1
|
Zhang R, Mei S, He G, Wei M, Chen L, Chen Z, Zhu M, Zhou B, Wang K, Cheng Z, Wang C, Zhu E, Chen C. Multi-omics analyses reveal fecal microbial community and metabolic alterations in finishing cattle fed probiotics-fermented distiller's grains diets. Microbiol Spectr 2025; 13:e0072124. [PMID: 40214255 PMCID: PMC12054032 DOI: 10.1128/spectrum.00721-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 03/13/2025] [Indexed: 05/08/2025] Open
Abstract
Distiller's grains (DG) are a potential source of animal feeds, and many studies have indicated positive regulatory roles of feeding DG diets in animal breeding. However, there is currently a dearth of research on the actions and underlying mechanisms of probiotics-fermented distiller's grains (FDG)-based diets in cattle breeding. This study aimed to assess the impact of integrating FDG into the diet of finishing cattle on their fecal microbial community and metabolites. Thirty Simmental crossbred cattle (local yellow cattle × Simmental cattle, 8.5 months old, 420.38 ± 68.11 kg) were selected and randomly divided into three dietary treatments, including the basal diet group (CON group), the FDG replacing 10% concentrate (FDG-10%) group, and the FDG replacing 20% concentrate (FDG-20%) group. 16S and ITS sequencing of fecal samples collected from each group on the 30th day of the formal feeding suggested that feeding FDG diets had little effect on the composition and diversity of fecal bacterial and fungal communities in finishing cattle. However, the relative abundance of cellulose-degrading bacteria, including the Christensenellaceae R-7 group and Ruminococcaceae family was significantly higher in both the FDG-20% vs CON comparison and the FDG-20% vs FDG-10% comparison. Besides, the FDG-10% group had a significant drop in the relative abundance of Aspergillus and a noteworthy increase in the relative abundance of Candida when compared to the CON group. Non-targeted metabolomics analysis showed that the addition of FDG modified the levels of organoheterocyclic compounds, lipids and lipid-like molecules, and benzenoids in the feces of finishing cattle and significantly enhanced the metabolic pathway of bile secretion. Further correlation analyses suggested a close association between the significantly differential fecal microbiota and metabolites. In conclusion, these results suggest that FDG supplementation has little effect on the structure and diversity of the fecal microbiota in finishing cattle, but alters intestinal metabolite profiles and influences bile secretion pathways by modulating the relative abundance of genera of fecal bacteria and fungi Christensenellaceae R-7 group, Lachnospiraceae_NK3A20_group, Mucor, and Candida. These findings provide a scientific theoretical basis for the use of FDG in animal feeds. IMPORTANCE Probiotics-fermented distiller's grains (FDG) are potential feed sources for livestock. Here, we investigated the effects of partially replacing concentrates with FDG on fecal bacterial and fungal community structure and metabolic profiles in finishing cattle. The results reveal that feeding FDG-based diets alters intestinal metabolite profiles and up-regulates bile secretion pathways through the regulation of relative abundance of certain fecal genera. These findings provide some new insights into clarifying the role and potential mechanisms of FDG diets and also offer a scientific basis for the development of FDG into functional feed resources.
Collapse
Affiliation(s)
- Rong Zhang
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Shihui Mei
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Guangxia He
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Miaozhan Wei
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Lan Chen
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Ze Chen
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Min Zhu
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Bijun Zhou
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Kaigong Wang
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Zhentao Cheng
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Chunmei Wang
- College of Animal Science, Guizhou University, Guiyang, China
| | - Erpeng Zhu
- College of Animal Science, Guizhou University, Guiyang, China
- Guizhou Provincial Animal Disease Research Laboratory, Guiyang, China
| | - Chao Chen
- College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
2
|
Hodson LE, Joseph Tholath P, Jacobs L, Pribut N, Pashikanti G, van der Westhuyzen AE, Laws D, Liotta DC. Mild and Chemoselective Triethylsilane-Mediated Debenzylation for Phosphate Synthesis. Org Lett 2025; 27:246-251. [PMID: 39718906 PMCID: PMC11731398 DOI: 10.1021/acs.orglett.4c04258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
The synthetic utility of tetrabenzyl pyrophosphate for achieving chemoselective phosphorylation of phenols, as well as primary, secondary, and tertiary alcohols, is reported here. Additionally, we introduce a rapid, mild, and chemoselective debenzylation procedure, enabling access to phosphates in the presence of redox sensitive groups. Finally, stoichiometrically controlled monodebenzylation provides a versatile platform for late-stage divergent synthesis of phosphodiester and phosphoramidate chemical libraries.
Collapse
Affiliation(s)
| | | | - Leon Jacobs
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Nicole Pribut
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Gouthami Pashikanti
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | | | - David Laws
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Dennis C. Liotta
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
3
|
Jamil SNH, Ali AH, Feroz SR, Lam SD, Agustar HK, Mohd Abd Razak MR, Latip J. Curcumin and Its Derivatives as Potential Antimalarial and Anti-Inflammatory Agents: A Review on Structure-Activity Relationship and Mechanism of Action. Pharmaceuticals (Basel) 2023; 16:609. [PMID: 37111366 PMCID: PMC10146798 DOI: 10.3390/ph16040609] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Curcumin, one of the major ingredients of turmeric (Curcuma longa), has been widely reported for its diverse bioactivities, including against malaria and inflammatory-related diseases. However, curcumin's low bioavailability limits its potential as an antimalarial and anti-inflammatory agent. Therefore, research on the design and synthesis of novel curcumin derivatives is being actively pursued to improve the pharmacokinetic profile and efficacy of curcumin. This review discusses the antimalarial and anti-inflammatory activities and the structure-activity relationship (SAR), as well as the mechanisms of action of curcumin and its derivatives in malarial treatment. This review provides information on the identification of the methoxy phenyl group responsible for the antimalarial activity and the potential sites and functional groups of curcumin for structural modification to improve its antimalarial and anti-inflammatory actions, as well as potential molecular targets of curcumin derivatives in the context of malaria and inflammation.
