1
|
Arabiotorre A, Bankaitis VA, Grabon A. Regulation of phosphoinositide metabolism in Apicomplexan parasites. Front Cell Dev Biol 2023; 11:1163574. [PMID: 37791074 PMCID: PMC10543664 DOI: 10.3389/fcell.2023.1163574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/11/2023] [Indexed: 10/05/2023] Open
Abstract
Phosphoinositides are a biologically essential class of phospholipids that contribute to organelle membrane identity, modulate membrane trafficking pathways, and are central components of major signal transduction pathways that operate on the cytosolic face of intracellular membranes in eukaryotes. Apicomplexans (such as Toxoplasma gondii and Plasmodium spp.) are obligate intracellular parasites that are important causative agents of disease in animals and humans. Recent advances in molecular and cell biology of Apicomplexan parasites reveal important roles for phosphoinositide signaling in key aspects of parasitosis. These include invasion of host cells, intracellular survival and replication, egress from host cells, and extracellular motility. As Apicomplexans have adapted to the organization of essential signaling pathways to accommodate their complex parasitic lifestyle, these organisms offer experimentally tractable systems for studying the evolution, conservation, and repurposing of phosphoinositide signaling. In this review, we describe the regulatory mechanisms that control the spatial and temporal regulation of phosphoinositides in the Apicomplexan parasites Plasmodium and T. gondii. We further discuss the similarities and differences presented by Apicomplexan phosphoinositide signaling relative to how these pathways are regulated in other eukaryotic organisms.
Collapse
Affiliation(s)
- Angela Arabiotorre
- Department of Cell Biology and Genetics, College of Medicine Texas A&M Health Sciences Center College Station, Bryan, TX, United States
| | - Vytas A. Bankaitis
- Department of Cell Biology and Genetics, College of Medicine Texas A&M Health Sciences Center College Station, Bryan, TX, United States
- Department of Biochemistry and Biophysics Texas A&M University College Station, College Station, TX, United States
- Department of Chemistry Texas A&M University College Station, College Station, TX, United States
| | - Aby Grabon
- Department of Cell Biology and Genetics, College of Medicine Texas A&M Health Sciences Center College Station, Bryan, TX, United States
| |
Collapse
|
2
|
Zimmermann A, Prieto-Vivas JE, Cautereels C, Gorkovskiy A, Steensels J, Van de Peer Y, Verstrepen KJ. A Cas3-base editing tool for targetable in vivo mutagenesis. Nat Commun 2023; 14:3389. [PMID: 37296137 PMCID: PMC10256805 DOI: 10.1038/s41467-023-39087-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
The generation of genetic diversity via mutagenesis is routinely used for protein engineering and pathway optimization. Current technologies for random mutagenesis often target either the whole genome or relatively narrow windows. To bridge this gap, we developed CoMuTER (Confined Mutagenesis using a Type I-E CRISPR-Cas system), a tool that allows inducible and targetable, in vivo mutagenesis of genomic loci of up to 55 kilobases. CoMuTER employs the targetable helicase Cas3, signature enzyme of the class 1 type I-E CRISPR-Cas system, fused to a cytidine deaminase to unwind and mutate large stretches of DNA at once, including complete metabolic pathways. The tool increases the number of mutations in the target region 350-fold compared to the rest of the genome, with an average of 0.3 mutations per kilobase. We demonstrate the suitability of CoMuTER for pathway optimization by doubling the production of lycopene in Saccharomyces cerevisiae after a single round of mutagenesis.
Collapse
Affiliation(s)
- Anna Zimmermann
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, 3001, Belgium
- Laboratory for Genetics and Genomics, Center of Microbial and Plant Genetics, Department M2S, KU Leuven, Gaston Geenslaan 1, 3001, Leuven, Belgium
| | - Julian E Prieto-Vivas
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, 3001, Belgium
- Laboratory for Genetics and Genomics, Center of Microbial and Plant Genetics, Department M2S, KU Leuven, Gaston Geenslaan 1, 3001, Leuven, Belgium
| | - Charlotte Cautereels
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, 3001, Belgium
- Laboratory for Genetics and Genomics, Center of Microbial and Plant Genetics, Department M2S, KU Leuven, Gaston Geenslaan 1, 3001, Leuven, Belgium
| | - Anton Gorkovskiy
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, 3001, Belgium
- Laboratory for Genetics and Genomics, Center of Microbial and Plant Genetics, Department M2S, KU Leuven, Gaston Geenslaan 1, 3001, Leuven, Belgium
| | - Jan Steensels
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, 3001, Belgium
- Laboratory for Genetics and Genomics, Center of Microbial and Plant Genetics, Department M2S, KU Leuven, Gaston Geenslaan 1, 3001, Leuven, Belgium
| | - Yves Van de Peer
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium.
