1
|
Van de Vliet L, Vackier T, Thevissen K, Decoster D, Steenackers HP. Imidazoles and Quaternary Ammonium Compounds as Effective Therapies against (Multidrug-Resistant) Bacterial Wound Infections. Antibiotics (Basel) 2024; 13:949. [PMID: 39452215 PMCID: PMC11505196 DOI: 10.3390/antibiotics13100949] [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: 09/18/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES The rise and spread of antimicrobial resistance complicates the treatment of bacterial wound pathogens, further increasing the need for newer, effective therapies. Azoles such as miconazole have shown promise as antibacterial compounds; however, they are currently only used as antifungals. Previous research has shown that combining azoles with quaternary ammonium compounds yields synergistic activity against fungal pathogens, but the effect on bacterial pathogens has not been studied yet. METHODS In this study, the focus was on finding active synergistic combinations of imidazoles and quaternary ammonium compounds against (multidrug-resistant) bacterial pathogens through checkerboard assays. Experimental evolution in liquid culture was used to evaluate the possible emergence of resistance against the most active synergistic combination. RESULTS Several promising synergistic combinations were identified against an array of Gram-positive pathogens: miconazole/domiphen bromide, ketoconazole/domiphen bromide, clotrimazole/domiphen bromide, fluconazole/domiphen bromide and miconazole/benzalkonium chloride. Especially, miconazole with domiphen bromide exhibits potential, as it has activity at a low concentration against a broad range of pathogens and shows an absence of strong resistance development over 11 cycles of evolution. CONCLUSIONS This study provides valuable insight into the possible combinations of imidazoles and quaternary ammonium compounds that could be repurposed for (topical) wound treatment. Miconazole with domiphen bromide shows the highest application potential as a possible future wound therapy. However, further research is needed into the mode of action of these compounds and their efficacy and toxicity in vivo.
Collapse
Affiliation(s)
- Lauren Van de Vliet
- MiCA Lab, Centre of Microbial and Plant Genetics (CMPG), Department Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium
| | - Thijs Vackier
- MiCA Lab, Centre of Microbial and Plant Genetics (CMPG), Department Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium
| | - Karin Thevissen
- CMPG-PFI (Plant-Fungus Interactions Group of Centre of Microbial and Plant Genetics), Department Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium
| | - David Decoster
- MiCA Lab, Centre of Microbial and Plant Genetics (CMPG), Department Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium
| | - Hans P. Steenackers
- MiCA Lab, Centre of Microbial and Plant Genetics (CMPG), Department Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
2
|
Hill S, Decorso I, Nezamololama N, Babaei Z, Rafferty SP. Catalytic Differences between Flavohemoglobins of Giardia intestinalis and E. coli. Pathogens 2024; 13:480. [PMID: 38921778 PMCID: PMC11206828 DOI: 10.3390/pathogens13060480] [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: 04/15/2024] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
The sole known heme enzyme of the parasitic protist Giardia intestinalis is a flavohemoglobin (gFlHb) that acts as a nitric oxide dioxygenase (NOD) and protects the organism from the free radical nitric oxide. To learn more about the properties of this enzyme, we measured its nitric oxide dioxygenase, NADH oxidase, and cytochrome c reductase activities and compared these to the activities of the E. coli flavohemoglobin (Hmp). The turnover number for the NOD activity of gFlHb (23 s-1) is about two-thirds of that of Hmp (34 s-1) at pH 6.5 and 37 °C. The two enzymes differ in their sensitivity towards molecules that act as heme ligands. For both gFlHb and Hmp, inhibition with miconazole, a large imidazole ligand, is adequately described by simple competitive inhibition, with KI = 10 μM and 0.27 μM for gFlHb and Hmp, respectively. Inhibition plots with the small ligand imidazole were biphasic, which is consistent with previous experiments with carbon monoxide as a probe that show that the active site of flavohemoglobins exists in two conformations. Interestingly, the largest difference is observed with nitrite, which, like imidazole, also shows a biphasic inhibition plot; however, nitrite inhibits gFlHb at sub-millimolar concentrations while Hmp is not significantly affected. NADH oxidase activity measured under aerobic conditions in the absence of nitric oxide for Hmp was more than twice the activity of gFlHb. The addition of 1 mM hydrogen peroxide in these assays stimulated the NADH oxidase activity of gFlHb but not Hmp. Both enzymes had nearly identical cytochrome c reductase activities but the extent of the contribution of indirect reduction by flavohemoglobin-generated superoxide was much lower with gFlHb (4% SOD-inhibited) than with Hmp (17% SOD-inhibited). Although the active sites of the two enzymes share the same highly conserved residues that are important for catalysis, differences in the distal ligand binding site may account for these differences in activity and sensitivity towards NOD inhibitors. The differences observed in the NADH oxidase and cytochrome c reductase assays suggest that gFlHb may have evolved to protect the protist, which lacks both superoxide dismutase and catalase, from the damaging effects of superoxide by minimizing its production and from peroxide by actively reducing it.
