1
|
Gupta A, Hassan MA, Ndugire W, Park J, Noor S, Nagaraj H, Chakraborty S, Rotello VM. Light-Triggered Bioorthogonal Nanozyme Hydrogels for Prodrug Activation and Treatment of Bacterial Biofilms. ACS APPLIED MATERIALS & INTERFACES 2025; 17:26361-26370. [PMID: 40275431 DOI: 10.1021/acsami.5c02074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Bioorthogonal nanozymes offer in situ activation of pro-dyes and prodrugs using abiotic chemical transformations. Bacterial infections, especially biofilm-associated infections, are extremely difficult to treat due to obstacles such as poor antibiotic penetration and the rising threat of antibiotic resistance. Spatiotemporal control of bioorthogonal catalysis provides a strategy for "on-demand" generation of therapeutics, effectively localizing therapeutic action and minimizing side effects. Here, we present the fabrication of visible-light-responsive alginate hydrogel beads embedded with bioorthogonal polyzymes (PZs). Exposure to a 405 nm light induces the reduction of Fe(III) to Fe(II), triggering the dissolution of the PZ-gel beads with concomitant release and activation of the polyzyme. This approach enabled the selective activation of a prodrug of Linezolid, a last-in-line antibiotic for Gram-positive bacterial infections, enabling the targeted eradication of multidrug-resistantStaphylococcus aureus biofilms. Overall, the use of alginate biomaterial along with noninvasive visible light offers a nontoxic platform for spatiotemporal release of antibiotics through bioorthogonal activation.
Collapse
Affiliation(s)
- Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Muhammad Aamir Hassan
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - William Ndugire
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Jungmi Park
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Sadaf Noor
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Soham Chakraborty
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
2
|
Hou XX, Wang S, Ma XX, Wen Y, Li ZJ, Liu XY, Zhang X, Zhang Y, Qin XY. Design, Synthesis, and Biological Evaluation of Novel Hydroxytyrosol Derivatives as Protectors for Vascular Endothelium Against Lipid Overload. Drug Des Devel Ther 2025; 19:2433-2452. [PMID: 40190807 PMCID: PMC11971996 DOI: 10.2147/dddt.s500670] [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: 11/09/2024] [Accepted: 03/18/2025] [Indexed: 04/09/2025] Open
Abstract
Background and Objective Hydroxytyrosol (HT) is reported to protect endothelial cells against metabolic overload through inhibiting inflammation. However, the hydrophilic nature of HT limits its oral bioavailability and biological efficiency. The aim of the study was to design and synthesize novel hybrid molecules to improve HT's biological efficiency. Materials and Methods A pharmacophore connection strategy was used to design and synthesize novel hybrid molecules by combining HT or its analogues with adamantane (ADM). Palmitic acid (PA) was used to induce lipid overload in HAEC cells, and P407 was used to induce acute hyperlipidemia in C57 mice. Results We found that DP-ADM, combining ADM and dopamine (a HT analogues), exhibited potent protective effects against metabolic overload-induced endothelial dysfunction. DP-ADM showed low toxicity and inhibited inflammation in response to PA overload in cultured endothelial cells. Additionally, it (30 mg/kg) decreased circulating lipids to an extent similar to HT in a mouse model of hyperlipidemia and was superior to HT in decreasing circulating inflammatory cytokine. It was also superior to HT in improving vascular endothelial function in mice with hyperlipidemia. Mechanistically, DP-ADM inactivated MAPK signaling, as evidenced by downregulated phosphorylation of p38 and Erk. Inhibition of MAPK or NF-κB abolished the anti-inflammatory effect of DP-ADM. Specifically, DP-ADM activated FoxO1 signaling and increased mitochondrial biogenesis in endothelial cells. Conclusion Overall, DP-ADM is a superior form of HT, highlighting its potential therapeutic use in improving endothelial function in metabolic diseases.
Collapse
Affiliation(s)
- Xi-Xi Hou
- College of Life Sciences, Northwest University, Xi’an, People’s Republic of China
- Department of Chemistry, School of Pharmacy, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Shuang Wang
- College of Life Sciences, Northwest University, Xi’an, People’s Republic of China
- Department of Recuperation and rehabilitation for flight personnel, School of aerospace medicine, fourth Military Medical University, Xi’an, People’s Republic of China
| | - Xiao-Xia Ma
- College of Life Sciences, Northwest University, Xi’an, People’s Republic of China
- Department of Chemistry, School of Pharmacy, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Ying Wen
- College of Life Sciences, Northwest University, Xi’an, People’s Republic of China
- Department of Recuperation and rehabilitation for flight personnel, School of aerospace medicine, fourth Military Medical University, Xi’an, People’s Republic of China
| | - Zhi-Jun Li
- School of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi’an, People’s Republic of China
| | - Xu-Yun Liu
- Center for Mitochondrial Biology and Medicine, the Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiao Tong University, Xi’an, People’s Republic of China
| | - Xing Zhang
- Department of Recuperation and rehabilitation for flight personnel, School of aerospace medicine, fourth Military Medical University, Xi’an, People’s Republic of China
| | - Yang Zhang
- Department of Medical Electronics, School of Biomedical Engineering, Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Xiang-Yang Qin
- Department of Chemistry, School of Pharmacy, Fourth Military Medical University, Xi’an, People’s Republic of China
| |
Collapse
|
3
|
Sundararaman SA, Miller JJ, Daley EC, O’Brien KA, Kasak P, Daniels AM, Edwards RL, Heidel KM, Bague DA, Wilson MA, Koelper AJ, Kourtoglou EC, White AD, August SA, Apple GA, Rouamba RW, Durand AJ, Esteb JJ, Muller FL, Johnson RJ, Hoops GC, Dowd CS, Odom John AR. Prodrug activation in malaria parasites mediated by an imported erythrocyte esterase, acylpeptide hydrolase (APEH). Proc Natl Acad Sci U S A 2025; 122:e2417682122. [PMID: 40035755 PMCID: PMC11912422 DOI: 10.1073/pnas.2417682122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
The continued emergence of antimalarial drug resistance highlights the need to develop new antimalarial therapies. Unfortunately, new drug development is often hampered by undesirable drug-like properties of lead compounds. Prodrug approaches temporarily mask undesirable compound features, improving bioavailability and target penetration. We have found that lipophilic diester prodrugs of phosphonic acid antibiotics, such as fosmidomycin (Fsm), exhibit significantly higher antimalarial potency than their parent compounds [R.L. Edwards et al., Sci. Rep. 7, 8400 (2017)]. However, the activating enzymes for these prodrugs were unknown. Here, we show that an erythrocyte enzyme, acylpeptide hydrolase (APEH), is the major activating enzyme of multiple lipophilic ester prodrugs. Surprisingly, this enzyme is taken up by the malaria parasite, Plasmodium falciparum, where it localizes to the parasite cytoplasm and retains enzymatic activity. Using a fluorogenic ester library, we characterize the structure-activity relationship of APEH and compare it to that of P. falciparum esterases. We show that parasite-internalized APEH plays an important role in the activation of substrates with branching at the alpha carbon, in keeping with its exopeptidase activity. Our findings highlight a mechanism for antimicrobial prodrug activation, relying on a host-derived enzyme to yield activation at a microbial target. Mutations in prodrug-activating enzymes are a common mechanism for antimicrobial drug resistance [E. S. Istvan et al., Nat. Commun. 8, 14240 (2017); K. M. V. Sindhe et al., mBio 11, e02640-19 (2020); J. H. Butler et al., Acs Infect Dis. 6, 2994-3003 (2020)]. Leveraging an internalized host enzyme would circumvent this, enabling the design of prodrugs with higher barriers to drug resistance.
Collapse
Affiliation(s)
- Sesh A. Sundararaman
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Justin J. Miller
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA19104
| | - Ellora C. Daley
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Kelsey A. O’Brien
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Paulina Kasak
- College of Health Professions, Thomas Jefferson University, Philadelphia, PA19107
| | - Abigail M. Daniels
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Rachel L. Edwards
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO63110
- Omniose, Saint Louis, MO63110
| | - Kenneth M. Heidel
- Department of Chemistry, George Washington University, Washington, DC20052
| | - Darean A. Bague
- Department of Chemistry, George Washington University, Washington, DC20052
| | - Madeleine A. Wilson
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Andrew J. Koelper
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Elexi C. Kourtoglou
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Alex D. White
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Sloan A. August
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Georgia A. Apple
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Regis W. Rouamba
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Anthony J. Durand
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - John J. Esteb
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | | | - R. Jeremy Johnson
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Geoffrey C. Hoops
- Department of Chemistry and Biochemistry, Butler University, Indianapolis, IN46208
| | - Cynthia S. Dowd
- Department of Chemistry, George Washington University, Washington, DC20052
| | - Audrey R. Odom John
- Department of Pediatrics, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
4
|
Akram F, Fatima T, Shabbir I, Haq IU, Ibrar R, Mukhtar H. Abridgement of Microbial Esterases and Their Eminent Industrial Endeavors. Mol Biotechnol 2025; 67:817-833. [PMID: 38461181 DOI: 10.1007/s12033-024-01108-7] [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: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/11/2024]
Abstract
Esterases are hydrolases that contribute to the hydrolysis of ester bonds into both water-soluble acyl esters and emulsified glycerol-esters containing short-chain acyl groups. They have garnered significant attention from biotechnologists and organic chemists due to their immense commercial value. Esterases, with their diverse and significant properties, have become highly sought after for various industrial applications. Synthesized ubiquitously by a wide range of living organisms, including animals, plants, and microorganisms, these enzymes have found microbial esterases to be the preferred choice in industrial settings. The cost-effective production of microbial esterases ensures higher yields, unaffected by seasonal variations. Their applications span diverse sectors, such as food manufacturing, leather tanneries, paper and pulp production, textiles, detergents, cosmetics, pharmaceuticals, biodiesel synthesis, bioremediation, and waste treatment. As the global trend shifts toward eco-friendly and sustainable practices, industrial processes are evolving with reduced waste generation, lower energy consumption, and the utilization of biocatalysts derived from renewable and unconventional raw materials. This review explores the background, structural characteristics, thermostability, and multifaceted roles of bacterial esterases in crucial industries, aiming to optimize and analyze their properties for continued successful utilization in diverse industrial processes. Additionally, recent advancements in esterase research are overviewed, showcasing novel techniques, innovations, and promising areas for further exploration.