- VIB Center for Plant Systems Biology, Ghent, Belgium.
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa.
- College of Horticulture, Academy for Advanced Interdisciplinary Studies, Nanjing Agricultural University, 210095, Nanjing, China.
| | - Kevin J Verstrepen
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, 3001, Belgium.
- Laboratory for Genetics and Genomics, Center of Microbial and Plant Genetics, Department M2S, KU Leuven, Gaston Geenslaan 1, 3001, Leuven, Belgium.
| |
Collapse
|
3
|
Chen XR, Poudel L, Hong Z, Johnen P, Katti S, Tripathi A, Nile AH, Green SM, Khan D, Schaaf G, Bono F, Bankaitis VA, Igumenova TI. Mechanisms by which small molecules of diverse chemotypes arrest Sec14 lipid transfer activity. J Biol Chem 2023; 299:102861. [PMID: 36603766 PMCID: PMC9898755 DOI: 10.1016/j.jbc.2022.102861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Phosphatidylinositol (PtdIns) transfer proteins (PITPs) enhance the activities of PtdIns 4-OH kinases that generate signaling pools of PtdIns-4-phosphate. In that capacity, PITPs serve as key regulators of lipid signaling in eukaryotic cells. Although the PITP phospholipid exchange cycle is the engine that stimulates PtdIns 4-OH kinase activities, the underlying mechanism is not understood. Herein, we apply an integrative structural biology approach to investigate interactions of the yeast PITP Sec14 with small-molecule inhibitors (SMIs) of its phospholipid exchange cycle. Using a combination of X-ray crystallography, solution NMR spectroscopy, and atomistic MD simulations, we dissect how SMIs compete with native Sec14 phospholipid ligands and arrest phospholipid exchange. Moreover, as Sec14 PITPs represent new targets for the development of next-generation antifungal drugs, the structures of Sec14 bound to SMIs of diverse chemotypes reported in this study will provide critical information required for future structure-based design of next-generation lead compounds directed against Sec14 PITPs of virulent fungi.
Collapse
Affiliation(s)
- Xiao-Ru Chen
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA
| | - Lokendra Poudel
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA
| | - Zebin Hong
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Philipp Johnen
- Institute for Crop Science and Resource Conservation, Universität Bonn, Bonn, Germany
| | - Sachin Katti
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA
| | - Ashutosh Tripathi
- Department of Cell Biology & Genetics, Texas A&M University, College Station, Texas, USA
| | - Aaron H Nile
- Department of Cell Biology & Genetics, Texas A&M University, College Station, Texas, USA
| | - Savana M Green
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA; Department of Cell Biology & Genetics, Texas A&M University, College Station, Texas, USA
| | - Danish Khan
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA
| | - Gabriel Schaaf
- Institute for Crop Science and Resource Conservation, Universität Bonn, Bonn, Germany
| | - Fulvia Bono
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Vytas A Bankaitis
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA; Department of Cell Biology & Genetics, Texas A&M University, College Station, Texas, USA.
| | - Tatyana I Igumenova
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas USA.
| |
Collapse
|
4
|
Tu J, Liu N, Huang Y, Yang W, Sheng C. Small molecules for combating multidrug-resistant superbug Candida auris infections. Acta Pharm Sin B 2022; 12:4056-4074. [PMID: 36386475 PMCID: PMC9643296 DOI: 10.1016/j.apsb.2022.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/09/2022] [Accepted: 07/25/2022] [Indexed: 01/12/2023] Open
Abstract
Candida auris is emerging as a major global threat to human health. C. auris infections are associated with high mortality due to intrinsic multi-drug resistance. Currently, therapeutic options for the treatment of C. auris infections are rather limited. We aim to provide a comprehensive review of current strategies, drug candidates, and lead compounds in the discovery and development of novel therapeutic agents against C. auris. The drug resistance profiles and mechanisms are briefly summarized. The structures and activities of clinical candidates, drug combinations, antifungal chemosensitizers, repositioned drugs, new targets, and new types of compounds will be illustrated in detail, and perspectives for guiding future research will be provided. We hope that this review will be helpful to prompting the drug development process to combat this fungal pathogen.