Collapse
Affiliation(s)
- Sarah Hill
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON K9J 7B8, Canada; (S.H.); (I.D.); (N.N.)
| | - Isabelle Decorso
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON K9J 7B8, Canada; (S.H.); (I.D.); (N.N.)
| | - Novin Nezamololama
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON K9J 7B8, Canada; (S.H.); (I.D.); (N.N.)
| | - Zahra Babaei
- Department of Parasitology & Mycology, Kerman University of Medical Sciences, Kerman 76169-13555, Iran;
| | - Steven Patrick Rafferty
- Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON K9J 7B8, Canada; (S.H.); (I.D.); (N.N.)
| |
Collapse
|
3
|
Alam N, Islam M, Najnin H, Shakya S, Khan IM, Hossain MW, Zaidi R. Design and characterization of a binary CT complex of imidazole-oxyresveratrol: exploring its pharmacological and computational aspects. J Biomol Struct Dyn 2024; 42:1319-1335. [PMID: 37054451 DOI: 10.1080/07391102.2023.2199088] [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: 12/13/2022] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
A new binary charge transfer (CT) complex between imidazole (IMZ) and oxyresveratrol (OXA) was synthesized and characterized experimentally and theoretically. The experimental work was carried out in solution and solid state in selected solvents such as chloroform (CHL), methanol (Me-OH), ethanol (Et-OH), and acetonitrile (AN). The newly synthesized CT complex (D1) has been characterized by various techniques such as UV-visible spectroscopy, FTIR, 1H-NMR, and powder-XRD. The 1:1 composition of D1 is confirmed by Jobs' method of continuous variation and spectrophotometric (at λmax 554 nm) methods at 298 K. The infrared spectra of D1 confirmed the existence of proton transfer hydrogen bond beside charge transfer interaction. These findings indicate that the cation and anion are joined together by the weak hydrogen bonding as N+-H-O-. Reactivity parameters strongly recommended that IMZ behaves as a good electron donor and OXA an efficient electron acceptor. Density functional theory (DFT) computations with basis set B3LYP/6-31G (d,p) was applied to support the experimental results. TD-DFT calculations gives HOMO (-5.12 eV) → LUMO (-1.14 eV) electronic energy gap (Δ E ) to be 3.80 eV. The bioorganic chemistry of D1 was well established after antioxidant, antimicrobial, and toxicity screening in Wistar rat. The type of interactions between HSA and D1 at the molecular level was studied through fluorescence spectroscopy. Binding constant along with the type of quenching mechanism, was investigated through the Stern-Volmer equation. Molecular docking demonstrated that D1 binds perfectly with human serum albumin and EGFR (1M17) and exposes free energy of binding (FEB) values of -295.2 and -283.3 kcal/mol, respectively. The D1 fits successfully into the minor groove of HAS and 1M17, the results of molecular docking show that the D1 binds perfectly with the HAS and 1M17, the higher value of binding energy shows stronger interaction between HAS and 1M17 with D1. Our synthesized complex shows good binding results with HAS compared to 1M17.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nisat Alam
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Maidul Islam
- Deparment of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Hasina Najnin
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Sonam Shakya
- Deparment of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Ishaat M Khan
- Deparment of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | | | - Rana Zaidi
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
4
|
Islam M, Khan IM, Shakya S, Alam N. Design, synthesis, characterizing and DFT calculations of a binary CT complex co-crystal of bioactive moieties in different polar solvents to investigate its pharmacological activity. J Biomol Struct Dyn 2023; 41:10813-10829. [PMID: 36579428 DOI: 10.1080/07391102.2022.2158937] [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: 05/30/2022] [Accepted: 12/10/2022] [Indexed: 12/30/2022]
Abstract
Imidazole (IM) and salicylic acid (SA) have a significant class among the medical compound. These are widely used as topical drugs like antifungal, antibacterial, anticancer, immunosuppressive agent, etc. These two bioactive organic moieties are combined by a weak hydrogen bond formed by hydrogen transfer. The charge transfer (CT) complex of acceptor (SA) and donor (IM), has been synthesized at room temperature in methanol and confirmed by signal-crystal XRD, conductance and UV-visible spectroscopy. The X-ray crystallography provides the original structural information of CT complex and displays the existence of N+-H--O- bond between IM and SA. The physical properties such as (ECT), (RN), (ID), (f), (D) and (Δ G0) along with molar extinction coefficient (εCT) and formation constant (KCT) were estimated through UV-visible spectroscopy. Job's method and Benesi-Hildebrand equation suggested 1:1 stoichiometry of ([IM]+[SA]-). The results indicate a complete transfer of hydrogen atom and CT complex formation with 1:1 molar ratio of IM and SA. Antimicrobial activity was veiled against different bacteria like Escherichia coli, Bacillus subtilis and Staphylococcus aureus; and different fungi as Fusarium oxysporum, Candida albicans and Aspergillus niger by disc diffusion method. CT complex was also tested for cytotoxic activity against lung cancer cell lines in comparison to breast cancer cell lines. Molecular docking provides the information of binding of [(IM)+(SA)-] with the cancer marker (1M17), which has substantial application for drug designing. The investigational studies were supplemented through time-dependent density functional theory (TD-DFT) using basis set B3LYP/6-311G**. Through DFT calculations, HOMO→LUMO electronic energy gap (Δ E ) was obtained.