Collapse
Affiliation(s)
- Siti Nur Hidayah Jamil
- Department of Chemical Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
| | - Amatul Hamizah Ali
- Department of Chemical Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
| | - Shevin Rizal Feroz
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
| | - Su Datt Lam
- Department of Applied Physics, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
| | - Hani Kartini Agustar
- Department of Earth Sciences and Environment, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
| | - Mohd Ridzuan Mohd Abd Razak
- Herbal Medicine Research Centre, Institute for Medical Research, National Institute of Health (NIH) Complex, Ministry of Health Malaysia, Shah Alam 40170, Selangor, Malaysia
| | - Jalifah Latip
- Department of Chemical Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia (UKM), Bangi 43600, Selangor, Malaysia
| |
Collapse
|
4
|
Kingston DGI, Cassera MB. Antimalarial Natural Products. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2022; 117:1-106. [PMID: 34977998 DOI: 10.1007/978-3-030-89873-1_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Natural products have made a crucial and unique contribution to human health, and this is especially true in the case of malaria, where the natural products quinine and artemisinin and their derivatives and analogues, have saved millions of lives. The need for new drugs to treat malaria is still urgent, since the most dangerous malaria parasite, Plasmodium falciparum, has become resistant to quinine and most of its derivatives and is becoming resistant to artemisinin and its derivatives. This volume begins with a short history of malaria and follows this with a summary of its biology. It then traces the fascinating history of the discovery of quinine for malaria treatment and then describes quinine's biosynthesis, its mechanism of action, and its clinical use, concluding with a discussion of synthetic antimalarial agents based on quinine's structure. The volume then covers the discovery of artemisinin and its development as the source of the most effective current antimalarial drug, including summaries of its synthesis and biosynthesis, its mechanism of action, and its clinical use and resistance. A short discussion of other clinically used antimalarial natural products leads to a detailed treatment of other natural products with significant antiplasmodial activity, classified by compound type. Although the search for new antimalarial natural products from Nature's combinatorial library is challenging, it is very likely to yield new antimalarial drugs. The chapter thus ends by identifying over ten natural products with development potential as clinical antimalarial agents.
Collapse
Affiliation(s)
- David G I Kingston
- Department of Chemistry and the Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA, 24061, USA.
| | - Maria Belen Cassera
- Department of Biochemistry and Molecular Biology, and Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
5
|
Cook A, MacLean H, St. Onge P, Newman SG. Nickel-Catalyzed Reductive Deoxygenation of Diverse C–O Bond-Bearing Functional Groups. ACS Catal 2021. [DOI: 10.1021/acscatal.1c03980] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Adam Cook
- Centre for Catalysis Research and Innovation, Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie, Ottawa, Ontario K1N 6N5, Canada
| | - Haydn MacLean
- Centre for Catalysis Research and Innovation, Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie, Ottawa, Ontario K1N 6N5, Canada
| | - Piers St. Onge
- Centre for Catalysis Research and Innovation, Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie, Ottawa, Ontario K1N 6N5, Canada
| | - Stephen G. Newman
- Centre for Catalysis Research and Innovation, Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
6
|
Leanse LG, Dong PT, Goh XS, Lu M, Cheng JX, Hooper DC, Dai T. Quinine Enhances Photo-Inactivation of Gram-Negative Bacteria. J Infect Dis 2020; 221:618-626. [PMID: 31565732 DOI: 10.1093/infdis/jiz487] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/24/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Antimicrobial resistance is a significant concern to public health, and there is a pressing need to develop novel antimicrobial therapeutic modalities. METHODS In this study, we investigated the capacity for quinine hydrochloride (Q-HCL) to enhance the antimicrobial effects of antimicrobial blue light ([aBL] 405 nm wavelength) against multidrug-resistant (MDR) Gram-negative bacteria in vitro and in vivo. RESULTS Our findings demonstrated the significant improvement in the inactivation of MDR Pseudomonas aeruginosa and Acinetobacter baumannii (planktonic cells and biofilms) when aBL was illuminated during Q-HCL exposure. Furthermore, the addition of Q-HCL significantly potentiated the antimicrobial effects of aBL in a mouse skin abrasion infection model. In addition, combined exposure of aBL and Q-HCL did not result in any significant apoptosis when exposed to uninfected mouse skin. CONCLUSIONS In conclusion, aBL in combination with Q-HCL may offer a novel approach for the treatment of infections caused by MDR bacteria.
Collapse
Affiliation(s)
- Leon G Leanse
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pu-Ting Dong
- Department of Chemistry, Boston University, Boston, Massachusetts, USA
| | - Xueping S Goh
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Min Lu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ji-Xin Cheng
- Department of Chemistry, Boston University, Boston, Massachusetts, USA.,Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - David C Hooper
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tianhong Dai
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Vongsvivut J, Pérez-Guaita D, Wood BR, Heraud P, Khambatta K, Hartnell D, Hackett MJ, Tobin MJ. Synchrotron macro ATR-FTIR microspectroscopy for high-resolution chemical mapping of single cells. Analyst 2019; 144:3226-3238. [DOI: 10.1039/c8an01543k] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Coupling synchrotron IR beam to an ATR element enhances spatial resolution suited for high-resolution single cell analysis in biology, medicine and environmental science.
Collapse
Affiliation(s)
| | | | - Bayden R. Wood
- Centre for Biospectroscopy
- Monash University
- Clayton
- Australia
| | - Philip Heraud
- Centre for Biospectroscopy
- Monash University
- Clayton
- Australia
- Department of Microbiology and Biomedicine Discovery Institute
| | - Karina Khambatta
- Curtin Institute for Functional Molecules and Interfaces
- School of Molecular and Life Sciences
- Curtin University
- Perth
- Australia
| | - David Hartnell
- Curtin Institute for Functional Molecules and Interfaces
- School of Molecular and Life Sciences
- Curtin University
- Perth
- Australia
| | - Mark J. Hackett
- Curtin Institute for Functional Molecules and Interfaces
- School of Molecular and Life Sciences
- Curtin University
- Perth
- Australia
| | - Mark J. Tobin
- Infrared Microspectroscopy (IRM) Beamline
- Australian Synchrotron
- Clayton
- Australia
| |
Collapse
|
8
|
Hisamatsu Y, Umezawa N, Yagi H, Kato K, Higuchi T. Design and synthesis of a 4-aminoquinoline-based molecular tweezer that recognizes protoporphyrin IX and iron(iii) protoporphyrin IX and its application as a supramolecular photosensitizer. Chem Sci 2018; 9:7455-7467. [PMID: 30319746 PMCID: PMC6180317 DOI: 10.1039/c8sc02133c] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/14/2018] [Indexed: 01/08/2023] Open
Abstract
We report on the design and synthesis of a new type of 4-aminoquinoline-based molecular tweezer 1 which forms a stable host-guest complex with protoporphyrin IX (PPIX) via multiple interactions in a DMSO and HEPES buffer (pH 7.4) mixed solvent system. The binding constant for the 1 : 1 complex (K 11) between 1 and PPIX is determined to be 4 × 106 M-1. Furthermore, 1 also forms a more stable complex with iron(iii) protoporphyrin IX (Fe(iii)PPIX), the K 11 value for which is one order of magnitude greater than that for PPIX, indicating that 1 could be used as a recognition unit of a synthetic heme sensor. On the other hand, the formation of the stable PPIX·1 complex (supramolecular photosensitizer) prompted us to apply it to photodynamic therapy (PDT). Cell staining experiments using the supramolecular photosensitizer and evaluations of its photocytotoxicity indicate that the PDT activity of PPIX is improved as the result of the formation of a complex with 1.