Collapse
Affiliation(s)
| | | | - Yahui Huang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wanzhen Yang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
5
|
Kong L, Deng Z, You D. Chemistry and biosynthesis of bacterial polycyclic xanthone natural products. Nat Prod Rep 2022; 39:2057-2095. [PMID: 36083257 DOI: 10.1039/d2np00046f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Covering: up to the end of 2021Bacterial polycyclic xanthone natural products (BPXNPs) are a growing family of natural xanthones featuring a pentangular architecture with various modifications to the tricyclic xanthone chromophore. Their structural diversities and various activities have fueled biosynthetic and chemical synthetic studies. Moreover, their more potent activities than the clinically used drugs make them potential candidates for the treatment of diseases. Future unraveling of structure activity relationships (SARs) will provide new options for the (bio)-synthesis of drug analogues with higher activities. This review summarizes the isolation, structural elucidation and biological activities and more importantly, the recent strategies for the microbial biosynthesis and chemical synthesis of BPXNPs. Regarding their biosynthesis, we discuss the recent progress in enzymes that synthesize tricyclic xanthone, the protein candidates for structural moieties (methylene dioxygen bridge and nitrogen heterocycle), tailoring enzymes for methylation and halogenation. The chemical synthesis part summarizes the recent methodology for the division synthesis and coupling construction of achiral molecular skeletons. Ultimately, perspectives on the biosynthetic study of BPXNPs are discussed.
Collapse
Affiliation(s)
- Lingxin Kong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Delin You
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
6
|
Bankaitis VA, Tripathi A, Chen XR, Igumenova TI. New strategies for combating fungal infections: Inhibiting inositol lipid signaling by targeting Sec14 phosphatidylinositol transfer proteins. Adv Biol Regul 2022; 84:100891. [PMID: 35240534 PMCID: PMC9149032 DOI: 10.1016/j.jbior.2022.100891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 12/13/2022]
Abstract
Virulent fungi represent a particularly difficult problem in the infectious disease arena as these organisms are eukaryotes that share many orthologous activities with their human hosts. The fact that these activities are often catalyzed by conserved proteins places additional demands on development of pharmacological strategies for specifically inhibiting target fungal activities without imposing undesirable secondary effects on the host. While deployment of a limited set of anti-mycotics has to date satisfied the clinical needs for treatment of fungal infections, the recent emergence of multi-drug resistant fungal 'superbugs' now poses a serious global health threat with rapidly diminishing options for treatment. This escalating infectious disease problem emphasizes the urgent need for development of new classes of anti-mycotics. In that regard, Sec14 phosphatidylinositol transfer proteins offer interesting possibilities for interfering with fungal phosphoinositide signaling with exquisite specificity and without targeting the highly conserved lipid kinases responsible for phosphoinositide production. Herein, we review the establishment of proof-of-principle that demonstrates the feasibility of such an approach. We also describe the lead compounds of four chemotypes that directly target fungal Sec14 proteins. The rules that pertain to the mechanism(s) of Sec14 inhibition by validated small molecule inhibitors, and the open questions that remain, are discussed - as are the challenges that face development of next generation Sec14-directed inhibitors.
