1
|
Chen F, Jiang Y, Xu Z, Zhao D, Li D, Yang H, Zhu S, Xu H, Peng S, Miao Z, Wang H, Tong M, Hou Y, Zhao Y. Design, synthesis and evaluation of novel LpxC inhibitors containing a hydrazone moiety as Gram-negative antibacterial agents. Eur J Med Chem 2024; 279:116892. [PMID: 39341094 DOI: 10.1016/j.ejmech.2024.116892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/31/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
LpxC inhibitors are new-type antibacterial agents developed in the last twenty years, mainly against Gram-negative bacteria infections. To enable the development of novel LpxC inhibitors with potent antibacterial activities, several series of compounds were designed and synthesized and their antibacterial activities were evaluated against E. coli ATCC25922, P. aeruginosa ATCC27853, P. aeruginosa clinical isolate PAE 22-1, K. pneumoniae ATCC700603, K. pneumoniae clinical isolate KPN+22-1 in vitro. Compound 6i exhibited significant antibacterial activities against above five Gram-negative bacteria except P. aeruginosa ATCC27853. Moreover, compound 6i exhibited moderate liver microsomal stability and a promising pharmacokinetic profile (AUC0-t = 1050 ng h mL-1, oral bioavailability of 13.3 %) in Sprague-Dawley rats, acceptable PPB, low risk of drug-drug interactions and non-cytotoxic activity against hepatic cell. Collectively, compound 6i could be a promising Gram-negative antibacterial agent for further investigation.
Collapse
Affiliation(s)
- Fei Chen
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Yufeng Jiang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Zidong Xu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Dong Zhao
- Yangtze River Pharmaceutical Group JiangSu Haici Biological Pharmaceutical Co., Ltd., No. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou, Jiangsu, 225326, China
| | - Dan Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Huiyuan Yang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Shenghong Zhu
- Yangtze River Pharmaceutical Group JiangSu Haici Biological Pharmaceutical Co., Ltd., No. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou, Jiangsu, 225326, China
| | - Haoyu Xu
- Yangtze River Pharmaceutical Group Co., Ltd., No. 1 South Yangtze River Road, Taizhou, Jiangsu, 225321, China
| | - Shan Peng
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou, 215104, China
| | - Zhenyu Miao
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou, 215104, China
| | - Han Wang
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou, 215104, China
| | - Minghui Tong
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou, 215104, China
| | - Yunlei Hou
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China.
| | - Yanfang Zhao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
2
|
Mielniczuk S, Hoff K, Baselious F, Li Y, Haupenthal J, Kany AM, Riedner M, Rohde H, Rox K, Hirsch AKH, Krimm I, Sippl W, Holl R. Development of Fragment-Based Inhibitors of the Bacterial Deacetylase LpxC with Low Nanomolar Activity. J Med Chem 2024; 67:17363-17391. [PMID: 39303295 PMCID: PMC11472313 DOI: 10.1021/acs.jmedchem.4c01262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
In a fragment-based approach using NMR spectroscopy, benzyloxyacetohydroxamic acid-derived inhibitors of the bacterial deacetylase LpxC bearing a substituent to target the uridine diphosphate-binding site of the enzyme were developed. By appending privileged fragments via a suitable linker, potent LpxC inhibitors with promising antibacterial activities could be obtained, like the one-digit nanomolar LpxC inhibitor (S)-13j [Ki (EcLpxC C63A) = 9.5 nM; Ki (PaLpxC): 5.6 nM]. To rationalize the observed structure-activity relationships, molecular docking and molecular dynamics studies were performed. Initial in vitro absorption-distribution-metabolism-excretion-toxicity (ADMET) studies of the most potent compounds have paved the way for multiparameter optimization of our newly developed isoserine-based amides.
Collapse
Affiliation(s)
- Sebastian Mielniczuk
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| | - Katharina Hoff
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| | - Fady Baselious
- Institute
of Pharmacy, Martin-Luther-University of
Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120 Halle (Saale), Germany
| | - Yunqi Li
- Team
“Small Molecules for Biological Targets”, Institut Convergence
Plascan, Centre de Recherche en Cancérologie de Lyon, INSERM
U1052-CNRS UMR5286, Centre Léon Bérard, Université
de Lyon, Université Claude Bernard
Lyon1, 69008 Lyon, France
- Shanghai
Key Laboratory of Regulatory Biology, The Institute of Biomedical
Sciences & School of Life Sciences, East China Normal University, 200241 Shanghai, China
| | - Jörg Haupenthal
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Andreas M. Kany
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Maria Riedner
- Technology
Platform Mass Spectrometry, Universität
Hamburg, Mittelweg 177, 20148 Hamburg, Germany
| | - Holger Rohde
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
- Institute
of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Anna K. H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Helmholtz International Lab for Anti-infectives, Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Isabelle Krimm
- Team
“Small Molecules for Biological Targets”, Institut Convergence
Plascan, Centre de Recherche en Cancérologie de Lyon, INSERM
U1052-CNRS UMR5286, Centre Léon Bérard, Université
de Lyon, Université Claude Bernard
Lyon1, 69008 Lyon, France
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther-University of
Halle-Wittenberg, Kurt-Mothes-Straße 3, 06120 Halle (Saale), Germany
| | - Ralph Holl
- Institute
of Organic Chemistry, Universität
Hamburg, Martin-Luther-King-Platz
6, 20146 Hamburg, Germany
- German
Center for Infection Research (DZIF), Partner
Site Hamburg-Lübeck-Borstel-Riems, 20146 Hamburg, Germany
| |
Collapse
|
3
|
Ostroumova OS, Efimova SS. Lipid-Centric Approaches in Combating Infectious Diseases: Antibacterials, Antifungals and Antivirals with Lipid-Associated Mechanisms of Action. Antibiotics (Basel) 2023; 12:1716. [PMID: 38136750 PMCID: PMC10741038 DOI: 10.3390/antibiotics12121716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
One of the global challenges of the 21st century is the increase in mortality from infectious diseases against the backdrop of the spread of antibiotic-resistant pathogenic microorganisms. In this regard, it is worth targeting antibacterials towards the membranes of pathogens that are quite conservative and not amenable to elimination. This review is an attempt to critically analyze the possibilities of targeting antimicrobial agents towards enzymes involved in pathogen lipid biosynthesis or towards bacterial, fungal, and viral lipid membranes, to increase the permeability via pore formation and to modulate the membranes' properties in a manner that makes them incompatible with the pathogen's life cycle. This review discusses the advantages and disadvantages of each approach in the search for highly effective but nontoxic antimicrobial agents. Examples of compounds with a proven molecular mechanism of action are presented, and the types of the most promising pharmacophores for further research and the improvement of the characteristics of antibiotics are discussed. The strategies that pathogens use for survival in terms of modulating the lipid composition and physical properties of the membrane, achieving a balance between resistance to antibiotics and the ability to facilitate all necessary transport and signaling processes, are also considered.
Collapse
Affiliation(s)
- Olga S. Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, St. Petersburg 194064, Russia;
| | | |
Collapse
|
4
|
Zhao J, Cochrane CS, Najeeb J, Gooden D, Sciandra C, Fan P, Lemaitre N, Newns K, Nicholas RA, Guan Z, Thaden JT, Fowler VG, Spasojevic I, Sebbane F, Toone EJ, Duncan C, Gammans R, Zhou P. Preclinical safety and efficacy characterization of an LpxC inhibitor against Gram-negative pathogens. Sci Transl Med 2023; 15:eadf5668. [PMID: 37556556 PMCID: PMC10785772 DOI: 10.1126/scitranslmed.adf5668] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
The UDP-3-O-(R-3-hydroxyacyl)-N-acetylglucosamine deacetylase LpxC is an essential enzyme in the biosynthesis of lipid A, the outer membrane anchor of lipopolysaccharide and lipooligosaccharide in Gram-negative bacteria. The development of LpxC-targeting antibiotics toward clinical therapeutics has been hindered by the limited antibiotic profile of reported non-hydroxamate inhibitors and unexpected cardiovascular toxicity observed in certain hydroxamate and non-hydroxamate-based inhibitors. Here, we report the preclinical characterization of a slow, tight-binding LpxC inhibitor, LPC-233, with low picomolar affinity. The compound is a rapid bactericidal antibiotic, unaffected by established resistance mechanisms to commercial antibiotics, and displays outstanding activity against a wide range of Gram-negative clinical isolates in vitro. It is orally bioavailable and efficiently eliminates infections caused by susceptible and multidrug-resistant Gram-negative bacterial pathogens in murine soft tissue, sepsis, and urinary tract infection models. It displays exceptional in vitro and in vivo safety profiles, with no detectable adverse cardiovascular toxicity in dogs at 100 milligrams per kilogram. These results establish the feasibility of developing oral LpxC-targeting antibiotics for clinical applications.