Collapse
Affiliation(s)
- Maidul Islam
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Ishaat M Khan
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Sonam Shakya
- Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, India
| | - Nisat Alam
- Department of Bio-chemistry, School of Chemical and Life Science, New Delhi, India
| |
Collapse
|
5
|
Kaviani N, Behrouz S, Jafari AA, Soltani Rad MN. Functionalization of Fe 3O 4@SiO 2 nanoparticles with Cu(i)-thiosemicarbazone complex as a robust and efficient heterogeneous nanocatalyst for N-arylation of N-heterocycles with aryl halides. RSC Adv 2023; 13:30293-30305. [PMID: 37849694 PMCID: PMC10577646 DOI: 10.1039/d3ra06327e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023] Open
Abstract
In this research, the functionalized silica-coated magnetite nanoparticles with Cu(i)-thiosemicarbazone complex (Fe3O4@SiO2-[CuL]) has been designed and synthesized as a magnetically retrievable nanocatalyst. Different techniques were employed to characterize the structure of Fe3O4@SiO2-[CuL] comprising FT-IR, FE-SEM, TEM, DLS, XRD, EDX, TGA, AAS, and VSM analysis. The catalytic performance of Fe3O4@SiO2-[CuL] was perused in Ullmann-type N-arylation of nucleobases, xanthines, and other N-heterocycles with diverse aryl halides which acquired the desired N-aryl products in good to excellent yields. Fe3O4@SiO2-[CuL] is a thermal and chemical stable, easy to prepare and recyclable, inexpensive, and ecofriendly catalyst that needs no additional additive or ligand as promoters. This catalyst could be separated without difficulty by a simple magnet and reused for at least seven sequential runs without a significant decline in its catalytic performance.
Collapse
Affiliation(s)
- Narjes Kaviani
- Department of Chemistry, Faculty of Science, Yazd University Yazd Iran
| | - Somayeh Behrouz
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| | - Abbas Ali Jafari
- Department of Chemistry, Faculty of Science, Yazd University Yazd Iran
| | - Mohammad Navid Soltani Rad
- Department of Chemistry, Shiraz University of Technology Shiraz 71555-313 Iran +98 71 3735 4520 +98 71 3735 4500
| |
Collapse
|
6
|
Mashood Ahamed FM, Shakya B, Shakya S. Synthesis and characterization of a novel Mannich base benzimidazole derivative to explore interaction with human serum albumin and antimicrobial property: experimental and theoretical approach. J Biomol Struct Dyn 2023; 41:8701-8714. [PMID: 36284457 DOI: 10.1080/07391102.2022.2136757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/09/2022] [Indexed: 10/31/2022]
Abstract
The novel Mannich base benzimidazole derivative (CB-1), 1-((1H-benzo[d]imidazol-1-yl)(3-chlorophenyl)methyl)-3-phenylurea) has been designed and synthesized by reacting benzimidazole, 3-chloro benzaldehyde, and N-Phenyl urea. CB-1 has been characterized by UV- Visible, FTIR, and 1H NMR. CB-1 was explored to study the interaction with the most abundant blood protein which involved in the role of transport of molecules (drugs), human serum albumin (HSA). Fluorescence results are evident for the presence of both dynamic and static quenching mechanisms in the binding of CB-1 to HSA. Antimicrobial screening were carried out against three bacteria and three fungi pathogens employing disc diffusion method. Molecular docking using AutoDock Vina tool further confirms the experimental binding interactions obtained from fluorescence. Density functional theory (DFT) with B3LYP/6-311G++ basis set was used for correlating theoretical data and obtaining optimized structures of CB-1 along with reactants with molecular electrostatic potential (MEP) map and HOMO→LUMO energy gap calculation. HIGHLIGHTSThe novel Mannich base benzimidazole derivative (CB-1) has been designed and synthesized by Mannich reaction.CB-1 has been characterized by UV- Visible, FTIR, and 1H NMR.Fluorescence quenching reveals that HSA binds to CB-1 via aromatic residues, which is corroborated by molecular docking.Antifungal and antibacterial activity was evaluated in comparison to Nystatin and Gentamicin standard drugs, respectively.DFT calculations support experimental data and provide HOMO-LUMO energy gap.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- F M Mashood Ahamed