Collapse
Affiliation(s)
- Yosuke Hisamatsu
- Graduate School of Pharmaceutical Sciences , Nagoya City University , 3-1 Tanabe-dori, Mizuho-ku , Nagoya 467-8603 , Japan . ;
| | - Naoki Umezawa
- Graduate School of Pharmaceutical Sciences , Nagoya City University , 3-1 Tanabe-dori, Mizuho-ku , Nagoya 467-8603 , Japan . ;
| | - Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences , Nagoya City University , 3-1 Tanabe-dori, Mizuho-ku , Nagoya 467-8603 , Japan . ;
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences , Nagoya City University , 3-1 Tanabe-dori, Mizuho-ku , Nagoya 467-8603 , Japan . ;
- Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science (IMS) , National Institutes of Natural Sciences , 5-1 Higashiyama, Myodaiji , Okazaki 444-8787 , Japan
| | - Tsunehiko Higuchi
- Graduate School of Pharmaceutical Sciences , Nagoya City University , 3-1 Tanabe-dori, Mizuho-ku , Nagoya 467-8603 , Japan . ;
| |
Collapse
|
9
|
Kacprzak K, Ruszkowski P, Valentini L, Huczyński A, Steverding D. Cytotoxic and trypanocidal activities of cinchona alkaloid derivatives. Chem Biol Drug Des 2018; 92:1778-1787. [PMID: 29877033 DOI: 10.1111/cbdd.13346] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/09/2018] [Accepted: 05/12/2018] [Indexed: 11/28/2022]
Abstract
A series of 27 cinchona alkaloid derivatives (1f-w, 2a-e and 3a-d) were investigated for their cytotoxic and trypanocidal activities using seven different cancer cell lines (KB, HeLa, MCF-7, A-549, Hep-G2, U-87 and HL-60), two normal cell lines (HDF and CHO) and bloodstream forms of Trypanosoma brucei brucei, respectively. Four compounds (1u, 1w, 2e and 3d) were identified with promising cytotoxic activity with 50% growth inhibition (GI50 ) values below 10 μM. Two (2e and 3d) of the four compounds also exhibited potent anti-trypanosomal activity with GI50 values of 0.3-0.4 μM. All four active compounds represented derivatives modified at their C-9 hydroxy group. With respect to anti-proliferative activity and selectivity, 2e (epi-N-quinidyl-N'-bis(3,5-trifluoromethyl)phenylthiourea) proved to be the most promising derivative for both cancer cells and bloodstream forms of T. b. brucei. The cytotoxic activity of compounds 1u, 1w, 2e and 3d was attributed to their ability to induce apoptosis in cancer cells. The results demonstrate the potential of cinchona alkaloid derivatives as novel anti-cancer and anti-trypanosome drug candidates.
Collapse
Affiliation(s)
- Karol Kacprzak
- Bioorganic Chemistry Department, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| | - Piotr Ruszkowski
- Department of Pharmacology, Poznan University of Medical Sciences, Poznan, Poland
| | - Luisa Valentini
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Adam Huczyński
- Bioorganic Chemistry Department, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| | | |
Collapse
|
10
|
Perez-Guaita D, Marzec KM, Hudson A, Evans C, Chernenko T, Matthäus C, Miljkovic M, Diem M, Heraud P, Richards JS, Andrew D, Anderson DA, Doerig C, Garcia-Bustos J, McNaughton D, Wood BR. Parasites under the Spotlight: Applications of Vibrational Spectroscopy to Malaria Research. Chem Rev 2018; 118:5330-5358. [DOI: 10.1021/acs.chemrev.7b00661] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- David Perez-Guaita
- Centre for Biospectroscopy, School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Katarzyna M. Marzec
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzyńskiego 14, Kraków 30-348, Poland
- Center for Medical Genomics (OMICRON), Jagiellonian University, Kopernika 7C, Krakow 31-034, Poland
| | - Andrew Hudson
- Department of Chemistry, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Corey Evans
- Department of Chemistry, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Tatyana Chernenko
- Becton Dickinson and Company, 2350 Qume Drive, San Jose, California 95131, United States
| | - Christian Matthäus
- Leibniz Institute of Photonic Technology, Albert Einstein Straße 9, Jena 07745, Germany
- Institute of Physical Chemistry and Abbe School of Photonics, Friedrich Schiller University, Helmholtz Weg 4, Jena 07743, Germany
| | - Milos Miljkovic
- Department of Mechanical Engineering, Tufts University, 200 Boston Avenue, Medford, Massachusetts 02155, United States
| | - Max Diem
- Laboratory for Spectral Diagnosis (LSpD), Department of Chemistry and Chemical Biology, Northeastern University, 316 Hurtig Hall, 360 Huntington Avenue, Boston, Massachusetts 02155, United States
| | - Philip Heraud
- Centre for Biospectroscopy, School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Jack S. Richards
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
- Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
- Department of Medicine, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Dean Andrew
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - David A. Anderson
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Christian Doerig
- Department of Microbiology and the Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Jose Garcia-Bustos
- Department of Microbiology and the Biomedical Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Don McNaughton
- Centre for Biospectroscopy, School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Bayden R. Wood
- Centre for Biospectroscopy, School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
11
|
Woodland JG, Hunter R, Smith PJ, Egan TJ. Shining new light on ancient drugs: preparation and subcellular localisation of novel fluorescent analogues of Cinchona alkaloids in intraerythrocytic Plasmodium falciparum. Org Biomol Chem 2018; 15:589-597. [PMID: 27785512 DOI: 10.1039/c6ob02110g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Fluorescent derivatives of the archetypal antimalarial quinine and its diastereomer, quinidine, suitable for cellular imaging have been synthesised by attaching the small extrinsic fluorophore, NBD. Interactions of these derivatives with ferriprotoporphyrin IX were evaluated to verify that insights generated by live-cell imaging were relevant to the parent molecules. These analogues are shown by confocal and super-resolution microscopy to accumulate selectively in Plasmodium falciparum. Localisation to the region corresponding to the digestive vacuole supports the putative primary role of these alkaloids as haemozoin inhibitors. Quantitative analysis revealed minimal accumulation within the nucleus, rejecting the disruption of DNA replication as a possible mode of action. While extensive localisation to phospholipid structures and associated organelles was observed, the analogues did not show evidence of association with neutral lipid bodies.
