play a role in the resistance to oxidative stress and is essential for viability in stationary phase. Environ Microbiol 2019; 21:814-826. [PMID: 30585380 DOI: 10.1111/1462-2920.14515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/13/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
The well-known role of antibiotics in killing sensitive organisms has been challenged by the effects they exert at subinhibitory concentrations. Unfortunately, there are very few published reports on the advantages these molecules may confer to their producers. This study describes the construction of a genetically verified deletion mutant of Streptomyces flaviscleroticus unable to synthesize chromomycin. This mutant was characterized by a rapid loss of viability in stationary phase that was correlated with high oxidative stress and altered antioxidant defences. Altered levels of key metabolites in the mutant signalled a redistribution of the glycolytic flux toward the PPP to generate NADPH to fight oxidative stress as well as reduction of ATP-phosphofructokinase and Krebs cycle enzymes activities. These changes were correlated with a shift in the preference for carbon utilization from glucose to amino acids. Remarkably, chromomycin at subinhibitory concentration increased longevity of the non-producer and restored most of the phenotypic features' characteristic of the wild type strain. Altogether these observations suggest that chromomycin may have antioxidant properties that would explain, at least in part, some of the phenotypes of the mutant. Our observations warrant reconsideration of the secondary metabolite definition and raise the possibility of crucial roles for their producers.
Collapse
Affiliation(s)
- Divya Prajapati
- Department of Bio-Chemistry, The Maharaja Sayajirao University of Baroda, Baroda, 390003, Gujarat, India
| | - Namita Kumari
- Center for Sickle Cell Disease, Howard University, Washington, DC, 20059
| | - Keyur Dave
- Cellcys Labs Pvt. Ltd., Mumbai, 400104, India
| | - Vaidehi Chatupale
- Department of Bio-Chemistry, The Maharaja Sayajirao University of Baroda, Baroda, 390003, Gujarat, India
| | - Jayashree Pohnerkar
- Department of Bio-Chemistry, The Maharaja Sayajirao University of Baroda, Baroda, 390003, Gujarat, India
| |
Collapse
|
40
|
Tolmie C, Smit MS, Opperman DJ. Native roles of Baeyer–Villiger monooxygenases in the microbial metabolism of natural compounds. Nat Prod Rep 2019; 36:326-353. [DOI: 10.1039/c8np00054a] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Baeyer–Villiger monooxygenases function in the primary metabolism of atypical carbon sources, as well as the synthesis of complex microbial metabolites.
Collapse
Affiliation(s)
- Carmien Tolmie
- Department of Biotechnology
- University of the Free State
- Bloemfontein
- South Africa
| | - Martha S. Smit
- Department of Biotechnology
- University of the Free State
- Bloemfontein
- South Africa
| | | |
Collapse
|
41
|
Vickman AE, Pohl NLB. Probing deoxysugar conformational preference: A comprehensive computational study investigating the effects of deoxygenation. Carbohydr Res 2018; 475:17-26. [PMID: 30771703 DOI: 10.1016/j.carres.2018.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/26/2018] [Accepted: 12/06/2018] [Indexed: 10/27/2022]
Abstract
Deoxysugars are intrinsic components in a number of antibiotics, antimicrobials, and therapeutic agents that often dictate receptor binding, improve efficacy, and provide a diverse toolbox in modifying glycoconjugate function due to an extensive number of unique isomers and inherent conformational flexibility. Hence, this work provides a comprehensive examination of the conformational effects associated with deoxygenation of the pyranose ring. Both the location and degree of deoxygenation were evaluated by interrogating the energetic landscape for a number of mono- and dideoxyhexopyranose derivatives using DFT methods (M05-2X/cc-pVTZ(-f)). Both anomeric forms and in some cases, the alternate chair form, have been investigated in the gas phase. As was documented in a preceding study, variation of the C-6 oxidation state has been shown to affect the anomeric preference of select glucose stereoisomers. Similar results were also observed for several deoxysugar isomers in this work, wherein the alternate anomer was favored upon reduction to the 6-deoxyhexose derivative or oxidation to the hexonic acid. Additionally, comparison of relative Gibbs free energies revealed C-3 deoxygenation imparts greater instability compared to C-2 or C-4 deoxygenation, as indicated by an increase in free energy for 3-deoxysugars. A polarizable continuum solvation model was also applied to empirically validate theoretical results for several deoxysugars, wherein good agreement with both carbon (σ = 1.6 ppm) and proton (σ = 0.20 ppm) NMR shifts was observed for the majority of isomers. Solvated and gas phase anomeric ratios were also calculated and compared favorably to reported literature values, although some discrepancies are noted.