Collapse
Affiliation(s)
- Vytas A Bankaitis
- Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, 77843-0014, USA; Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA.
| | - Ashutosh Tripathi
- Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, 77843-0014, USA
| | - Xiao-Ru Chen
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA
| | - Tatyana I Igumenova
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA
| |
Collapse
|
7
|
Ihsan A, Khera RA, Iqbal J, Asgher M. Binding interaction of benzamide derivatives as inhibitors of DNA gyrase and Sec14p using Molegro Virtual Docker based on binding free energy. Z PHYS CHEM 2022. [DOI: 10.1515/zpch-2021-3119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The docking simulation of benzamide derivatives as ligands and protein targets (DNA–gyrase) was performed and Sec14p binding mode interaction was predicted based on binding free energy analysis. Software Molegro Virtual Docking (MVD) was used to visualize the ligand–protein binding interactions. The results indicated the prevalence of steric or hydrophobic interactions among all the benzamide ligands besides hydrogen bonding or electrostatic interactions. The compounds B2, B4 against DNA gyrase, and compounds B3, B5 against Sec14p showed an uncompetitive pattern of inhibition as compared with the reference molecule. While compounds B1, B5 exhibited the best MolDock scores, i.e., −109.736 and −114.391 kcal/mol respectively for DNA gyrase, also compounds B1 and B2 against Sec14p displayed −100.105 and −119.451 kcal/mol sequentially. It was evident from the comparison of MolDock score for both the bacterial and fungal protein receptors that all the ligands were found to be more potent against DNA gyrase than Sec14p. However, only compound B2 with MolDock score −119.451 kcal/mol showed exceptional activity against Sec14p and was predicted to have potency as a lead compound to find a new anti-fungal therapeutic agent. Docking studies further highlighted the unique interactions such as tail-end hydrophobic rings of benzamide inhibitors with catalytically important amino acid residues, allowing flexibility in binding to both the receptors different from other inhibitors. These findings showed us that B1, B2 against Staphylococcus aureus and B5 against Saccharomyces cerevisiae could be leading compounds to discover new multidrug-resistant strains.
Collapse
Affiliation(s)
- Anaum Ihsan
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Rasheed Ahmad Khera
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Javed Iqbal
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Muhammad Asgher
- Department of Biochemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| |
Collapse
|
8
|
Emerging Prospects for Combating Fungal Infections by Targeting Phosphatidylinositol Transfer Proteins. Int J Mol Sci 2021; 22:ijms22136754. [PMID: 34201733 PMCID: PMC8269425 DOI: 10.3390/ijms22136754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/27/2022] Open
Abstract
The emergence of fungal “superbugs” resistant to the limited cohort of anti-fungal agents available to clinicians is eroding our ability to effectively treat infections by these virulent pathogens. As the threat of fungal infection is escalating worldwide, this dwindling response capacity is fueling concerns of impending global health emergencies. These developments underscore the urgent need for new classes of anti-fungal drugs and, therefore, the identification of new targets. Phosphoinositide signaling does not immediately appear to offer attractive targets due to its evolutionary conservation across the Eukaryota. However, recent evidence argues otherwise. Herein, we discuss the evidence identifying Sec14-like phosphatidylinositol transfer proteins (PITPs) as unexplored portals through which phosphoinositide signaling in virulent fungi can be chemically disrupted with exquisite selectivity. Recent identification of lead compounds that target fungal Sec14 proteins, derived from several distinct chemical scaffolds, reveals exciting inroads into the rational design of next generation Sec14 inhibitors. Development of appropriately refined next generation Sec14-directed inhibitors promises to expand the chemical weaponry available for deployment in the shifting field of engagement between fungal pathogens and their human hosts.
Collapse
|
9
|
Heard SC, Wu G, Winter JM. Antifungal natural products. Curr Opin Biotechnol 2021; 69:232-241. [PMID: 33640596 DOI: 10.1016/j.copbio.2021.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 12/19/2022]
Abstract
Natural products are specialized small molecules produced in Nature and play pivotal roles in many cellular processes. These compounds possess exquisite chemical diversity and represent some of the most important pharmaceutical agents in human health care. With the rampant rise of fungal pathogens that are becoming resistant to nearly all clinically available antibiotics, there is an increased urgency to find new antifungal therapies with novel modes of action. To meet this need, we must be able to quickly identify new bioactive chemical scaffolds within complex natural extracts, determine their mechanisms of action, and generate appreciable yields for preclinical studies. In this review, we will highlight naturally derived antifungal agents of clinical importance as well as those with strong potential as leads in drug development.