Collapse
Affiliation(s)
- John G Woodland
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch, 7701, South Africa.
| | - Roger Hunter
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch, 7701, South Africa.
| | - Peter J Smith
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, 7925, South Africa
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch, 7701, South Africa.
| |
Collapse
|
12
|
Synthesis of novel quinine analogs and evaluation of their effects on Trypanosoma cruzi. Future Med Chem 2018; 10:391-408. [PMID: 29380636 DOI: 10.4155/fmc-2017-0184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM Chagas disease is a tropical disease caused by the hemoflagellate protozoan Trypanosoma cruzi. There is no vaccine for Chagas disease and available drugs (e.g., benznidazole) are effective only during the acute phase, displaying a variable curative activity in the established chronic form of the disease. New leads with high efficacy and better toxicity profiles are urgently required. Materials & methods: A library of novel quinine derivatives was synthesized using Heck chemistry and evaluated against the various developmental forms of T. cruzi. RESULTS AND CONCLUSION Several novel quinine analogs with trypanocidal activity have been identified with the para-nitro-substituted derivative displaying a submicromolar IC50, which is 83-times lower than quinine and three-times lower than benznidazole. Transmission electron microscopy analysis demonstrated that these compounds induced a marked vacuolization of the kinetoplast of intracellular amastigotes and cell-derived trypomastigotes.
Collapse
|
13
|
Olafson KN, Nguyen TQ, Vekilov PG, Rimer JD. Deconstructing Quinoline-Class Antimalarials to Identify Fundamental Physicochemical Properties of Beta-Hematin Crystal Growth Inhibitors. Chemistry 2017; 23:13638-13647. [PMID: 28833627 DOI: 10.1002/chem.201702251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Indexed: 11/12/2022]
Abstract
A versatile approach to control crystallization involves the use of modifiers, which are additives that interact with crystal surfaces and alter their growth rates. Elucidating a modifier's binding specificity to anisotropic crystal surfaces is a ubiquitous challenge that is critical to their design. In this study, we select hematin, a byproduct of malaria parasites, as a model system to examine the complementarity of modifiers (i.e., antimalarial drugs) to β-hematin crystal surfaces. We divide two antimalarials, chloroquine and amodiaquine, into segments consisting of a quinoline base, common to both drugs, and side chains that differentiate their modes of action. Using a combination of scanning probe microscopy, bulk crystallization, and analytical techniques, we show that the base and side chain work synergistically to reduce the rate of hematin crystallization. In contrast to general observations that modifiers retain their function upon segmentation, we show that the constituents do not act as modifiers. A systematic study of quinoline isomers and analogues shows how subtle rearrangement and removal of functional moieties can create effective constituents from previously ineffective modifiers, along with tuning their inhibitory modes of action. These findings highlight the importance of specific functional moieties in drug compounds, leading to an improved understanding of modifier-crystal interactions that could prove to be applicable to the design of new antimalarials.
Collapse
Affiliation(s)
- Katy N Olafson
- Department of Chemical and Biomolecular Engineering, University of Houston, 4726 Calhoun Road, Houston, TX 77204, USA
| | - Tam Q Nguyen
- Department of Chemical and Biomolecular Engineering, University of Houston, 4726 Calhoun Road, Houston, TX 77204, USA
| | - Peter G Vekilov
- Department of Chemical and Biomolecular Engineering, University of Houston, 4726 Calhoun Road, Houston, TX 77204, USA.,Department of Chemistry, University of Houston, 3585 Cullen Boulevard, Houston, TX 77204, USA
| | - Jeffrey D Rimer
- Department of Chemical and Biomolecular Engineering, University of Houston, 4726 Calhoun Road, Houston, TX 77204, USA.,Department of Chemistry, University of Houston, 3585 Cullen Boulevard, Houston, TX 77204, USA
| |
Collapse
|
14
|
Dodd EL, Tazoo D, Bohle DS. Solution and Solid State Correlations of Antimalarial Drug Actions: NMR and Crystallographic Studies of Drug Interactions with a Heme Model. Inorg Chem 2017. [PMID: 28650618 DOI: 10.1021/acs.inorgchem.7b00526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Solution NMR has been used in tandem with a diamagnetic non-iron heme model compound as a simple and effective tool to rapidly probe the structures of the bound complexes formed between the metalloporphyrin and antimalarial drugs from the 4-aminoquinoline, 4-methylenehydroxylquinoline, and 8-aminoquinoline subfamilies. The ability of gallium(III) protoporphyrin IX to mimic heme chemistry is exploited. The 4-aminoquinolines quinacrine and amodiaquine and two novel 3-halo chloroquine analogues are found to bind to the metalloporphyrin through hydrogen-bonding and stacking interactions, while halofantrine and the 4-methylenehydroxylquinolines, quinine and mefloquine bind through the alcohol group of the drug. In each case, detailed structural information is available from the NMR assessment. The mefloquine model is confirmed crystallographically. The 8-aminoquinoline primaquine does not interact strongly. These tools show promise for future applications in assessing antimalarials in preclinical development for heme-binding drug targets.
