1
|
Li M, Xu J, Zang H, Peng WR, Hong LL, Xu C, Li W, Jiao WH, Lin HW, Zhou Y. Anti-Inflammatory α-Pyrone Polyethers Generated by Interrupting the Enolized 1,3-Diketone Formation in the Polyketide Synthases of Calcium-Ionophore Ionomycin. Org Lett 2025; 27:1806-1811. [PMID: 39965537 DOI: 10.1021/acs.orglett.4c04711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Carboxyl polyethers from Actinomycetes have diverse bioactivities, being biosynthesized by type-I polyketide synthetases. Of them, ionomycin, the commercial calcium ionophore, bears a unique enolized 1,3-diketone moiety. Herein, mutagenetic analysis of ionomycin biosynthesis revealed a rare dehydrogenase gene inoJ putatively involved in enol moiety formation. Interruption of the enolized 1,3-diketone formation by inactivating a ketoreductase domain yielded the first natural polyether with an α-pyrone ring, which was demonstrated with significant anti-inflammatory activity in a mouse colitis model.
Collapse
Affiliation(s)
- Mengjia Li
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Xu
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Haixia Zang
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wen-Rui Peng
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Li-Li Hong
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chang Xu
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wanlu Li
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei-Hua Jiao
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hou-Wen Lin
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yongjun Zhou
- Research Center for Marine Drugs, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
2
|
Fiorilla S, Tasso F, Clemente N, Trisciuoglio T, Boldorini R, Carini R. Monensin Inhibits Triple-Negative Breast Cancer in Mice by a Na +-Dependent Cytotoxic Action Unrelated to Cytostatic Effects. Cells 2025; 14:185. [PMID: 39936977 PMCID: PMC11817484 DOI: 10.3390/cells14030185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/11/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Triple-negative breast cancer (TNBC) represents the most aggressive breast cancer subtype, defined by its limited therapeutic options and poor outcomes. This study investigated the therapeutic potential of targeting Na+ homeostasis in TNBC cells to induce TNBC inhibition. For this purpose, BALB/c mice were inoculated with 4T1-Luc2 breast cancer cells and treated with the Na+ ionophore monensin (8 mg/kg) or vehicle alone. Tumor development and cellular Na+ content were assessed using vivo live imaging techniques, while intracellular Na+ variations and cytotoxicity were evaluated through live cell analysis. Monensin treatment increased Na+ levels in cancerous tissues and reduced TNBC mass (monensin: 0.146 ± 0.06; vehicle: 0.468 ± 0.2 cm3; p < 0.001). This treatment induced extensive necrosis in TNBC tumors while preserving the structural and functional integrity of healthy organs and maintaining the proliferative activity of both tumor and normal tissues. Monensin did not alter the expression of proliferating nuclear antigen (PCNA) in 4T1-Luc2 cells but triggered cytotoxicity preceded by intracellular Na+ accumulation. Na+-free conditions prevented both Na+ accumulation and 4T1-Luc2 cell death. Thus, monensin exerts its antitumor effects in TNBC through a Na+-dependent and tumor-specific cytotoxic mechanism, without inducing cytostatic effects on normal or transformed tissues. Collectively, these findings underscore the potential of Na+ ionophores as promising therapeutic agents for TNBC.
Collapse
Affiliation(s)
| | | | | | | | | | - Rita Carini
- Department of Health Science, Università del Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy; (S.F.); (F.T.); (N.C.); (T.T.); (R.B.)
| |
Collapse
|
3
|
Saller BS, Wöhrle S, Fischer L, Dufossez C, Ingerl IL, Kessler S, Mateo-Tortola M, Gorka O, Lange F, Cheng Y, Neuwirt E, Marada A, Koentges C, Urban C, Aktories P, Reuther P, Giese S, Kirschnek S, Mayer C, Pilic J, Falquez-Medina H, Oelgeklaus A, Deepagan VG, Shojaee F, Zimmermann JA, Weber D, Tai YH, Crois A, Ciminski K, Peyronnet R, Brandenburg KS, Wu G, Baumeister R, Heimbucher T, Rizzi M, Riedel D, Helmstädter M, Buescher J, Neumann K, Misgeld T, Kerschensteiner M, Walentek P, Kreutz C, Maurer U, Rambold AS, Vince JE, Edlich F, Malli R, Häcker G, Kierdorf K, Meisinger C, Köttgen A, Jakobs S, Weber ANR, Schwemmle M, Groß CJ, Groß O. Acute suppression of mitochondrial ATP production prevents apoptosis and provides an essential signal for NLRP3 inflammasome activation. Immunity 2025; 58:90-107.e11. [PMID: 39571574 DOI: 10.1016/j.immuni.2024.10.012] [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: 02/13/2024] [Revised: 08/15/2024] [Accepted: 10/24/2024] [Indexed: 01/18/2025]
Abstract
How mitochondria reconcile roles in functionally divergent cell death pathways of apoptosis and NLRP3 inflammasome-mediated pyroptosis remains elusive, as is their precise role in NLRP3 activation and the evolutionarily conserved physiological function of NLRP3. Here, we have shown that when cells were challenged simultaneously, apoptosis was inhibited and NLRP3 activation prevailed. Apoptosis inhibition by structurally diverse NLRP3 activators, including nigericin, imiquimod, extracellular ATP, particles, and viruses, was not a consequence of inflammasome activation but rather of their effects on mitochondria. NLRP3 activators turned out as oxidative phosphorylation (OXPHOS) inhibitors, which we found to disrupt mitochondrial cristae architecture, leading to trapping of cytochrome c. Although this effect was alone not sufficient for NLRP3 activation, OXPHOS inhibitors became triggers of NLRP3 when combined with resiquimod or Yoda-1, suggesting that NLRP3 activation requires two simultaneous cellular signals, one of mitochondrial origin. Therefore, OXPHOS and apoptosis inhibition by NLRP3 activators provide stringency in cell death decisions.
Collapse
Affiliation(s)
- Benedikt S Saller
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Svenja Wöhrle
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Larissa Fischer
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Clara Dufossez
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Isabella L Ingerl
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Kessler
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Maria Mateo-Tortola
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Felix Lange
- Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Clinic for Neurology, University Medical Center of Göttingen, Göttingen, Germany
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Emilia Neuwirt
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Adinarayana Marada
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Koentges
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Chiara Urban
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philipp Aktories
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Peter Reuther
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sebastian Giese
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Kirschnek
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Carolin Mayer
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Johannes Pilic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Hugo Falquez-Medina
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Aline Oelgeklaus
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Veerasikku Gopal Deepagan
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Farzaneh Shojaee
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Julia A Zimmermann
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Damian Weber
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Yi-Heng Tai
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians Universität München, Munich, Germany; Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany; Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Anna Crois
- Faculty of Biology, University of Freiburg, Freiburg, Germany; Institute for Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kevin Ciminski
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Remi Peyronnet
- Institute for Experimental Cardiovascular Medicine, Faculty of Medicine, University Heart Center Freiburg - Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Katharina S Brandenburg
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Gang Wu
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ralf Baumeister
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thomas Heimbucher
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Department of Rheumatology and Clinical Immunology and Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Riedel
- Laboratory for Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Martin Helmstädter
- EMcore, Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Joerg Buescher
- Metabolomics and FACS Core Facilities, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Konstantin Neumann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians Universität München, Munich, Germany; Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Walentek
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Clemens Kreutz
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Medical Biometry and Statistics, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ulrich Maurer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute for Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angelika S Rambold
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Frank Edlich
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Chris Meisinger
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Stefan Jakobs
- Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Clinic for Neurology, University Medical Center of Göttingen, Göttingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy TNM, Göttingen, Germany
| | - Alexander N R Weber
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany; Clusters of Excellence EXC-2180 (iFIT) and -2124 (CMFI), University of Tübingen, Tübingen, Germany
| | - Martin Schwemmle
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Christina J Groß
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
4
|
Borcher Møller SL, Rasmussen MH, Li J, Svenningsen EB, Wørmer GJ, Tørring T, Poulsen TB. The Biological Activities of Polyether Ionophore Antibiotic Routiennocin is Independent of Absolute Stereochemistry. Chembiochem 2024; 25:e202400013. [PMID: 38329925 DOI: 10.1002/cbic.202400013] [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: 01/05/2024] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/10/2024]
Abstract
Carboxylic polyether ionophores (CPIs) are among the most prevalent agricultural antibiotics (notably in the US) and these compounds have been in use for decades. The potential to reposition CPIs beyond veterinary use, e. g. through chemical modifications to enhance their selectivity window, is an exciting challenge and opportunity, considering their general resilience towards resistance development. Given the very large societal impact of these somewhat controversial compounds, it is surprising that many aspects of their mechanisms and activities in cells remain unclear. Here, we report comparative biological activities of the CPI routiennocin and two stereoisomers, including its enantiomer. We used an efficient convergent synthesis strategy to access the compounds and conducted a broad survey of antibacterial activities against planktonic cells and biofilms as well as the compounds' effects on mammalian cells, the latter assessed both via standard cell viability assays and broad morphological profiling. Interestingly, similar bioactivity of the enantiomeric pair was observed across all assays, strongly suggesting that chiral interactions do not play a decisive role in the mode of action. Overall, our findings are consistent with a mechanistic model involving highly dynamic behaviour of CPIs in biological membranes.