Collapse
Affiliation(s)
- Stephanie C Heard
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| | - Guangwei Wu
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA; Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, and Jiangsu Key Lab of Biomass-Based Green Fuels and Chemicals, Nanjing Forestry University, Nanjing 210037, China.
| | - Jaclyn M Winter
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
10
|
Zhang F, Zhao M, Braun DR, Ericksen SS, Piotrowski JS, Nelson J, Peng J, Ananiev GE, Chanana S, Barns K, Fossen J, Sanchez H, Chevrette MG, Guzei IA, Zhao C, Guo L, Tang W, Currie CR, Rajski SR, Audhya A, Andes DR, Bugni TS. A marine microbiome antifungal targets urgent-threat drug-resistant fungi. Science 2020; 370:974-978. [PMID: 33214279 PMCID: PMC7756952 DOI: 10.1126/science.abd6919] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/29/2022]
Abstract
New antifungal drugs are urgently needed to address the emergence and transcontinental spread of fungal infectious diseases, such as pandrug-resistant Candida auris. Leveraging the microbiomes of marine animals and cutting-edge metabolomics and genomic tools, we identified encouraging lead antifungal molecules with in vivo efficacy. The most promising lead, turbinmicin, displays potent in vitro and mouse-model efficacy toward multiple-drug-resistant fungal pathogens, exhibits a wide safety index, and functions through a fungal-specific mode of action, targeting Sec14 of the vesicular trafficking pathway. The efficacy, safety, and mode of action distinct from other antifungal drugs make turbinmicin a highly promising antifungal drug lead to help address devastating global fungal pathogens such as C. auris.
Collapse
Affiliation(s)
- Fan Zhang
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Miao Zhao
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Doug R Braun
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Spencer S Ericksen
- Small Molecule Screening Facility, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | | | | | - Jian Peng
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Gene E Ananiev
- Small Molecule Screening Facility, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Shaurya Chanana
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Kenneth Barns
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Jen Fossen
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Hiram Sanchez
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Marc G Chevrette
- Department of Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery and Department of Plant Pathology, University of Wisconsin-Madison, Madison, WI, USA
| | - Ilia A Guzei
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Changgui Zhao
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Le Guo
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Weiping Tang
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Cameron R Currie
- Department of Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott R Rajski
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA
| | - Anjon Audhya
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - David R Andes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
11
|
Zhu Y, Zou R, Sha H, Lu Y, Zhang Y, Wu J, Feng J, Wang D. Lipid metabolism-related proteins of relevant evolutionary and lymphoid interest (PRELI) domain containing family proteins in cancer. Am J Transl Res 2020; 12:6015-6026. [PMID: 33194011 PMCID: PMC7653579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/11/2020] [Indexed: 06/11/2023]
Abstract
Metabolic reprogramming of tumor cells plays a critical role in the tumor microenvironment, including disorder of lipid metabolism. Recently, lipid metabolism has received increasing attention in cancer research. The proteins of relevant evolutionary and lymphoid interest (PRELI) domain containing family contains 6 proteins. Functionally, the PRELI-like family proteins were mainly involved in mitochondrial lipid transport and correlated with several types of diseases and malignant tumors. Here we review current knowledge of the functions, structures, biological functions and underlying mechanisms of the PRELI-like family proteins in cancer progression, which provide insights into the clinical translational application.
Collapse
Affiliation(s)
- Yue Zhu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Renrui Zou
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Huanhuan Sha
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Ya Lu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Yuan Zhang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Jianzhong Wu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| | - Dongfeng Wang
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research Nanjing, Jiangsu, China
| |
Collapse
|
12
|
Parthasarathy A, Mantravadi PK, Kalesh K. Detectives and helpers: Natural products as resources for chemical probes and compound libraries. Pharmacol Ther 2020; 216:107688. [PMID: 32980442 DOI: 10.1016/j.pharmthera.2020.107688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
About 70% of the drugs in use are derived from natural products, either used directly or in chemically modified form. Among all possible small molecules (not greater than 5 kDa), only a few of them are biologically active. Natural product libraries may have a higher rate of finding "hits" than synthetic libraries, even with the use of fewer compounds. This is due to the complementarity between the "chemical space" of small molecules and biological macromolecules such as proteins, DNA and RNA, in addition to the three-dimensional complexity of NPs. Chemical probes are molecules which aid in the elucidation of the biological mechanisms behind the action of drugs or drug-like molecules by binding with macromolecular/cellular interaction partners. Probe development and application have been spurred by advancements in photoaffinity label synthesis, affinity chromatography, activity based protein profiling (ABPP) and instrumental methods such as cellular thermal shift assay (CETSA) and advanced/hyphenated mass spectrometry (MS) techniques, as well as genome sequencing and bioengineering technologies. In this review, we restrict ourselves to a survey of natural products (including peptides/mini-proteins and excluding antibodies), which have been applied largely in the last 5 years for the target identification of drugs/drug-like molecules used in research on infectious diseases, and the description of their mechanisms of action.