Collapse
Affiliation(s)
- Alison E Vickman
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA
| | - Nicola L B Pohl
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
42
|
Molphy Z, Montagner D, Bhat SS, Slator C, Long C, Erxleben A, Kellett A. A phosphate-targeted dinuclear Cu(II) complex combining major groove binding and oxidative DNA cleavage. Nucleic Acids Res 2018; 46:9918-9931. [PMID: 30239938 PMCID: PMC6212767 DOI: 10.1093/nar/gky806] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/05/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022] Open
Abstract
Free radical generation is an inevitable consequence of aerobic existence and is implicated in a wide variety of pathological conditions including cancer, cardiovascular disease, ageing and neurodegenerative disorder. Free radicals can, however, be used to our advantage since their production is catalysed by synthetic inorganic molecules-termed artificial metallonucleases-that cut DNA strands by oxidative cleavage reactions. Here, we report the rational design and DNA binding interactions of a novel di-Cu2+ artificial metallonuclease [Cu2(tetra-(2-pyridyl)-NMe-naphthalene)Cl4] (Cu2TPNap). Cu2TPNap is a high-affinity binder of duplex DNA with an apparent binding constant (Kapp) of 107 M(bp)-1. The agent binds non-intercalatively in the major groove causing condensation and G-C specific destabilization. Artificial metallonuclease activity occurs in the absence of exogenous reductant, is dependent on superoxide and hydrogen peroxide, and gives rise to single strand DNA breaks. Pre-associative molecular docking studies with the 8-mer d(GGGGCCCC)2, a model for poly[d(G-C)2], identified selective major groove incorporation of the complex with ancillary Cu2+-phosphate backbone binding. Molecular mechanics methods then showed the d(GGGGCCCC)2 adduct to relax about the complex and this interaction is supported by UV melting experiments where poly[d(G-C)2] is selectively destabilized.
Collapse
Affiliation(s)
- Zara Molphy
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Diego Montagner
- Department of Chemistry, Maynooth University, Maynooth, Kildare, Ireland
| | - Satish S Bhat
- School of Chemistry, National University of Ireland Galway, Galway, Ireland
| | - Creina Slator
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Conor Long
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Andrea Erxleben
- School of Chemistry, National University of Ireland Galway, Galway, Ireland
- Synthesis and Solid-State Pharmaceutical Centre, School of Chemistry, National University of Ireland Galway, Galway, Ireland
| | - Andrew Kellett
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
- Synthesis and Solid-State Pharmaceutical Centre, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
43
|
Kalwat MA, Hwang IH, Macho J, Grzemska MG, Yang JZ, McGlynn K, MacMillan JB, Cobb MH. Chromomycin A 2 potently inhibits glucose-stimulated insulin secretion from pancreatic β cells. J Gen Physiol 2018; 150:1747-1757. [PMID: 30352794 PMCID: PMC6279362 DOI: 10.1085/jgp.201812177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Drugs that target insulin secretion are useful to understand β cell function and the pathogenesis of diabetes. Kalwat et al. investigate an aureolic acid that inhibits insulin secretion and reveal that it disrupts Wnt signaling, interferes with gene expression, and suppresses Ca2+ influx in β cells. Modulators of insulin secretion could be used to treat diabetes and as tools to investigate β cell regulatory pathways in order to increase our understanding of pancreatic islet function. Toward this goal, we previously used an insulin-linked luciferase that is cosecreted with insulin in MIN6 β cells to perform a high-throughput screen of natural products for chronic effects on glucose-stimulated insulin secretion. In this study, using multiple phenotypic analyses, we found that one of the top natural product hits, chromomycin A2 (CMA2), potently inhibited insulin secretion by at least three potential mechanisms: disruption of Wnt signaling, interference of β cell gene expression, and partial suppression of Ca2+ influx. Chronic treatment with CMA2 largely ablated glucose-stimulated insulin secretion even after washout, but it did not inhibit glucose-stimulated generation of ATP or Ca2+ influx. However, by using the KATP channel opener diazoxide, we uncovered defects in depolarization-induced Ca2+ influx that may contribute to the suppressed secretory response. Glucose-responsive ERK1/2 and S6 phosphorylation were also disrupted by chronic CMA2 treatment. By querying the FUSION bioinformatic database, we revealed that the phenotypic effects of CMA2 cluster with a number of Wnt–GSK3 pathway-related genes. Furthermore, CMA2 consistently decreased GSK3β phosphorylation and suppressed activation of a β-catenin activity reporter. CMA2 and a related compound, mithramycin, are known to have DNA interaction properties, possibly abrogating transcription factor binding to critical β cell gene promoters. We observed that CMA2 but not mithramycin suppressed expression of PDX1 and UCN3. However, neither expression of INSI/II nor insulin content was affected by chronic CMA2. The mechanisms of CMA2-induced insulin secretion defects may involve components both proximal and distal to Ca2+ influx. Therefore, CMA2 is an example of a chemical that can simultaneously disrupt β cell function through both noncytotoxic and cytotoxic mechanisms. Future therapeutic applications of CMA2 and similar aureolic acid analogues should consider their potential effects on pancreatic islet function.