- PG and Research Department of Chemistry, Jamal Mohamed College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, India
| | - Barkha Shakya
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Sonam Shakya
- Department of Chemistry, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
7
|
Cortat Y, Zobi F. Resurgence and Repurposing of Antifungal Azoles by Transition Metal Coordination for Drug Discovery. Pharmaceutics 2023; 15:2398. [PMID: 37896159 PMCID: PMC10609764 DOI: 10.3390/pharmaceutics15102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Coordination compounds featuring one or more antifungal azole (AA) ligands constitute an interesting family of candidate molecules, given their medicinal polyvalence and the viability of drug complexation as a strategy to improve and repurpose available medications. This review reports the work performed in the field of coordination derivatives of AAs synthesized for medical purposes by discussing the corresponding publications and emphasizing the most promising compounds discovered so far. The resulting overview highlights the efficiency of AAs and their metallic species, as well as the potential still lying in this research area.
Collapse
Affiliation(s)
| | - Fabio Zobi
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland;
| |
Collapse
|
8
|
Biswas S, Chowdhury T, Dutta K, Saha S, Das D. Biochemical Resistivity against Free Radicals and Microbes: Cooperative Action of Zn(II)/Imidazole in Phosphoesterase-Mediated Cell Death. ACS APPLIED BIO MATERIALS 2023; 6:3278-3290. [PMID: 37565455 DOI: 10.1021/acsabm.3c00365] [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] [Indexed: 08/12/2023]
Abstract
This work delivers a targeted synthesis of four isostructural O-substituted imidazole-based zinc(II) complexes, namely, [Zn2(L1)2(I)2](DMF) (1), [Zn2(L2)2(I)2](DMF) (2), [Zn2(L1)2(Br)2] (3), and [Zn2(L2)2(Br)2] (4), derived from homologous Schiff-base ligands HL1 and HL2 to explore their impact on free radicals, microbes, and dephosphorylation of phosphoesters. The antioxidant activity of all complexes was checked by various radical scavenging assays (ABTS+•, DPPH•, and H2O2 radical quenching). Among them, complex 2 showed superior radical quenching activity, as indicated by its lowest EC50 value and thus maximum antioxidative capability. Again, antibacterial assays against several Gram-positive and Gram-negative bacteria were conducted to evaluate the zone of inhibition. The minimum bactericidal concentration and minimum inhibitory concentration values from the microdilution method for all complexes revealed complex 3 to have maximum potency against Gram-positive bacteria. The P-O bond hydrolysis in the phospholipid chain caused by the hydrolytic phosphoesterase activity of the Zn(II)-complexes plays a crucial role in cell membrane rupture. A model substrate 4-PNPP was used to explain the potency of monomeric Zn(II) complex (3) for cell penetration over dimeric one (2) with a proper mechanism. Furthermore, a heme model substrate, Fe(TPP)Cl, has been introduced with the most potent complex 3 and has spectrophotometric evidence for covalent interaction with imidazole and Fe(III) that can disrupt the nitric oxide dioxygenase function of flavohemoglobin, leading to bacterial cell death. To our knowledge, this is the first case to report a novel mechanism of antimicrobial action where both the metal and the ligand are cooperatively involved in bacterial cell death. The main goal of this work is to invent multifunctional therapeutics as well as the proper chemical rationalization of biological processes using mechanistic approaches, which includes investigating the roles of halides, imidazoles, and solution-phase structural variations of complexes..