Collapse
Affiliation(s)
- Erin L Dodd
- Department of Chemistry, McGill University , Montreal, H3A OB8, Quebec, Canada
| | - Dagobert Tazoo
- Department of Chemistry, McGill University , Montreal, H3A OB8, Quebec, Canada
| | - D Scott Bohle
- Department of Chemistry, McGill University , Montreal, H3A OB8, Quebec, Canada
| |
Collapse
|
15
|
|
16
|
Gildenhuys J, Sammy CJ, Müller R, Streltsov VA, le Roex T, Kuter D, de Villiers KA. Alkoxide coordination of iron(III) protoporphyrin IX by antimalarial quinoline methanols: a key interaction observed in the solid-state and solution. Dalton Trans 2015; 44:16767-77. [PMID: 26335948 DOI: 10.1039/c5dt02671g] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The quinoline methanol antimalarial drug mefloquine is a structural analogue of the Cinchona alkaloids, quinine and quinidine. We have elucidated the single crystal X-ray diffraction structure of the complexes formed between racemic erythro mefloquine and ferriprotoporphyrin IX (Fe(iii)PPIX) and show that alkoxide coordination is a key interaction in the solid-state. Mass spectrometry confirms the existence of coordination complexes of quinine, quinidine and mefloquine to Fe(iii)PPIX in acetonitrile. The length of the iron(iii)-O bond in the quinine and quinidine complexes as determined by Extended X-ray Absorption Fine Structure (EXAFS) spectroscopy unequivocally confirms that coordination of the quinoline methanol compounds to Fe(iii)PPIX occurs in non-aqueous aprotic solution via their benzylic alkoxide functional group. UV-visible spectrophotometric titrations of the low-spin bis-pyridyl-Fe(iii)PPIX complex with each of the quinoline methanol compounds results in the displacement of a single pyridine molecule and subsequent formation of a six-coordinate pyridine-Fe(iii)PPIX-drug complex. We propose that formation of the drug-Fe(iii)PPIX coordination complexes is favoured in a non-aqueous environment, such as that found in lipid bodies or membranes in the malaria parasite, and that their existence may contribute to the mechanism of haemozoin inhibition or other toxicity effects that lead ultimately to parasite death. In either case, coordination is a key interaction to be considered in the design of novel antimalarial drug candidates.
Collapse
Affiliation(s)
- Johandie Gildenhuys
- Stellenbosch University, Department of Chemistry and Polymer Science, Private Bag X1, Matieland, 7602, Stellenbosch, South Africa.
| | | | | | | | | | | | | |
Collapse
|
17
|
Wicht KJ, Combrinck JM, Smith PJ, Egan TJ. Bayesian models trained with HTS data for predicting β-haematin inhibition and in vitro antimalarial activity. Bioorg Med Chem 2015; 23:5210-7. [PMID: 25573118 PMCID: PMC4475507 DOI: 10.1016/j.bmc.2014.12.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 11/29/2022]
Abstract
A large quantity of high throughput screening (HTS) data for antimalarial activity has become available in recent years. This includes both phenotypic and target-based activity. Realising the maximum value of these data remains a challenge. In this respect, methods that allow such data to be used for virtual screening maximise efficiency and reduce costs. In this study both in vitro antimalarial activity and inhibitory data for β-haematin formation, largely obtained from publically available sources, has been used to develop Bayesian models for inhibitors of β-haematin formation and in vitro antimalarial activity. These models were used to screen two in silico compound libraries. In the first, the 1510 U.S. Food and Drug Administration approved drugs available on PubChem were ranked from highest to lowest Bayesian score based on a training set of β-haematin inhibiting compounds active against Plasmodium falciparum that did not include any of the clinical antimalarials or close analogues. The six known clinical antimalarials that inhibit β-haematin formation were ranked in the top 2.1% of compounds. Furthermore, the in vitro antimalarial hit-rate for this prioritised set of compounds was found to be 81% in the case of the subset where activity data are available in PubChem. In the second, a library of about 5000 commercially available compounds (Aldrich(CPR)) was virtually screened for ability to inhibit β-haematin formation and then for in vitro antimalarial activity. A selection of 34 compounds was purchased and tested, of which 24 were predicted to be β-haematin inhibitors. The hit rate for inhibition of β-haematin formation was found to be 25% and a third of these were active against P. falciparum, corresponding to enrichments estimated at about 25- and 140-fold relative to random screening, respectively.
Collapse
Affiliation(s)
- Kathryn J Wicht
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Jill M Combrinck
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa; Division of Pharmacology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Peter J Smith
- Division of Pharmacology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.
| |
Collapse
|
18
|
Jones RA, Panda SS, Hall CD. Quinine conjugates and quinine analogues as potential antimalarial agents. Eur J Med Chem 2015; 97:335-55. [PMID: 25683799 DOI: 10.1016/j.ejmech.2015.02.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/08/2015] [Accepted: 02/04/2015] [Indexed: 10/24/2022]
Abstract
Malaria is a tropical disease, prevalent in Southeast Asia and Africa, resulting in over half a million deaths annually; efforts to develop new antimalarial agents are therefore particularly important. Quinine continues to play a role in the fight against malaria, but quinoline derivatives are more widely used. Drugs based on the quinoline scaffold include chloroquine and primaquine, which are able to act against the blood and liver stages of the parasite's life cycle. The purpose of this review is to discuss reported biologically active compounds based on either the quinine or quinoline scaffold that may have enhanced antimalarial activity. The review emphasises hybrid molecules, and covers advances made in the last five years. The review is divided into three sections: modifications to the quinine scaffold, modifications to aminoquinolines and finally metal-containing antimalarial compounds.
Collapse
Affiliation(s)
- Rachel A Jones
- Center for Heterocyclic Compounds, University of Florida, Department of Chemistry, Gainesville, FL 32611-7200, USA.