Collapse
Affiliation(s)
- Jinshi Zhao
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Javaria Najeeb
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
- Current address: Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David Gooden
- Department of Chemistry, Duke University, Durham, NC 27708, USA
- Small Molecule Synthesis Facility, Duke University, Durham, NC 27708, USA
| | - Carly Sciandra
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ping Fan
- Pharmacokinetics/Pharmacodynamics (PK/PD) Core Laboratory, Duke Cancer Institute, Durham, NC 27710, USA
| | - Nadine Lemaitre
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d’Infection et d’Immunité de Lille, Lille, France
| | - Kate Newns
- Departments of Pharmacology and Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
- Current address: Envision Pharma Group, Philadelphia, PA 19109, USA
| | - Robert A. Nicholas
- Departments of Pharmacology and Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Ziqiang Guan
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joshua T. Thaden
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Vance G. Fowler
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ivan Spasojevic
- Pharmacokinetics/Pharmacodynamics (PK/PD) Core Laboratory, Duke Cancer Institute, Durham, NC 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Florent Sebbane
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d’Infection et d’Immunité de Lille, Lille, France
| | - Eric J. Toone
- Department of Chemistry, Duke University, Durham, NC 27708, USA
- Current address: Breakthrough Energy Ventures, 4110 Carillon Point Kirkland, WA 98033 USA
| | | | | | - Pei Zhou
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Chemistry, Duke University, Durham, NC 27708, USA
| |
Collapse
|
5
|
Kumar Pal S, Kumar S. LpxC (UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase) inhibitors: A long path explored for potent drug design. Int J Biol Macromol 2023; 234:122960. [PMID: 36565833 DOI: 10.1016/j.ijbiomac.2022.12.179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Microbial infections are becoming resistant to traditional antibiotics. As novel resistance mechanisms are developed and disseminated across the world, our ability to treat the most common infectious diseases is becoming increasingly compromised. As existing antibiotics are losing their effectiveness, especially treatment of bacterial infections, is difficult. In order to combat this issue, it is of utmost importance to identify novel pharmacological targets or antibiotics. LpxC, a zinc-dependent metalloamidase that catalyzes the committed step in the biosynthesis of lipid A (endotoxin) in bacteria, is a prime candidate for drug/therapeutic target. So far, the rate-limiting metallo-amidase LpxC has been the most-targeted macromolecule in the Raetz pathway. This is because it is important for the growth of these bacterial infections. This review showcases on the research done to develop efficient drugs in this area before and after the 2015.
Collapse
Affiliation(s)
- Sudhir Kumar Pal
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Sanjit Kumar
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
6
|
Niu Z, Lei P, Wang Y, Wang J, Yang J, Zhang J. Small molecule LpxC inhibitors against gram-negative bacteria: Advances and future perspectives. Eur J Med Chem 2023; 253:115326. [PMID: 37023679 DOI: 10.1016/j.ejmech.2023.115326] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/18/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Uridine diphosphate-3-O-(hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) is a metalloenzyme with zinc ions as cofactors and is a key enzyme in the essential structural outer membrane lipid A synthesis commitment step of gram-negative bacteria. As LpxC is extremely homologous among different Gram-negative bacteria, it is conserved in almost all gram-negative bacteria, which makes LpxC a promising target. LpxC inhibitors have been reported extensively in recent years, such as PF-5081090 and CHIR-090 were found to have broad-spectrum antibiotic activity against P. aeruginosa and E. coli. They are mainly classified into hydroxamate inhibitors and non-hydroxamate inhibitors based on their structure, but no LpxC inhibitors have been marketed due to safety and activity issues. This review, therefore, focuses on small molecule inhibitors of LpxC against gram-negative pathogenic bacteria and covers recent advances in LpxC inhibitors, focusing on their structural optimization process, structure-activity relationships, and future directions, with the aim of providing ideas for the development of LpxC inhibitors and clinical research.
Collapse
|
7
|
Wimmer S, Hoff K, Martin B, Grewer M, Denni L, Lascorz Massanet R, Raimondi MV, Bülbül EF, Melesina J, Hotop SK, Haupenthal J, Rohde H, Heisig P, Hirsch AKH, Brönstrup M, Sippl W, Holl R. Synthesis, biological evaluation, and molecular docking studies of aldotetronic acid-based LpxC inhibitors. Bioorg Chem 2023; 131:106331. [PMID: 36587505 DOI: 10.1016/j.bioorg.2022.106331] [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: 09/14/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
In order to develop novel inhibitors of the bacterial deacetylase LpxC bearing a substituent to target the UDP binding site of the enzyme, a series of aldotetronic acid-based hydroxamic acids was accessed in chiral pool syntheses starting from 4,6-O-benzylidene-d-glucose and l-arabinitol. The synthesized hydroxamic acids were tested for LpxC inhibitory activity in vitro, revealing benzyl ether 17a ((2S,3S)-4-(benzyloxy)-N,3-dihydroxy-2-[(4-{[4-(morpholinomethyl)phenyl]ethynyl}benzyl)oxy]butanamide) as the most potent LpxC inhibitor. This compound was additionally tested for antibacterial activity against a panel of clinically relevant Gram-negative bacteria, bacterial uptake, and susceptibility to efflux pumps. Molecular docking studies were performed to rationalize the observed structure-activity relationships.
Collapse
Affiliation(s)
- Stefan Wimmer
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Katharina Hoff
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Benedikt Martin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149 Münster, Germany
| | - Martin Grewer
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149 Münster, Germany
| | - Laura Denni
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Raquel Lascorz Massanet
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Maria Valeria Raimondi
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Emre F Bülbül
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Jelena Melesina
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Sven-Kevin Hotop
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Holger Rohde
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany; Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Heisig
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Bundesstr. 45, 20146 Hamburg, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; Helmholtz International Lab for Anti-infectives, Germany; Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany; Helmholtz International Lab for Anti-infectives, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Ralph Holl
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany.
| |
Collapse
|
8
|
Dreger A, Hoff K, Agoglitta O, Bülbül EF, Melesina J, Sippl W, Holl R. Synthesis, biological evaluation, and molecular docking studies of deoxygenated C-glycosides as LpxC inhibitors. Bioorg Chem 2021; 117:105403. [PMID: 34758434 DOI: 10.1016/j.bioorg.2021.105403] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/23/2021] [Accepted: 10/02/2021] [Indexed: 11/29/2022]
Abstract
The bacterial deacetylase LpxC is a promising target for the development of novel antibiotics being selectively active against Gram-negative bacteria. In chiral pool syntheses starting from d- and l-ribose, a series regio- and stereoisomeric monohydroxytetrahydrofuran derivatives was synthesized and tested for LpxC inhibitory and antibacterial activities. Molecular docking studies were performed to rationalize the obtained structure-activity relationships. The (2S,3R,5R)-configured 3-hydroxytetrahydrofuran derivative ent-8 ((2S,3R,5R)-N,3-Dihydroxy-5-(4-{[4-(morpholinomethyl)phenyl]ethynyl}phenyl)tetrahydrofuran-2-carboxamide) was found to be the most potent LpxC inhibitor (Ki = 3.5 µM) of the synthesized series of monohydroxytetrahydrofuran derivatives and to exhibit the highest antibacterial activity against E. coli BL21(DE3) and the D22 strain.
Collapse
Affiliation(s)
- Alexander Dreger
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Katharina Hoff
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Oriana Agoglitta
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Emre F Bülbül
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Jelena Melesina
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Ralph Holl
- Institute of Organic Chemistry, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany.
| |
Collapse
|
9
|
Zhao Q, Xin L, Liu Y, Liang C, Li J, Jian Y, Li H, Shi Z, Liu H, Cao W. Current Landscape and Future Perspective of Oxazolidinone Scaffolds Containing Antibacterial Drugs. J Med Chem 2021; 64:10557-10580. [PMID: 34260235 DOI: 10.1021/acs.jmedchem.1c00480] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The widespread use of antibiotics has made the problem of bacterial resistance increasingly serious, and the study of new drug-resistant bacteria has become the main direction of antibacterial drug research. Among antibiotics, the fully synthetic oxazolidinone antibacterial drugs linezolid and tedizolid have been successfully marketed and have achieved good clinical treatment effects. Oxazolidinone antibacterial drugs have good pharmacokinetic and pharmacodynamic characteristics and unique antibacterial mechanisms, and resistant bacteria are sensitive to them. This Perspective focuses on reviewing oxazolidinones based on the structural modification of linezolid and new potential oxazolidinone drugs in the past 10 years, mainly describing their structure, antibacterial activity, safety, druggability, and so on, and discusses their structure-activity relationships, providing insight into the reasonable design of safer and more potent oxazolidinone antibacterial drugs.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Liang Xin
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China.,Xi'an Xuri Shengchang Pharmaceutical Technology Co., Ltd., High-tech Zone, Xi'an 710075, P. R. China
| | - Yuzhi Liu
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Chengyuan Liang
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Jingyi Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Yanlin Jian
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Han Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China
| | - Zhenfeng Shi
- Department of Urology Surgery Center, Xinjiang Uyghur People's Hospital, Urumqi 830002, P. R. China
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Hengqin New Area, Zhuhai 519030, P. R. China
| | - Wenqiang Cao
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Hengqin New Area, Zhuhai 519030, P. R. China
| |
Collapse
|
10
|
Abstract
A short de novo synthesis of an l-lemonose thioglycoside is described starting from d-threonine. The synthesis leverages a Dieckmann condensation and Stork-Danheiser transposition to arrive at a key vinylogous ester intermediate on gram scale. Ensuing 1,2-addition diastereoselectively establishes the C3 tetra-substituted center and subsequent glycal hydration allows for anomeric functionalization to the thioglycoside. 1H and NOESY NMR analyses reveal that the major α-anomer of thioglycoside deviates from the expected 1C4 conformation.