Collapse
Affiliation(s)
| | - Michelle H Rasmussen
- Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000, Aarhus C, Denmark
| | - Jun Li
- Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000, Aarhus C, Denmark
| | - Esben B Svenningsen
- Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000, Aarhus C, Denmark
| | - Gustav J Wørmer
- Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000, Aarhus C, Denmark
| | - Thomas Tørring
- Department of Biological and Chemical Engineering, Gustav Wieds Vej 10, DK-8000, Aarhus C, Denmark
| | - Thomas B Poulsen
- Department of Chemistry, Aarhus University, Langelandsgade 140, DK-8000, Aarhus C, Denmark
| |
Collapse
|
5
|
Clemente N, Baroni S, Fiorilla S, Tasso F, Reano S, Borsotti C, Ruggiero MR, Alchera E, Corrazzari M, Walker G, Follenzi A, Crich SG, Carini R. Boosting intracellular sodium selectively kills hepatocarcinoma cells and induces hepatocellular carcinoma tumor shrinkage in mice. Commun Biol 2023; 6:574. [PMID: 37248274 DOI: 10.1038/s42003-023-04946-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/16/2023] [Indexed: 05/31/2023] Open
Abstract
Pharmacological treatments for advanced hepatocellular carcinoma (HCC) have a partial efficacy. Augmented Na+ content and water retention are observed in human cancers and offer unexplored targets for anticancer therapies. Na+ levels are evaluated upon treatments with the antibiotic cation ionophore Monensin by fluorimetry, ICP-MS, 23Na-MRI, NMR relaxometry, confocal or time-lapse analysis related to energy production, water fluxes and cell death, employing both murine and human HCC cell lines, primary murine hepatocytes, or HCC allografts in NSG mice. Na+ levels of HCC cells and tissue are 8-10 times higher than that of healthy hepatocytes and livers. Monensin further increases Na+ levels in HCC cells and in HCC allografts but not in primary hepatocytes and in normal hepatic and extrahepatic tissue. The Na+ increase is associated with energy depletion, mitochondrial Na+ load and inhibition of O2 consumption. The Na+ increase causes an enhancement of the intracellular water lifetime and death of HCC cells, and a regression and necrosis of allograft tumors, without affecting the proliferating activity of either HCCs or healthy tissues. These observations indicate that HCC cells are, unlike healthy cells, energetically incapable of compensating and surviving a pharmacologically induced Na+ load, highlighting Na+ homeostasis as druggable target for HCC therapy.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Simona Baroni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126, Torino, Italy
| | - Simone Fiorilla
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Francesco Tasso
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Simone Reano
- Department of Department of Translational Medicine, Unit of Muscle Biology, Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Chiara Borsotti
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Maria Rosaria Ruggiero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126, Torino, Italy
| | - Elisa Alchera
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS, Ospedale San Raffaele, Milan, Italy
| | - Marco Corrazzari
- Department of Health Science and Interdisciplinary Research Center of Autoimmune Disease (IRCAD), Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Gillian Walker
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Antonia Follenzi
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Simonetta Geninatti Crich
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126, Torino, Italy.
| | - Rita Carini
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy.
| |
Collapse
|
6
|
Secondary Metabolites and Biosynthetic Gene Clusters Analysis of Deep-Sea Hydrothermal Vent-Derived Streptomyces sp. SCSIO ZS0520. Mar Drugs 2022; 20:md20060393. [PMID: 35736196 PMCID: PMC9228677 DOI: 10.3390/md20060393] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 01/27/2023] Open
Abstract
Streptomyces sp. SCSIO ZS0520 is a deep-sea hydrothermal vent-derived actinomycete. Our previous metabolism investigation showed that Streptomyces sp. SCSIO ZS0520 is a producer of cytotoxic actinopyrones. Here, another four types of secondary metabolites were identified, including six salinomycin isomers (2–7), the macrolide elaiophylin (8), the triterpene N-acetyl-aminobacteriohopanetriol (9), and the pyrone minipyrone (10). Among them, compounds 2–6 and 10 are new compounds. To understand the biosynthetic pathway of these compounds, a bioinformatic analysis of the whole genome was carried out, which identified 34 secondary metabolite biosynthetic gene clusters. Next, the biosynthetic pathways responsive to four types of products were deduced on the basis of gene function predictions and structure information. Taken together, these findings prove the metabolite potential of ZS0520 and lay the foundations to solve the remaining biosynthetic issues in four types of marine natural products.
Collapse
|
7
|
Sun Q, Huang M, Wei Y. Diversity of the reaction mechanisms of SAM-dependent enzymes. Acta Pharm Sin B 2021; 11:632-650. [PMID: 33777672 PMCID: PMC7982431 DOI: 10.1016/j.apsb.2020.08.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 08/08/2020] [Indexed: 02/08/2023] Open
Abstract
S-adenosylmethionine (SAM) is ubiquitous in living organisms and is of great significance in metabolism as a cofactor of various enzymes. Methyltransferases (MTases), a major group of SAM-dependent enzymes, catalyze methyl transfer from SAM to C, O, N, and S atoms in small-molecule secondary metabolites and macromolecules, including proteins and nucleic acids. MTases have long been a hot topic in biomedical research because of their crucial role in epigenetic regulation of macromolecules and biosynthesis of natural products with prolific pharmacological moieties. However, another group of SAM-dependent enzymes, sharing similar core domains with MTases, can catalyze nonmethylation reactions and have multiple functions. Herein, we mainly describe the nonmethylation reactions of SAM-dependent enzymes in biosynthesis. First, we compare the structural and mechanistic similarities and distinctions between SAM-dependent MTases and the non-methylating SAM-dependent enzymes. Second, we summarize the reactions catalyzed by these enzymes and explore the mechanisms. Finally, we discuss the structural conservation and catalytical diversity of class I-like non-methylating SAM-dependent enzymes and propose a possibility in enzymes evolution, suggesting future perspectives for enzyme-mediated chemistry and biotechnology, which will help the development of new methods for drug synthesis.