| | - Siva S Panda
- Center for Heterocyclic Compounds, University of Florida, Department of Chemistry, Gainesville, FL 32611-7200, USA
| | - C Dennis Hall
- Center for Heterocyclic Compounds, University of Florida, Department of Chemistry, Gainesville, FL 32611-7200, USA
| |
Collapse
|
19
|
Teixeira C, Vale N, Pérez B, Gomes A, Gomes JRB, Gomes P. "Recycling" classical drugs for malaria. Chem Rev 2014; 114:11164-220. [PMID: 25329927 DOI: 10.1021/cr500123g] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Cátia Teixeira
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal.,CICECO, Departamento de Química, Universidade de Aveiro , P-3810-193 Aveiro, Portugal
| | - Nuno Vale
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| | - Bianca Pérez
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| | - Ana Gomes
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| | - José R B Gomes
- CICECO, Departamento de Química, Universidade de Aveiro , P-3810-193 Aveiro, Portugal
| | - Paula Gomes
- Centro de Investigação em Química da Universidade do Porto, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto , P-4169-007 Porto, Portugal
| |
Collapse
|
20
|
Sanchez CP, Liu CH, Mayer S, Nurhasanah A, Cyrklaff M, Mu J, Ferdig MT, Stein WD, Lanzer M. A HECT ubiquitin-protein ligase as a novel candidate gene for altered quinine and quinidine responses in Plasmodium falciparum. PLoS Genet 2014; 10:e1004382. [PMID: 24830312 PMCID: PMC4022464 DOI: 10.1371/journal.pgen.1004382] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 04/01/2014] [Indexed: 11/18/2022] Open
Abstract
The emerging resistance to quinine jeopardizes the efficacy of a drug that has been used in the treatment of malaria for several centuries. To identify factors contributing to differential quinine responses in the human malaria parasite Plasmodium falciparum, we have conducted comparative quantitative trait locus analyses on the susceptibility to quinine and also its stereoisomer quinidine, and on the initial and steady-state intracellular drug accumulation levels in the F1 progeny of a genetic cross. These data, together with genetic screens of field isolates and laboratory strains associated differential quinine and quinidine responses with mutated pfcrt, a segment on chromosome 13, and a novel candidate gene, termed MAL7P1.19 (encoding a HECT ubiquitin ligase). Despite a strong likelihood of association, episomal transfections demonstrated a role for the HECT ubiquitin-protein ligase in quinine and quinidine sensitivity in only a subset of genetic backgrounds, and here the changes in IC50 values were moderate (approximately 2-fold). These data show that quinine responsiveness is a complex genetic trait with multiple alleles playing a role and that more experiments are needed to unravel the role of the contributing factors. Quinine, a natural product from cinchona bark, has been used in the treatment of malaria for centuries. Unfortunately, a progressive loss in responsiveness of the human malaria parasite Plasmodium falciparum to quinine has been observed, particularly in Southeast Asia, where cases of quinine treatment failure regularly occur. To better understand how P. falciparum defends itself against the cytotoxic activity of quinine, we have conducted comparative linkage analyses in the F1 progeny of a genetic cross where we assessed the susceptibility and the amount of intracellular accumulation of quinine and of its stereoisomer quinidine. These data identified a novel candidate gene encoding a HECT ubiquitin-protein ligase that might contribute to altered quinine responsiveness. The identification of this novel gene might improve the surveillance of quinine-resistant malaria parasites in the field and aid the preservation of this valuable antimalarial drug.
Collapse
Affiliation(s)
- Cecilia P. Sanchez
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Chia-Hao Liu
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Sybille Mayer
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Astutiati Nurhasanah
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Laboratory for the Development of Agroindustrial and Biomedical Technology (LAPTIAB), Tangerang Selatan, Indonesia
| | - Marek Cyrklaff
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Michael T. Ferdig
- The Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Wilfred D. Stein
- Biological Chemistry, Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Michael Lanzer
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
21
|
Gorka AP, de Dios A, Roepe PD. Quinoline drug-heme interactions and implications for antimalarial cytostatic versus cytocidal activities. J Med Chem 2013; 56:5231-46. [PMID: 23586757 DOI: 10.1021/jm400282d] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Historically, the most successful molecular target for antimalarial drugs has been heme biomineralization within the malarial parasite digestive vacuole. Heme released from catabolized host red blood cell hemoglobin is toxic, so malarial parasites crystallize heme to nontoxic hemozoin. For years it has been accepted that a number of effective quinoline antimalarial drugs (e.g., chloroquine, quinine, amodiaquine) function by preventing hemozoin crystallization. However, recent studies over the past decade have revealed a surprising molecular diversity in quinoline-heme molecular interactions. This diversity shows that even closely related quinoline drugs may have quite different molecular pharmacology. This paper reviews the molecular diversity and highlights important implications for understanding quinoline antimalarial drug resistance and for future drug design.
Collapse
Affiliation(s)
- Alexander P Gorka
- Department of Chemistry, Department of Biochemistry, Cellular, and Molecular Biology, and Center for Infectious Diseases, Georgetown University , 37th and O Streets, NW, Washington, D.C. 20057, United States
| | | | | |
Collapse
|
22
|
Relative to quinine and quinidine, their 9-epimers exhibit decreased cytostatic activity and altered heme binding but similar cytocidal activity versus Plasmodium falciparum. Antimicrob Agents Chemother 2012; 57:365-74. [PMID: 23114754 DOI: 10.1128/aac.01234-12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The 9-epimers of quinine (QN) and quinidine (QD) are known to exhibit poor cytostatic potency against P. falciparum (Karle JM, Karle IL, Gerena L, Milhous WK, Antimicrob. Agents Chemother. 36:1538-1544, 1992). We synthesized 9-epi-QN (eQN) and 9-epi-QD (eQD) via Mitsunobu esterification-saponification and evaluated both cytostatic and cytocidal antimalarial activities. Relative to the cytostatic activity of QN and QD, we observed a large decrease in cytostatic activity (higher 50% inhibitory concentration [IC(50)s]) against QN-sensitive strain HB3, QN-resistant strain Dd2, and QN-hypersensitive strain K76I, consistent with previous work. However, we observed relatively small changes in cytocidal activity (the 50% lethal dose), similar to observations with chloroquine (CQ) analogues with a wide range of IC(50)s (see the accompanying paper [A. P. Gorka, J. N. Alumasa, K. S. Sherlach, L. M. Jacobs, K. B. Nickley, J. P. Brower, A. C. de Dios, and P. D. Roepe, Antimicrob. Agents Chemother. 57:356-364, 2013]). Compared to QN and QD, the 9-epimers had significantly reduced hemozoin inhibition efficiency and did not affect pH-dependent aggregation of ferriprotoporphyrin IX (FPIX) heme. Magnetic susceptibility measurements showed that the 9-epimers perturb FPIX monomer-dimer equilibrium in favor of monomer, and UV-visible (VIS) titrations showed that eQN and eQD bind monomer with similar affinity relative to QN and QD. However, unique ring proton shifts in the presence of zinc(II) protoporphyrin IX (ZnPIX) indicate that binding of the 9-epimers to monomeric heme is via a distinct geometry. We isolated eQN- and eQD-FPIX complexes formed under aqueous conditions and analyzed them by mass, fluorescence, and UV-VIS spectroscopies. The 9-epimers produced low-fluorescent adducts with a 2:1 stoichiometry (drug to FPIX) which did not survive electrospray ionization, in contrast to QN and QD complexes. The data offer important insight into the relevance of heme interactions as a drug target for cytostatic versus cytocidal dosages of quinoline antimalarial drugs and further elucidate a surprising structural diversity of quinoline antimalarial drug-heme complexes.