Collapse
Affiliation(s)
- Michael A Kalwat
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - In Hyun Hwang
- Department of Pharmacy, Woosuk University, Wanju, South Korea
| | - Jocelyn Macho
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA
| | - Magdalena G Grzemska
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jonathan Z Yang
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kathleen McGlynn
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - John B MacMillan
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, Santa Cruz, CA
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
44
|
Shinde D, Albino D, Zoma M, Mutti A, Mapelli SN, Civenni G, Kokanovic A, Merulla J, Perez-Escuredo J, Costales P, Morìs F, Catapano CV, Carbone GM. Transcriptional Reprogramming and Inhibition of Tumor-propagating Stem-like Cells by EC-8042 in ERG-positive Prostate Cancer. Eur Urol Oncol 2018; 2:415-424. [PMID: 31277777 DOI: 10.1016/j.euo.2018.08.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND The TMPRSS2-ERG gene fusion is the most frequent genetic rearrangement in prostate cancers and results in broad transcriptional reprogramming and major phenotypic changes. Interaction and cooperation of ERG and SP1 may be instrumental in sustaining the tumorigenic and metastatic phenotype and could represent a potential vulnerability in ERG fusion-positive tumors. OBJECTIVE To test the activity of EC-8042, a compound able to block SP1, in cellular and mouse models of ERG-positive prostate cancer. DESIGN, SETTING, AND PARTICIPANTS We evaluated the activity of EC-8042 in cell cultures and ERG/PTEN transgenic/knockout mice that provide reliable models for testing novel therapeutics in this specific disease context. Using a new protocol to generate tumor spheroids from ERG/PTEN mice, we also examined the effects of EC-8042 on tumor-propagating stem-like cancer cells with high self-renewal and tumorigenic capabilities. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The efficacy of EC-8042 was determined by measuring the proliferative capacity and target gene expression in cell cultures, invasive and metastatic capabilities in chick chorioallantoic membrane assays, and tumor development in mice. Significance was determined using statistical test. RESULTS AND LIMITATIONS EC-8042 blocked transcription of ERG-regulated genes and reverted the invasive and metastatic phenotype of VCaP cells. EC-8042 blocked the expansion of stem-like tumor cells in tumor spheroids from VCaP cells and mouse-derived tumors. In ERG/PTEN mice, systemic treatment with EC-8042 inhibited ERG-regulated gene transcription, tumor progression, and tumor-propagating stem-like tumor cells. CONCLUSIONS Our data support clinical testing of EC-8042 for the treatment of ERG-positive prostate cancer in precision medicine approaches. PATIENT SUMMARY In this study, EC-8042, a novel compound with a favorable pharmacological and toxicological profile, exhibited relevant activity in cell cultures and in vivo in a genetically engineered mouse model that closely recapitulates the features of clinically aggressive ERG-positive prostate cancer. Our data indicate that further evaluation of EC-8042 in clinical trials is warranted.