Collapse
Affiliation(s)
- Eric D Huseman
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Steven D Townsend
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
11
|
Zhou P, Hong J. Structure- and Ligand-Dynamics-Based Design of Novel Antibiotics Targeting Lipid A Enzymes LpxC and LpxH in Gram-Negative Bacteria. Acc Chem Res 2021; 54:1623-1634. [PMID: 33720682 DOI: 10.1021/acs.accounts.0c00880] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bacterial infections caused by multi-drug-resistant Gram-negative pathogens pose a serious threat to public health. Gram-negative bacteria are characterized by the enrichment of lipid A-anchored lipopolysaccharide (LPS) or lipooligosaccharide (LOS) in the outer leaflet of their outer membrane. Constitutive biosynthesis of lipid A via the Raetz pathway is essential for bacterial viability and fitness in the human host. The inhibition of early-stage lipid A enzymes such as LpxC not only suppresses the growth of Pseudomonas aeruginosa, Klebsiella pneumoniae, Enterobacter spp., and other clinically important Gram-negative pathogens but also sensitizes these bacteria to other antibiotics. The inhibition of late-stage lipid A enzymes such as LpxH is uniquely advantageous because it has an extra mechanism of bacterial killing through the accumulation of toxic lipid A intermediates, rendering LpxH inhibition additionally lethal to Acinetobacter baumannii. Because essential enzymes of the Raetz pathway have never been exploited by commercial antibiotics, they are excellent targets for the development of novel antibiotics against multi-drug-resistant Gram-negative infections.This Account describes the ongoing research on characterizing the structure and inhibition of LpxC and LpxH, the second and fourth enzymes of the Raetz pathway of lipid A biosynthesis, in the laboratories of Dr. Pei Zhou and Dr. Jiyong Hong at Duke University. Our studies have elucidated the molecular basis of LpxC inhibition by the first broad-spectrum inhibitor, CHIR-090, as well as the mechanism underlying its spectrum of activity. Such an analysis has provided a molecular explanation for the broad-spectrum antibiotic activity of diacetylene-based LpxC inhibitors. Through the structural and biochemical investigation of LpxC inhibition by diacetylene LpxC inhibitors and the first nanomolar LpxC inhibitor, L-161,240, we have elucidated the intrinsic conformational and dynamics difference in individual LpxC enzymes near the active site. A similar approach has been taken to investigate LpxH inhibition, leading to the establishment of the pharmacophore model of LpxH inhibitors and subsequent structural elucidation of LpxH in complex with its first reported small-molecule inhibitor based on a sulfonyl piperazine scaffold.Intriguingly, although our crystallographic analysis of LpxC- and LpxH-inhibitor complexes detected only a single inhibitor conformation in the crystal lattice, solution NMR studies revealed the existence of multiple ligand conformations that together delineate a cryptic ligand envelope expanding the ligand-binding footprint beyond that observed in the crystal structure. By harnessing the ligand dynamics information and structural insights, we demonstrate the feasibility to design potent LpxC and LpxH inhibitors by merging multiple ligand conformations. Such an approach has enabled us to rationally design compounds with significantly enhanced potency in enzymatic assays and outstanding antibiotic activities in vitro and in animal models of bacterial infection. We anticipate that continued efforts with structure and ligand dynamics-based lead optimization will ultimately lead to the discovery of LpxC- and LpxH-targeting clinical antibiotics against a broad range of Gram-negative pathogens.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| |
Collapse
|
12
|
Fan S, Li D, Yan M, Feng X, Lv G, Wu G, Jin Y, Wang Y, Yang Z. The Complex Structure of Protein AaLpxC from Aquifex aeolicus with ACHN-975 Molecule Suggests an Inhibitory Mechanism at Atomic-Level against Gram-Negative Bacteria. Molecules 2021; 26:molecules26051451. [PMID: 33800069 PMCID: PMC7962117 DOI: 10.3390/molecules26051451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
New drugs with novel antibacterial targets for Gram-negative bacterial pathogens are desperately needed. The protein LpxC is a vital enzyme for the biosynthesis of lipid A, an outer membrane component of Gram-negative bacterial pathogens. The ACHN-975 molecule has high enzymatic inhibitory capacity against the infectious diseases, which are caused by multidrug-resistant bacteria, but clinical research was halted because of its inflammatory response in previous studies. In this work, the structure of the recombinant UDP-3-O-(R-3-hydroxymyristol)-N-acetylglucosamine deacetylase from Aquifex aeolicus in complex with ACHN-975 was determined to a resolution at 1.21 Å. According to the solved complex structure, ACHN-975 was docked into the AaLpxC’s active site, which occupied the site of AaLpxC substrate. Hydroxamate group of ACHN-975 forms five-valenced coordination with resides His74, His226, Asp230, and the long chain part of ACHN-975 containing the rigid alkynyl groups docked in further to interact with the hydrophobic area of AaLpxC. We employed isothermal titration calorimetry for the measurement of affinity between AaLpxC mutants and ACHN-975, and the results manifest the key residues (His74, Thr179, Tyr212, His226, Asp230 and His253) for interaction. The determined AaLpxC crystal structure in complex with ACHN-975 is expected to serve as a guidance and basis for the design and optimization of molecular structures of ACHN-975 analogues to develop novel drug candidates against Gram-negative bacteria.
Collapse
Affiliation(s)
- Shuai Fan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
| | - Danyang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
| | - Maocai Yan
- School of Pharmacy, Jining Medical University, Rizhao, Shandong 276800, China;
| | - Xiao Feng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
| | - Guangxin Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
| | - Guangteng Wu
- ArNuXon Pharm-Sci Co., Ltd., Beijing 100085, China;
| | - Yuanyuan Jin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
- Correspondence: (Y.W.); (Z.Y.)
| | - Zhaoyong Yang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; (S.F.); (D.L.); (X.F.); (G.L.); (Y.J.)
- Correspondence: (Y.W.); (Z.Y.)
| |
Collapse
|
13
|
Troudi A, Pagès JM, Brunel JM. Chemical Highlights Supporting the Role of Lipid A in Efficient Biological Adaptation of Gram-Negative Bacteria to External Stresses. J Med Chem 2021; 64:1816-1834. [PMID: 33538159 DOI: 10.1021/acs.jmedchem.0c02185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The outer membrane (OM) of Gram-negative bacteria provides an efficient barrier against external noxious compounds such as antimicrobial agents. Associated with drug target modification, it contributes to the overall failure of chemotherapy. In the complex OM architecture, Lipid A plays an essential role by anchoring the lipopolysaccharide in the membrane and ensuring the spatial organization between lipids, proteins, and sugars. Currently, the targets of almost all antibiotics are intracellularly located and require translocation across membranes. We report herein an integrated view of Lipid A synthesis, membrane assembly, a structure comparison at the molecular structure level of numerous Gram-negative bacterial species, as well as its recent use as a target for original antibacterial molecules. This review paves the way for a new vision of a key membrane component that acts during bacterial adaptation to environmental stresses and for the development of new weapons against microbial resistance to usual antibiotics.
Collapse
Affiliation(s)
- Azza Troudi
- UMR-MD1, U1261, Aix Marseille Université, INSERM, SSA, MCT, 13385 Marseille, France.,Laboratory of Microorganisms and Active Biomolecules, Department of Biology, Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis 1008, Tunisia
| | - Jean Marie Pagès
- UMR-MD1, U1261, Aix Marseille Université, INSERM, SSA, MCT, 13385 Marseille, France
| | - Jean Michel Brunel
- UMR-MD1, U1261, Aix Marseille Université, INSERM, SSA, MCT, 13385 Marseille, France
| |
Collapse
|
14
|
Antibacterial activity of xylose-derived LpxC inhibitors - Synthesis, biological evaluation and molecular docking studies. Bioorg Chem 2020; 107:104603. [PMID: 33429229 DOI: 10.1016/j.bioorg.2020.104603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022]
Abstract
LpxC inhibitors represent a promising class of novel antibiotics selectively combating Gram-negative bacteria. In chiral pool syntheses starting from D- and L-xylose, a series of four 2r,3c,4t-configured C-furanosidic LpxC inhibitors was obtained. The synthesized hydroxamic acids were tested for antibacterial and LpxC inhibitory activity, the acquired biological data were compared with those of previously synthesized C-furanosides, and molecular docking studies were performed to rationalize the observed structure-activity relationships. Additionally, bacterial uptake and susceptibility to efflux pump systems were investigated for the most promising stereoisomers.