Collapse
Affiliation(s)
- Fatima Akram
- Institute of Industrial Biotechnology, Government College University, Lahore, 54000, Pakistan.
| | - Taseer Fatima
- Institute of Industrial Biotechnology, Government College University, Lahore, 54000, Pakistan
| | - Ifrah Shabbir
- Institute of Industrial Biotechnology, Government College University, Lahore, 54000, Pakistan
| | - Ikram Ul Haq
- Institute of Industrial Biotechnology, Government College University, Lahore, 54000, Pakistan
- Pakistan Academy of Sciences, Islamabad, Pakistan
| | - Ramesha Ibrar
- Institute of Industrial Biotechnology, Government College University, Lahore, 54000, Pakistan
| | - Hamid Mukhtar
- Institute of Industrial Biotechnology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
5
|
de Souza MM, Gini ALR, Moura JA, Scarim CB, Chin CM, dos Santos JL. Prodrug Approach as a Strategy to Enhance Drug Permeability. Pharmaceuticals (Basel) 2025; 18:297. [PMID: 40143076 PMCID: PMC11946379 DOI: 10.3390/ph18030297] [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: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025] Open
Abstract
Absorption and permeability are critical physicochemical parameters that must be balanced to achieve optimal drug uptake. These key factors are closely linked to the maximum absorbable dose required to provide appropriate plasma levels of drugs. Among the various strategies employed to enhance drug solubility and permeability, prodrug design stands out as a highly effective and versatile approach for improving physicochemical properties and enabling the optimization of biopharmaceutical and pharmacokinetic parameters while mitigating adverse effects. Prodrugs are compounds with reduced or no activity that, through bio-reversible chemical or enzymatic processes, release an active parental drug. The application of this technology has led to significant advancements in drug optimization during the design phase, and it offers broad potential for further development. Notably, approximately 13% of the drugs approved by the U.S. Food and Drug Administration (FDA) between 2012 and 2022 were prodrugs. In this review article, we will explore the application of prodrug strategies to enhance permeability, describing examples of market drugs. We also describe the use of the prodrug approach to optimize PROteolysis TArgeting Chimeras (PROTACs) permeability by using conjugation technologies. We will highlight some new technologies in prodrugs to enrich permeability properties, contributing to developing new effective and safe prodrugs.
Collapse
Affiliation(s)
- Mateus Mello de Souza
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Ana Luísa Rodriguez Gini
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Jhonnathan Alves Moura
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| | - Cauê Benito Scarim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Union of the Colleges of the Great Lakes (UNILAGO), School of Medicine, Advanced Research Center in Medicine (CEPAM), Sao Jose do Rio Preto 15030-070, SP, Brazil
| | - Jean Leandro dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| |
Collapse
|
6
|
Madikonda AK, Ajayakumar A, Nadendla S, Banothu J, Muripiti V. Esterase-responsive nanoparticles (ERN): A targeted approach for drug/gene delivery exploits. Bioorg Med Chem 2024; 116:118001. [PMID: 39556942 DOI: 10.1016/j.bmc.2024.118001] [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: 09/02/2024] [Revised: 10/29/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
Nanoparticles are being developed to enhance drug delivery to cancer tumors, leveraging advantages such as the enhanced permeability and retention (EPR) effect. However, traditional nanoparticles often face challenges with low specificity for cancer cells, leading to inefficient delivery and unwanted side effects. Esterase-responsive nanoparticles offer a maximum targeted approach to tumor cells because they release their therapeutic payload at the tumor site under the influence of esterase activity. This review explores the role of esterase-responsive nanoparticles in drug and gene delivery, examines esterase prodrug therapy, and discusses prostate-specific membrane antigen (PSMA) targets esterase-responsive nanoparticles in prostate cancer treatment. Additionally, we reviewed the current research progress and future potential of esterase-responsive nanoparticles in enhancing drug and gene delivery.
Collapse
Affiliation(s)
- Ashok Kumar Madikonda
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Amritha Ajayakumar
- Department of Biochemistry & Molecular Biology, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Sudeena Nadendla
- Department of Chemistry, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India
| | - Janardhan Banothu
- Department of Chemistry, National Institute of Technology Calicut, Kozhikode 673601, Kerala, India
| | - Venkanna Muripiti
- Department of Education, Central University of Kerala, Tejaswini Hills, Periye, Kasaragod 671320, Kerala, India.
| |
Collapse
|
7
|
Bannon MS, Ellena JF, Gourishankar AS, Marsh SR, Trevisan-Silva D, Sherman NE, Jourdan LJ, Gourdie RG, Letteri RA. Multi-site esterification: a tunable, reversible strategy to tailor therapeutic peptides for delivery. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2024; 9:1215-1227. [PMID: 39281343 PMCID: PMC11395315 DOI: 10.1039/d4me00072b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024]
Abstract
Peptides are naturally potent and selective therapeutics with massive potential; however, low cell membrane permeability limits their clinical implementation, particularly for hydrophilic, anionic peptides with intracellular targets. To overcome this limitation, esterification of anionic carboxylic acids on therapeutic peptides can simultaneously increase hydrophobicity and net charge to facilitate cell internalization, whereafter installed esters can be cleaved hydrolytically to restore activity. To date, however, most esterified therapeutics contain either a single esterification site or multiple esters randomly incorporated on multiple sites. This investigation provides molecular engineering insight into how the number and position of esters installed onto the therapeutic peptide α carboxyl terminus 11 (αCT11, RPRPDDLEI) with 4 esterification sites affect hydrophobicity and the hydrolysis process that reverts the peptide to its original form. After installing methyl esters onto αCT11 using Fischer esterification, we isolated 5 distinct products and used 2D nuclear magnetic resonance spectroscopy, reverse-phase high performance liquid chromatography, and mass spectrometry to determine which residues were esterified in each and the resulting increase in hydrophobicity. We found esterifying the C-terminal isoleucine to impart the largest increase in hydrophobicity. Monitoring ester hydrolysis showed the C-terminal isoleucine ester to be the most hydrolytically stable, followed by the glutamic acid, whereas esters on aspartic acids hydrolyze rapidly. LC-MS revealed the formation of transient intramolecular aspartimides prior to hydrolysis to carboxylic acids. In vitro proof-of-concept experiments showed esterifying αCT11 to increase cell migration into a scratch, highlighting the potential of multi-site esterification as a tunable, reversible strategy to enable the delivery of therapeutic peptides.
Collapse
Affiliation(s)
- Mark S Bannon
- Department of Chemical Engineering, University of Virginia Charlottesville VA 22903 USA +1 434 243 3628
| | - Jeffrey F Ellena
- Biomolecular Magnetic Resonance Facility, School of Medicine, University of Virginia Charlottesville VA 22903 USA
| | - Aditi S Gourishankar
- Department of Chemical Engineering, University of Virginia Charlottesville VA 22903 USA +1 434 243 3628
| | - Spencer R Marsh
- Fralin Biomedical Institute, Virginia Tech Carillion School of Medicine Roanoke VA 24016 USA
| | - Dilza Trevisan-Silva
- Biomolecular Analysis Facility, School of Medicine, University of Virginia Charlottesville VA 22903 USA
| | - Nicholas E Sherman
- Biomolecular Analysis Facility, School of Medicine, University of Virginia Charlottesville VA 22903 USA
| | - L Jane Jourdan
- Fralin Biomedical Institute, Virginia Tech Carillion School of Medicine Roanoke VA 24016 USA
| | - Robert G Gourdie
- Fralin Biomedical Institute, Virginia Tech Carillion School of Medicine Roanoke VA 24016 USA
| | - Rachel A Letteri
- Department of Chemical Engineering, University of Virginia Charlottesville VA 22903 USA +1 434 243 3628
| |
Collapse
|
8
|
Pertino MW, F. de la Torre A, Schmeda-Hirschmann G, Vega Gómez C, Rolón M, Coronel C, Rojas de Arias A, Molina-Torres CA, Vera-Cabrera L, Viveros-Valdez E. Exploring Benzo[h]chromene Derivatives as Agents against Protozoal and Mycobacterial Infections. Pharmaceuticals (Basel) 2024; 17:1375. [PMID: 39459014 PMCID: PMC11510191 DOI: 10.3390/ph17101375] [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: 10/01/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: In this study, the efficacy of benzo[h]chromene derivatives as antiprotozoal and antimycobacterial agents was explored. Methods: A total of twenty compounds, including benzo[h]chromene alkyl diesters and benzo[h]chromene-triazole derivatives, were synthesized and tested against Trypanosoma cruzi, Leishmania braziliensis, L. infantum, and strains of Mycobacterium abscessus and Mycobacterium intracellulare LIID-01. Notably, compounds 1a, 1b, 2a, and 3f exhibited superior activity against Trypanosoma cruzi, with IC50 values of 19.2, 37.3, 68.7, and 24.7 µM, respectively, outperforming the reference drug benznidazole (IC50: 54.7 µM). Results: Compounds 1b and 3f showed excellent selectivity indices against Leishmania braziliensis, with SI values of 19 and 18, respectively, suggesting they could be potential alternatives to the commonly used, but more selective, miltefosine (IC50: 64.0 µM, SI: 43.0). Additionally, compounds 1a, 1b, and 3f were most effective against Leishmania infantum, with IC50 values of 24.9, 30.5, and 46.6 µM, respectively. Compounds 3f and 3h were particularly potent against various Mycobacterium abscessus strains, highlighting their significance given the inherent resistance of these bacteria to standard antimicrobials. Conclusions: The sensitivity of Mycobacterium intracellulare LIID-01 to these compounds also underscored their potential in managing infections by the Mycobacterium avium-intracellulare complex.