Collapse
|
23
|
Cytostatic versus cytocidal activities of chloroquine analogues and inhibition of hemozoin crystal growth. Antimicrob Agents Chemother 2012; 57:356-64. [PMID: 23114783 DOI: 10.1128/aac.01709-12] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We report an improved, nonhazardous, high-throughput assay for in vitro quantification of antimalarial drug inhibition of β-hematin (hemozoin) crystallization performed under conditions that are more physiological relative to previous assays. The assay uses the differential detergent solubility of crystalline and noncrystalline forms of heme and is optimized via the use of lipid catalyst. Using this assay, we quantify the effect of pH on the crystal growth-inhibitory activities of current quinoline antimalarials, evaluate the catalytic efficiencies of different lipids, and test for a possible correlation between hemozoin inhibition by drugs versus their antiplasmodial activity. Consistent with several previous reports, we found a good correlation between hemozoin inhibition potency versus cytostatic antiplasmodial potency (50% inhibitory concentration) for a series of chloroquine (CQ) analogues. However, we found no correlation between hemozoin inhibition potency and cytocidal antiplasmodial potency (50% lethal dose) for the same drugs, suggesting that cellular targets for these two layers of 4-aminoquinoline drug activity differ. This important concept is also explored further for QN and its stereoisomers in the accompanying paper (A. P. Gorka, K. S. Sherlach, A. C. de Dios, and P. D. Roepe, Antimicrob. Agents Chemother. 57:365-374, 2013).
Collapse
|
24
|
Haynes RK, Cheu KW, Chan HW, Wong HN, Li KY, Tang MMK, Chen MJ, Guo ZF, Guo ZH, Sinniah K, Witte AB, Coghi P, Monti D. Interactions between artemisinins and other antimalarial drugs in relation to the cofactor model--a unifying proposal for drug action. ChemMedChem 2012; 7:2204-26. [PMID: 23112085 DOI: 10.1002/cmdc.201200383] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 09/30/2012] [Indexed: 01/14/2023]
Abstract
Artemisinins are proposed to act in the malaria parasite cytosol by oxidizing dihydroflavin cofactors of redox-active flavoenzymes, and under aerobic conditions by inducing their autoxidation. Perturbation of redox homeostasis coupled with the generation of reactive oxygen species (ROS) ensues. Ascorbic acid-methylene blue (MB), N-benzyl-1,4-dihydronicotinamide (BNAH)-MB, BNAH-lumiflavine, BNAH-riboflavin (RF), and NADPH-FAD-E. coli flavin reductase (Fre) systems at pH 7.4 generate leucomethylene blue (LMB) and reduced flavins that are rapidly oxidized in situ by artemisinins. These oxidations are inhibited by the 4-aminoquinolines piperaquine (PPQ), chloroquine (CQ), and others. In contrast, the arylmethanols lumefantrine, mefloquine (MFQ), and quinine (QN) have little or no effect. Inhibition correlates with the antagonism exerted by 4-aminoquinolines on the antimalarial activities of MB, RF, and artemisinins. Lack of inhibition correlates with the additivity/synergism between the arylmethanols and artemisinins. We propose association via π complex formation between the 4-aminoquinolines and LMB or the dihydroflavins; this hinders hydride transfer from the reduced conjugates to the artemisinins. The arylmethanols have a decreased tendency to form π complexes, and so exert no effect. The parallel between chemical reactivity and antagonism or additivity/synergism draws attention to the mechanism of action of all drugs described herein. CQ and QN inhibit the formation of hemozoin in the parasite digestive vacuole (DV). The buildup of heme-Fe(III) results in an enhanced efflux from the DV into the cytosol. In addition, the lipophilic heme-Fe(III) complexes of CQ and QN that form in the DV are proposed to diffuse across the DV membrane. At the higher pH of the cytosol, the complexes decompose to liberate heme-Fe(III) . The quinoline or arylmethanol reenters the DV, and so transfers more heme-Fe(III) out of the DV. In this way, the 4-aminoquinolines and arylmethanols exert antimalarial activities by enhancing heme-Fe(III) and thence free Fe(III) concentrations in the cytosol. The iron species enter into redox cycles through reduction of Fe(III) to Fe(II) largely mediated by reduced flavin cofactors and likely also by NAD(P)H-Fre. Generation of ROS through oxidation of Fe(II) by oxygen will also result. The cytotoxicities of artemisinins are thereby reinforced by the iron. Other aspects of drug action are emphasized. In the cytosol or DV, association by π complex formation between pairs of lipophilic drugs must adversely influence the pharmacokinetics of each drug. This explains the antagonism between PPQ and MFQ, for example. The basis for the antimalarial activity of RF mirrors that of MB, wherein it participates in redox cycling that involves flavoenzymes or Fre, resulting in attrition of NAD(P)H. The generation of ROS by artemisinins and ensuing Fenton chemistry accommodate the ability of artemisinins to induce membrane damage and to affect the parasite SERCA PfATP6 Ca(2+) transporter. Thus, the effect exerted by artemisinins is more likely a downstream event involving ROS that will also be modulated by mutations in PfATP6. Such mutations attenuate, but cannot abrogate, antimalarial activities of artemisinins. Overall, parasite resistance to artemisinins arises through enhancement of antioxidant defense mechanisms.
Collapse
Affiliation(s)
- Richard K Haynes
- Department of Chemistry, Institute of Molecular Technology for Drug Discovery and Synthesis, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bohórquez EB, Chua M, Meshnick SR. Quinine localizes to a non-acidic compartment within the food vacuole of the malaria parasite Plasmodium falciparum. Malar J 2012; 11:350. [PMID: 23088166 PMCID: PMC3520729 DOI: 10.1186/1475-2875-11-350] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 10/18/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The naturally fluorescent compound quinine has long been used to treat malaria infections. Although some evidence suggests that quinine acts in the parasite food vacuole, the mechanism of action of quinine has not yet been resolved. The Plasmodium falciparum multidrug resistance (pfmdr1) gene encodes a food vacuolar membrane transporter and has been linked with parasite resistance to quinine. The effect of multiple pfmdr1 copies on the subcellular localization of quinine was explored. METHODS Fluorescence microscopy was used to evaluate the subcellular localization of quinine in parasites containing different pfmdr1 copy numbers to determine if copy number of the gene affects drug localization. The acidotropic dye LysoTracker Red was used to label the parasite food vacuole. Time-lapse images were taken to determine quinine localization over time following quinine exposure. RESULTS Regardless of pfmdr1 copy number, quinine overlapped with haemozoin but did not colocalize with LysoTracker Red, which labeled the acidic parasite food vacuole. CONCLUSIONS Quinine localizes to a non-acidic compartment within the food vacuole possibly haemozoin. Pfmdr1 copy number does not affect quinine subcellular localization.