Collapse
Affiliation(s)
- Sneha Biswas
- Department of Chemistry, University College of Science, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Tania Chowdhury
- Department of Chemistry, University College of Science, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Koushik Dutta
- Department of Polymer Science & Technology, University of Calcutta, 92, A.P.C. Road, Kolkata- 700009 West Bengal, India
| | - Sayan Saha
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Debasis Das
- Department of Chemistry, University College of Science, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| |
Collapse
|
9
|
Sari S, Sabuncuoğlu S, Koçak Aslan E, Avci A, Kart D, Özdemir Z, Acar MF, Sayoğlu B, Alagöz MA, Karakurt A, Dalkara S. Azoles containing naphthalene with activity against Gram-positive bacteria: in vitro studies and in silico predictions for flavohemoglobin inhibition. J Biomol Struct Dyn 2022; 40:10220-10229. [PMID: 34139139 DOI: 10.1080/07391102.2021.1940285] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Azoles are first-line drugs used in fungal infections. Topical antifungals, such as miconazole and econazole, are known to be active against Gram-positive bacteria, which was reported to result from bacterial flavohemoglobin (flavoHb) inhibition. Dual antibacterial/antifungal action is believed to have benefits for antimicrobial chemotherapy. In this study, we tested antibacterial effects of an in-house library of naphthalene-bearing azoles, some of which were reported as potent antifungals, in an attempt to find dual-acting hits. Several potent derivatives were obtained against the Gram-positive bacteria, Enterococcus faecalis and Staphylococcus aureus. 9 was active at a minimum inhibitor concentration (MIC) less than 1 µg/ml against E. faecalis and S. aureus, and 10 against S. aureus. 16 was also potent against E. faecalis and S. aureus (MIC = 1 and 2 µg/ml, respectively). Six more were active against S. aureus with MIC ≤ 4 µg/ml. In vitro cytotoxicity studies showed that the active compounds were safe for healthy cells within their MIC ranges. According to the calculated descriptors, the library was found within the drug-like chemical space and free of pan-assay interference compounds (PAINS). Molecular docking studies suggested that the compounds might be bacterial flavohemoglobin (flavoHb) inhibitors and the azole and naphthalene rings were important pharmacophores, which was further supported by pharmacophore modeling study. As a result, the current study presents several non-toxic azole derivatives with antibacterial effects. In addition to their previously reported antifungal properties, they could set a promising starting point for the future design of dual acting antimicrobials. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suat Sari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Suna Sabuncuoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ebru Koçak Aslan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ahmet Avci
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Didem Kart
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Zeynep Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inönü University, Malatya, Turkey
| | - M Fahir Acar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Burcu Sayoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - M Abdullah Alagöz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inönü University, Malatya, Turkey
| | - Arzu Karakurt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inönü University, Malatya, Turkey
| | - Sevim Dalkara
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
10
|
Pech-Santiago EO, Argüello-García R, Vázquez C, Saavedra E, González-Hernández I, Jung-Cook H, Rafferty SP, Ortega-Pierres MG. Giardia duodenalis: Flavohemoglobin is involved in drug biotransformation and resistance to albendazole. PLoS Pathog 2022; 18:e1010840. [PMID: 36166467 PMCID: PMC9514659 DOI: 10.1371/journal.ppat.1010840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/28/2022] [Indexed: 12/12/2022] Open
Abstract
Giardia duodenalis causes giardiasis, a major diarrheal disease in humans worldwide whose treatment relies mainly on metronidazole (MTZ) and albendazole (ABZ). The emergence of ABZ resistance in this parasite has prompted studies to elucidate the molecular mechanisms underlying this phenomenon. G. duodenalis trophozoites convert ABZ into its sulfoxide (ABZSO) and sulfone (ABZSOO) forms, despite lacking canonical enzymes involved in these processes, such as cytochrome P450s (CYP450s) and flavin-containing monooxygenases (FMOs). This study aims to identify the enzyme responsible for ABZ metabolism and its role in ABZ resistance in G. duodenalis. We first determined that the iron-containing cofactor heme induces higher mRNA expression levels of flavohemoglobin (gFlHb) in Giardia trophozoites. Molecular docking analyses predict favorable interactions of gFlHb with ABZ, ABZSO and ABZSOO. Spectral analyses of recombinant gFlHb in the presence of ABZ, ABZSO and ABZSOO showed high affinities for each of these compounds with Kd values of 22.7, 19.1 and 23.8 nM respectively. ABZ and ABZSO enhanced gFlHb NADH oxidase activity (turnover number 14.5 min-1), whereas LC-MS/MS analyses of the reaction products showed that gFlHb slowly oxygenates ABZ into ABZSO at a much lower rate (turnover number 0.01 min-1). Further spectroscopic analyses showed that ABZ is indirectly oxidized to ABZSO by superoxide generated from the NADH oxidase activity of gFlHb. In a similar manner, the superoxide-generating enzyme xanthine oxidase was able to produce ABZSO in the presence of xanthine and ABZ. Interestingly, we find that gFlHb mRNA expression is lower in albendazole-resistant clones compared to those that are sensitive to this drug. Furthermore, all albendazole-resistant clones transfected to overexpress gFlHb displayed higher susceptibility to the drug than the parent clones. Collectively these findings indicate a role for gFlHb in ABZ conversion to its sulfoxide and that gFlHb down-regulation acts as a passive pharmacokinetic mechanism of resistance in this parasite.