Collapse
|
15
|
Structural basis of the UDP-diacylglucosamine pyrophosphohydrolase LpxH inhibition by sulfonyl piperazine antibiotics. Proc Natl Acad Sci U S A 2020; 117:4109-4116. [PMID: 32041866 PMCID: PMC7049123 DOI: 10.1073/pnas.1912876117] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The UDP-2,3-diacylglucosamine pyrophosphate hydrolase LpxH is an essential lipid A biosynthetic enzyme that is conserved in the majority of gram-negative bacteria. It has emerged as an attractive novel antibiotic target due to the recent discovery of an LpxH-targeting sulfonyl piperazine compound (referred to as AZ1) by AstraZeneca. However, the molecular details of AZ1 inhibition have remained unresolved, stymieing further development of this class of antibiotics. Here we report the crystal structure of Klebsiella pneumoniae LpxH in complex with AZ1. We show that AZ1 fits snugly into the L-shaped acyl chain-binding chamber of LpxH with its indoline ring situating adjacent to the active site, its sulfonyl group adopting a sharp kink, and its N-CF3-phenyl substituted piperazine group reaching out to the far side of the LpxH acyl chain-binding chamber. Intriguingly, despite the observation of a single AZ1 conformation in the crystal structure, our solution NMR investigation has revealed the presence of a second ligand conformation invisible in the crystalline state. Together, these distinct ligand conformations delineate a cryptic inhibitor envelope that expands the observed footprint of AZ1 in the LpxH-bound crystal structure and enables the design of AZ1 analogs with enhanced potency in enzymatic assays. These designed compounds display striking improvement in antibiotic activity over AZ1 against wild-type K. pneumoniae, and coadministration with outer membrane permeability enhancers profoundly sensitizes Escherichia coli to designed LpxH inhibitors. Remarkably, none of the sulfonyl piperazine compounds occupies the active site of LpxH, foretelling a straightforward path for rapid optimization of this class of antibiotics.
Collapse
|
16
|
Deng X, Song M. Synthesis, antibacterial and anticancer activity, and docking study of aminoguanidines containing an alkynyl moiety. J Enzyme Inhib Med Chem 2020; 35:354-364. [PMID: 31851531 PMCID: PMC6968633 DOI: 10.1080/14756366.2019.1702654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Two series of aminoguanidines containing an alkynyl moiety were designed, synthesised, and screened for antibacterial and anticancer activities. Generally, the series 3a-3j with a 1,2-diphenylethyne exhibited better antibacterial activity than the other series (6a-6k) holding 1,4-diphenylbuta-1,3-diyne moiety antibacterial activity. Most compounds in series 3a-3j showed potent growth inhibition against the tested bacterial strains, with minimum inhibitory concentration (MIC) values in the range 0.25-8 µg/mL. Compound 3g demonstrated rapid and persistent bactericidal activity at 2 × MIC. The resistance study revealed that resistance of the tested bacteria towards 3g is not easily developed. Molecular docking studies revealed that compounds 3g and 6e bind strongly to the LpxC and FabH enzymes. Moreover, excellent activity of selected compounds against the growth of cancer cell lines A549 and SGC7901 was also observed, with IC50 values in the range 0.30-4.57 µg/mL. These findings indicate that compounds containing the aminoguanidine moiety are promising candidates for the development of new antibacterial and anticancer agents.
Collapse
Affiliation(s)
- Xianqing Deng
- Basic Medical and Pharmacy College, Jinggangshan University, Ji'an, China
| | - Mingxia Song
- Basic Medical and Pharmacy College, Jinggangshan University, Ji'an, China
| |
Collapse
|
17
|
Propargylglycine-based antimicrobial compounds are targets of TolC-dependent efflux systems in Escherichia coli. Bioorg Med Chem Lett 2020; 30:126875. [DOI: 10.1016/j.bmcl.2019.126875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 11/19/2022]
|
18
|
Surivet JP, Panchaud P, Specklin JL, Diethelm S, Blumstein AC, Gauvin JC, Jacob L, Masse F, Mathieu G, Mirre A, Schmitt C, Lange R, Tidten-Luksch N, Gnerre C, Seeland S, Herrmann C, Seiler P, Enderlin-Paput M, Mac Sweeney A, Wicki M, Hubschwerlen C, Ritz D, Rueedi G. Discovery of Novel Inhibitors of LpxC Displaying Potent in Vitro Activity against Gram-Negative Bacteria. J Med Chem 2019; 63:66-87. [PMID: 31804826 DOI: 10.1021/acs.jmedchem.9b01604] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
UDP-3-O-((R)-3-hydroxymyristoyl)-N-glucosamine deacetylase (LpxC) is as an attractive target for the discovery and development of novel antibacterial drugs to address the critical medical need created by multidrug resistant Gram-negative bacteria. By using a scaffold hopping approach on a known family of methylsulfone hydroxamate LpxC inhibitors, several hit series eliciting potent antibacterial activities against Enterobacteriaceae and Pseudomonas aeruginosa were identified. Subsequent hit-to-lead optimization, using cocrystal structures of inhibitors bound to Pseudomonas aeruginosa LpxC as guides, resulted in the discovery of multiple chemical series based on (i) isoindolin-1-ones, (ii) 4,5-dihydro-6H-thieno[2,3-c]pyrrol-6-ones, and (iii) 1,2-dihydro-3H-pyrrolo[1,2-c]imidazole-3-ones. Synthetic methods, antibacterial activities and relative binding affinities, as well as physicochemical properties that allowed compound prioritization are presented. Finally, in vivo properties of lead molecules which belong to the most promising pyrrolo-imidazolone series, such as 18d, are discussed.
Collapse
Affiliation(s)
- Jean-Philippe Surivet
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Philippe Panchaud
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Jean-Luc Specklin
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Stefan Diethelm
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | | | | | - Loïc Jacob
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Florence Masse
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Gaëlle Mathieu
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Azely Mirre
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Christine Schmitt
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Roland Lange
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Naomi Tidten-Luksch
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Carmela Gnerre
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Swen Seeland
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Charlyse Herrmann
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Peter Seiler
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Michel Enderlin-Paput
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Aengus Mac Sweeney
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Micha Wicki
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | | | - Daniel Ritz
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| | - Georg Rueedi
- Idorsia Pharmaceuticals Ltd. , Hegenheimermattweg 91 , CH-4123 Allschwil , Switzerland
| |
Collapse
|
19
|
Mutations Reducing In Vitro Susceptibility to Novel LpxC Inhibitors in Pseudomonas aeruginosa and Interplay of Efflux and Nonefflux Mechanisms. Antimicrob Agents Chemother 2019; 64:AAC.01490-19. [PMID: 31658970 DOI: 10.1128/aac.01490-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/22/2019] [Indexed: 11/20/2022] Open
Abstract
Upregulated expression of efflux pumps, lpxC target mutations, LpxC protein overexpression, and mutations in fabG were previously shown to mediate single-step resistance to the LpxC inhibitor CHIR-090 in P. aeruginosa Single-step selection experiments using three recently described LpxC inhibitors (compounds 2, 3, and 4) and mutant characterization showed that these mechanisms affect susceptibility to additional novel LpxC inhibitors. Serial passaging of P. aeruginosa wild-type and efflux pump-defective strains using the LpxC inhibitor CHIR-090 or compound 1 generated substantial shifts in susceptibility and underscored the interplay of efflux and nonefflux mechanisms. Whole-genome sequencing of CHIR-090 passage mutants identified efflux pump overexpression, fabG mutations, and novel mutations in fabF1 and in PA4465 as determinants of reduced susceptibility. Two new lpxC mutations, encoding A214V and G208S, that reduce susceptibility to certain LpxC inhibitors were identified in these studies, and we show that these and other target mutations differentially affect different LpxC inhibitor scaffolds. Lastly, the combination of target alteration (LpxCA214V) and upregulated expression of LpxC was shown to be tolerated in P. aeruginosa and could mediate significant decreases in susceptibility.
Collapse
|
20
|
González-Bello C. Recently developed synthetic compounds with anti-infective activity. Curr Opin Pharmacol 2019; 48:17-23. [DOI: 10.1016/j.coph.2019.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
|
21
|
Ding S, Ji J, Zhang M, Yang Y, Wang R, Zhu X, Wang L, Zhong Y, Gao L, Lu M, Liu J, Chen Y. Exploration of the structure–activity relationship and druggability of novel oxazolidinone‐based compounds as Gram‐negative antibacterial agents. Arch Pharm (Weinheim) 2019; 352:e1900129. [DOI: 10.1002/ardp.201900129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Shi Ding
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Jing‐Chao Ji
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Ming‐Juan Zhang
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Yu‐She Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Rui Wang
- Central Hospital affiliated to Shenyang Medical College Shenyang Liaoning China
| | - Xing‐Long Zhu
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Li‐Hong Wang
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Yi Zhong
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Le Gao
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Man Lu
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Ju Liu
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| | - Ye Chen
- Key Laboratory of New Drug Research and Development of Liaoning Province, College of Pharmacy Liaoning University Shenyang Liaoning China
| |
Collapse
|
22
|
Suaifan GA, Mohammed AA. Fluoroquinolones structural and medicinal developments (2013–2018): Where are we now? Bioorg Med Chem 2019; 27:3005-3060. [DOI: 10.1016/j.bmc.2019.05.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 12/11/2022]
|
23
|
Dreger A, Kharwb O, Agoglitta O, Bülbül EF, Melesina J, Sippl W, Holl R. Chiral Pool Synthesis, Biological Evaluation and Molecular Docking Studies ofC‐Furanosidic LpxC Inhibitors. ChemMedChem 2019; 14:871-886. [DOI: 10.1002/cmdc.201900068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Alexander Dreger
- Department of Chemistry, Institute of Organic ChemistryUniversity of Hamburg Martin-Luther-King Platz 6 20146 Hamburg Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems Germany
| | - Omar Kharwb
- Institute of Pharmaceutical and Medicinal ChemistryUniversity of Münster Corrensstr. 48 48149 Münster Germany
| | - Oriana Agoglitta
- Department of Chemistry, Institute of Organic ChemistryUniversity of Hamburg Martin-Luther-King Platz 6 20146 Hamburg Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems Germany
- NRW Graduate School of ChemistryUniversity of Münster Germany
| | - Emre F. Bülbül
- Institute of PharmacyMartin Luther University of Halle-Wittenberg Wolfgang-Langenbeck Str. 4 06120 Halle/Saale Germany
| | - Jelena Melesina
- Institute of PharmacyMartin Luther University of Halle-Wittenberg Wolfgang-Langenbeck Str. 4 06120 Halle/Saale Germany
| | - Wolfgang Sippl
- Institute of PharmacyMartin Luther University of Halle-Wittenberg Wolfgang-Langenbeck Str. 4 06120 Halle/Saale Germany
| | - Ralph Holl
- Department of Chemistry, Institute of Organic ChemistryUniversity of Hamburg Martin-Luther-King Platz 6 20146 Hamburg Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems Germany
| |
Collapse
|
24
|
Williams JC, Sheldon JR, Imlay HD, Dutter BF, Draelos MM, Skaar EP, Sulikowski GA. Synthesis of the Siderophore Coelichelin and Its Utility as a Probe in the Study of Bacterial Metal Sensing and Response. Org Lett 2019; 21:679-682. [PMID: 30645132 PMCID: PMC6474248 DOI: 10.1021/acs.orglett.8b03857] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A convergent total synthesis of the siderophore coelichelin is described. The synthetic route also provided access to acetyl coelichelin and other congeners of the parent siderophore. The synthetic products were evaluated for their ability to bind ferric iron and promote growth of a siderophore-deficient strain of the Gram-negative bacterium Pseudomonas aeruginosa under iron restriction conditions. The results of these studies indicate coelichelin and several derivatives serve as ferric iron delivery vehicles for P. aeruginosa.