Collapse
Affiliation(s)
- Mariano Walter Pertino
- Instituto de Química de Recursos Naturales, Universidad de Talca, Campus Lircay, Talca 3480094, Chile;
| | - Alexander F. de la Torre
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | | | - Celeste Vega Gómez
- Centro para el Desarrollo de la Investigación Científica (CEDIC), Manduvirá 635, Asunción CP 1255, Paraguay; (C.V.G.); (M.R.); (C.C.); (A.R.d.A.)
| | - Miriam Rolón
- Centro para el Desarrollo de la Investigación Científica (CEDIC), Manduvirá 635, Asunción CP 1255, Paraguay; (C.V.G.); (M.R.); (C.C.); (A.R.d.A.)
| | - Cathia Coronel
- Centro para el Desarrollo de la Investigación Científica (CEDIC), Manduvirá 635, Asunción CP 1255, Paraguay; (C.V.G.); (M.R.); (C.C.); (A.R.d.A.)
| | - Antonieta Rojas de Arias
- Centro para el Desarrollo de la Investigación Científica (CEDIC), Manduvirá 635, Asunción CP 1255, Paraguay; (C.V.G.); (M.R.); (C.C.); (A.R.d.A.)
| | - Carmen A. Molina-Torres
- Servicios de Dermatología, Hospital Universitario “José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico; (C.A.M.-T.); (L.V.-C.)
| | - Lucio Vera-Cabrera
- Servicios de Dermatología, Hospital Universitario “José E. González”, Universidad Autónoma de Nuevo León, Monterrey 64460, NL, Mexico; (C.A.M.-T.); (L.V.-C.)
| | - Ezequiel Viveros-Valdez
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, NL, Mexico;
| |
Collapse
|
9
|
Sundararaman SA, Miller JJ, Daley EC, O'Brien KA, Kasak P, Daniels AM, Edwards RL, Heidel KM, Bague DA, Wilson MA, Koelper AJ, Kourtoglou EC, White AD, August SA, Apple GA, Rouamba RW, Durand AJ, Esteb JJ, Muller FL, Johnson RJ, Hoops GC, Dowd CS, Odom John AR. Prodrug activation in malaria parasites mediated by an imported erythrocyte esterase, acylpeptide hydrolase (APEH). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610542. [PMID: 39257815 PMCID: PMC11383709 DOI: 10.1101/2024.08.30.610542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The continued emergence of antimalarial drug resistance highlights the need to develop new antimalarial therapies. Unfortunately, new drug development is often hampered by poor drug-like properties of lead compounds. Prodrugging temporarily masks undesirable compound features, improving bioavailability and target penetration. We have found that lipophilic diester prodrugs of phosphonic acid antibiotics, such as fosmidomycin, exhibit significantly higher antimalarial potency than their parent compounds (1). However, the activating enzymes for these prodrugs were unknown. Here, we show that an erythrocyte enzyme, acylpeptide hydrolase (APEH) is the major activating enzyme of multiple lipophilic ester prodrugs. Surprisingly, this enzyme is taken up by the malaria parasite, Plasmodium falciparum, where it localizes to the parasite cytoplasm and retains enzymatic activity. Using a novel fluorogenic ester library, we characterize the structure activity relationship of APEH, and compare it to that of P. falciparum esterases. We show that parasite-internalized APEH plays an important role in the activation of substrates with branching at the alpha carbon, in keeping with its exopeptidase activity. Our findings highlight a novel mechanism for antimicrobial prodrug activation, relying on a host-derived enzyme to yield activation at a microbial target. Mutations in prodrug activating enzymes are a common mechanism for antimicrobial drug resistance (2-4). Leveraging an internalized host enzyme would circumvent this, enabling the design of prodrugs with higher barriers to drug resistance.
Collapse
Affiliation(s)
- S A Sundararaman
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - J J Miller
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, USA
| | - E C Daley
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - K A O'Brien
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - P Kasak
- College of Health Professions, Thomas Jefferson University, Philadelphia, PA, USA
| | - A M Daniels
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, USA
| | - R L Edwards
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, Missouri, USA
- Omniose, Saint Louis, MO, USA
| | - K M Heidel
- Department of Chemistry, George Washington University, 800 22nd Street NW, Washington DC, USA
| | - D A Bague
- Department of Chemistry, George Washington University, 800 22nd Street NW, Washington DC, USA
| | - M A Wilson
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - A J Koelper
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - E C Kourtoglou
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - A D White
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - S A August
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - G A Apple
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - R W Rouamba
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - A J Durand
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - J J Esteb
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - F L Muller
- Lindonlight Collective, Houston, TX, USA
| | - R J Johnson
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - G C Hoops
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave, Indianapolis, IN, USA
| | - C S Dowd
- Department of Chemistry, George Washington University, 800 22nd Street NW, Washington DC, USA
| | - A R Odom John
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Goss AL, Shudick RE, Johnson RJ. Shifting Mycobacterial Serine Hydrolase Activity Visualized Using Multi-Layer In-Gel Activity Assays. Molecules 2024; 29:3386. [PMID: 39064965 PMCID: PMC11279797 DOI: 10.3390/molecules29143386] [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: 06/20/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The ability of Mycobacterium tuberculosis to derive lipids from the host, store them intracellularly, and then break them down into energy requires a battery of serine hydrolases. Serine hydrolases are a large, diverse enzyme family with functional roles in dormant, active, and reactivating mycobacterial cultures. To rapidly measure substrate-dependent shifts in mycobacterial serine hydrolase activity, we combined a robust mycobacterial growth system of nitrogen limitation and variable carbon availability with nimble in-gel fluorogenic enzyme measurements. Using this methodology, we rapidly analyzed a range of ester substrates, identified multiple hydrolases concurrently, observed functional enzyme shifts, and measured global substrate preferences. Within every growth condition, mycobacterial hydrolases displayed the full, dynamic range of upregulated, downregulated, and constitutively active hydrolases independent of the ester substrate. Increasing the alkyl chain length of the ester substrate also allowed visualization of distinct hydrolase activity likely corresponding with lipases most responsible for lipid breakdown. The most robust expression of hydrolase activity was observed under the highest stress growth conditions, reflecting the induction of multiple metabolic pathways scavenging for energy to survive under this high stress. The unique hydrolases present under these high-stress conditions could represent novel drug targets for combination treatment with current front-line therapeutics. Combining diverse fluorogenic esters with in-gel activity measurements provides a rapid, customizable, and sensitive detection method for mycobacterial serine hydrolase activity.
Collapse
Affiliation(s)
| | | | - R. Jeremy Johnson
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave., Indianapolis, IN 46208, USA
| |
Collapse
|
11
|
Singh A, Ottavi S, Krieger I, Planck K, Perkowski A, Kaneko T, Davis AM, Suh C, Zhang D, Goullieux L, Alex A, Roubert C, Gardner M, Preston M, Smith DM, Ling Y, Roberts J, Cautain B, Upton A, Cooper CB, Serbina N, Tanvir Z, Mosior J, Ouerfelli O, Yang G, Gold BS, Rhee KY, Sacchettini JC, Fotouhi N, Aubé J, Nathan C. Redirecting raltitrexed from cancer cell thymidylate synthase to Mycobacterium tuberculosis phosphopantetheinyl transferase. SCIENCE ADVANCES 2024; 10:eadj6406. [PMID: 38489355 PMCID: PMC10942122 DOI: 10.1126/sciadv.adj6406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024]
Abstract
There is a compelling need to find drugs active against Mycobacterium tuberculosis (Mtb). 4'-Phosphopantetheinyl transferase (PptT) is an essential enzyme in Mtb that has attracted interest as a potential drug target. We optimized a PptT assay, used it to screen 422,740 compounds, and identified raltitrexed, an antineoplastic antimetabolite, as the most potent PptT inhibitor yet reported. While trying unsuccessfully to improve raltitrexed's ability to kill Mtb and remove its ability to kill human cells, we learned three lessons that may help others developing antibiotics. First, binding of raltitrexed substantially changed the configuration of the PptT active site, complicating molecular modeling of analogs based on the unliganded crystal structure or the structure of cocrystals with inhibitors of another class. Second, minor changes in the raltitrexed molecule changed its target in Mtb from PptT to dihydrofolate reductase (DHFR). Third, the structure-activity relationship for over 800 raltitrexed analogs only became interpretable when we quantified and characterized the compounds' intrabacterial accumulation and transformation.