Collapse
Affiliation(s)
- Edar O. Pech-Santiago
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Raúl Argüello-García
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Citlali Vázquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Iliana González-Hernández
- Laboratorio de Neuropsicofarmacología, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Ciudad de México, México
| | - Helgi Jung-Cook
- Laboratorio de Neuropsicofarmacología, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Ciudad de México, México
| | | | - M. Guadalupe Ortega-Pierres
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
- * E-mail:
| |
Collapse
|
11
|
Liu J, Yin J, Li Y, Li D, Wu J, Wang C, Wang C, Yin F, Yang B, Zhang W. High nitrite-nitrogen stress intensity drives nitrite anaerobic oxidation to nitrate and inhibits methanogenesis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 832:155109. [PMID: 35398130 DOI: 10.1016/j.scitotenv.2022.155109] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 06/14/2023]
Abstract
Nitrite is an important intermediate in nitrogen metabolism. We explored the effect of nitrite-nitrogen stress intensity (NNSI) on nitrite metabolism and methanogenesis in anaerobic digestion. The results showed that the NNSI regulated microbial diversity, composition, and functions, and microbial community assembly was primarily shaped by stochastic processes. Moreover, the NNSI was negatively correlated with α-diversity and positively correlated with non-metric multi-dimensional scaling distance. Denitrification gradually increased with increasing NNSI; however, methanogenesis was gradually inhibited, which was primarily due to the inhibition of the aceticlastic methanogenesis pathway (i.e., Methanosaeta) and methylotrophic methanogenesis pathway (i.e., Candidatus_Methanofastidiosum). High NNSI (1882 ± 98.99 mg/L NO2--N) promoted nitrite anaerobic oxidation to nitrate and was favorable for dissimilatory nitrate reduction to ammonia (DNRA). We present evidence for the microbial transformation of nitrite under anaerobic conditions, with potential geochemical and evolutionary importance. As nitrogen oxides were already present on early Earth, our finding presents the possibility of a nitrogen cycle before the evolution of oxygenic photosynthesis.
Collapse
Affiliation(s)
- Jianfeng Liu
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China; Engineering and Research Center of Sustainable Development and Utilization of Bioenergy, Ministry of Education, Yunnan Normal University, Kunming 650500, PR China; Jilin Dongsheng Institute of Biomass Energy Engineering, Tonghua 134118, PR China; DongMing Agriculture and Animal Husbandry Development (Group) Co., Ltd., Tonghua 134118, PR China
| | - Jiao Yin
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China
| | - Yanshuang Li
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China
| | - Dingjin Li
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China
| | - Jiaxuan Wu
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China
| | - Chengxian Wang
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China; Engineering and Research Center of Sustainable Development and Utilization of Bioenergy, Ministry of Education, Yunnan Normal University, Kunming 650500, PR China
| | - Changmei Wang
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China; Engineering and Research Center of Sustainable Development and Utilization of Bioenergy, Ministry of Education, Yunnan Normal University, Kunming 650500, PR China; Jilin Dongsheng Institute of Biomass Energy Engineering, Tonghua 134118, PR China
| | - Fang Yin
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China; Engineering and Research Center of Sustainable Development and Utilization of Bioenergy, Ministry of Education, Yunnan Normal University, Kunming 650500, PR China; Jilin Dongsheng Institute of Biomass Energy Engineering, Tonghua 134118, PR China
| | - Bin Yang
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China; Engineering and Research Center of Sustainable Development and Utilization of Bioenergy, Ministry of Education, Yunnan Normal University, Kunming 650500, PR China
| | - Wudi Zhang
- Yunnan Research Center of Biogas Technology and Engineering, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, PR China; Engineering and Research Center of Sustainable Development and Utilization of Bioenergy, Ministry of Education, Yunnan Normal University, Kunming 650500, PR China; Jilin Dongsheng Institute of Biomass Energy Engineering, Tonghua 134118, PR China; DongMing Agriculture and Animal Husbandry Development (Group) Co., Ltd., Tonghua 134118, PR China.