Collapse
Affiliation(s)
- Jade C. Williams
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jessica R. Sheldon
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Hunter D. Imlay
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Brendan F. Dutter
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Matthew M. Draelos
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Gary A. Sulikowski
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
25
|
Chen AY, Adamek RN, Dick BL, Credille CV, Morrison CN, Cohen SM. Targeting Metalloenzymes for Therapeutic Intervention. Chem Rev 2019; 119:1323-1455. [PMID: 30192523 PMCID: PMC6405328 DOI: 10.1021/acs.chemrev.8b00201] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metalloenzymes are central to a wide range of essential biological activities, including nucleic acid modification, protein degradation, and many others. The role of metalloenzymes in these processes also makes them central for the progression of many diseases and, as such, makes metalloenzymes attractive targets for therapeutic intervention. Increasing awareness of the role metalloenzymes play in disease and their importance as a class of targets has amplified interest in the development of new strategies to develop inhibitors and ultimately useful drugs. In this Review, we provide a broad overview of several drug discovery efforts focused on metalloenzymes and attempt to map out the current landscape of high-value metalloenzyme targets.
Collapse
Affiliation(s)
- Allie Y Chen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Rebecca N Adamek
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Benjamin L Dick
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Cy V Credille
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Christine N Morrison
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Seth M Cohen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
26
|
González-Bello C. The Inhibition of Lipid A Biosynthesis-The Antidote Against Superbugs? ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Concepción González-Bello
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS); Departamento de Química Orgánica; Universidade de Santiago de Compostela; calle Jenaro de la Fuente s/n 15782 Santiago de Compostela Spain
| |
Collapse
|
27
|
Interplay of Klebsiella pneumoniae fabZ and lpxC Mutations Leads to LpxC Inhibitor-Dependent Growth Resulting from Loss of Membrane Homeostasis. mSphere 2018; 3:3/5/e00508-18. [PMID: 30381354 PMCID: PMC6211225 DOI: 10.1128/msphere.00508-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Emergence of antibiotic resistance has prompted efforts to identify and optimize novel inhibitors of antibacterial targets such as LpxC. This enzyme catalyzes the first committed step of lipid A synthesis, which is necessary to generate lipopolysaccharide and ultimately the Gram-negative protective outer membrane. Investigation of this pathway and its interrelationship with inner membrane (phospholipid) biosynthesis or other pathways is therefore highly important to the fundamental understanding of Gram-negative bacteria and by extension to antibiotic discovery. Here we exploited the availability of a novel LpxC inhibitor to engender the generation of K. pneumoniae resistant mutants whose growth depends on chemical inhibition of LpxC. Inhibitor dependency resulted from the interaction of different resistance mutations and was based on loss of normal cellular mechanisms required to establish membrane homeostasis. This study provides new insights into the importance of this process in K. pneumoniae and how it may be linked to novel biosynthetic pathway inhibitors. Tight coordination of inner and outer membrane biosynthesis is very important in Gram-negative bacteria. Biosynthesis of the lipid A moiety of lipopolysaccharide, which comprises the outer leaflet of the outer membrane has garnered interest for Gram-negative antibacterial discovery. In particular, several potent inhibitors of LpxC (the first committed step of the lipid A pathway) are described. Here we show that serial passaging of Klebsiella pneumoniae in increasing levels of an LpxC inhibitor yielded mutants that grew only in the presence of the inhibitor. These strains had mutations in fabZ and lpxC occurring together (encoding either FabZR121L/LpxCV37G or FabZF51L/LpxCV37G). K. pneumoniae mutants having only LpxCV37G or LpxCV37A or various FabZ mutations alone were less susceptible to the LpxC inhibitor and did not require LpxC inhibition for growth. Western blotting revealed that LpxCV37G accumulated to high levels, and electron microscopy of cells harboring FabZR121L/LpxCV37G indicated an extreme accumulation of membrane in the periplasm when cells were subcultured without LpxC inhibitor. Significant accumulation of detergent-like lipid A pathway intermediates that occur downstream of LpxC (e.g., lipid X and disaccharide monophosphate [DSMP]) was also seen. Taken together, our results suggest that redirection of lipid A pathway substrate by less active FabZ variants, combined with increased activity from LpxCV37G was overdriving the lipid A pathway, necessitating LpxC chemical inhibition, since native cellular maintenance of membrane homeostasis was no longer functioning. IMPORTANCE Emergence of antibiotic resistance has prompted efforts to identify and optimize novel inhibitors of antibacterial targets such as LpxC. This enzyme catalyzes the first committed step of lipid A synthesis, which is necessary to generate lipopolysaccharide and ultimately the Gram-negative protective outer membrane. Investigation of this pathway and its interrelationship with inner membrane (phospholipid) biosynthesis or other pathways is therefore highly important to the fundamental understanding of Gram-negative bacteria and by extension to antibiotic discovery. Here we exploited the availability of a novel LpxC inhibitor to engender the generation of K. pneumoniae resistant mutants whose growth depends on chemical inhibition of LpxC. Inhibitor dependency resulted from the interaction of different resistance mutations and was based on loss of normal cellular mechanisms required to establish membrane homeostasis. This study provides new insights into the importance of this process in K. pneumoniae and how it may be linked to novel biosynthetic pathway inhibitors.
Collapse
|
28
|
Disrupting Gram-Negative Bacterial Outer Membrane Biosynthesis through Inhibition of the Lipopolysaccharide Transporter MsbA. Antimicrob Agents Chemother 2018; 62:AAC.01142-18. [PMID: 30104274 DOI: 10.1128/aac.01142-18] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/04/2018] [Indexed: 12/24/2022] Open
Abstract
There is a critical need for new antibacterial strategies to counter the growing problem of antibiotic resistance. In Gram-negative bacteria, the outer membrane (OM) provides a protective barrier against antibiotics and other environmental insults. The outer leaflet of the outer membrane is primarily composed of lipopolysaccharide (LPS). Outer membrane biogenesis presents many potentially compelling drug targets as this pathway is absent in higher eukaryotes. Most proteins involved in LPS biosynthesis and transport are essential; however, few compounds have been identified that inhibit these proteins. The inner membrane ABC transporter MsbA carries out the first essential step in the trafficking of LPS to the outer membrane. We conducted a biochemical screen for inhibitors of MsbA and identified a series of quinoline compounds that kill Escherichia coli through inhibition of its ATPase and transport activity, with no loss of activity against clinical multidrug-resistant strains. Identification of these selective inhibitors indicates that MsbA is a viable target for new antibiotics, and the compounds we identified serve as useful tools to further probe the LPS transport pathway in Gram-negative bacteria.
Collapse
|
29
|
Haque A, Al-Balushi RA, Al-Busaidi IJ, Khan MS, Raithby PR. Rise of Conjugated Poly-ynes and Poly(Metalla-ynes): From Design Through Synthesis to Structure-Property Relationships and Applications. Chem Rev 2018; 118:8474-8597. [PMID: 30112905 DOI: 10.1021/acs.chemrev.8b00022] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Conjugated poly-ynes and poly(metalla-ynes) constitute an important class of new materials with potential application in various domains of science. The key factors responsible for the diverse usage of these materials is their intriguing and tunable chemical and photophysical properties. This review highlights fascinating advances made in the field of conjugated organic poly-ynes and poly(metalla-ynes) incorporating group 4-11 metals. This includes several important aspects of conjugated poly-ynes viz. synthetic protocols, bonding, electronic structure, nature of luminescence, structure-property relationships, diverse applications, and concluding remarks. Furthermore, we delineated the future directions and challenges in this particular area of research.