Collapse
Affiliation(s)
- Amrita Singh
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Samantha Ottavi
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Inna Krieger
- Department of Biochemistry and Biophysics, Texas Agricultural and Mechanical University, College Station, TX 77843, USA
| | - Kyle Planck
- Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Andrew Perkowski
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Takushi Kaneko
- Global Alliance for TB Drug Development, New York, NY 10005, USA
| | | | - Christine Suh
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| | - David Zhang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| | | | - Alexander Alex
- AMG Consultants Limited, Camburgh House, 27 New Dover Road, Canterbury, Kent, CT1 3DN, UK
- Evenor Consulting Limited, The New Barn, Mill Lane, Eastry, Kent CT13 0JW, UK
| | | | - Mark Gardner
- AMG Consultants Limited, Camburgh House, 27 New Dover Road, Canterbury, Kent, CT1 3DN, UK
| | - Marian Preston
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Dave M. Smith
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, UK
| | - Yan Ling
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Julia Roberts
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Bastien Cautain
- Evotec ID (Lyon), SAS 40 Avenue Tony Garnier, Lyon 69001, France
| | - Anna Upton
- Evotec ID (Lyon), SAS 40 Avenue Tony Garnier, Lyon 69001, France
| | | | - Natalya Serbina
- Global Alliance for TB Drug Development, New York, NY 10005, USA
| | - Zaid Tanvir
- Global Alliance for TB Drug Development, New York, NY 10005, USA
| | - John Mosior
- Department of Biochemistry and Biophysics, Texas Agricultural and Mechanical University, College Station, TX 77843, USA
| | - Ouathek Ouerfelli
- Organic Synthesis Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Guangli Yang
- Organic Synthesis Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ben S. Gold
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Kyu Y. Rhee
- Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - James C. Sacchettini
- Department of Biochemistry and Biophysics, Texas Agricultural and Mechanical University, College Station, TX 77843, USA
| | - Nader Fotouhi
- Global Alliance for TB Drug Development, New York, NY 10005, USA
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York 10021, USA
| |
Collapse
|
12
|
Wang Y, Xie F, He Z, Che L, Chen X, Yuan Y, Liu C. Senescence-Targeted and NAD +-Dependent SIRT1-Activated Nanoplatform to Counteract Stem Cell Senescence for Promoting Aged Bone Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304433. [PMID: 37948437 DOI: 10.1002/smll.202304433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/03/2023] [Indexed: 11/12/2023]
Abstract
Age-related bone defects are a leading cause of disability and mortality in elderly individuals, and targeted therapy to delay the senescence of bone marrow-derived mesenchymal stem cells (MSCs) has emerged as a promising strategy to rejuvenate bone regeneration in aged scenarios. More specifically, activating the nicotinamide adenine dinucleotide (NAD+)-dependent sirtuin 1 (SIRT1) pathway is demonstrated to effectively counteract MSC senescence and thus promote osteogenesis. Herein, based on an inventively identified senescent MSC-specific surface marker Kremen1, a senescence-targeted and NAD+ dependent SIRT1 activated nanoplatform is fabricated with a dual delivery of resveratrol (RSV) (SIRT1 promoter) and nicotinamide riboside (NR, NAD+ precursor). This targeting nanoplatform exhibits a strong affinity for senescent MSCs through conjugation with anti-Kremen1 antibodies and enables specifically responsive release of NR and RSV in lysosomes via senescence-associated β-galactosidase-stimulated enzymatic hydrolysis of the hydrophilic chain. Furthermore, this nanoplatform performs well in promoting aged bone formation both in vitro and in vivo by boosting NAD+, activating SIRT1, and delaying MSC senescence. For the first time, a novel senescent MSC-specific surface marker is identified and aged bone repair is rejuvenated by delaying senescence of MSCs using an active targeting platform. This discovery opens up new insights for nanotherapeutics aimed at age-related diseases.
Collapse
Affiliation(s)
- Ying Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Fangru Xie
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Zirui He
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Lingbin Che
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, P. R. China
| | - Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| |
Collapse
|
13
|
Xu X, Jia Z, Chen N, Lele SM, Arash S, Reinhardt RA, Killeen AC, Wang D. The Development of Thermoresponsive Polymeric Simvastatin Prodrug for the Treatment of Experimental Periodontitis in Rats. Mol Pharm 2023; 20:5631-5645. [PMID: 37772991 DOI: 10.1021/acs.molpharmaceut.3c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Periodontitis (PD) is a severe inflammatory gum pathology that damages the periodontal soft tissue and bone. It is highly prevalent in the US, affecting more than 47% of adults. Besides routine scaling and root planing, there are few effective treatments for PD. Developed as an effective treatment for hyperlipidemia, simvastatin (SIM) is also known for its well-established anti-inflammatory and osteogenic properties, suggesting its potential utility in treating PD. Its clinical translation, however, has been impeded by its poor water-solubility, lack of osteotropicity, and side effects (e.g., hepatoxicity) associated with systemic exposure. To address these challenges, an N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer-based thermoresponsive polymeric prodrug of SIM (ProGel-SIM) was developed as a local therapy for PD. Its aqueous solution is free-flowing at 4 °C and transitions into a hydrogel at ∼30 °C, allowing for easy local application and retention. After a thorough characterization of its physicochemical properties, ProGel-SIM was administered weekly into the periodontal pocket of an experimental rat model of PD. At 3 weeks post initiation of the treatment, the animals were euthanized with palate isolated for μ-CT and histological analyses. When compared to dose equivalent simvastatin acid (SMA, active form of SIM) treatment, the rats in the ProGel-SIM treated group showed significantly higher periodontal bone volume (0.34 mm3 vs 0.20 mm3, P = 0.0161) and less neutrophil (PMN) infiltration (P < 0.0001) and IL-1β secretion (P = 0.0036). No measurable side effect was observed. Collectively, these results suggest that ProGel-SIM may be developed as a promising drug candidate for the effective clinical treatment of PD.
Collapse
|
14
|
Sun C, Liu B, Zhou F, Zheng Q, Dai C, Wei W, Liao G, Sun Y. Assessment of Purity, Stability, and Pharmacokinetics of NGP-1, a Novel Prodrug of GS441254 with Potential Anti-SARS-CoV-2 Activity, Using Liquid Chromatography. Molecules 2023; 28:5634. [PMID: 37570604 PMCID: PMC10420250 DOI: 10.3390/molecules28155634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
SARS-CoV-2 is a highly contagious and pathogenic virus that first appeared in late December 2019 and caused a global pandemic in a short period. The virus is a single-stranded RNA virus belonging to the Coronaviridae family. Numerous treatments have been developed and tested in response to the pandemic, particularly antiviral drugs. Among them, GS441524 (GS441), a nucleoside antiviral drug, has demonstrated promising results in inhibiting SARS-CoV-2. Nevertheless, the limited oral bioavailability of GS441 restricts its application to patients with the virus. In this study, a novel prodrug of GS441 (NGP-1) with an isobutyl ester and cyclic carbonate structure was designed and synthesized. Its purity and the stability in different artificial digestive juices of NGP-1 was determined with HPLC-DAD methods. The pharmacokinetics of NGP-1 and GS441 were studied in rats via gavage administration. A new LC-MS/MS method was developed to quantitatively analyze GS441 in plasma samples. The results showed that the ka, Cmax, and MRT of converted GS441 from NGP-1 were 5.9, 3, and 2.5 times greater than those of GS441 alone. The Frel of NGP-1 was approximately four-fold that of GS441, with an AUC0-∞ of 9716.3 h·ng mL-1. As a prodrug of GS441, NGP-1 increased its lipophilicity, absorption, and bioavailability, indicating that it holds promise in improving the clinical efficacy of anti-SARS-CoV-2 medications.
Collapse
Affiliation(s)
- Chen Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| | - Bo Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| | - Fengzhi Zhou
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| | - Qianqian Zheng
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| | - Chunmei Dai
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| | - Wei Wei
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| | - Guochao Liao
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yuqi Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
15
|
Schemenauer D, Pool EH, Raynor SN, Ruiz GP, Goehring LM, Koelper AJ, Wilson MA, Durand AJ, Kourtoglou EC, Larsen EM, Lavis LD, Esteb JJ, Hoops GC, Johnson RJ. Sequence and Structural Motifs Controlling the Broad Substrate Specificity of the Mycobacterial Hormone-Sensitive Lipase LipN. ACS OMEGA 2023; 8:13252-13264. [PMID: 37065048 PMCID: PMC10099132 DOI: 10.1021/acsomega.3c00534] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/24/2023] [Indexed: 06/19/2023]
Abstract
Mycobacterium tuberculosis has a complex life cycle transitioning between active and dormant growth states depending on environmental conditions. LipN (Rv2970c) is a conserved mycobacterial serine hydrolase with regulated catalytic activity at the interface between active and dormant growth conditions. LipN also catalyzes the xenobiotic degradation of a tertiary ester substrate and contains multiple conserved motifs connected with the ability to catalyze the hydrolysis of difficult tertiary ester substrates. Herein, we expanded a library of fluorogenic ester substrates to include more tertiary and constrained esters and screened 33 fluorogenic substrates for activation by LipN, identifying its unique substrate signature. LipN preferred short, unbranched ester substrates, but had its second highest activity against a heteroaromatic five-membered oxazole ester. Oxazole esters are present in multiple mycobacterial serine hydrolase inhibitors but have not been tested widely as ester substrates. Combined structural modeling, kinetic measurements, and substitutional analysis of LipN showcased a fairly rigid binding pocket preorganized for catalysis of short ester substrates. Substitution of diverse amino acids across the binding pocket significantly impacted the folded stability and catalytic activity of LipN with two conserved motifs (HGGGW and GDSAG) playing interconnected, multidimensional roles in regulating its substrate specificity. Together this detailed substrate specificity profile of LipN illustrates the complex interplay between structure and function in mycobacterial hormone-sensitive lipase homologues and indicates oxazole esters as promising inhibitor and substrate scaffolds for mycobacterial hydrolases.
Collapse
Affiliation(s)
- Daniel
E. Schemenauer
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Emily H. Pool
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Stephanie N. Raynor
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Gabriela P. Ruiz
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Leah M. Goehring
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Andrew J. Koelper
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Madeleine A. Wilson
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Anthony J. Durand
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Elexi C. Kourtoglou
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Erik M. Larsen
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Luke D. Lavis
- Howard
Hughes Medical Institute, Janelia Research Campus, Ashburn, Virginia 20147, United States
| | - John J. Esteb
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - Geoffrey C. Hoops
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| | - R. Jeremy Johnson
- Department
of Chemistry and Biochemistry, Butler University, Indianapolis, Indiana 46208, United States
| |
Collapse
|
16
|
Jörgensen AM, Wibel R, Bernkop-Schnürch A. Biodegradable Cationic and Ionizable Cationic Lipids: A Roadmap for Safer Pharmaceutical Excipients. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206968. [PMID: 36610004 DOI: 10.1002/smll.202206968] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Cationic and ionizable cationic lipids are broadly applied as auxiliary agents, but their use is associated with adverse effects. If these excipients are rapidly degraded to endogenously occurring metabolites such as amino acids and fatty acids, their toxic potential can be minimized. So far, synthesized and evaluated biodegradable cationic and ionizable cationic lipids already showed promising results in terms of functionality and safety. Within this review, an overview about the different types of such biodegradable lipids, the available building blocks, their synthesis and cleavage by endogenous enzymes is provided. Moreover, the relationship between the structure of the lipids and their toxicity is described. Their application in drug delivery systems is critically discussed and placed in context with the lead compounds used in mRNA vaccines. Moreover, their use as preservatives is reviewed, guidance for their design is provided, and an outlook on future developments is given.