Collapse
Affiliation(s)
- Anutthaman Parthasarathy
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, 85 Lomb Memorial Dr, Rochester, NY 14623, USA
| | | | - Karunakaran Kalesh
- Department of Chemistry, Durham University, Lower Mount Joy, South Road, Durham DH1 3LE, UK.
| |
Collapse
|
13
|
Tranter D, Filipuzzi I, Lochmann T, Knapp B, Kellosalo J, Estoppey D, Pistorius D, Meissner A, Paavilainen VO, Hoepfner D. Kendomycin Cytotoxicity against Bacterial, Fungal, and Mammalian Cells Is Due to Cation Chelation. JOURNAL OF NATURAL PRODUCTS 2020; 83:965-971. [PMID: 32182062 PMCID: PMC7497661 DOI: 10.1021/acs.jnatprod.9b00826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Indexed: 06/10/2023]
Abstract
Kendomycin is a small-molecule natural product that has gained significant attention due to reported cytotoxicity against pathogenic bacteria and fungi as well as a number of cancer cell lines. Despite significant biomedical interest and attempts to reveal its mechanism of action, the cellular target of kendomycin remains disputed. Herein it is shown that kendomycin induces cellular responses indicative of cation stress comparable to the effects of established iron chelators. Furthermore, addition of excess iron and copper attenuated kendomycin cytotoxicity in bacteria, yeast, and mammalian cells. Finally, NMR analysis demonstrated a direct interaction with cations, corroborating a close link between the observed kendomycin polypharmacology across different species and modulation of iron and/or copper levels.
Collapse
Affiliation(s)
- Dale Tranter
- Institute
of Biotechnology, University of Helsinki, Helsinki, 00014, Finland
| | - Ireos Filipuzzi
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Thomas Lochmann
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Britta Knapp
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Juho Kellosalo
- Institute
of Biotechnology, University of Helsinki, Helsinki, 00014, Finland
| | - David Estoppey
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Dominik Pistorius
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Axel Meissner
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| | | | - Dominic Hoepfner
- Novartis
Institutes for BioMedical Research, 4056 Basel, Switzerland
| |
Collapse
|
14
|
Abstract
Invasive fungal diseases pose a serious threat, and new drugs are urgently needed. In this issue of Cell Chemical Biology, Pries et al. (2018) identified benzamide- and picolinamide-based small-molecule inhibitors with antifungal properties, including some active against pathogenic Candida species. These compounds target an essential component of the fungal secretion machinery, suggesting a new approach to antifungal development.
Collapse
Affiliation(s)
- Julianne Teresa Djordjevic
- Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Sydney Medical School - Westmead, University of Sydney, Westmead, NSW 2145, Australia; Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, NSW, Australia.