Collapse
|
8
|
Doboszewska U, Wlaź P, Nowak G, Młyniec K. Targeting zinc metalloenzymes in coronavirus disease 2019. Br J Pharmacol 2020; 177:4887-4898. [PMID: 32671829 PMCID: PMC7405164 DOI: 10.1111/bph.15199] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/22/2020] [Accepted: 07/05/2020] [Indexed: 12/15/2022] Open
Abstract
Several lines of evidence support a link between the essential element zinc and the coronavirus disease 2019 (COVID-19). An important fact is that zinc is present in proteins of humans and of viruses. Some zinc sites in viral enzymes may serve as drug targets and may liberate zinc ions, thus leading to changes in intracellular concentration of zinc ions, while increased intracellular zinc may induce biological effects in both the host and the virus. Drugs such as chloroquine may contribute to increased intracellular zinc. Moreover, clinical trials on the use of zinc alone or in addition to other drugs in the prophylaxis/treatment of COVID-19 are ongoing. Thereby, we aim to discuss the rationale for targeting zinc metalloenzymes as a new strategy for the treatment of COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Urszula Doboszewska
- Department of PharmacobiologyJagiellonian University Medical CollegeKrakówPoland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological SciencesMaria Curie‐Skłodowska UniversityLublinPoland
| | - Gabriel Nowak
- Department of PharmacobiologyJagiellonian University Medical CollegeKrakówPoland
- Laboratory of Trace Elements Neurobiology, Department of Neurobiology, Maj Institute of PharmacologyPolish Academy of SciencesKrakówPoland
| | - Katarzyna Młyniec
- Department of PharmacobiologyJagiellonian University Medical CollegeKrakówPoland
| |
Collapse
|
9
|
Chen J, Huang X, Li N, Liu B, Ma Z, Ling J, Yang W, Li T. Narasin inhibits tumor metastasis and growth of ERα‑positive breast cancer cells by inactivation of the TGF‑β/SMAD3 and IL‑6/STAT3 signaling pathways. Mol Med Rep 2020; 22:5113-5124. [PMID: 33174044 PMCID: PMC7646975 DOI: 10.3892/mmr.2020.11624] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 08/27/2020] [Indexed: 12/20/2022] Open
Abstract
Treatment of human estrogen receptor (ER)-positive breast cancer (ER+ BC) using conventional chemotherapy remains a challenge and is often ineffective as a result of tumor metastasis. The present study aimed to investigate the ability of narasin, an ionophore antibiotic, to potentially inhibit tumor metastasis and growth in human ER+ BC. Narasin was found to have significant inhibitory abilities on cell proliferation, migration and invasion in ER+ BC cell lines MCF-7 and T47D compared with the triple-negative BC cell MDA-MB-231. For the in vivo studies, narasin effectively decreased the number of tumor metastasis nodules, tumor volume and weight without apparent toxicity in human MCF-7 nude mouse left ventricle injection tumor metastasis and xenograft models. Mechanistically, it demonstrated that exposure to TGF-β or IL-6 induced the expression of epithelial-mesenchymal transition (EMT) markers in ER+ BC cell lines. On the contrary, narasin dose-dependently reversed EMT by increasing the expression of E-cadherin and decreasing the expression of N-cadherin, vimentin, β-catenin and zinc finger E-box-binding homeobox 1 at the protein and gene expression levels. Gene microarray, molecular docking and western blotting were performed to demonstrate that those protein and gene expression levels are regulated by the inactivation of the TGF-β/phosphorylated (p)-SMAD3 and IL-6/p-STAT3 signaling pathways. Taken together, these findings indicated that narasin may be a promising candidate that can be further optimized for the treatment of human ER+ BC.
Collapse
Affiliation(s)
- Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xieping Huang
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Na Li
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Boxia Liu
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Zhanbing Ma
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Jun Ling
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Wenjun Yang
- School of Basic Medical Sciences, Ningxia Medical University, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Tao Li
- Department of Oncology, General Hospital of The Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
10
|
González-Rubio S, García-Gómez D, Ballesteros-Gómez A, Rubio S. A new sample treatment strategy based on simultaneous supramolecular solvent and dispersive solid-phase extraction for the determination of ionophore coccidiostats in all legislated foodstuffs. Food Chem 2020; 326:126987. [PMID: 32416421 DOI: 10.1016/j.foodchem.2020.126987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 11/28/2022]
Abstract
A single-step sample treatment, for the simultaneous extraction and clean-up for the determination of ionophore coccidiostats in EU legislated foodstuffs, is here proposed. The treatment is based on the combination of: (i) a supramolecular solvent with restricted access properties (SUPRAS-RAM), spontaneously formed by the addition of hexanol, water and THF to the sample; and (ii) dispersive solid phase extraction (dSPE). The SUPRAS-RAM extract was directly compatible with LC-MS/MS and no further re-extraction, evaporation or cleanup procedures were necessary. SUPRAS-RAM efficiently extracted the ionophores (recoveries in milk, eggs, fat, liver, kidney, and chicken and beef muscle were in the range 71-112%) and removed proteins and carbohydrates, whereas dSPE removed fats and other lipophilic compounds. The method was validated following the European Commission Decision 2002/657/EC. Detection limits (0.004-0.07 µg kg-1) were far below the maximum residue limits (1-150 µg kg-1). Method analytical and operational characteristics were suitable for routine determination of ionophores.
Collapse
Affiliation(s)
- S González-Rubio
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica IUIQFN, Universidad de Córdoba, Campus de Rabanales, Edificio Marie Curie (anexo), E-14071 Córdoba, Spain
| | - D García-Gómez
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica IUIQFN, Universidad de Córdoba, Campus de Rabanales, Edificio Marie Curie (anexo), E-14071 Córdoba, Spain.
| | - A Ballesteros-Gómez
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica IUIQFN, Universidad de Córdoba, Campus de Rabanales, Edificio Marie Curie (anexo), E-14071 Córdoba, Spain
| | - S Rubio
- Departamento de Química Analítica, Instituto Universitario de Investigación en Química Fina y Nanoquímica IUIQFN, Universidad de Córdoba, Campus de Rabanales, Edificio Marie Curie (anexo), E-14071 Córdoba, Spain
| |
Collapse
|
11
|
Golbek TW, Schmüser L, Rasmussen MH, Poulsen TB, Weidner T. Lasalocid Acid Antibiotic at a Membrane Surface Probed by Sum Frequency Generation Spectroscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:3184-3192. [PMID: 32069059 DOI: 10.1021/acs.langmuir.9b03752] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Carboxyl polyether ionophores (CPIs) are widely used as veterinary antibiotics and to increase food utilization in ruminating animals. Furthermore, CPIs can target drug-resistant bacteria, but detailed knowledge about their mode-of-action is needed to develop agents with a reasonable therapeutic index. It has been suggested that ionophores bind to membranes and incur large structural changes to shield a bound ion from the hydrophobic environment of the lipid bilayer for transport. One crucial piece of information is missing, however: Is it necessary for the free ionophore to adsorb on the membrane surface before interacting with a cation to facilitate cross-membrane ion transport? To answer this question, we applied sum-frequency generation (SFG) vibrational spectroscopy and surface tensiometry to identify the interaction between the prototypical CPI lasalocid acid (LA) and a model membrane. Observed changes in the surface pressure demonstrate that the free LA undergoes a self-assembly process with the lipid monolayer. Spectra taken from the lipid monolayer show that the free acid inserts partially into the lipid monolayer and then after complexation with sodium chloride disrupts the lipid monolayer. Overall, this study strongly suggests that this must be the crucial step of LA and metal ion complexation that allows the ionophore to traverse a lipid membrane.