Collapse
Affiliation(s)
- Dheeraj Shinde
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Domenico Albino
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Marita Zoma
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Azzurra Mutti
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Sarah N Mapelli
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Gianluca Civenni
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Aleksandra Kokanovic
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Jessica Merulla
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | | | | | | | - Carlo V Catapano
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Giuseppina M Carbone
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland.
| |
Collapse
|
45
|
Ogawara H. Comparison of Strategies to Overcome Drug Resistance: Learning from Various Kingdoms. Molecules 2018; 23:E1476. [PMID: 29912169 PMCID: PMC6100412 DOI: 10.3390/molecules23061476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 11/16/2022] Open
Abstract
Drug resistance, especially antibiotic resistance, is a growing threat to human health. To overcome this problem, it is significant to know precisely the mechanisms of drug resistance and/or self-resistance in various kingdoms, from bacteria through plants to animals, once more. This review compares the molecular mechanisms of the resistance against phycotoxins, toxins from marine and terrestrial animals, plants and fungi, and antibiotics. The results reveal that each kingdom possesses the characteristic features. The main mechanisms in each kingdom are transporters/efflux pumps in phycotoxins, mutation and modification of targets and sequestration in marine and terrestrial animal toxins, ABC transporters and sequestration in plant toxins, transporters in fungal toxins, and various or mixed mechanisms in antibiotics. Antibiotic producers in particular make tremendous efforts for avoiding suicide, and are more flexible and adaptable to the changes of environments. With these features in mind, potential alternative strategies to overcome these resistance problems are discussed. This paper will provide clues for solving the issues of drug resistance.
Collapse
Affiliation(s)
- Hiroshi Ogawara
- HO Bio Institute, Yushima-2, Bunkyo-ku, Tokyo 113-0034, Japan.
- Department of Biochemistry, Meiji Pharmaceutical University, Noshio-2, Kiyose, Tokyo 204-8588, Japan.
| |
Collapse
|
46
|
Sun L, Zeng J, Cui P, Wang W, Yu D, Zhan J. Manipulation of two regulatory genes for efficient production of chromomycins in Streptomyces reseiscleroticus. J Biol Eng 2018; 12:9. [PMID: 29977332 PMCID: PMC5992853 DOI: 10.1186/s13036-018-0103-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/30/2018] [Indexed: 11/23/2022] Open
Abstract
Background Regulatory genes play critical roles in natural product biosynthetic pathways. Chromomycins are promising anticancer natural products from actinomycetes. This study is aimed to create an efficient strain for production of these molecules by manipulating the regulatory genes. Results A putative but silent chromomycin biosynthetic gene cluster was discovered in Streptomyces reseiscleroticus. Heterologous expression of the ketosynthase, chain length factor, and acyl carrier protein in Streptomyces lividans confirmed that they are responsible for the assembly of a decaketide. Two regulatory genes are present in this gene cluster, including SARP-type activator SrcmRI and PadR-like repressor SrcmRII. Either overexpression of SrcmRI or disruption of SrcmRII turned on the biosynthetic pathway of chromomycins. The production titers of chromomycin A3/A2 in R5 agar in these two strains reached 8.9 ± 1.2/13.2 ± 1.6 and 49.3 ± 4.3/53.3 ± 3.6 mg/L, respectively. An engineered strain was then constructed with both SrcmRII disruption and SrcmRI overexpression, which produced chromomycins A3 and A2 in R5 agar at 69.4 ± 7.6 and 81.7 ± 7.2 mg/L, respectively. Optimization of the culture conditions further increased the titers of chromomycins A3 and A2 respectively to 145.1 ± 15.3 and 158.3 ± 15.4 mg/L in liquid fermentation. Conclusions This work revealed the synergistic effect of manipulation of pathway repressor and activator genes in the engineering of a natural product biosynthetic pathway. The resulting engineered strain showed the highest production titers of chromomycins by a strain of Streptomyces, providing an efficient way to produce these pharmaceutically valuable molecules. Electronic supplementary material The online version of this article (10.1186/s13036-018-0103-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lei Sun
- 1Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322-4105 USA
| | - Jia Zeng
- 1Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322-4105 USA
| | - Peiwu Cui