| | - Sophie Lev
- Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Sydney Medical School - Westmead, University of Sydney, Westmead, NSW 2145, Australia; Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, NSW, Australia
| |
Collapse
|
15
|
Luo B, Li D, Zhang AL, Gao JM. Synthesis, Antifungal Activities and Molecular Docking Studies of Benzoxazole and Benzothiazole Derivatives. Molecules 2018; 23:molecules23102457. [PMID: 30257495 PMCID: PMC6222379 DOI: 10.3390/molecules23102457] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 11/16/2022] Open
Abstract
Based on benzoxazole and benzothiazole scaffold as an important pharmacophore, two series of 2-(aryloxymethyl) benzoxazole and benzothiazole derivatives were synthesized and their antifungal effects against eight phytopathogenic fungi were evaluated. Compounds 5a, 5b, 5h, and 5i exhibited significant antifungal activities against most of the pathogens tested. Especially 5a, 5b, 5h, 5i, 5j, and 6h inhibited the growth of F. solani with IC50 of 4.34⁻17.61 μg/mL, which were stronger than that of the positive control, hymexazol (IC50 of 38.92 μg/mL). 5h was the most potent inhibitor (IC50 of 4.34 μg/mL) against F. Solani, which was about nine times more potent than hymexazol. Most of the test compounds displayed significant antifungal effects against B. cinerea (IC50 of 19.92⁻77.41 μg/mL), among them, 5a was the best one (IC50 of 19.92 μg/mL). The structure-activity relationships (SARs) were compared and analyzed. The result indicates that the electron-drawing ability and position of the substituents have a significant impact on biological activities. Furthermore, docking studies were carried out on the lipid transfer protein sec14p from S. cerevisiae, and preliminarily verified the antifungal activities. Taken together, these results provide 2-(phenoxymethyl)benzo[d]oxazole as an encouraging framework that could lead to the development of potent novel antifungal agents.
Collapse
Affiliation(s)
- Bo Luo
- College of Life Sciences, Xinyang Normal University, Tea Plant Biology Key Laboratory of Henan Province, Xinyang 464000, China.
- Shaanxi Key Labotory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China.
| | - Ding Li
- Shaanxi Key Labotory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China.
| | - An-Ling Zhang
- Shaanxi Key Labotory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China.
| | - Jin-Ming Gao
- Shaanxi Key Labotory of Natural Products & Chemical Biology, Shaanxi Engineering Center of Bioresource Chemistry & Sustainable Utilization, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
16
|
Diaz P, Horne E, Xu C, Hamel E, Wagenbach M, Petrov RR, Uhlenbruck B, Haas B, Hothi P, Wordeman L, Gussio R, Stella N. Modified carbazoles destabilize microtubules and kill glioblastoma multiform cells. Eur J Med Chem 2018; 159:74-89. [PMID: 30268825 DOI: 10.1016/j.ejmech.2018.09.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 11/26/2022]
Abstract
Small molecules that target microtubules (MTs) represent promising therapeutics to treat certain types of cancer, including glioblastoma multiform (GBM). We synthesized modified carbazoles and evaluated their antitumor activity in GBM cells in culture. Modified carbazoles with an ethyl moiety linked to the nitrogen of the carbazole and a carbonyl moiety linked to distinct biaromatic rings exhibited remarkably different killing activities in human GBM cell lines and patient-derived GBM cells, with IC50 values from 67 to >10,000 nM. Measures of the activity of modified carbazoles with tubulin and microtubules coupled to molecular docking studies show that these compounds bind to the colchicine site of tubulin in a unique low interaction space that inhibits tubulin assembly. The modified carbazoles reported here represent novel chemical tools to better understand how small molecules disrupt MT functions and kill devastating cancers such as GBM.
Collapse
Affiliation(s)
- Philippe Diaz
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA; DermaXon LLC, 32 Campus Drive, Missoula, MT, 59812, USA.