Collapse
Affiliation(s)
| | - Lars Schmüser
- Department of Chemistry, Aarhus University, 8000 Aarhus, Denmark
| | | | - Thomas B Poulsen
- Department of Chemistry, Aarhus University, 8000 Aarhus, Denmark
| | - Tobias Weidner
- Department of Chemistry, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
12
|
Liu H, Lin S, Jacobsen KM, Poulsen TB. Chemische Synthesen und chemische Biologie von Carboxylpolyether‐Ionophoren: Aktuelle Entwicklungen. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201812982] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Han Liu
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Dänemark
| | - Shaoquan Lin
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Dänemark
| | - Kristian M. Jacobsen
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Dänemark
| | - Thomas B. Poulsen
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Dänemark
| |
Collapse
|
13
|
Liu H, Lin S, Jacobsen KM, Poulsen TB. Chemical Syntheses and Chemical Biology of Carboxyl Polyether Ionophores: Recent Highlights. Angew Chem Int Ed Engl 2019; 58:13630-13642. [PMID: 30793459 DOI: 10.1002/anie.201812982] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Indexed: 12/21/2022]
Abstract
A central goal of chemical biology is to develop molecular probes that enable fundamental studies of cellular systems. In the hierarchy of bioactive molecules, the so-called ionophore class occupies an unflattering position in the lower branches, with typical labels being "non-specific" and "toxic". In fact, the mere possibility that a candidate molecule possesses "ionophore activity" typically prompts its removal from further studies; ionophores-from a chemical genetics perspective-are molecular outlaws. In stark contrast to this overall poor reputation of ionophores, synthetic chemistry owes some of its most amazing achievements to studies of ionophore natural products, in particular the carboxyl polyethers renowned for their intricate molecular structures. These compounds have for decades been academic battlegrounds where new synthetic methodology is tested and retrosynthetic tactics perfected. Herein, we review the most exciting recent advances in carboxyl polyether ionophore (CPI) synthesis and in addition discuss the burgeoning field of CPI chemical biology.
Collapse
Affiliation(s)
- Han Liu
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000, Aarhus C, Denmark
| | - Shaoquan Lin
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000, Aarhus C, Denmark
| | - Kristian M Jacobsen
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000, Aarhus C, Denmark
| | - Thomas B Poulsen
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000, Aarhus C, Denmark
| |
Collapse
|
14
|
Noack S, Chapman HD, Selzer PM. Anticoccidial drugs of the livestock industry. Parasitol Res 2019; 118:2009-2026. [PMID: 31152233 PMCID: PMC6611755 DOI: 10.1007/s00436-019-06343-5] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
Coccidiosis is a parasitic disease of a wide variety of animals caused by coccidian protozoa. The coccidia are responsible for major economic losses of the livestock industry. For example, the annual cost due to coccidiosis to the global poultry industry has been estimated to exceed US$ 3 billion annually. Currently available drugs for the control of this disease are either polyether ionophorous antibiotics that are derived from fermentation products, or synthetic compounds, produced by chemical synthesis. Unfortunately, no new drugs in either category have been approved for use for decades. Resistance has been documented for all those of the drugs currently employed and therefore the discovery of novel drugs with unique modes of action is imperative if chemotherapy is to remain the principal means to control this disease. This chapter aims to give an overview of the efficacy and mode of action of the current compounds used to control coccidiosis in livestock and provides a brief outlook of research needs for the future.
Collapse
Affiliation(s)
- Sandra Noack
- Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany
| | - H David Chapman
- Department of Poultry Science, University of Arkansas, Fayetteville, AR, USA
| | - Paul M Selzer
- Boehringer Ingelheim Vetmedica GmbH, Ingelheim am Rhein, Germany.
| |
Collapse
|
15
|
Moura J, Freitas T, Cruz R, Pereira R, Silva A, Santos A, da Silva J, Luz-Lima C, Coutinho H. Antibacterial properties and modulation analysis of antibiotic activity of NaCe(MoO4)2 microcrystals. Microb Pathog 2019; 126:258-262. [DOI: 10.1016/j.micpath.2018.11.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/12/2018] [Indexed: 11/16/2022]
|
16
|
Zhu Z, Li H, Yu P, Guo Y, Luo S, Chen Z, Mao X, Guan W, Li Y. SlnR is a positive pathway-specific regulator for salinomycin biosynthesis in Streptomyces albus. Appl Microbiol Biotechnol 2016; 101:1547-1557. [DOI: 10.1007/s00253-016-7918-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/23/2016] [Accepted: 10/04/2016] [Indexed: 01/15/2023]
|
17
|
Jiang C, Qi Z, Kang Q, Liu J, Jiang M, Bai L. Formation of the Δ 18,19Double Bond and Bis(spiroacetal) in Salinomycin Is Atypically Catalyzed by SlnM, a Methyltransferase-like Enzyme. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201503561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
18
|
Jiang C, Qi Z, Kang Q, Liu J, Jiang M, Bai L. Formation of the Δ(18,19) Double Bond and Bis(spiroacetal) in Salinomycin Is Atypically Catalyzed by SlnM, a Methyltransferase-like Enzyme. Angew Chem Int Ed Engl 2015; 54:9097-100. [PMID: 26096919 PMCID: PMC4744726 DOI: 10.1002/anie.201503561] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Indexed: 11/30/2022]
Abstract
Salinomycin is a widely used polyether coccidiostat and was recently found to have antitumor activities. However, the mechanism of its biosynthesis remained largely speculative until now. Reported herein is the identification of an unprecedented function of SlnM, homologous to O‐methyltransferases, by correlating its activity with the formation of the Δ18,19 double bond and bis(spiroacetal). Detailed in vivo and in vitro investigations revealed that SlnM, using positively charged S‐adenosylmethionine (SAM) or sinefungin as the cofactor, catalyzed the spirocyclization‐coupled dehydration of C19 in a highly atypical fashion to yield salinomycin.
Collapse
Affiliation(s)
- Chunyan Jiang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd. Shanghai 200240 (China)
| | - Zhen Qi
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd. Shanghai 200240 (China)
| | - Qianjin Kang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd. Shanghai 200240 (China)
| | - Jing Liu
- Institute of Health Sciences, School of Chemistry and Chemical Engineering, Anhui University, Hefei 230601 (China)
| | - Ming Jiang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd. Shanghai 200240 (China)
| | - Linquan Bai
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Rd. Shanghai 200240 (China).
| |
Collapse
|
19
|
Cybulski W, Radko L, Rzeski W. Cytotoxicity of monensin, narasin and salinomycin and their interaction with silybin in HepG2, LMH and L6 cell cultures. Toxicol In Vitro 2014; 29:337-44. [PMID: 25500126 DOI: 10.1016/j.tiv.2014.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 11/25/2014] [Accepted: 11/26/2014] [Indexed: 12/27/2022]
Abstract
The cytotoxic effect of monensin, narasin and salinomycin followed by their co-action with silybin in the cell line cultures of human hepatoma (HepG2), chicken hepatoma (LMH) or rat myoblasts (L6) have been investigated. The effective concentration of the studied ionophoric polyethers has been assessed within two biochemical endpoints: mitochondrial activity (MTT assay) and membrane integrity (LDH assay) after 24h incubation of each compound and farther, the cytotoxicity influenced in course of their interaction with silybin was determined. The most affected endpoints were found for inhibition of mitochondrial activity of the hepatoma cell lines and their viability depended on concentration of the ionophoric polyether, as well as on the cell line tested. The rat myoblasts were more sensitive target for cellular membrane damage when compared to inhibition of mitochondrial activity. An interaction between the ionophoric polyethers and silybin resulted a considerable cytotoxicity decrease within all studied cell lines; the combination index (CI) showed differences of interaction mode and dependence on cell culture, concentration of silybin, as well as the assay used. The obtained results are of interest in respect to recent findings on applicability of salinomycin and monensin for human therapy.