| |
Collapse
|
12
|
de Sousa LP, Cipriano MAP, Freitas SDS, Carazzolle MF, da Silva MJ, Mondego JMC. Genomic and physiological evaluation of two root associated Pseudomonas from Coffea arabica. Microbiol Res 2022; 263:127129. [PMID: 35907286 DOI: 10.1016/j.micres.2022.127129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 10/17/2022]
Abstract
Many Pseudomonas species promote plant growth and colonize a wide range of environments. The annotation of a Coffea arabica ESTs database revealed a considerable number of Pseudomonas sequences. To evaluate the genomic and physiology of Pseudomonas that inhabit coffee plants, fluorescent Pseudomonas from C. arabica root environment were isolated. Two of them had their genomes sequenced; one from rhizospheric soil, named as MNR3A, and one from internal part of the root, named as EMN2. In parallel, we performed biochemical and physiological experiments to confirm genomic analyses results. Interestingly, EMN2 has achromobactin and aerobactin siderophore receptors, but does not have the genes responsible for the production of these siderophores, suggesting an interesting bacterial competition strategy. The two bacterial isolates were able to degrade and catabolize plant phenolic compounds for their own benefit. Surprisingly, MNR3A and EMN2 do not contain caffeine methylases that are responsible for the catabolism of caffeine. In fact, bench experiments confirm that the bacteria did not metabolize caffeine, but were resistant and chemically attracted to it. Furthermore, both bacteria, most especially MNR3A, were able to increase growth of lettuce plants. Our results indicate MNR3A as a potential plant growth promoting bacteria.
Collapse
Affiliation(s)
- Leandro Pio de Sousa
- Instituto Agronômico de Campinas, IAC, Campinas, SP, Brazil; UNICAMP, Programa de Pós-graduação em Genética e Biologia Molecular, Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
13
|
Zheng Z, Liu H, Shi Y, Liu Z, Teng H, Deng S, Wei L, Wang Y, Zhang F. Comparative transcriptome analysis reveals the resistance regulation mechanism and fungicidal activity of the fungicide phenamacril in Fusarium oxysporum. Sci Rep 2022; 12:11081. [PMID: 35773469 PMCID: PMC9247061 DOI: 10.1038/s41598-022-15188-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/20/2022] [Indexed: 12/21/2022] Open
Abstract
Fusarium oxysporum (Fo) is an important species complex of soil-borne pathogenic fungi that cause vascular wilt diseases of agricultural crops and some opportunistic diseases of humans. The fungicide phenamacril has been extensively reported to have antifungal activity against Fusarium graminearum and Fusarium fujikuroi. In this study, we found that the amino acid substitutions (V151A and S418T) in Type I myosin FoMyo5 cause natural low resistance to phenamacril in the plant pathogenic Fo isolates. Therefore, we compared the transcriptomes of two phenamacril-resistant Fo isolates FoII5, Fo1st and one phenamacril-sensitive isolate Fo3_a after 1 μg/mL phenamacril treatment. Among the 2728 differentially expressed genes (DEGs), 14 DEGs involved in oxidation–reduction processes and MFS transporters, were significantly up-regulated in phenamacril-resistant isolates. On the other hand, 14 DEGs involved in ATP-dependent RNA helicase and ribosomal biogenesis related proteins, showed significantly down-regulated expression in both phenamacril-resistant and -sensitive isolates. These results indicated that phenamacril not only seriously affected the cytoskeletal protein binding and ATPase activity of sensitive isolate, but also suppressed ribosome biogenesis in all the isolates. Hence, this study helps us better understand resistance regulation mechanism and fungicidal activity of phenamacril and provide reference for the development of new fungicides to control Fo.
Collapse
Affiliation(s)
- Zhitian Zheng
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, People's Republic of China.
| | - Huaqi Liu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, People's Republic of China
| | - Yunyong Shi
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, People's Republic of China
| | - Zao Liu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, People's Republic of China
| | - Hui Teng
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, People's Republic of China
| | - Sheng Deng
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, People's Republic of China.
| | - Lihui Wei
- Institute of Plant Protection, Key Lab of Food Quality and Safety of Jiangsu Province-State, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, People's Republic of China
| | - Yunpeng Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, 223003, People's Republic of China.
| | - Feng Zhang
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| |
Collapse
|
14
|
Foley EL, Hvitved AN, Eich RF, Olson JS. Mechanisms of nitric oxide reactions with Globins using mammalian myoglobin as a model system. J Inorg Biochem 2022; 233:111839. [DOI: 10.1016/j.jinorgbio.2022.111839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/15/2022]
|
15
|
Kobayashi K, Igarashi J, Kozawa T. Interdomain Electron Transfer in Flavohemoglobin from
Candida norvegensis
with Antibiotic Azole Compounds. FEBS Lett 2022; 596:938-946. [DOI: 10.1002/1873-3468.14327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Kazuo Kobayashi
- Institute of Scientific and Industrial Research Osaka University Mihogaoka 8‐1 Ibaraki Osaka 567‐0047 Japan
| | - Jotaro Igarashi
- Fukushima Medical University Hikaraigaoka 1 Fukushima 960‐1295 Japan
| | - Takahiro Kozawa
- Institute of Scientific and Industrial Research Osaka University Mihogaoka 8‐1 Ibaraki Osaka 567‐0047 Japan
| |
Collapse
|
16
|
Hussein AHM, El-Adasy ABA, El-Saghier AM, Olish M, Abdelmonsef AH. Synthesis, characterization, in silico molecular docking, and antibacterial activities of some new nitrogen-heterocyclic analogues based on a p-phenolic unit. RSC Adv 2022; 12:12607-12621. [PMID: 35496342 PMCID: PMC9041306 DOI: 10.1039/d2ra01794f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/18/2022] [Indexed: 12/30/2022] Open
Abstract
Compounds 6a and 6b (with pyrimidine moiety, amide linkage, and phenolic substrate) might be potent bacterial flavohemoglobin (flavoHB) inhibitors and they could set a promising starting point for future design of antibacterial agents.