Collapse
Affiliation(s)
- Elaine B Bohórquez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Campus Box 7435, Chapel Hill, NC 27599-7435, USA
| | | | | |
Collapse
|
26
|
Griffin CE, Hoke JM, Samarakoon U, Duan J, Mu J, Ferdig MT, Warhurst DC, Cooper RA. Mutation in the Plasmodium falciparum CRT protein determines the stereospecific activity of antimalarial cinchona alkaloids. Antimicrob Agents Chemother 2012; 56:5356-64. [PMID: 22869567 PMCID: PMC3457399 DOI: 10.1128/aac.05667-11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 07/30/2012] [Indexed: 11/20/2022] Open
Abstract
The Cinchona alkaloids are quinoline aminoalcohols that occur as diastereomer pairs, typified by (-)-quinine and (+)-quinidine. The potency of (+)-isomers is greater than the (-)-isomers in vitro and in vivo against Plasmodium falciparum malaria parasites. They may act by the inhibition of heme crystallization within the parasite digestive vacuole in a manner similar to chloroquine. Earlier studies showed that a K76I mutation in the digestive vacuole-associated protein, PfCRT (P. falciparum chloroquine resistance transporter), reversed the normal potency order of quinine and quinidine toward P. falciparum. To further explore PfCRT-alkaloid interactions in the malaria parasite, we measured the in vitro susceptibility of eight clonal lines of P. falciparum derived from the 106/1 strain, each containing a unique pfcrt allele, to four Cinchona stereoisomer pairs: quinine and quinidine; cinchonidine and cinchonine; hydroquinine and hydroquinidine; 9-epiquinine and 9-epiquinidine. Stereospecific potency of the Cinchona alkaloids was associated with changes in charge and hydrophobicity of mutable PfCRT amino acids. In isogenic chloroquine-resistant lines, the IC(50) ratio of (-)/(+) CA pairs correlated with side chain hydrophobicity of the position 76 residue. Second-site PfCRT mutations negated the K76I stereospecific effects: charge-change mutations C72R or Q352K/R restored potency patterns similar to the parent K76 line, while V369F increased susceptibility to the alkaloids and nullified stereospecific differences between alkaloid pairs. Interactions between key residues of the PfCRT channel/transporter with (-) and (+) alkaloids are stereospecifically determined, suggesting that PfCRT binding plays an important role in the antimalarial activity of quinine and other Cinchona alkaloids.
Collapse
Affiliation(s)
- Carol E. Griffin
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA
| | - Jonathan M. Hoke
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA
| | - Upeka Samarakoon
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Junhui Duan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Michael T. Ferdig
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - David C. Warhurst
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Roland A. Cooper
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California, USA
| |
Collapse
|
27
|
Dinio T, Gorka AP, McGinniss A, Roepe PD, Morgan JB. Investigating the activity of quinine analogues versus chloroquine resistant Plasmodium falciparum. Bioorg Med Chem 2012; 20:3292-7. [PMID: 22512909 PMCID: PMC3345081 DOI: 10.1016/j.bmc.2012.03.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 03/16/2012] [Accepted: 03/21/2012] [Indexed: 11/23/2022]
Abstract
Plasmodium falciparum, the deadliest malarial parasite species, has developed resistance against nearly all man-made antimalarial drugs within the past century. However, quinine (QN), the first antimalarial drug, remains efficacious worldwide. Some chloroquine resistant (CQR) P. falciparum strains or isolates show mild cross resistance to QN, but many do not. Further optimization of QN may provide a well-tolerated therapy with improved activity versus CQR malaria. Thus, using the Heck reaction, we have pursued a structure-activity relationship study, including vinyl group modifications of QN. Certain derivatives show good antiplasmodial activity in QN-resistant and QN-sensitive strains, with lower IC(50) values relative to QN.
Collapse
Affiliation(s)
- Theresa Dinio
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, Dobo Hall, 601 S. College Road, Wilmington NC 28403
| | - Alexander P. Gorka
- Department of Chemistry, Department of Biochemistry and Cellular and Molecular Biology, and Center for Infectious Disease, Georgetown University, 37 and O Streets, Washington DC 20057
| | - Andrew McGinniss
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, Dobo Hall, 601 S. College Road, Wilmington NC 28403
| | - Paul D. Roepe
- Department of Chemistry, Department of Biochemistry and Cellular and Molecular Biology, and Center for Infectious Disease, Georgetown University, 37 and O Streets, Washington DC 20057
| | - Jeremy B. Morgan
- Department of Chemistry and Biochemistry, University of North Carolina, Wilmington, Dobo Hall, 601 S. College Road, Wilmington NC 28403
| |
Collapse
|
28
|
de Villiers KA, Gildenhuys J, le Roex T. Iron(III) protoporphyrin IX complexes of the antimalarial Cinchona alkaloids quinine and quinidine. ACS Chem Biol 2012; 7:666-71. [PMID: 22276975 DOI: 10.1021/cb200528z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The antimalarial properties of the Cinchona alkaloids quinine and quinidine have been known for decades. Surprisingly, 9-epiquinine and 9-epiquinidine are almost inactive. A lack of definitive structural information has precluded a clear understanding of the relationship between molecular structure and biological activity. In the current study, we have determined by single crystal X-ray diffraction the structures of the complexes formed between quinine and quinidine and iron(III) protoporphyrin IX (Fe(III)PPIX). Coordination of the alkaloid to the Fe(III) center is a key feature of both complexes, and further stability is provided by an intramolecular hydrogen bond formed between a propionate side chain of Fe(III)PPIX and the protonated quinuclidine nitrogen atom of either alkaloid. These interactions are believed to be responsible for inhibiting the incorporation of Fe(III)PPIX into crystalline hemozoin during its in vivo detoxification. It is also possible to rationalize the greater activity of quinidine compared to that of quinine.
Collapse
Affiliation(s)
- Katherine A. de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Stellenbosch
7602, South Africa
| | - Johandie Gildenhuys
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Stellenbosch
7602, South Africa
| | - Tanya le Roex
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Stellenbosch
7602, South Africa
| |
Collapse
|