Collapse
Affiliation(s)
- Wojciech Cybulski
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantow 57, 24-100 Pulawy, Poland.
| | - Lidia Radko
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantow 57, 24-100 Pulawy, Poland.
| | - Wojciech Rzeski
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland; Department of Medical Biology, Institute of Agricultural Medicine, Jaczewskiego 2, 20-950 Lublin, Poland.
| |
Collapse
|
20
|
Clarke L, Fodey TL, Crooks SR, Moloney M, O'Mahony J, Delahaut P, O'Kennedy R, Danaher M. A review of coccidiostats and the analysis of their residues in meat and other food. Meat Sci 2014; 97:358-74. [DOI: 10.1016/j.meatsci.2014.01.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/18/2013] [Accepted: 01/10/2014] [Indexed: 10/25/2022]
|
21
|
Pankiewicz R. 2D NMR, FT-IR, ESI MS studies and DFT, PM5 semiempirical calculations of new benzoic semduramicin anhydride and their complexes with selected monovalent cations. J Mol Struct 2014. [DOI: 10.1016/j.molstruc.2013.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Polyether ionophores—promising bioactive molecules for cancer therapy. Bioorg Med Chem Lett 2012; 22:7002-10. [DOI: 10.1016/j.bmcl.2012.09.046] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/13/2012] [Accepted: 09/15/2012] [Indexed: 01/06/2023]
|
23
|
Szkudlarek-Mikho M, Saunders RA, Yap SF, Ngeow YF, Chin KV. Salinomycin, a polyether ionophoric antibiotic, inhibits adipogenesis. Biochem Biophys Res Commun 2012; 428:487-93. [PMID: 23123626 DOI: 10.1016/j.bbrc.2012.10.080] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 10/23/2012] [Indexed: 12/16/2022]
Abstract
The polyether ionophoric antibiotics including monensin, salinomycin, and narasin, are widely used in veterinary medicine and as food additives and growth promoters in animal husbandry including poultry farming. Their effects on human health, however, are not fully understood. Recent studies showed that salinomycin is a cancer stem cell inhibitor. Since poultry consumption has risen sharply in the last three decades, we asked whether the consumption of meat tainted with growth promoting antibiotics might have effects on adipose cells. We showed in this report that the ionophoric antibiotics inhibit the differentiation of preadipocytes into adipocytes. The block of differentiation is not due to the induction of apoptosis nor the inhibition of cell proliferation. In addition, salinomycin also suppresses the transcriptional activity of the CCAAT/enhancer binding proteins and the peroxisome proliferator-activated receptor γ. These results suggest that the ionophoric antibiotics can be exploited as novel anti-obesity therapeutics and as pharmacological probes for the study of adipose biology. Further, the pharmacological effects of salinomycin could be a harbinger of its toxicity on the adipose tissue and other susceptible target cells in cancer therapy.
Collapse
Affiliation(s)
- Maria Szkudlarek-Mikho
- Department of Medicine, Biochemistry and Cancer Biology, Center for Diabetes and Endocrine Research, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | | | | | | | | |
Collapse
|
24
|
Cytostatic and cytotoxic properties of monensic acid and its biometal(II) complexes against human tumor / non-tumor cell lines. OPEN CHEM 2012. [DOI: 10.2478/s11532-012-0071-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe anticancer activity of monensic acid (MonH) and its biometal(II) complexes [M(Mon)2(H2O)2](M = Mg, Ca, Mn, Co, Ni, Zn) was evaluated against cultured human permanent cell lines established from glioblastoma multiforme (8MGBA) and cancers of the lung (A549), breast (MCF-7), uterine cervix (HeLa) and liver (HepG2). The viability and proliferation of the non-tumor human embryonic cell line Lep3 was also tested. The investigations were carried out using a thiazolyl blue tetrazolium bromide test, neutral red uptake cytotoxicity assay, crystal violet staining, colony forming method and double staining with acridin orange and propidium iodide. The results obtained reveal that the compounds applied at concentrations of 0.5–25 µg mL−1 for 24–72 h decrease the viability and proliferation of the treated cells in a time- and concentration-dependent manner. The metal(II) complexes studied (especially those of Co(II), Ni(II) and Zn(II)) have been found to express stronger cytotoxic and cytostatic activities than the non-coordinated monensic acid. The non-tumor human cell line showed strong chemosensitivity towards compounds tested comparable to that of cultured human tumor cell lines.
Collapse
|
25
|
Huczyński A, Janczak J, Łowicki D, Brzezinski B. Monensin A acid complexes as a model of electrogenic transport of sodium cation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2108-19. [DOI: 10.1016/j.bbamem.2012.04.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/17/2012] [Accepted: 04/23/2012] [Indexed: 11/25/2022]
|
26
|
Wu G, Zhu J. NMR studies of alkali metal ions in organic and biological solids. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2012; 61:1-70. [PMID: 22340207 DOI: 10.1016/j.pnmrs.2011.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Accepted: 05/31/2011] [Indexed: 05/31/2023]
Affiliation(s)
- Gang Wu
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, Ontario, Canada.
| | | |
Collapse
|
27
|
Cloning and characterization of the polyether salinomycin biosynthesis gene cluster of Streptomyces albus XM211. Appl Environ Microbiol 2011; 78:994-1003. [PMID: 22156425 DOI: 10.1128/aem.06701-11] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Salinomycin is widely used in animal husbandry as a food additive due to its antibacterial and anticoccidial activities. However, its biosynthesis had only been studied by feeding experiments with isotope-labeled precursors. A strategy with degenerate primers based on the polyether-specific epoxidase sequences was successfully developed to clone the salinomycin gene cluster. Using this strategy, a putative epoxidase gene, slnC, was cloned from the salinomycin producer Streptomyces albus XM211. The targeted replacement of slnC and subsequent trans-complementation proved its involvement in salinomycin biosynthesis. A 127-kb DNA region containing slnC was sequenced, including genes for polyketide assembly and release, oxidative cyclization, modification, export, and regulation. In order to gain insight into the salinomycin biosynthesis mechanism, 13 gene replacements and deletions were conducted. Including slnC, 7 genes were identified as essential for salinomycin biosynthesis and putatively responsible for polyketide chain release, oxidative cyclization, modification, and regulation. Moreover, 6 genes were found to be relevant to salinomycin biosynthesis and possibly involved in precursor supply, removal of aberrant extender units, and regulation. Sequence analysis and a series of gene replacements suggest a proposed pathway for the biosynthesis of salinomycin. The information presented here expands the understanding of polyether biosynthesis mechanisms and paves the way for targeted engineering of salinomycin activity and productivity.
Collapse
|
28
|
The biosynthesis of the polyether antibiotic nanchangmycin is controlled by two pathway-specific transcriptional activators. Arch Microbiol 2011; 194:415-26. [PMID: 22109812 DOI: 10.1007/s00203-011-0768-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 09/28/2011] [Accepted: 10/24/2011] [Indexed: 10/15/2022]
Abstract
The nanchangmycin (NAN) produced by Streptomyces nanchangensis NS3226 is a polyether antibiotic resembling monensin in their gene clusters and the chemical structures. They can inhibit gram-positive bacteria and be a growth promoter for ruminants. Within the nanchangmycin gene cluster (nan), we identified that two SARP-family regulatory genes, nanR1 and nanR2, were both required to activate the transcription of all nan polyketide genes. Overexpression of NanR1 and NanR2 in wild-type increase NAN yields by at least three folds. Bioinformatic analysis of the immediate upstream DNA sequence of each nan gene and quantitative real-time RT-PCR analysis of the nan operons identified five putative SARP binding sites. Moreover, deletion of an AraC-family repressor gene nanR4 increased expression of NanR1 and R2 and led to a threefold increase in NAN production.
Collapse
|
29
|
Ivanova J, Pantcheva IN, Mitewa M, Simova S, Tanabe M, Osakada K. Cd(II) and Pb(II) complexes of the polyether ionophorous antibiotic salinomycin. Chem Cent J 2011; 5:52. [PMID: 21906282 PMCID: PMC3184049 DOI: 10.1186/1752-153x-5-52] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/09/2011] [Indexed: 12/13/2022] Open
Abstract
Background The natural polyether ionophorous antibiotics are used for the treatment of coccidiosis in poultry and ruminants. They are effective agents against infections caused by Gram-positive microorganisms. On the other hand, it was found that some of these compounds selectively bind lead(II) ions in in vivo experiments, despite so far no Pb(II)-containing compounds of defined composition have been isolated and characterized. To assess the potential of polyether ionophores as possible antidotes in the agriculture, a detailed study on their in vitro complexation with toxic metal ions is required. In the present paper we report for the first time the preparation and the structure elucidation of salinomycin complexes with ions of cadmium(II) and lead(II). Results New metal(II) complexes of the polyether ionophorous antibiotic salinomycin with Cd(II) and Pb(II) ions were prepared and structurally characterized by IR, FAB-MS and NMR techniques. The spectroscopic information and elemental analysis data reveal that sodium salinomycin (SalNa) undergoes a reaction with heavy metal(II) ions to form [Cd(Sal)2(H2O)2] (1) and [Pb(Sal)(NO3)] (2), respectively. Abstraction of sodium ions from the cavity of the antibiotic is occurring during the complexation reaction. Salinomycin coordinates with cadmium(II) ions as a bidentate monoanionic ligand through the deprotonated carboxylic moiety and one of the hydroxyl groups to yield 1. Two salinomycin anions occupy the equatorial plane of the Cd(II) center, while two water molecules take the axial positions of the inner coordination sphere of the metal(II) cation. Complex 2 consists of monoanionic salinomycin acting in polydentate coordination mode in a molar ratio of 1: 1 to the metal ion with one nitrate ion for charge compensation. Conclusion The formation of the salinomycin heavy metal(II) complexes indicates a possible antidote activity of the ligand in case of chronic/acute intoxications likely to occur in the stock farming.