- 2TCM and Ethnomedicine Innovation & Development Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| | - Wei Wang
- 2TCM and Ethnomedicine Innovation & Development Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| | - Dayu Yu
- Hangzhou Viablife Biotech Co., Ltd., 1 Jingyi Road, Yuhang District, Hangzhou, Zhejiang 311113 China
| | - Jixun Zhan
- 1Department of Biological Engineering, Utah State University, 4105 Old Main Hill, Logan, UT 84322-4105 USA.,2TCM and Ethnomedicine Innovation & Development Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208 China
| |
Collapse
|
47
|
Choi MC, Choi WH. Mithramycin A Alleviates Osteoarthritic Cartilage Destruction by Inhibiting HIF-2α Expression. Int J Mol Sci 2018; 19:ijms19051411. [PMID: 29747385 PMCID: PMC5983647 DOI: 10.3390/ijms19051411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA) is the most common and increasing joint disease worldwide. Current treatment for OA is limited to control of symptoms. The purpose of this study was to determine the effect of specificity protein 1 (SP1) inhibitor Mithramycin A (MitA) on chondrocyte catabolism and OA pathogenesis and to explore the underlying molecular mechanisms involving SP1 and other key factors that are critical for OA. Here, we show that MitA markedly inhibited expressions of matrix-degrading enzymes induced by pro-inflammatory cytokine interleukin-1β (IL-1β) in mouse primary chondrocytes. Intra-articular injection of MitA into mouse knee joint alleviated OA cartilage destruction induced by surgical destabilization of the medial meniscus (DMM). However, modulation of SP1 level in chondrocyte and mouse cartilage did not alter catabolic gene expression or cartilage integrity, respectively. Instead, MitA significantly impaired the expression of HIF-2α known to be critical for OA pathogenesis. Such reduction in expression of HIF-2α by MitA was caused by inhibition of NF-κB activation, at least in part. These results suggest that MitA can alleviate OA pathogenesis by suppressing NF-κB-HIF-2α pathway, thus providing insight into therapeutic strategy for OA.
Collapse
Affiliation(s)
- Moon-Chang Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
| | - Woo Hee Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
| |
Collapse
|
48
|
Murase H, Noguchi T, Sasaki S. Evaluation of simultaneous binding of Chromomycin A 3 to the multiple sites of DNA by the new restriction enzyme assay. Bioorg Med Chem Lett 2018; 28:1832-1835. [PMID: 29657103 DOI: 10.1016/j.bmcl.2018.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 11/19/2022]
Abstract
Chromomycin A3 (CMA3) is an aureolic acid-type antitumor antibiotic. CMA3 forms dimeric complexes with divalent cations, such as Mg2+, which strongly binds to the GC rich sequence of DNA to inhibit DNA replication and transcription. In this study, the binding property of CMA3 to the DNA sequence containing multiple GC-rich binding sites was investigated by measuring the protection from hydrolysis by the restriction enzymes, AccII and Fnu4HI, for the center of the CGCG site and the 5'-GC↓GGC site, respectively. In contrast to the standard DNase I footprinting method, the DNA substrates are fully hydrolyzed by the restriction enzymes, therefore, the full protection of DNA at all the cleavable sites indicates that CMA3 simultaneously binds to all the binding sites. The restriction enzyme assay has suggested that CMA3 has a high tendency to bind the successive CGCG sites and the CGG repeat.
Collapse
Affiliation(s)
- Hirotaka Murase
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tomoharu Noguchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shigeki Sasaki
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
49
|
Bandi R, Chalapala S, Chandrasekaran S. 2-Deoxyglycosyl 3-benzoylpropionates as novel donors for the direct and stereoselective synthesis of 2-deoxy-glycosides. Org Biomol Chem 2018. [DOI: 10.1039/c8ob00216a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lewis acid mediated stereoselective synthesis of 2-deoxy-O-glycosides has been demonstrated using 2-deoxyglycosyl 3-benzoylpropionates as novel glycosyl donors.
Collapse
Affiliation(s)
- Ramakrishna Bandi
- Department of Organic Chemistry
- Indian Institute of Science
- Bangalore 560 012
- India
| | - Sudharani Chalapala
- Department of Organic Chemistry
- Indian Institute of Science
- Bangalore 560 012
- India
| | | |
Collapse
|
50
|
Novakova R, Núñez LE, Homerova D, Knirschova R, Feckova L, Rezuchova B, Sevcikova B, Menéndez N, Morís F, Cortés J, Kormanec J. Increased heterologous production of the antitumoral polyketide mithramycin A by engineered Streptomyces lividans TK24 strains. Appl Microbiol Biotechnol 2017; 102:857-869. [DOI: 10.1007/s00253-017-8642-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022]
|