Collapse
Affiliation(s)
- Ashanul Haque
- Department of Chemistry , Sultan Qaboos University , P.O. Box 36, Al-Khod 123 , Sultanate of Oman
| | - Rayya A Al-Balushi
- Department of Chemistry , Sultan Qaboos University , P.O. Box 36, Al-Khod 123 , Sultanate of Oman
| | - Idris Juma Al-Busaidi
- Department of Chemistry , Sultan Qaboos University , P.O. Box 36, Al-Khod 123 , Sultanate of Oman
| | - Muhammad S Khan
- Department of Chemistry , Sultan Qaboos University , P.O. Box 36, Al-Khod 123 , Sultanate of Oman
| | - Paul R Raithby
- Department of Chemistry , University of Bath , Claverton Down , Bath BA2 7AY , U.K
| |
Collapse
|
30
|
Listunov D, Joly E, Duhayon C, Saffon-Merceron N, Fabing I, Génisson Y, Maraval V, Chauvin R. Methinylogation Approach in Chiral Pharmacophore Design: from Alkynyl- to Allenyl-carbinol Warheads against Tumor Cells. ChemMedChem 2018; 13:1711-1722. [DOI: 10.1002/cmdc.201800284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Indexed: 11/11/2022]
Affiliation(s)
| | - Etienne Joly
- UMR CNRS 5089, IPBS (Institut de Pharmacologie et de Biologie Structurale); 205 Route de Narbonne 31077 Toulouse cedex France
| | - Carine Duhayon
- LCC-CNRS; Université de Toulouse, CNRS, UPS; Toulouse France
| | | | - Isabelle Fabing
- UMR CNRS 5068, LSPCMIB; Université de Toulouse, Université Paul Sabatier; 118 Route de Narbonne 31062 Toulouse cedex 9 France
| | - Yves Génisson
- UMR CNRS 5068, LSPCMIB; Université de Toulouse, Université Paul Sabatier; 118 Route de Narbonne 31062 Toulouse cedex 9 France
| | - Valérie Maraval
- LCC-CNRS; Université de Toulouse, CNRS, UPS; Toulouse France
| | - Remi Chauvin
- LCC-CNRS; Université de Toulouse, CNRS, UPS; Toulouse France
| |
Collapse
|
31
|
Wang T, Yu Y, Liang X, Luo S, He Z, Sun Z, Jiang Y, Omsland A, Zhou P, Song L. Lipid A Has Significance for Optimal Growth of Coxiella burnetii in Macrophage-Like THP-1 Cells and to a Lesser Extent in Axenic Media and Non-phagocytic Cells. Front Cell Infect Microbiol 2018; 8:192. [PMID: 29938202 PMCID: PMC6002510 DOI: 10.3389/fcimb.2018.00192] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/22/2018] [Indexed: 02/02/2023] Open
Abstract
Lipid A is an essential basal component of lipopolysaccharide of most Gram-negative bacteria. Inhibitors targeting LpxC, a conserved enzyme in lipid A biosynthesis, are antibiotic candidates against Gram-negative pathogens. Here we report the characterization of the role of lipid A in Coxiella burnetii growth in axenic media, monkey kidney cells (BGMK and Vero), and macrophage-like THP-1 cells by using a potent LpxC inhibitor -LPC-011. We first determined the susceptibility of C. burnetii LpxC to LPC-011 in a surrogate E. coli model. In E. coli, the minimum inhibitory concentration (MIC) of LPC-011 against C. burnetii LpxC is < 0.05 μg/mL, a value lower than the inhibitor's MIC against E. coli LpxC. Considering the inhibitor's problematic pharmacokinetic properties in vivo and Coxiella's culturing time up to 7 days, the stability of LPC-011 in cell cultures was assessed. We found that regularly changing inhibitor-containing media was required for sustained inhibition of C. burnetii LpxC in cells. Under inhibitor treatment, Coxiella has reduced growth yields in axenic media and during replication in non-phagocytic cells, and has a reduced number of productive vacuoles in such cells. Inhibiting lipid A biosynthesis in C. burnetii by the inhibitor was shown in a phase II strain transformed with chlamydial kdtA. This exogenous KdtA enzyme modifies Coxiella lipid A with an α-Kdo-(2 → 8)-α-Kdo epitope that can be detected by anti-chlamydia genus antibodies. In inhibitor-treated THP-1 cells, Coxiella shows severe growth defects characterized by poor vacuole formation and low growth yields. Coxiella progenies prepared from inhibitor-treated cells retain the capability of normally infecting all tested cells in the absence of the inhibitor, which suggests a dispensable role of lipid A for infection and early vacuole development. In conclusion, our data suggest that lipid A has significance for optimal development of Coxiella-containing vacuoles, and for robust multiplication of C. burnetii in macrophage-like THP-1 cells. Unlike many bacteria, C. burnetii replication in axenic media and non-phagocytic cells was less dependent on normal lipid A biosynthesis.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yonghui Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaofei Liang
- Department of Chemistry, Duke University, Durham, NC, United States
| | - Shengdong Luo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zemin He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zhihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Anders Omsland
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Pei Zhou
- Department of Biochemistry, Duke University Medical Center, Durham, NC, United States
| | - Lihua Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
32
|
Žalubovskis R, Winum JY. Inhibitors of Selected Bacterial Metalloenzymes. Curr Med Chem 2018; 26:2690-2714. [PMID: 29611472 DOI: 10.2174/0929867325666180403154018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/14/2018] [Accepted: 03/19/2018] [Indexed: 12/26/2022]
Abstract
The utilization of bacterial metalloenzymes, especially ones not having mammalian (human) counterparts, has drawn attention to develop novel antibacterial agents to overcome drug resistance and especially multidrug resistance. In this review, we focus on the recent achievements on the development of inhibitors of bacterial enzymes peptide deformylase (PDF), metallo-β-lactamase (MBL), methionine aminopeptidase (MetAP) and UDP-3-O-acyl- N-acetylglucosamine deacetylase (LpxC). The state of the art of the design and investigation of inhibitors of bacterial metalloenzymes is presented, and challenges are outlined and discussed.
Collapse
Affiliation(s)
- Raivis Žalubovskis
- Latvian Institute of Organic Synthesis, Riga, Latvia.,Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Latvia
| | - Jean-Yves Winum
- Institut des Biomolecules Max Mousseron, Universite de Montpellier, France
| |
Collapse
|
33
|
Bohl HO, Shi K, Lee JK, Aihara H. Crystal structure of lipid A disaccharide synthase LpxB from Escherichia coli. Nat Commun 2018; 9:377. [PMID: 29371662 PMCID: PMC5785501 DOI: 10.1038/s41467-017-02712-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/20/2017] [Indexed: 11/23/2022] Open
Abstract
Most Gram-negative bacteria are surrounded by a glycolipid called lipopolysaccharide (LPS), which forms a barrier to hydrophobic toxins and, in pathogenic bacteria, is a virulence factor. During LPS biosynthesis, a membrane-associated glycosyltransferase (LpxB) forms a tetra-acylated disaccharide that is further acylated to form the membrane anchor moiety of LPS. Here we solve the structure of a soluble and catalytically competent LpxB by X-ray crystallography. The structure reveals that LpxB has a glycosyltransferase-B family fold but with a highly intertwined, C-terminally swapped dimer comprising four domains. We identify key catalytic residues with a product, UDP, bound in the active site, as well as clusters of hydrophobic residues that likely mediate productive membrane association or capture of lipidic substrates. These studies provide the basis for rational design of antibiotics targeting a crucial step in LPS biosynthesis.
Collapse
Affiliation(s)
- Heather O Bohl
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - John K Lee
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
- Bristol-Myers Squibb, Redwood City, CA, 94063, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA.
| |
Collapse
|
34
|
Ding S, Dai RY, Wang WK, Cao Q, Lan LF, Zhou XL, Yang YS. Design, synthesis and structure-activity relationship evaluation of novel LpxC inhibitors as Gram-negative antibacterial agents. Bioorg Med Chem Lett 2018; 28:94-102. [DOI: 10.1016/j.bmcl.2017.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/28/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
|
35
|
Malkowski SN, Dishuck CF, Lamanilao GG, Embry CP, Grubb CS, Cafiero M, Peterson LW. Design, Modeling and Synthesis of 1,2,3-Triazole-Linked Nucleoside-Amino Acid Conjugates as Potential Antibacterial Agents. Molecules 2017; 22:molecules22101682. [PMID: 28994722 PMCID: PMC6151744 DOI: 10.3390/molecules22101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 02/01/2023] Open
Abstract
Copper-catalyzed azide-alkyne cycloadditions (CuAAC or click chemistry) are convenient methods to easily couple various pharmacophores or bioactive molecules. A new series of 1,2,3-triazole-linked nucleoside-amino acid conjugates have been designed and synthesized in 57–76% yields using CuAAC. The azido group was introduced on the 5′-position of uridine or the acyclic analogue using the tosyl-azide exchange method and alkylated serine or proparylglycine was the alkyne. Modeling studies of the conjugates in the active site of LpxC indicate they have promise as antibacterial agents.