Collapse
Affiliation(s)
- Juliana Ivanova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad, G, Bontchev Str,, build, 9, 1113 Sofia, Bulgaria.
| | | | | | | | | | | |
Collapse
|
30
|
Huczyński A, Ratajczak-Sitarz M, Stefańska J, Katrusiak A, Brzezinski B, Bartl F. Reinvestigation of the structure of monensin A phenylurethane sodium salt based on X-ray crystallographic and spectroscopic studies, and its activity against hospital strains of methicillin-resistant S. epidermidis and S. aureus. J Antibiot (Tokyo) 2011; 64:249-56. [PMID: 21224863 DOI: 10.1038/ja.2010.167] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Monensin A phenylurethane sodium salt (MON-UR1-Na) crystals were studied by the X-ray, NMR, FT-IR and PM5 semi-empirical methods. The X-ray data show that the compound forms a pseudocyclic structure, stabilized by three intramolecular hydrogen bonds, and the sodium cation coordinated by five oxygen atoms in the hydrophilic sphere. The NMR and FT-IR data demonstrate that this pseudocyclic structure is also conserved in CH(2)Cl(2) solution. This structure of MON-UR1-Na is significantly different than the ones previously proposed by Westley et al. and Tanaka et al. The semi-empirical calculations of the MON-UR1-Na structures indicate that the one of the crystal is the most energetically favorable one. Other parameters, such as the size, chemical and biological nature of the urethane substituent, and especially the free carbonyl urethane group, may have a role in the biological activity of MON-UR1-Na. The in vitro microbiological tests provide evidence that MON-UR1-Na shows higher antibacterial activity against human pathogenic bacteria, including antibiotic-resistant Staphylococcus aureus and Staphylococcus epidermidis than the parent unmodified antibiotic-Monensin A.
Collapse
Affiliation(s)
- Adam Huczyński
- Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland.
| | | | | | | | | | | |
Collapse
|
31
|
Pantcheva IN, Ivanova J, Zhorova R, Mitewa M, Simova S, Mayer-Figge H, Sheldrick WS. Nickel(II) and zinc(II) dimonensinates: Single crystal X-ray structure, spectral properties and bactericidal activity. Inorganica Chim Acta 2010. [DOI: 10.1016/j.ica.2010.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Kowalska E, Phopase J, Gathergood N. Synthesis, X-ray Crystal Studies and Metal Picrates Extraction Properties of Lipophilic Benzocrown Ethers. Aust J Chem 2010. [DOI: 10.1071/ch10112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The preparation of 16 macrocycles based on 2-(2-hydroxyethoxy) phenol with both aliphatic and aromatic linkers has been achieved. Macrocycles varying in ring sizes (23–28 atoms), number of aromatic groups (2–4), and donor atoms (6–10 including oxygen and nitrogen) were synthesized. Binding affinities were also assessed by extraction studies against a series of metal picrates. X-ray crystal structures were solved for four macrocycles.
Collapse
|
33
|
Matsumori N, Murata M. 3D structures of membrane-associated small molecules as determined in isotropic bicelles. Nat Prod Rep 2010; 27:1480-92. [DOI: 10.1039/c0np00002g] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
34
|
Pantcheva IN, Zhorova R, Mitewa M, Simova S, Mayer-Figge H, Sheldrick WS. First solid state alkaline-earth complexes of monensic acid A (MonH): crystal structure of [M(Mon)2(H (2)O)2] (M = Mg, Ca), spectral properties and cytotoxicity against aerobic Gram-positive bacteria. Biometals 2009; 23:59-70. [PMID: 19768636 DOI: 10.1007/s10534-009-9269-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 09/03/2009] [Indexed: 11/26/2022]
Abstract
Alkaline-earth metal complexes of the monoanionic form of the polyether ionophore monensin A were isolated for the first time in solid state and were structurally characterized using various spectroscopic methods (IR, NMR, FAB-MS). The stoichiometric reaction of monensic acid (MonH) with M(2+) (M = Mg, Ca) in the presence of an organic base leads to the formation of mononuclear complexes of composition [M(Mon)(2)(H(2)O)(2)]. The structures of magnesium (1) and calcium (2) monensin complexes in the solid state were established by single crystal X-ray crystallography. The complexes crystallize as [Mg(Mon)(2)(H(2)O)(2)]x5MeCN (1) and [Ca(Mon)(2)(H(2)O)(2)]xH(2)Ox5MeCN (2) in the monoclinic P21 space group. The alkaline-earth metal ion is placed in a distorted octahedral environment, defined by two monensin anions acting as bidentate ligands in the equatorial plane of the complex as well as by two water molecules occupying the axial positions of the inner coordination sphere. The bactericidal activity of 1 and 2 was evaluated against aerobic Gram-positive microorganisms applying the double layer agar hole diffusion method.
Collapse
Affiliation(s)
- Ivayla N Pantcheva
- Laboratory on Biocoordination and Bioanalytical Chemistry, Department of Analytical Chemistry, Faculty of Chemistry, Sofia University, 1164 Sofia, Bulgaria.
| | | | | | | | | | | |
Collapse
|
35
|
Łowicki D, Huczyński A, Stefańska J, Brzezinski B. Syntheses, structural and antimicrobial studies of a new N-allylamide of monensin A and its complexes with monovalent metal cations. Tetrahedron 2009. [DOI: 10.1016/j.tet.2009.06.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
36
|
Pantcheva IN, Dorkov P, Atanasov VN, Mitewa M, Shivachev BL, Nikolova RP, Mayer-Figge H, Sheldrick WS. Crystal structure and properties of the copper(II) complex of sodium monensin A. J Inorg Biochem 2009; 103:1419-24. [PMID: 19748125 DOI: 10.1016/j.jinorgbio.2009.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 05/06/2009] [Accepted: 05/07/2009] [Indexed: 11/30/2022]
Abstract
The preparation and structural characterization of a new copper(II) complex of the polyether ionophorous antibiotic sodium monensin A (MonNa) are described. Sodium monensin A binds Cu(II) to produce a heterometallic complex of composition [Cu(MonNa)(2)Cl(2)].H(2)O, 1. The crystallographic data of 1 show that the complex crystallizes in monoclinic space group C2 with Cu(II) ion adopting a distorted square-planar geometry. Copper(II) coordinates two anionic sodium monensin ligands and two chloride anions producing a neutral compound. The sodium ion remains in the inner cavity of the ligand retaining its sixfold coordination with oxygen atoms. Replacement of crystallization water by acetonitrile is observed in the crystal structure of the complex 1. Copper(I) salt of the methyl ester of MonNa, 2, was identified by X-ray crystallography as a side product of the reaction of MonNa with Cu(II). Compound 2, [Me-MonNa][H-MonNa][CuCl(2)]Cl, crystallizes in monoclinic space group C2 with the same coordination pattern of the sodium cation but contains a chlorocuprate(I) counter [CuCl(2)](-), which is linear and not coordinated by sodium monensin A. The antibacterial and antioxidant properties as two independent activities of 1 were studied. Compound 1 is effective against aerobic Gram(+)-microorganisms Bacillus subtilis, Bacillus mycoides and Sarcina lutea. Complex 1 shows SOD-like activity comparable with that of the copper(II) ion.