| | - Eric Horne
- Stella Therapeutics, Inc., Pacific Northwest Research Institute, 720 Broadway, Seattle, WA, 98122, USA
| | - Cong Xu
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Ernest Hamel
- Screening Technologies Branch (EH) and Computational Drug Development Group (RG), Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Michael Wagenbach
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Ravil R Petrov
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Benjamin Uhlenbruck
- Department of Biomedical and Pharmaceutical Sciences, The University of Montana, 32 Campus Drive, Missoula, MT, 59812, USA
| | - Brian Haas
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Parvinder Hothi
- Ivy Center for Advance Brain Tumor Treatment, Swedish Neuroscience Institute, 550 17th Ave, Seattle, WA, 98122, USA
| | - Linda Wordeman
- Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA
| | - Rick Gussio
- Screening Technologies Branch (EH) and Computational Drug Development Group (RG), Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Nephi Stella
- Stella Therapeutics, Inc., Pacific Northwest Research Institute, 720 Broadway, Seattle, WA, 98122, USA; Department of Pharmacology (CX, BH and NS), Department of Physiology and Biophysics (MW and LW), Department of Psychiatry and Behavioral Sciences (NS), The University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
17
|
Target Identification and Mechanism of Action of Picolinamide and Benzamide Chemotypes with Antifungal Properties. Cell Chem Biol 2018; 25:279-290.e7. [PMID: 29307839 DOI: 10.1016/j.chembiol.2017.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/18/2017] [Accepted: 12/06/2017] [Indexed: 11/20/2022]
Abstract
Invasive fungal infections are accompanied by high mortality rates that range up to 90%. At present, only three different compound classes are available for use in the clinic, and these often suffer from low bioavailability, toxicity, and drug resistance. These issues emphasize an urgent need for novel antifungal agents. Herein, we report the identification of chemically versatile benzamide and picolinamide scaffolds with antifungal properties. Chemogenomic profiling and biochemical assays with purified protein identified Sec14p, the major phosphatidylinositol/phosphatidylcholine transfer protein in Saccharomyces cerevisiae, as the sole essential target for these compounds. A functional variomics screen identified resistance-conferring residues that localized to the lipid-binding pocket of Sec14p. Determination of the X-ray co-crystal structure of a Sec14p-compound complex confirmed binding in this cavity and rationalized both the resistance-conferring residues and the observed structure-activity relationships. Taken together, these findings open new avenues for rational compound optimization and development of novel antifungal agents.
Collapse
|
18
|
Liu N, Tu J, Dong G, Wang Y, Sheng C. Emerging New Targets for the Treatment of Resistant Fungal Infections. J Med Chem 2018; 61:5484-5511. [DOI: 10.1021/acs.jmedchem.7b01413] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Na Liu
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Jie Tu
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Guoqiang Dong
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Yan Wang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, People’s Republic of China
| |
Collapse
|
19
|
Filipuzzi I, Thomas JR, Pries V, Estoppey D, Salcius M, Studer C, Schirle M, Hoepfner D. Direct Interaction of Chivosazole F with Actin Elicits Cell Responses Similar to Latrunculin A but Distinct from Chondramide. ACS Chem Biol 2017; 12:2264-2269. [PMID: 28796488 DOI: 10.1021/acschembio.7b00385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The microbial metabolite Chivosazole F has been described to affect the cytoskeleton and to inhibit actin polymerization in vitro. Applying orthogonal genomic and proteomics approaches, we now show for the first time that Chivosazole F exerts its effect by directly interacting with actin and demonstrate the cellular impact of Chivosazole F in an unbiased, genome-wide context in yeast and in mammalian cells. Furthermore, mutation-based resistance mapping identifies two SNPs located in the putative Chivosazole F binding site of actin. Comparing chemogenomic profiles and responses to the Chivosazole F-resistant SNPs shows a partially conserved mechanism of action for Chivosazole F and Latrunculin A, but clear divergence from Chondramide. In addition, C14orf80 is an evolutionarily highly conserved ORF, lacking any functional annotation. As editing of C14orf80 leads to Chivosazole F hyper-resistance, we propose a function for this gene product in counteracting perturbation of actin filaments.
Collapse
Affiliation(s)
- Ireos Filipuzzi
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum
1 Novartis Campus, CH-4056 Basel, Switzerland
| | - Jason Ray Thomas
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Verena Pries
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum
1 Novartis Campus, CH-4056 Basel, Switzerland
| | - David Estoppey
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum
1 Novartis Campus, CH-4056 Basel, Switzerland
| | - Michael Salcius
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Christian Studer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum
1 Novartis Campus, CH-4056 Basel, Switzerland
| | - Markus Schirle
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Dominic Hoepfner
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum
1 Novartis Campus, CH-4056 Basel, Switzerland
| |
Collapse
|
20
|
Two low complexity ultra-high throughput methods to identify diverse chemically bioactive molecules using Saccharomyces cerevisiae. Microbiol Res 2017; 199:10-18. [DOI: 10.1016/j.micres.2017.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/17/2017] [Accepted: 02/19/2017] [Indexed: 11/21/2022]
|