Collapse
Affiliation(s)
- Arne Matteo Jörgensen
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck, 6020, Austria
| | - Richard Wibel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck, 6020, Austria
| |
Collapse
|
17
|
Rafeeq H, Hussain A, Shabbir S, Ali S, Bilal M, Sher F, Iqbal HMN. Esterases as emerging biocatalysts: Mechanistic insights, genomic and metagenomic, immobilization, and biotechnological applications. Biotechnol Appl Biochem 2022; 69:2176-2194. [PMID: 34699092 DOI: 10.1002/bab.2277] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/20/2021] [Indexed: 02/05/2023]
Abstract
Esterase enzymes are a family of hydrolases that catalyze the breakdown and formation of ester bonds. Esterases have gained a prominent position in today's world's industrial enzymes market. Due to their unique biocatalytic attributes, esterases contribute to environmentally sustainable design approaches, including biomass degradation, food and feed industry, dairy, clothing, agrochemical (herbicides, insecticides), bioremediation, biosensor development, anticancer, antitumor, gene therapy, and diagnostic purposes. Esterases can be isolated by a diverse range of mammalian tissues, animals, and microorganisms. The isolation of extremophilic esterases increases the interest of researchers in the extraction and utilization of these enzymes at the industrial level. Genomic, metagenomic, and immobilization techniques have opened innovative ways to extract esterases and utilize them for a longer time to take advantage of their beneficial activities. The current study discusses the types of esterases, metagenomic studies for exploring new esterases, and their biomedical applications in different industrial sectors.
Collapse
Affiliation(s)
- Hamza Rafeeq
- Department of Biochemistry, Riphah International University, Faisalabad, Pakistan
| | - Asim Hussain
- Department of Biochemistry, Riphah International University, Faisalabad, Pakistan
| | - Sumaira Shabbir
- Department of Zoology, Wildlife, and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Sabir Ali
- Department of Biochemistry, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Farooq Sher
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| |
Collapse
|
18
|
Pais JP, Policarpo M, Pires D, Francisco AP, Madureira AM, Testa B, Anes E, Constantino L. Fluoroquinolone Derivatives in the Treatment of Mycobacterium tuberculosis Infection. Pharmaceuticals (Basel) 2022; 15:ph15101213. [PMID: 36297325 PMCID: PMC9609866 DOI: 10.3390/ph15101213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 12/01/2022] Open
Abstract
Tuberculosis (TB) is currently one of the leading causes of death due to infective agents, and the growing rate of multidrug-resistant tuberculosis (MDR TB) cases poses an emergent public health threat. Fluoroquinolones are commonly used in the treatment of both MDR TB and drug-sensitive tuberculosis patients who are intolerant to first-line antitubercular agents. Unfortunately, these drugs have mild side effects, relevant to the prolonged treatment regimens and diminished bioavailability due to binding of metal ions. Moreover, the resistance to fluoroquinolones is also on the rise, a characteristic of extensively drug-resistant TB (XDR TB). Here, we developed esters as prodrugs of the fluoroquinolones levofloxacin and ciprofloxacin, with long-chain fatty alcohols. Both the alcohols and the quinolone have previously shown antimycobacterial activity and the aim was to develop esters with improved lipophilicity and capable of delivering the free acid inside mycobacterial cells. The carboxylic acid group of fluoroquinolones is essential to the mode of action but is also responsible for many of its side effects and metal-chelating properties. The synthesis, stability in biological media, and antibacterial activity were evaluated, the latter not only against Mycobacterium tuberculosis but also against other clinically relevant bacterial species, since the parent compounds display a broad spectrum of activity. The biological results show a reduction in the antitubercular activity of the synthesized derivatives, probably due to deficient activation of the ester prodrug. Despite this, it was found that the derivatives exhibit bioactivity against other fluoroquinolone-resistant bacteria, indicating a different mode of action and suggesting that it may be worthwhile to research further modifications to the carboxylic acid group. This might lead to new compounds that are efficient against resistant strains. This idea that the compounds may act by a different mechanism of action was further supported by a brief computer investigation that demonstrated the potential lack of selectivity of the esters to the fluoroquinolone target.
Collapse
Affiliation(s)
- João Pedro Pais
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Margarida Policarpo
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - David Pires
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ana Paula Francisco
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ana Margarida Madureira
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | | | - Elsa Anes
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Luís Constantino
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: ; Tel.: +35-19-6548-8519
| |
Collapse
|
19
|
Benzoic Acid Derivatives as Prodrugs for the Treatment of Tuberculosis. Pharmaceuticals (Basel) 2022; 15:ph15091118. [PMID: 36145340 PMCID: PMC9502840 DOI: 10.3390/ph15091118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022] Open
Abstract
One interesting approach to fight tuberculosis is the use of prodrugs that often have shown improved biological activities over drugs with poor absorption or difficulty to cross membranes. Previous studies demonstrate that weak acids such as benzoic acid, present antimycobacterial activity. Moreover, esters of those acids revealed to be a viable alternative since they may diffuse more easily through the cell membranes. Previously we showed that mycobacteria can easily activate benzoic acid esters by conversion to the corresponding acid. Since Zhang postulated that the activity of the acids can be dependent on their pKa, we set up to synthesize a library of benzoates with different electron withdrawing groups (4-chloro, 2,6-dichloro, 3,5-dichloro, 4-nitro, and 3,5 dinitro), to modulate pKa of the liberated acid and different alkoxy substituents (propyl, hexyl, and phenyl) to modulate their lipophilicity, and tested the activity of the esters and the corresponding free acids against mycobacteria. We also studied the activation of the esters by mycobacterial enzymes and the stability of the compounds in buffer and plasma. We concluded that all the benzoates in our study can be activated by mycobacterial enzymes and that the phenyl and hexyl esters presented higher activity than the corresponding free acids, with the nitrobenzoates, and especially the dinitrobenzoates, showing very interesting antitubercular activity that deserve further exploration. Our results did not show a correlation between the activity and the pKa of the acids.
Collapse
|
20
|
Quinine Esters with 1,2-Azole, Pyridine and Adamantane Fragments. Molecules 2022; 27:molecules27113476. [PMID: 35684414 PMCID: PMC9182173 DOI: 10.3390/molecules27113476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
An efficient method of producing quinine derivatives via reaction of acylation with 4,5-dichloroisothiazole-3-, 5-arylisoxazole-3-, adamantane- and hydrochlorides of pyridine-3- and pyridine-4-carbonyl chlorides was developed. All synthesized compounds were tested for antiviral, antimicrobial and analgesic activity. The most pronounced antibacterial activity was shown by the compounds 2e, 3b, 3c and 3e with isoxazole and pyridine fragments. It was found that most of the tested compounds showed significant analgesic activity reducing the pain response of animals to the irritating effect of acetic acid.
Collapse
|
21
|
Identification, Characterization, and Preliminary X-ray Diffraction Analysis of a Novel Esterase (ScEst) from Staphylococcus chromogenes. CRYSTALS 2022. [DOI: 10.3390/cryst12040546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ester prodrugs can develop novel antibiotics and have potential therapeutic applications against multiple drug-resistant bacteria. The antimicrobial activity of these prodrugs is activated after being cleaved by the esterases produced by the pathogen. Here, novel esterase ScEst originating from Staphylococcus chromogenes NCTC10530, which causes dairy cow mastitis, was identified, characterized, and analyzed using X-ray crystallography. The gene encoding ScEst was cloned into the pVFT1S vector and overexpressed in E. coli. The recombinant ScEst protein was obtained by affinity and size-exclusion purification. ScEst showed substrate preference for the short chain length of acyl derivatives. It was crystallized in an optimized solution composed of 0.25 M ammonium citrate tribasic (pH 7.0) and 20% PEG 3350 at 296 K. A total of 360 X-ray diffraction images were collected at a 1.66 Å resolution. ScEst crystal belongs to the space group of P212121 with the unit cell parameters of a = 50.23 Å, b = 68.69 Å, c = 71.15 Å, and α = β = γ = 90°. Structure refinement after molecular replacement is under progress. Further biochemical studies will elucidate the hydrolysis mechanism of ScEst. Overall, this study is the first to report the functional characterization of an esterase from Staphylococcus chromogenes, which is potentially useful in elaborating its hydrolysis mechanism.