Collapse
Affiliation(s)
- Ivayla N Pantcheva
- Department of Analytical Chemistry, Faculty of Chemistry, Sofia University, 1, J. Bourchier Blvd., 1164 Sofia, Bulgaria
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Structural and antimicrobial studies of a new N-phenylamide of monensin A complex with sodium chloride. J Mol Struct 2009. [DOI: 10.1016/j.molstruc.2009.01.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Takada Y, Matsuo K, Kataoka T. Gramicidin A directly inhibits mammalian Na+/K+-ATPase. Mol Cell Biochem 2008; 319:99-103. [DOI: 10.1007/s11010-008-9882-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 07/03/2008] [Indexed: 11/29/2022]
|
39
|
Huczyński A, Stefańska J, Przybylski P, Brzezinski B, Bartl F. Synthesis and antimicrobial properties of monensin A esters. Bioorg Med Chem Lett 2008; 18:2585-9. [PMID: 18375122 DOI: 10.1016/j.bmcl.2008.03.038] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 03/12/2008] [Accepted: 03/14/2008] [Indexed: 11/25/2022]
Abstract
The esters (2-10) of the ionophore antibiotic Monensin (1) were synthesized by four different methods, which are discussed in detail. These new esters were characterized by various spectroscopic techniques and subsequently tested in the face of their antimicrobial properties. Three derivatives (3, 8 and 10) showed activity against Gram-positive bacteria. Additionally derivative (10) exhibited a relatively low antifungal activity against Candida in contrast to Monensin A.
Collapse
Affiliation(s)
- Adam Huczyński
- Faculty of Chemistry, A. Mickiewicz University, Grunwaldzka 6, 60-780 Poznan, Poland
| | | | | | | | | |
Collapse
|
40
|
Dorkov P, Pantcheva IN, Sheldrick WS, Mayer-Figge H, Petrova R, Mitewa M. Synthesis, structure and antimicrobial activity of manganese(II) and cobalt(II) complexes of the polyether ionophore antibiotic Sodium Monensin A. J Inorg Biochem 2008; 102:26-32. [PMID: 17692921 DOI: 10.1016/j.jinorgbio.2007.06.033] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 06/20/2007] [Accepted: 06/25/2007] [Indexed: 11/16/2022]
Abstract
Mononuclear neutral manganese(II) and cobalt(II) complexes with the antibiotic Sodium Monensin A (Mon-Na, 1b) were synthesized and characterized. The crystal structures of M(Mon-Na)2Cl2.H2O (M=Mn, 2; M=Co, 3) were determined by X-ray crystallography. The complexes crystallize in monoclinic space group C2 with a tetrahedrally coordinated transition metal attached to oxygen atoms of deprotonated carboxyl groups of two Sodium Monensin molecules and two chloride ions. The sodium ion remains in the cavity of the ligand and cannot be replaced by Mn(II) or Co(II). The complexes were additionally characterized by different spectroscopic techniques (UV-Visible, EPR, FAB-MS). A preferable octahedral environment around the transition metal centers is observed in polar solvents while the complexes retain their tetrahedral structure in non-polar media. The antimicrobial activity of 1b, 2 and 3 was tested against Gram(+) and Gram(-) bacteria.
Collapse
Affiliation(s)
- Petar Dorkov
- Department of Analytical Chemistry, Faculty of Chemistry, Sofia University, 1, J. Bourchier Blvd., 1164 Sofia, Bulgaria
| | | | | | | | | | | |
Collapse
|
41
|
Matsumori N, Morooka A, Murata M. Conformation and Location of Membrane-Bound Salinomycin−Sodium Complex Deduced from NMR in Isotropic Bicelles. J Am Chem Soc 2007; 129:14989-95. [DOI: 10.1021/ja075024l] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nobuaki Matsumori
- Contribution from the Department of Chemistry, Graduate School of Science, Osaka University, Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Atsushi Morooka
- Contribution from the Department of Chemistry, Graduate School of Science, Osaka University, Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Michio Murata
- Contribution from the Department of Chemistry, Graduate School of Science, Osaka University, Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
42
|
Huczyński A, Przybylski P, Brzezinski B. NMR, FTIR, ESI-MS and semiempirical study of a new 2-(2-hydroxyethoxy)ethyl ester of monensin A and its complexes with alkali metal cations. Tetrahedron 2007. [DOI: 10.1016/j.tet.2007.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
Liu T, You D, Valenzano C, Sun Y, Li J, Yu Q, Zhou X, Cane DE, Deng Z. Identification of NanE as the thioesterase for polyether chain release in nanchangmycin biosynthesis. ACTA ACUST UNITED AC 2006; 13:945-55. [PMID: 16984884 DOI: 10.1016/j.chembiol.2006.07.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2006] [Revised: 07/17/2006] [Accepted: 07/17/2006] [Indexed: 11/19/2022]
Abstract
The polyketide synthase (PKS) for the biosynthesis of the polyether nanchangmycin lacks an apparent thioesterase comparable to the type I thioesterase domains of the modular PKSs responsible for macrolide biosynthesis. Three candidate polyether chain-releasing factors were examined. Both the putative CR domain and the NanE protein appeared to be genetically relevant. Among the three heterologously expressed soluble proteins (recombinant CR domain, the ACP-CR didomain, and NanE) tested, only NanE hydrolyzed the polyether-SNAC. By contrast, recombinant DEBS TE from the erythromycin pathway, and the recombinant MonAX, a type II TE associated with the polyether monensin biosynthesis for which a homolog has not been detected in the nanchangmycin cluster, hydrolyzed a diketide-SNAC but not the polyether-SNAC. We could thus conclude that NanE is a dedicated thioesterase mediating the specific release of the polyether chain during nanchangmycin biosynthesis.
Collapse
Affiliation(s)
- Tiangang Liu
- Laboratory of Microbial Metabolism and School of Life Science & Biotechnology, Shanghai Jiaotong University, Shanghai 200030, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Deng Z, Bai L. Antibiotic biosynthetic pathways and pathway engineering--a growing research field in China. Nat Prod Rep 2006; 23:811-27. [PMID: 17003911 DOI: 10.1039/b611140h] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This review describes the recent research activities in China in relation to studies on antibiotic biosynthetic pathways and pathway engineering in actinomycetes. 75 references are cited.
Collapse
Affiliation(s)
- Zixin Deng
- Laboratory of Microbial Metabolism and School of Life Science & Biotechnology, Shanghai Jiaotong University, Shanghai, 200030, China.
| | | |
Collapse
|
45
|
Huczyński A, Przybylski P, Brzezinski B, Bartl F. Spectroscopic, mass spectrometry, and semiempirical investigation of a new ester of Monensin A with ethylene glycol and its complexes with monovalent metal cations. Biopolymers 2006; 82:491-503. [PMID: 16518855 DOI: 10.1002/bip.20502] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A new ester of Monensin A with ethylene glycol (MON2) has been synthesized by a new method and its ability to form complexes with Li+, Na+, and K+ cations has been studied by ESI MS, 1H and 13C NMR, FT-IR, and PM5 semiempirical methods. It is demonstrated that MON2 forms stable complexes of 1:1 stoichiometry with monovalent metal cations. The structures of the complexes are stabilized by intramolecular hydrogen bonds in which the OH groups are always involved. In the structure of MON2 the oxygen atom of the C=O ester group is involved in very weak bifurcated intramolecular hydrogen bonds with two hydroxyl groups, whereas in the complexes of MON2 with monovalent metal cations the C=O ester group is not engaged in any intramolecular hydrogen bonds. The structures of the MON2 and its complexes with Li+, Na+, and K+ cations are visualized and discussed in detail.