Collapse
Affiliation(s)
- Sarah N Malkowski
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Carolyn F Dishuck
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Gene G Lamanilao
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Carter P Embry
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Christopher S Grubb
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Mauricio Cafiero
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| | - Larryn W Peterson
- Department of Chemistry, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| |
Collapse
|
36
|
Affiliation(s)
- Dmitrii V. Kalinin
- Institut für Organische Chemie, Universität Hamburg, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Ralph Holl
- Institut für Organische Chemie, Universität Hamburg, Hamburg, Germany
- German Center for Infection Research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems
| |
Collapse
|
37
|
Piizzi G, Parker DT, Peng Y, Dobler M, Patnaik A, Wattanasin S, Liu E, Lenoir F, Nunez J, Kerrigan J, McKenney D, Osborne C, Yu D, Lanieri L, Bojkovic J, Dzink-Fox J, Lilly MD, Sprague ER, Lu Y, Wang H, Ranjitkar S, Xie L, Wang B, Glick M, Hamann LG, Tommasi R, Yang X, Dean CR. Design, Synthesis, and Properties of a Potent Inhibitor of Pseudomonas aeruginosa Deacetylase LpxC. J Med Chem 2017; 60:5002-5014. [PMID: 28549219 DOI: 10.1021/acs.jmedchem.7b00377] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Over the past several decades, the frequency of antibacterial resistance in hospitals, including multidrug resistance (MDR) and its association with serious infectious diseases, has increased at alarming rates. Pseudomonas aeruginosa is a leading cause of nosocomial infections, and resistance to virtually all approved antibacterial agents is emerging in this pathogen. To address the need for new agents to treat MDR P. aeruginosa, we focused on inhibiting the first committed step in the biosynthesis of lipid A, the deacetylation of uridyldiphospho-3-O-(R-hydroxydecanoyl)-N-acetylglucosamine by the enzyme LpxC. We approached this through the design, synthesis, and biological evaluation of novel hydroxamic acid LpxC inhibitors, exemplified by 1, where cytotoxicity against mammalian cell lines was reduced, solubility and plasma-protein binding were improved while retaining potent anti-pseudomonal activity in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - David McKenney
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Colin Osborne
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Donghui Yu
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Leanne Lanieri
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Jade Bojkovic
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - JoAnn Dzink-Fox
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Maria-Dawn Lilly
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | | | | | | | - Srijan Ranjitkar
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Lili Xie
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | | | | | | | | | - Xia Yang
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| | - Charles R Dean
- Infectious Diseases Area, Novartis Institutes for BioMedical Research , Emeryville, California 94608, United States
| |
Collapse
|
38
|
Cram ED, Rockey DD, Dolan BP. Chlamydia spp. development is differentially altered by treatment with the LpxC inhibitor LPC-011. BMC Microbiol 2017; 17:98. [PMID: 28438125 PMCID: PMC5402638 DOI: 10.1186/s12866-017-0992-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chlamydia species are obligate intracellular bacteria that infect a broad range of mammalian hosts. Members of related genera are pathogens of a variety of vertebrate and invertebrate species. Despite the diversity of Chlamydia, all species contain an outer membrane lipooligosaccharide (LOS) that is comprised of a genus-conserved, and genus-defining, trisaccharide 3-deoxy-D-manno-oct-2-ulosonic acid Kdo region. Recent studies with lipopolysaccharide inhibitors demonstrate that LOS is important for the C. trachomatis developmental cycle during RB- > EB differentiation. Here, we explore the effects of one of these inhibitors, LPC-011, on the developmental cycle of five chlamydial species. RESULTS Sensitivity to the drug varied in some of the species and was conserved between others. We observed that inhibition of LOS biosynthesis in some chlamydial species induced formation of aberrant reticulate bodies, while in other species, no change was observed to the reticulate body. However, loss of LOS production prevented completion of the chlamydial reproductive cycle in all species tested. In previous studies we found that C. trachomatis and C. caviae infection enhances MHC class I antigen presentation of a model self-peptide. We find that treatment with LPC-011 prevents enhanced host-peptide presentation induced by infection with all chlamydial-species tested. CONCLUSIONS The data demonstrate that LOS synthesis is necessary for production of infectious progeny and inhibition of LOS synthesis induces aberrancy in certain chlamydial species, which has important implications for the use of LOS synthesis inhibitors as potential antibiotics.
Collapse
Affiliation(s)
- Erik D Cram
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, Corvallis, OR, 97331, USA.
| | - Daniel D Rockey
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, Corvallis, OR, 97331, USA
| | - Brian P Dolan
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, Corvallis, OR, 97331, USA
| |
Collapse
|
39
|
Zhang J, Chan A, Lippa B, Cross JB, Liu C, Yin N, Romero JAC, Lawrence J, Heney R, Herradura P, Goss J, Clark C, Abel C, Zhang Y, Poutsiaka KM, Epie F, Conrad M, Mahamoon A, Nguyen K, Chavan A, Clark E, Li TC, Cheng RK, Wood M, Andersen OA, Brooks M, Kwong J, Barker J, Parr IB, Gu Y, Ryan MD, Coleman S, Metcalf CA. Structure-based discovery of LpxC inhibitors. Bioorg Med Chem Lett 2017; 27:1670-1680. [DOI: 10.1016/j.bmcl.2017.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 10/20/2022]
|
40
|
|
41
|
Zhou P, Zhao J. Structure, inhibition, and regulation of essential lipid A enzymes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:1424-1438. [PMID: 27940308 DOI: 10.1016/j.bbalip.2016.11.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
Abstract
The Raetz pathway of lipid A biosynthesis plays a vital role in the survival and fitness of Gram-negative bacteria. Research efforts in the past three decades have identified individual enzymes of the pathway and have provided a mechanistic understanding of the action and regulation of these enzymes at the molecular level. This article reviews the discovery, biochemical and structural characterization, and regulation of the essential lipid A enzymes, as well as continued efforts to develop novel antibiotics against Gram-negative pathogens by targeting lipid A biosynthesis. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Biochemistry, Duke University Medical Center, Research Drive, DUMC 3711, Durham, NC 27710, USA.
| | - Jinshi Zhao
- Department of Biochemistry, Duke University Medical Center, Research Drive, DUMC 3711, Durham, NC 27710, USA
| |
Collapse
|
42
|
Paek SM, Jeong M, Jo J, Heo YM, Han YT, Yun H. Recent Advances in Substrate-Controlled Asymmetric Induction Derived from Chiral Pool α-Amino Acids for Natural Product Synthesis. Molecules 2016; 21:E951. [PMID: 27455209 PMCID: PMC6274556 DOI: 10.3390/molecules21070951] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/07/2016] [Accepted: 07/18/2016] [Indexed: 11/20/2022] Open
Abstract
Chiral pool α-amino acids have been used as powerful tools for the total synthesis of structurally diverse natural products. Some common naturally occurring α-amino acids are readily available in both enantiomerically pure forms. The applications of the chiral pool in asymmetric synthesis can be categorized prudently as chiral sources, devices, and inducers. This review specifically examines recent advances in substrate-controlled asymmetric reactions induced by the chirality of α-amino acid templates in natural product synthesis research and related areas.
Collapse
Affiliation(s)
- Seung-Mann Paek
- College of Pharmacy, Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju daero, Jinju 52828, Korea.
| | - Myeonggyo Jeong
- College of Pharmacy, Pusan National University, Busan 46241, Korea.
| | - Jeyun Jo
- College of Pharmacy, Pusan National University, Busan 46241, Korea.
| | - Yu Mi Heo
- College of Pharmacy, Research Institute of Pharmaceutical Science, Gyeongsang National University, Jinju daero, Jinju 52828, Korea.
| | - Young Taek Han
- College of Pharmacy, Dankook University, Cheonan 31116, Korea.
| | - Hwayoung Yun
- College of Pharmacy, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
43
|
Erwin AL. Antibacterial Drug Discovery Targeting the Lipopolysaccharide Biosynthetic Enzyme LpxC. Cold Spring Harb Perspect Med 2016; 6:cshperspect.a025304. [PMID: 27235477 DOI: 10.1101/cshperspect.a025304] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The enzyme LpxC (UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase) is broadly conserved across Gram-negative bacteria and is essential for synthesis of lipid A, the membrane anchor of the lipopolysaccharides (LPSs), which are a major component of the outer membrane in nearly all Gram-negative bacteria. LpxC has been the focus of target-directed antibiotic discovery projects in numerous pharmaceutical and academic groups for more than 20 years. Despite intense effort, no LpxC inhibitor has been approved for therapeutic use, and only one has yet reached human studies. This article will summarize the history of LpxC as a drug target and the parallel history of research on LpxC biology. Both academic and industrial researchers have used LpxC inhibitors as tool compounds, leading to increased understanding of the differing mechanisms for regulation of LPS synthesis in Escherichia coli and Pseudomonas aeruginosa.
Collapse
|
44
|
Henderson JC, Zimmerman SM, Crofts AA, Boll JM, Kuhns LG, Herrera CM, Trent MS. The Power of Asymmetry: Architecture and Assembly of the Gram-Negative Outer Membrane Lipid Bilayer. Annu Rev Microbiol 2016; 70:255-78. [PMID: 27359214 DOI: 10.1146/annurev-micro-102215-095308] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Determining the chemical composition of biological materials is paramount to the study of natural phenomena. Here, we describe the composition of model gram-negative outer membranes, focusing on the predominant assembly, an asymmetrical bilayer of lipid molecules. We also give an overview of lipid biosynthetic pathways and molecular mechanisms that organize this material into the outer membrane bilayer. An emphasis is placed on the potential of these pathways as targets for antibiotic development. We discuss deviations in composition, through bacterial cell surface remodeling, and alternative modalities to the asymmetric lipid bilayer. Outer membrane lipid alterations of current microbiological interest, such as lipid structures found in commensal bacteria, are emphasized. Additionally, outer membrane components could potentially be engineered to develop vaccine platforms. Observations related to composition and assembly of gram-negative outer membranes will continue to generate novel discoveries, broaden biotechnologies, and reveal profound mysteries to compel future research.