Collapse
|
22
|
de Souza HMR, Guedes JS, Freitas RHCN, Gelves LGV, Fokoue HH, Sant’Anna CMR, Barreiro EJ, Lima LM. Comparative chemical and biological hydrolytic stability of homologous esters and isosteres. J Enzyme Inhib Med Chem 2022; 37:718-727. [PMID: 35156494 PMCID: PMC8856110 DOI: 10.1080/14756366.2022.2027933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Hygor M. R. de Souza
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro, Brasil
- Pós-graduação em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Jéssica S. Guedes
- Pós-graduação em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Rosana H. C. N. Freitas
- Pós-graduação em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Luis G. V. Gelves
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro, Brasil
| | - Harold H. Fokoue
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro, Brasil
- Instituto de Tecnologia em Fármacos – Farmanguinhos/FIOCRUZ, Rio de Janeiro, Brasil
| | - Carlos Mauricio R. Sant’Anna
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro, Brasil
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Brasil
| | - Eliezer J. Barreiro
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro, Brasil
- Pós-graduação em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Lidia M. Lima
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio®), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro, Brasil
- Pós-graduação em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
23
|
Sequence Analysis and Preliminary X-ray Crystallographic Analysis of an Acetylesterase (LgEstI) from Lactococcus garvieae. CRYSTALS 2021. [DOI: 10.3390/cryst12010046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A gene encoding LgEstI was cloned from a bacterial fish pathogen, Lactococcus garvieae. Sequence and bioinformatic analysis revealed that LgEstI is close to the acetyl esterase family and had maximum similarity to a hydrolase (UniProt: Q5UQ83) from Acanthamoeba polyphaga mimivirus (APMV). Here, we present the results of LgEstI overexpression and purification, and its preliminary X-ray crystallographic analysis. The wild-type LgEstI protein was overexpressed in Escherichia coli, and its enzymatic activity was tested using p-nitrophenyl of varying lengths. LgEstI protein exhibited higher esterase activity toward p-nitrophenyl acetate. To better understand the mechanism underlying LgEstI activity and subject it to protein engineering, we determined the high-resolution crystal structure of LgEstI. First, the wild-type LgEstI protein was crystallized in 0.1 M Tris-HCl buffer (pH 7.1), 0.2 M calcium acetate hydrate, and 19% (w/v) PEG 3000, and the native X-ray diffraction dataset was collected up to 2.0 Å resolution. The crystal structure was successfully determined using a molecular replacement method, and structure refinement and model building are underway. The upcoming complete structural information of LgEstI may elucidate the substrate-binding mechanism and provide novel strategies for subjecting LgEstI to protein engineering.
Collapse
|
24
|
Dutta B, Nigam VK, Panja AS, Shrivastava S, Bandopadhyay R. Statistical optimisation of esterase from Salinicoccus roseus strain RF1H and its potential application in synthetic dye decolorisation. BIOCATAL BIOTRANSFOR 2021. [DOI: 10.1080/10242422.2021.2010718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Bhramar Dutta
- Department of Botany, The University of Burdwan, Bardhaman, India
| | - Vinod Kumar Nigam
- Department of Bio-Engineering, Birla Institute of Technology, Ranchi, India
| | - Anindya Sundar Panja
- Post-Graduate Department of Biotechnology and Biochemistry, Oriental Institute of Science and Technology, Burdwan, India
| | | | | |
Collapse
|
25
|
Banerjee S, Maiti TK, Roy RN. Enzyme producing insect gut microbes: an unexplored biotechnological aspect. Crit Rev Biotechnol 2021; 42:384-402. [PMID: 34612103 DOI: 10.1080/07388551.2021.1942777] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
To explore the unmapped biotechnologically important microbial platforms for human welfare, the insect gut system is such a promising arena. Insects, the inhabitant of all ecological niches, harbor a healthy diversified microbial population in their versatile gut environment. This deep-rooted symbiotic relationship between insects and gut microbes is the result of several indispensable microbial performances that include: enzyme production, detoxification of plant defense compounds and insecticides, maintenance of life cycle, host fertility, bioremediation, pest biocontrol, production of antimicrobial compounds, and in addition provide vitamins, amino acids, and lactic acids to their hosts. Insects have developed such symbiotic interactions with different microorganisms for nutritional benefits like the digestion of dietary compounds by the production of several key hydrolytic enzymes viz: amylase, cellulase, lignocellulase, protease, lipase, xylanase, pectinase, chitinase, laccase, etc. The nutritional enrichment offered by these microbes to insects may be the key factor in the evolutionary attainment of this group. Around one million insect species are grouped under 31 orders, however, only ten of such groups' have been studied in relation to enzyme-producing gut microbes. Moreover, insect gut symbionts are a potential source of biotechnologically active biomolecules as these microbes go through a course of selection pressures in their host gut environment. As symbiosis has pronounced potential regarding the production of novel compounds, especially enzymes with multidimensional industrial capabilities, so there are ample scopes to explore this treasure box for human welfare. Biological significance as well as industrially compatible capabilities can categorize these insect gut symbionts as an unexplored biotechnological aspect.
Collapse
Affiliation(s)
- Sandipan Banerjee
- Microbiology Research Laboratory, Department of Botany, Dr. B. N. Dutta Smriti Mahavidyalaya, Hatgobindapur, Burdwan, India.,Mycology and Plant Pathology Laboratory, Department of Botany, Visva-Bharati University, Santiniketan, India
| | | | - Raj Narayan Roy
- Microbiology Research Laboratory, Department of Botany, Dr. B. N. Dutta Smriti Mahavidyalaya, Hatgobindapur, Burdwan, India
| |
Collapse
|
26
|
Mohanty H, Pachpute S, Yadav RP. Mechanism of drug resistance in bacteria: efflux pump modulation for designing of new antibiotic enhancers. Folia Microbiol (Praha) 2021; 66:727-739. [PMID: 34431062 DOI: 10.1007/s12223-021-00910-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/10/2021] [Indexed: 11/21/2022]
Abstract
Drug resistance has now become a serious concern in the domain of microbial infection. Bacteria are becoming smarter by displaying a variety of mechanisms during drug resistance. It is not only helping bacteria to adapt nicely in adverse environment but it also makes a smart system for better availability of nutritional status for microorganisms. In this domain, pathogenic bacteria are extensively studied and their mechanism for drug resistance is well explored. The common modes in bacterial resistance include degradation of antibiotics by enzymes, antibiotic target modification or inactivation by enzymatic actions, complete replacement of antibiotic targets, quorum sensing (QS) mechanism, and efflux pump-based extrusion of antibiotics. In this review, various mechanisms of drug resistance in bacteria have been highlighted with giving the importance of efflux pumps. This can be explored as a knowledge source for the management of a variety of bacterial infections, related disease and vibrant clue for next-generation drug development.
Collapse
Affiliation(s)
- Harshita Mohanty
- MGMIHS OMICS Research Center, MGM Central Research Laboratory, MGM Medical College and Hospital, MGM Institute of Health Sciences, Sector 1, Kamothe, Navi Mumbai-410209, Maharashtra, India.,Department of Molecular Biology, MGM School of Biomedical Sciences, MGM Institute of Health Sciences, Sector 1, Kamothe, Navi Mumbai-410209, Maharashtra, India
| | - Samir Pachpute
- Department of Medical Microbiology, MGM Medical College and Hospital, MGM Institute of Health Sciences, Sector 1, Kamothe, Navi Mumbai-410209, Maharashtra, India
| | - Raman P Yadav
- MGMIHS OMICS Research Center, MGM Central Research Laboratory, MGM Medical College and Hospital, MGM Institute of Health Sciences, Sector 1, Kamothe, Navi Mumbai-410209, Maharashtra, India. .,Department of Molecular Biology, MGM School of Biomedical Sciences, MGM Institute of Health Sciences, Sector 1, Kamothe, Navi Mumbai-410209, Maharashtra, India.
| |
Collapse
|
27
|
Knepp ZJ, Ghaner A, Root KT. Purification and refolding protocol for cold-active recombinant esterase AaSGNH1 from Aphanizomenon flos-aquae expressed as insoluble inclusion bodies. Prep Biochem Biotechnol 2021; 52:394-403. [PMID: 34355672 DOI: 10.1080/10826068.2021.1952601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microbial esterases are a highly desirable tool for numerous biosynthetic and biotechnological applications requiring ester bond cleavage. Once identified, microbial esterases are often produced recombinantly in Escherichia coli to enhance yield and ease of purification. In this study a polyhistidine-tagged SGNH esterase gene (AaSGNH1), originating from the cyanobacterium Aphanizomenon flos-aquae, was cloned into an over-expression plasmid and expressed in BL21(DE3) cells. The recombinant esterase enzyme was produced as inactive inclusion bodies which were insoluble in 8 M urea but readily solubilized by the detergent Empigen BB®. Crucially, the procurement of active enzyme required controlled removal of detergent during column chromatography and dialysis steps. The refolded esterase was characterized with respect to its ability to catalyze the cleavage of p-nitrophenol esters of different chain lengths (C2, C8, C16). In addition, the temperature and pH optima were determined and it was found that the enzyme was most active at low temperatures (5-15 °C) and under alkaline conditions (pH 8-10). It was found that the kinetic properties of AaSGNH1 were remarkably similar to other SGNH esterases described thereby validating that the protein was effectively refolded. Overall, this study provides a simple strategy for isolating cold-active recombinant esterase enzyme when expressed as inclusion bodies.
Collapse
Affiliation(s)
- Zachary J Knepp
- Department of Chemistry, Lock Haven University, Lock Haven, PA, USA
| | - Ashlea Ghaner
- Department of Chemistry, Lock Haven University, Lock Haven, PA, USA
| | - Kyle T Root
- Department of Chemistry, Lock Haven University, Lock Haven, PA, USA
| |
Collapse
|
28
|
Park JE, Jeong GS, Lee HW, Kim H. Molecular Characterization of Novel Family IV and VIII Esterases from a Compost Metagenomic Library. Microorganisms 2021; 9:microorganisms9081614. [PMID: 34442693 PMCID: PMC8399190 DOI: 10.3390/microorganisms9081614] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022] Open
Abstract
Two novel esterase genes, est8L and est13L, were isolated and identified from a compost metagenomic library. The encoded Est8L and Est13L had molecular masses of 33,181 and 44,913 Da consisting of 314 and 411 amino acids, respectively, without signal peptides. Est8L showed the highest identity (32.9%) to a hyper-thermophilic carboxylesterase AFEST from Archaeoglobus fulgidus compared to other esterases reported and was classified to be a novel member of family IV esterases with conserved regions such as HGGG, DY, GXSXG, DPL, and GXIH. Est13L showed the highest identity (98.5%) to the family VIII esterase Est7K from the metagenome library. Est8L and Est13L had the highest activities for p-nitrophenyl butyrate (C4) and p-nitrophenyl caproate (C6), respectively, and Est13L showed a broad substrate specificity for p-nitrophenyl substrates. Est8L and Est13L effectively hydrolyzed glyceryl tributyrate. The optimum temperatures for activities of Est8L and Est13L were identical (40 °C), and the optimum pH values were 9.0 and 10.0, respectively. Est13L showed higher thermostability than Est8L. Sephacryl S-200 HR chromatography showed that the native form of Est8L was a dimer. Interestingly, Est13L was found to be a tetramer, contrary to other family VIII esterases reported. Est8L was inhibited by 30% isopropanol, methanol, and acetonitrile; however, Est13L was activated to 182.9% and 356.1%, respectively, by 30% isopropanol and methanol. Est8L showed enantioselectivity for the S-form, but Est13L showed no enantioselectivity. These results show that intracellular Est8L and/or Est13L are oligomeric in terms of native forms and can be used for pharmaceutical and industrial applications with organic solvents under alkaline conditions.