Collapse
Affiliation(s)
- Adam Huczyński
- Faculty of Chemistry, A. Mickiewicz University, Grunwaldzka 6, 60-780 Poznan, Poland
| | | | | | | |
Collapse
|
46
|
Huczyñski A, Przybylski P, Brzezinski B, Bartl F. Spectroscopic and Semiempirical Studies of a Proton Channel Formed by the Methyl Ester of Monensin A. J Phys Chem B 2006; 110:15615-23. [PMID: 16884286 DOI: 10.1021/jp062160o] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Monensin A is an ionophore able to carry protons and cations through the cell membrane. Its methyl ester (MON1) and its hydrates have been studied in acetonitrile, and its deuterated analogue by Fourier transform infrared (FTIR) and (1)H and (13)C NMR spectroscopies as well as by vapor pressure osmotic and PM5 semiempirical methods. Interestingly, these hydrates show new and unexpected biophysical and biochemical properties. The formation of the hydrates starts with a transfer of a proton from the O(IV)-H hydroxyl group of MON1 to an oxygen atom of a water molecule, which is subsequently hydrated by other water molecules forming the (MON1 + 3H(2)O) species. This hydrate exhibits a ringlike structure in which the water molecules form an almost linear hydrogen-bonded chain. Within this chain, the excess proton fluctuates very fast inside the water cluster as indicated by a continuous absorption in the FTIR spectra. The formation of the (MON1 + 3H(2)O) species is accompanied by a self-assembly process, leading to the formation of a proton channel made up of eight (MON1 + 3H(2)O) units with a length of 60 A, in which the proton can fluctuate over the whole distance. Semiempirical calculations suggest that due to the hydrophobic surface the channel can be incorporated readily in a lipid bilayer. This hypothetical new channel is thought to be able to transport protons through the cell membrane. Thus it is a suitable model for studying proton-transfer processes, and in addition, it may open interesting new fields of application.
Collapse
Affiliation(s)
- Adam Huczyñski
- Faculty of Chemistry, A. Mickiewicz University, Grunwaldzka 6, 60-780 Poznan, Poland
| | | | | | | |
Collapse
|
47
|
Huczyński A, Przybylski P, Brzezinski B, Bartl F. Monensin A methyl ester complexes with Li+, Na+, and K+ cations studied by ESI-MS, 1H- and 13C-NMR, FTIR, as well as PM5 semiempirical method. Biopolymers 2006; 81:282-94. [PMID: 16312019 DOI: 10.1002/bip.20415] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Monensin A methyl ester (MON1) was synthesized by a new method and its ability to form complexes with Li+, Na+, and K+ cations was studied by electrospray ionization-mass spectroscopy (ESI-MS), 1H and 13C nuclear magnetic resonance (NMR), Fourier transform infrared (FTIR), and PM5 semiempirical methods. It is shown that MON1 with monovalent metal cations forms stable complexes of 1:1 stoichiometry. The structures of the complexes are stabilized by intramolecular hydrogen bonds in which the OH groups are always involved. In the structure of MON1, the oxygen atom of the C=O ester group is involved in very weak bifurcated intramolecular hydrogen bonds with two hydroxyl groups, whereas in the complexes of MON1 with monovalent metal cations the C=O ester group is not engaged in any intramolecular hydrogen bonds. Furthermore, it is demonstrated that the strongest intramolecular hydrogen bonds are formed within the MON1-Li+ complex structure. The structures of the MON1 and its complexes with Li+, Na+, and K+ cations are visualized and discussed in detail.
Collapse
Affiliation(s)
- Adam Huczyński
- Faculty of Chemistry, A. Mickiewicz University, Grunwaldzka 6, 60-780 Poznan, Poland
| | | | | | | |
Collapse
|
48
|
Vázquez-Hernández M, Rosquete-Pina GA, Juaristi E. Salt Effects on the Conformational Behavior of 5-Carboxy- and 5-Hydroxy-1,3-dioxane1. J Org Chem 2004; 69:9063-72. [PMID: 15609939 DOI: 10.1021/jo048900b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The varied and essential involvement of metal ions and inorganic salts in biological and chemical processes motivated the present study where 5-carboxy- and 5-hydroxy-1,3-dioxanes are used as model frameworks for the evaluation of the conformational behavior of oxygen-containing receptors in the presence of Li(+), Na(+), K(+), Ag(+), Mg(2+), Ca(2+), Ba(2+), and Zn(2+). Thus, the position of equilibria, established by means of BF(3), between diastereomeric cis- and trans-5-substituted-2-phenyl-1,3-dioxanes, in solvent THF and in the presence of 0, 1, and 5 equiv of salt, has been determined. The observed Delta G(o) degrees values for the conformational equilibria of 5-carboxy-1,3-dioxane show that Ag(+), Li(+), and Ca(2+) complexation leads to increased stability of the axial isomer. In the case of the 5-hydroxy-1,3-dioxane, Mg(2+), Ag(+), and Zn(2+) are the metal ions that stabilize the axial conformer of the heterocycle upon association. Interpretation of the experimental observations was based on DFT molecular modeling studies at the Becke3LYP/6-31G* and Becke3LYP/6-31+G** levels of theory. Although gas-phase calculations give Delta E values that are too large when modeling equilibria involving ionic species in polar solution, the computational results confirm the structural and energetic consequences of metal cation coordination to the oxygen atom in carbonyls or ethers. The results derived from the present study contribute to our understanding of the chemical processes involved in molecular recognition and physiological events.
Collapse
Affiliation(s)
- Maribel Vázquez-Hernández
- Departamento de Química, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, 07000 México, D.F., México
| | | | | |
Collapse
|
49
|
Fischer HP, Brunner NA, Wieland B, Paquette J, Macko L, Ziegelbauer K, Freiberg C. Identification of antibiotic stress-inducible promoters: a systematic approach to novel pathway-specific reporter assays for antibacterial drug discovery. Genome Res 2004; 14:90-8. [PMID: 14707172 PMCID: PMC314284 DOI: 10.1101/gr.1275704] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
As present antibiotics therapy becomes increasingly ineffectual, new technologies are required to identify and develop novel classes of antibacterial agents. An attractive alternative to the classical target-based approach is the use of promoter-inducible reporter assays for high-throughput screening. The wide usage of these assays is, however, limited by the small number of specifically responding promoters that are known at present. This work describes a novel approach for identifying genetic regulators that are suitable for the design of pathway-specific assays. The basis for the proposed strategy is a large set of antibiotics-triggered expression profiles ("Reference Compendium"). Pattern recognition algorithms applied to the expression data pinpoint the relevant transcription-factor-binding sites in whole-genome sequences. Using this technique, we constructed a fatty-acid-pathway-specific reporter assay that is based on a novel stress-inducible promoter. In a proof-of-principle experiment, this assay was shown to enable screening for new small-molecule inhibitors of bacterial growth.
Collapse
MESH Headings
- 5' Flanking Region/drug effects
- 5' Flanking Region/genetics
- Amino Acid Sequence
- Anti-Bacterial Agents/pharmacology
- Bacillus/drug effects
- Bacillus/genetics
- Binding Sites/genetics
- Cell Extracts/chemistry
- Chromosome Mapping
- Consensus Sequence
- Conserved Sequence
- Drug Design
- Drug Evaluation, Preclinical/methods
- Drug Resistance, Bacterial/genetics
- Fatty Acids/biosynthesis
- Gene Expression Profiling/methods
- Gene Expression Regulation, Bacterial/drug effects
- Gene Expression Regulation, Bacterial/genetics
- Genes, Regulator/drug effects
- Genes, Regulator/genetics
- Genes, Reporter/drug effects
- Genes, Reporter/genetics
- Genome, Bacterial
- Molecular Sequence Data
- Operon/genetics
- Promoter Regions, Genetic/drug effects
- Transcription Factors/genetics
Collapse
|
50
|
Aspinall HC, Greeves N, McIver EG. Modular chiral polyether podands and their lanthanide complexes. Tetrahedron 2003. [DOI: 10.1016/j.tet.2003.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|