Collapse
Affiliation(s)
- Jeremy C Henderson
- Department of Molecular Biosciences, The University of Texas at Austin, Texas 78712
| | - Shawn M Zimmerman
- Department of Infectious Diseases, The University of Georgia, Athens, Georgia 30602;
| | - Alexander A Crofts
- Department of Molecular Biosciences, The University of Texas at Austin, Texas 78712
| | - Joseph M Boll
- Department of Molecular Biosciences, The University of Texas at Austin, Texas 78712
| | - Lisa G Kuhns
- Department of Infectious Diseases, The University of Georgia, Athens, Georgia 30602;
| | - Carmen M Herrera
- Department of Infectious Diseases, The University of Georgia, Athens, Georgia 30602;
| | - M Stephen Trent
- Department of Infectious Diseases, The University of Georgia, Athens, Georgia 30602;
| |
Collapse
|
45
|
Kurasaki H, Tsuda K, Shinoyama M, Takaya N, Yamaguchi Y, Kishii R, Iwase K, Ando N, Nomura M, Kohno Y. LpxC Inhibitors: Design, Synthesis, and Biological Evaluation of Oxazolidinones as Gram-negative Antibacterial Agents. ACS Med Chem Lett 2016; 7:623-8. [PMID: 27326338 DOI: 10.1021/acsmedchemlett.6b00057] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/05/2016] [Indexed: 02/02/2023] Open
Abstract
Herein we report a scaffold-hopping approach to identify a new scaffold with a zinc binding headgroup. Structural information was used to give novel oxazolidinone-based LpxC inhibitors. In particular, the most potent compound, 23j, showed a low efflux ratio, nanomolar potencies against E. coli LpxC enzyme, and excellent antibacterial activity against E. coli and K. pneumoniae. Computational docking was used to predict the interaction between 23j and E. coli LpxC, suggesting that the interactions with C207 and C63 contribute to the strong activity. These results provide new insights into the design of next-generation LpxC inhibitors.
Collapse
Affiliation(s)
- Haruaki Kurasaki
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Kosuke Tsuda
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Mariko Shinoyama
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Noriko Takaya
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Yuko Yamaguchi
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Ryuta Kishii
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Kazuhiko Iwase
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Naoki Ando
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Masahiro Nomura
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| | - Yasushi Kohno
- Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., 1848 Nogi, Nogi-machi, Shimotsuga-gun, Tochigi 329-0114, Japan
| |
Collapse
|
46
|
Liang X, Gopalaswamy R, Navas F, Toone EJ, Zhou P. A Scalable Synthesis of the Difluoromethyl-allo-threonyl Hydroxamate-Based LpxC Inhibitor LPC-058. J Org Chem 2016; 81:4393-8. [PMID: 27128325 DOI: 10.1021/acs.joc.6b00589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The difluoromethyl-allo-threonyl hydroxamate-based compound LPC-058 is a potent inhibitor of UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) in Gram-negative bacteria. A scalable synthesis of this compound is described. The key step in the synthetic sequence is a transition metal/base-catalyzed aldol reaction of methyl isocyanoacetate and difluoroacetone, giving rise to 4-(methoxycarbonyl)-5,5-disubstituted 2-oxazoline. A simple NMR-based determination of enantiomeric purity is also described.
Collapse
Affiliation(s)
- Xiaofei Liang
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Ramesh Gopalaswamy
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Frank Navas
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States
| | - Eric J Toone
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States.,Department of Biochemistry, Duke University Medical Center , Durham, North Carolina 27710, United States
| | - Pei Zhou
- Department of Chemistry, Duke University , Durham, North Carolina 27708, United States.,Department of Biochemistry, Duke University Medical Center , Durham, North Carolina 27710, United States
| |
Collapse
|
47
|
Tangherlini G, Torregrossa T, Agoglitta O, Köhler J, Melesina J, Sippl W, Holl R. Synthesis and biological evaluation of enantiomerically pure glyceric acid derivatives as LpxC inhibitors. Bioorg Med Chem 2016; 24:1032-44. [DOI: 10.1016/j.bmc.2016.01.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/13/2016] [Accepted: 01/15/2016] [Indexed: 10/22/2022]
|
48
|
Lee CJ, Liang X, Wu Q, Najeeb J, Zhao J, Gopalaswamy R, Titecat M, Sebbane F, Lemaitre N, Toone EJ, Zhou P. Drug design from the cryptic inhibitor envelope. Nat Commun 2016; 7:10638. [PMID: 26912110 PMCID: PMC4773385 DOI: 10.1038/ncomms10638] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 01/05/2016] [Indexed: 11/09/2022] Open
Abstract
Conformational dynamics plays an important role in enzyme catalysis, allosteric regulation of protein functions and assembly of macromolecular complexes. Despite these well-established roles, such information has yet to be exploited for drug design. Here we show by nuclear magnetic resonance spectroscopy that inhibitors of LpxC--an essential enzyme of the lipid A biosynthetic pathway in Gram-negative bacteria and a validated novel antibiotic target--access alternative, minor population states in solution in addition to the ligand conformation observed in crystal structures. These conformations collectively delineate an inhibitor envelope that is invisible to crystallography, but is dynamically accessible by small molecules in solution. Drug design exploiting such a hidden inhibitor envelope has led to the development of potent antibiotics with inhibition constants in the single-digit picomolar range. The principle of the cryptic inhibitor envelope approach may be broadly applicable to other lead optimization campaigns to yield improved therapeutics.
Collapse
Affiliation(s)
- Chul-Jin Lee
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Xiaofei Liang
- Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
| | - Qinglin Wu
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Javaria Najeeb
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Jinshi Zhao
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Ramesh Gopalaswamy
- Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
| | - Marie Titecat
- Inserm, Univ. Lille, CHU Lille, Institut Pasteur de Lille, CNRS, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Florent Sebbane
- Inserm, Univ. Lille, CHU Lille, Institut Pasteur de Lille, CNRS, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Nadine Lemaitre
- Inserm, Univ. Lille, CHU Lille, Institut Pasteur de Lille, CNRS, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Eric J Toone
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA.,Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
| | - Pei Zhou
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710, USA.,Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
49
|
Characterization of an Acinetobacter baumannii lptD Deletion Strain: Permeability Defects and Response to Inhibition of Lipopolysaccharide and Fatty Acid Biosynthesis. J Bacteriol 2015; 198:731-41. [PMID: 26668262 DOI: 10.1128/jb.00639-15] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/04/2015] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED Lipid A on the Gram-negative outer membrane (OM) is synthesized in the cytoplasm by the Lpx pathway and translocated to the OM by the Lpt pathway. Some Acinetobacter baumannii strains can tolerate the complete loss of lipopolysaccharide (LPS) resulting from the inactivation of early LPS pathway genes such as lpxC. Here, we characterized a mutant deleted for lptD, which encodes an OM protein that mediates the final translocation of fully synthesized LPS to the OM. Cells lacking lptD had a growth defect comparable to that of an lpxC deletion mutant under the growth conditions tested but were more sensitive to hydrophobic antibiotics, revealing a more significant impact on cell permeability from impaired LPS translocation than from the loss of LPS synthesis. Consistent with this, ATP leakage and N-phenyl-1-naphthylamine (NPN) fluorescence assays indicated a more severe impact of lptD deletion than of lpxC deletion on inner and outer membrane permeability, respectively. Targeted liquid chromatography-mass spectrometry (LCMS) analysis of LPS intermediates from UDP-3-O-R-3-hydroxylauroyl-N-acetyl-α-d-glucosamine through lipid IV(A) showed that the loss of LptD caused an accumulation of lipid IV(A). This suggested that pathway intermediate accumulation or mislocalization caused by the blockage of later LPS pathway steps impacts envelope integrity. Supporting this notion, chemical inhibition of lipid A precursor enzymes, including LpxC and FabB/F, in the lptD deletion strain partially rescued growth and permeability defects. IMPORTANCE New antibiotics to treat Gram-negative bacterial infections are urgently needed. Inhibition of LPS biosynthesis is attractive because this would impact viability and cell permeability. Therefore, a better understanding of this pathway is important, especially in strains such as A. baumannii ATCC 19606, where LPS biosynthesis is not essential in vitro. We show that ATCC 19606 also survives the loss of the final translocation of LPS into the OM (lptD deletion). Intriguingly, this impaired cell envelope integrity more than the loss of LPS biosynthesis (lpxC deletion), presumably due to the accumulation of toxic intermediates. Supporting this, chemical inhibition of LPS biosynthesis partially reversed this permeability defect. This extends our understanding of the LPS machinery and provides insights into potential interrelationships of the target steps along this important pathway.
Collapse
|
50
|
Giguère D. Surface polysaccharides from Acinetobacter baumannii : Structures and syntheses. Carbohydr Res 2015; 418:29-43. [DOI: 10.1016/j.carres.2015.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 09/30/2015] [Accepted: 10/03/2015] [Indexed: 12/31/2022]
|