Collapse
Affiliation(s)
| | | | | | - Hoon Kim
- Correspondence: ; Tel.: +82-617503751
| |
Collapse
|
29
|
Characterization of a Novel Family IV Esterase Containing a Predicted CzcO Domain and a Family V Esterase with Broad Substrate Specificity from an Oil-Polluted Mud Flat Metagenomic Library. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11135905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Two novel esterase genes, est2L and est4L, were identified from a previously constructed metagenomic library derived from an oil-polluted mud flat sample. The encoded Est2L and Est4L were composed of 839 and 267 amino acids, respectively, without signal peptides. Est2L was a unique fusion type of protein composed of two domains: a domain of the CzcO superfamily, associated with a cationic diffusion promoter with CzcD, and a domain of the acetylesterase superfamily, belonging to family IV with conserved motifs, such as HGG, GXSAG, and GXPP. Est2L was the first fused esterase with a CzcO domain. Est4L belonged to family V with GXS, GXSMGG, and PTL motifs. Native Est2L and Est4L were found to be in dimeric and tetrameric forms, respectively. Est2L and Est4L showed the highest activities at 60 °C and 50 °C, respectively, and at a pH of 10.0. Est2L preferred short length substrates, especially p-nitrophenyl (pNP)-acetate, with moderate butyrylcholinesterase activity, whereas Est4L showed the highest activity with pNP-decanoate and had broad specificity. Significant effects were not observed in Est2L from Co2+ and Zn2+, although Est2L contains the domain CzcD. Est2L and Est4L showed high stabilities in 30% methanol and 1% Triton X-100. These enzymes could be used for a variety of applications, such as detergent and mining processing under alkaline conditions.
Collapse
|
30
|
Li CH, Huang R, Makabenta JM, Schmidt-Malan S, Patel R, Rotello VM. In situ Generation of Antibiotics using Bioorthogonal "Nanofactories". Microbiol Insights 2021; 14:1178636121997121. [PMID: 33707951 PMCID: PMC7907933 DOI: 10.1177/1178636121997121] [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] [Received: 12/20/2020] [Accepted: 01/26/2021] [Indexed: 02/02/2023] Open
Abstract
Prodrug strategies use chemical modifications to improve the pharmacokinetic properties and therefore therapeutic effects of parent drugs. Traditional prodrug approaches use endogenous enzymes for activation. Bioorthogonal catalysis uses processes that endogenous enzymes cannot access, providing a complementary strategy for prodrug uncaging. Site-selective activation of prodrugs to drugs (uncaging) using synthetic catalysts is a promising strategy for localized drug activation. We discuss here recent studies that incorporate metal catalysts into polymers and nanoparticle scaffolds to provide biocompatible "enzyme-like" catalysts that can penetrate bacterial biofilms and activate prodrug antibiotics in situ, affording a new strategy to treat bacterial biofilm infections with the potential for reduced off-target effects.
Collapse
Affiliation(s)
- Cheng-Hsuan Li
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| | | | - Suzannah Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
31
|
Wang P, Li RQ, Wang L, Yang WT, Zou QH, Xiao D. Proteomic Analyses of Acinetobacter baumannii Clinical Isolates to Identify Drug Resistant Mechanism. Front Cell Infect Microbiol 2021; 11:625430. [PMID: 33718272 PMCID: PMC7943614 DOI: 10.3389/fcimb.2021.625430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022] Open
Abstract
Acinetobacter baumannii is one of the main causes of nosocomial infections. Increasing numbers of multidrug-resistant Acinetobacter baumannii cases have been reported in recent years, but its antibiotic resistance mechanism remains unclear. We studied 9 multidrug-resistant (MDR) and 10 drug-susceptible Acinetobacter baumannii clinical isolates using Label free, TMT labeling approach and glycoproteomics analysis to identify proteins related to drug resistance. Our results showed that 164 proteins exhibited different expressions between MDR and drug-susceptible isolates. These differential proteins can be classified into six groups: a. proteins related to antibiotic resistance, b. membrane proteins, membrane transporters and proteins related to membrane formation, c. Stress response-related proteins, d. proteins related to gene expression and protein translation, e. metabolism-related proteins, f. proteins with unknown function or other functions containing biofilm formation and virulence. In addition, we verified seven proteins at the transcription level in eight clinical isolates by using quantitative RT-PCR. Results showed that four of the selected proteins have positive correlations with the protein level. This study provided an insight into the mechanism of antibiotic resistance of multidrug-resistant Acinetobacter baumannii.
Collapse
Affiliation(s)
- Ping Wang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ren-Qing Li
- Institute for Control of Infectious Diseases and Endemic Diseases, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Lei Wang
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wen-Tao Yang
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qing-Hua Zou
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Di Xiao
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
32
|
Gwynne L, Williams GT, Yan K, Gardiner JE, Hilton KLF, Patenall BL, Hiscock JR, Maillard J, He X, James TD, Sedgwick AC, Jenkins ATA. The Evaluation of Ester Functionalised TCF‐Based Fluorescent Probes for the Detection of Bacterial Species. Isr J Chem 2021. [DOI: 10.1002/ijch.202000105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Lauren Gwynne
- Department of Chemistry University of Bath BA2 7AY Bath UK
| | - George T. Williams
- Department of Chemistry University of Bath BA2 7AY Bath UK
- School of Physical Sciences University of Kent CT2 7NH Canterbury UK
| | - Kai‐Cheng Yan
- Department of Chemistry University of Bath BA2 7AY Bath UK
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | | | - Kira L. F. Hilton
- School of Physical Sciences University of Kent CT2 7NH Canterbury UK
| | | | | | - Jean‐Yves Maillard
- School of Pharmacy and Pharmaceutical Sciences Cardiff University CF10 3NB Cardiff UK
| | - Xiao‐Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry East China University of Science and Technology 130 Meilong Road Shanghai 200237 China
| | - Tony D. James
- Department of Chemistry University of Bath BA2 7AY Bath UK
- School of Chemistry and Chemical Engineering Henan Normal University Xinxiang 453007 P. R. China
| | - Adam C. Sedgwick
- Department of Chemistry The University of Texas at Austin 105 East 24th Street A5300 Austin, Texas 78712–1224 USA
| | | |
Collapse
|
33
|
Krátký M, Konečná K, Brokešová K, Maixnerová J, Trejtnar F, Vinšová J. Optimizing the structure of (salicylideneamino)benzoic acids: Towards selective antifungal and anti-staphylococcal agents. Eur J Pharm Sci 2021; 159:105732. [PMID: 33493669 DOI: 10.1016/j.ejps.2021.105732] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/04/2021] [Accepted: 01/18/2021] [Indexed: 01/08/2023]
Abstract
An increasing resistance of human pathogenic bacteria and fungi has become a global health problem. Based on previous reports of 4-(salicylideneamino)benzoic acids, we designed, synthesised and evaluated their me-too analogues as potential antimicrobial agents. Forty imines derived from substituted salicylaldehydes and aminobenzoic acids, 4-aminobenzoic acid esters and 4-amino-N-phenylbenzamide were designed using molecular hybridization and prodrug strategies. The target compounds were synthesized with high yields and characterized by spectral methods. They were investigated against a panel of Gram-positive and Gram-negative bacteria, mycobacteria, yeasts and moulds. The most active imines were tested to determine their cytotoxicity and selectivity in HepG2 cells. Dihalogenosalicylaldehydes-based derivatives showed potent broad-spectrum antimicrobial properties, particularly against Gram-positive bacteria including methicillin-resistant Staphylococcus aureus (minimum inhibitory concentrations, MIC, from 7.81 µM) and Enterococcus faecalis (MIC of ≥15.62 µM), yeasts (MIC from 7.81 µM) and Trichophyton interdigitale mould (MIC of ≥3.90 µM). Methyl 4-[(2-hydroxy-3,5-diiodobenzylidene)amino]benzoate 4h exhibited excellent in vitro activity along with low toxicity to mammalian cells. This compound is selective for staphylococci, Candida spp. and Trichophyton interdigitale. In addition, this imine was evaluated as a potential inhibitor of Gram-positive biofilms. The successful approach used provided some promising derivatives with more advantageous properties than the parent 4-(salicylideneamino)benzoic acids.
Collapse
Affiliation(s)
- Martin Krátký
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic.
| | - Klára Konečná
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Kateřina Brokešová
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Jana Maixnerová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - František Trejtnar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05, Hradec Králové, Czech Republic
| | - Jarmila Vinšová
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| |
Collapse
|
34
|
Butler JH, Baptista RP, Valenciano AL, Zhou B, Kissinger JC, Tumwebaze PK, Rosenthal PJ, Cooper RA, Yue JM, Cassera MB. Resistance to Some But Not Other Dimeric Lindenane Sesquiterpenoid Esters Is Mediated by Mutations in a Plasmodium falciparum Esterase. ACS Infect Dis 2020; 6:2994-3003. [PMID: 32970404 PMCID: PMC11075783 DOI: 10.1021/acsinfecdis.0c00487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Unique lindenane sesquiterpenoid dimers from Chloranthecae spp. were recently identified with promising in vitro antiplasmodial activity and potentially novel mechanisms of action. To gain mechanistic insights to this new class of natural products, in vitro selection of Plasmodium falciparum resistance to the most active antiplasmodial compound, chlorajaponilide C, was explored. In all selected resistant clones, the half-maximal effective concentration (EC50) of chlorajaponilide C increased >250-fold, and whole genome sequencing revealed mutations in the recently discovered P. falciparum prodrug activation and resistance esterase (PfPARE). Chlorajaponilide C was highly potent (mean EC50 = 1.6 nM, n = 34) against fresh Ugandan P. falciparum isolates. The analysis of the structure-resistance relationships revealed that in vitro potency of a subset of lindenane sesquiterpenoid dimers was not mediated by PfPARE mutations. Thus, chlorajaponilide C, but not some related compounds, required parasite esterase activity for in vitro potency, and those compounds serve as the foundation for development of potent and selective antimalarials.
Collapse
Affiliation(s)
- Joshua H Butler
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602, United States
- Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, Georgia 30602, United States
| | - Rodrigo P Baptista
- Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, Georgia 30602, United States
| | - Ana L Valenciano
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602, United States
- Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, Georgia 30602, United States
| | - Bin Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100864, People's Republic of China
| | - Jessica C Kissinger
- Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, Georgia 30602, United States
| | | | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, California 94110, United States
| | - Roland A Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California 94901, United States
| | - Jian-Min Yue
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100864, People's Republic of China
| | - Maria B Cassera
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602, United States
- Center for Tropical and Emerging Global Diseases (CTEGD), University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
35
|
Brillault J, Tewes F. Control of the Lung Residence Time of Highly Permeable Molecules after Nebulization: Example of the Fluoroquinolones. Pharmaceutics 2020; 12:pharmaceutics12040387. [PMID: 32340298 PMCID: PMC7238242 DOI: 10.3390/pharmaceutics12040387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/29/2022] Open
Abstract
Pulmonary drug delivery is a promising strategy to treat lung infectious disease as it allows for a high local drug concentration and low systemic side effects. This is particularly true for low-permeability drugs, such as tobramycin or colistin, that penetrate the lung at a low rate after systemic administration and greatly benefit from lung administration in terms of the local drug concentration. However, for relatively high-permeable drugs, such as fluoroquinolones (FQs), the rate of absorption is so high that the pulmonary administration has no therapeutic advantage compared to systemic or oral administration. Formulation strategies have thus been developed to decrease the absorption rate and increase FQs’ residence time in the lung after inhalation. In the present review, some of these strategies, which generally consist of either decreasing the lung epithelium permeability or decreasing the release rate of FQs into the epithelial lining fluid after lung deposition, are presented in regards to their clinical aspects.
Collapse
Affiliation(s)
- Julien Brillault
- INSERM U-1070, Pôle Biologie Santé, 86000 Poitiers, France
- UFR Médecine-Pharmacie, Université de Poitiers, 86073 Poitiers, France
- Correspondence: (J.B.); (F.T.)
| | - Frédéric Tewes
- INSERM U-1070, Pôle Biologie Santé, 86000 Poitiers, France
- UFR Médecine-Pharmacie, Université de Poitiers, 86073 Poitiers, France
- Correspondence: (J.B.); (F.T.)
| |
Collapse
|
36
|
Juhás M, Kučerová L, Horáček O, Janďourek O, Kubíček V, Konečná K, Kučera R, Bárta P, Janoušek J, Paterová P, Kuneš J, Doležal M, Zitko J. N-Pyrazinoyl Substituted Amino Acids as Potential Antimycobacterial Agents-The Synthesis and Biological Evaluation of Enantiomers. Molecules 2020; 25:E1518. [PMID: 32230728 PMCID: PMC7181131 DOI: 10.3390/molecules25071518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 11/22/2022] Open
Abstract
Tuberculosis is an infectious disease caused by Mycobacterium tuberculosis (Mtb), each year causing millions of deaths. In this article, we present the synthesis and biological evaluations of new potential antimycobacterial compounds containing a fragment of the first-line antitubercular drug pyrazinamide (PZA), coupled with methyl or ethyl esters of selected amino acids. The antimicrobial activity was evaluated on a variety of (myco)bacterial strains, including Mtb H37Ra, M. smegmatis, M. aurum, Staphylococcus aureus, Pseudomonas aeruginosa, and fungal strains, including Candida albicans and Aspergillus flavus. Emphasis was placed on the comparison of enantiomer activities. None of the synthesized compounds showed any significant activity against fungal strains, and their antibacterial activities were also low, the best minimum inhibitory concentration (MIC) value was 31.25 µM. However, several compounds presented high activity against Mtb. Overall, higher activity was seen in derivatives containing ʟ-amino acids. Similarly, the activity seems tied to the more lipophilic compounds. The most active derivative contained phenylglycine moiety (PC-ᴅ/ʟ-Pgl-Me, MIC < 1.95 µg/mL). All active compounds possessed low cytotoxicity and good selectivity towards Mtb. To the best of our knowledge, this is the first study comparing the activities of the ᴅ- and ʟ-amino acid derivatives of pyrazinamide as potential antimycobacterial compounds.
Collapse
Affiliation(s)
- Martin Juhás
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Lucie Kučerová
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Ondřej Horáček
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Ondřej Janďourek
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Vladimír Kubíček
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Klára Konečná
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Radim Kučera
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Pavel Bárta
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Jiří Janoušek
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Pavla Paterová
- University Hospital Hradec Králové, Department of Clinical Microbiology, Sokolská 581, 500 05 Hradec Králové, Czech Republic;
| | - Jiří Kuneš
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Martin Doležal
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| | - Jan Zitko
- Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové, Czech Republic; (L.K.); (O.H.); (O.J.); (V.K.); (K.K.); (R.K.); (P.B.); (J.J.); (J.K.); (M.D.)
| |
Collapse
|
37
|
Nemes D, Kovács R, Nagy F, Tóth Z, Herczegh P, Borbás A, Kelemen V, Pfliegler WP, Rebenku I, Hajdu PB, Fehér P, Ujhelyi Z, Fenyvesi F, Váradi J, Vecsernyés M, Bácskay I. Comparative biocompatibility and antimicrobial studies of sorbic acid derivates. Eur J Pharm Sci 2020; 143:105162. [DOI: 10.1016/j.ejps.2019.105162] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/24/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022]
|
38
|
Synthesis and Biological Evaluation of Lipophilic Nucleoside Analogues as Inhibitors of Aminoacyl-tRNA Synthetases. Antibiotics (Basel) 2019; 8:antibiotics8040180. [PMID: 31600972 PMCID: PMC6963541 DOI: 10.3390/antibiotics8040180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 11/16/2022] Open
Abstract
Emerging antibiotic resistance in pathogenic bacteria and reduction of compounds in the existing antibiotics discovery pipeline is the most critical concern for healthcare professionals. A potential solution aims to explore new or existing targets/compounds. Inhibition of bacterial aminoacyl-tRNA synthetase (aaRSs) could be one such target for the development of antibiotics. The aaRSs are a group of enzymes that catalyze the transfer of an amino acid to their cognate tRNA and therefore play a pivotal role in translation. Thus, selective inhibition of these enzymes could be detrimental to microbes. The 5′-O-(N-(L-aminoacyl)) sulfamoyladenosines (aaSAs) are potent inhibitors of the respective aaRSs, however due to their polarity and charged nature they cannot cross the bacterial membranes. In this work, we increased the lipophilicity of these existing aaSAs in an effort to promote their penetration through the bacterial membrane. Two strategies were followed, either attaching a (permanent) alkyl moiety at the adenine ring via alkylation of the N6-position or introducing a lipophilic biodegradable prodrug moiety at the alpha-terminal amine, totaling eight new aaSA analogues. All synthesized compounds were evaluated in vitro using either a purified Escherichiacoli aaRS enzyme or in presence of total cellular extract obtained from E. coli. The prodrugs showed comparable inhibitory activity to the parent aaSA analogues, indicating metabolic activation in cellular extracts, but had little effect on bacteria. During evaluation of the N6-alkylated compounds against different microbes, the N6-octyl containing congener 6b showed minimum inhibitory concentration (MIC) of 12.5 µM against Sarcina lutea while the dodecyl analogue 6c displayed MIC of 6.25 µM against Candidaalbicans.
Collapse
|
39
|
Mancini G, Bouda M, Gamrat JM, Tomsho JW. Synthesis and Antimicrobial Evaluation of γ-Borono Phosphonate Compounds in Escherichia coli and Mycobacterium smegmatis. ACS OMEGA 2019; 4:14551-14559. [PMID: 31528809 PMCID: PMC6740193 DOI: 10.1021/acsomega.9b01774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/02/2019] [Indexed: 06/10/2023]
Abstract
Drug resistance in bacteria is a serious threat, and drugs with novel modes of action are constantly needed. Fosmidomycin is a naturally occurring antibiotic that inhibits the nonmevalonate pathway via inhibition of the enzyme 1-deoxylulose-5-phosphate reductoisomerase (DXR). This work is the first report in which a boronic acid is evaluated as an isostere of the retrohydroxamate moiety of fosmidomycin. We report the novel synthesis of a γ-borono phosphonate analog of fosmidomycin and its corresponding prodrugs. We evaluate the inhibition of DXR and the antimicrobial activity of γ-borono phosphonate compounds against Escherichia coli wild type, E. coli Δglycerol-3-phosphate transporter, and Mycobacterium smegmatis. Despite its structural similarities, the γ-borono phosphonate compound shows antimicrobial activity against E. coli with a mechanism of action that is different from fosmidomycin. This was proven with an underutilized method for studying in vitro inhibition of the MEP pathway in E. coli via isopentenyl pyrophosphate chemical rescue. These results indicate that these compounds may serve as a promising scaffold for developing a new class of antimicrobial agents.
Collapse
|