Collapse
Affiliation(s)
| | | | - Ahmed M. El-Saghier
- Chemistry Department, Faculty of Science, Sohag University, 82524 Sohag, Egypt
| | - M. Olish
- Chemistry Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | | |
Collapse
|
17
|
Gardner PR. Ordered Motions in the Nitric-Oxide Dioxygenase Mechanism of Flavohemoglobin and Assorted Globins with Tightly Coupled Reductases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1414:45-96. [PMID: 36520413 DOI: 10.1007/5584_2022_751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nitric-oxide dioxygenases (NODs) activate and combine O2 with NO to form nitrate. A variety of oxygen-binding hemoglobins with associated partner reductases or electron donors function as enzymatic NODs. Kinetic and structural investigations of the archetypal two-domain microbial flavohemoglobin-NOD have illuminated an allosteric mechanism that employs selective tunnels for O2 and NO, gates for NO and nitrate, transient O2 association with ferric heme, and an O2 and NO-triggered, ferric heme spin crossover-driven, motion-controlled, and dipole-regulated electron-transfer switch. The proposed mechanism facilitates radical-radical coupling of ferric-superoxide with NO to form nitrate while preventing suicidal ferrous-NO formation. Diverse globins display the structural and functional motifs necessary for a similar allosteric NOD mechanism. In silico docking simulations reveal monomeric erythrocyte hemoglobin alpha-chain and beta-chain intrinsically matched and tightly coupled with NADH-cytochrome b5 oxidoreductase and NADPH-cytochrome P450 oxidoreductase, respectively, forming membrane-bound flavohemoglobin-like mammalian NODs. The neuroprotective neuroglobin manifests a potential NOD role in a close-fitting ternary complex with membrane-bound NADH-cytochrome b5 oxidoreductase and cytochrome b5. Cytoglobin interfaces weakly with cytochrome b5 for O2 and NO-regulated electron-transfer and coupled NOD activity. The mechanistic model also provides insight into the evolution of O2 binding cooperativity in hemoglobin and a basis for the discovery of allosteric NOD inhibitors.
Collapse
|
18
|
Wan X, Brynildsen MP. Robustness of nitric oxide detoxification to nitrogen starvation in Escherichia coli requires RelA. Free Radic Biol Med 2021; 176:286-297. [PMID: 34624482 DOI: 10.1016/j.freeradbiomed.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/04/2021] [Indexed: 01/18/2023]
Abstract
Reactive nitrogen species and nutrient deprivation are two elements of the immune response used to eliminate pathogens within phagosomes. Concomitantly, pathogenic bacteria have evolved defense systems to cope with phagosomal stressors, which include enzymes that detoxify nitric oxide (•NO) and respond to nutrient scarcity. A deeper understanding of how those defense systems are deployed under adverse conditions that contain key elements of phagosomes will facilitate targeting of those systems for therapeutic purposes. Here we investigated how Escherichia coli detoxifies •NO in the absence of useable nitrogen, because nitrogen availability is limited in phagosomes due to the removal of nitrogenous compounds (e.g., amino acids). We hypothesized that nitrogen starvation would impair •NO detoxification by E. coli because it depresses translation rates and the main E. coli defense enzyme, Hmp, is synthesized in response to •NO. However, we found that E. coli detoxifies •NO at the same rate regardless of whether useable nitrogen was present. We confirmed that the nitrogen in •NO and its autoxidation products could not be used by E. coli under our experimental conditions, and discovered that •NO eliminated differences in carbon and oxygen consumption between nitrogen-replete and nitrogen-starved cultures. Interestingly, E. coli does not consume measurable extracellular nitrogen during •NO stress despite the need to translate defense enzymes. Further, we found that RelA, which responds to uncharged tRNA, was required to observe the robustness of •NO detoxification to nitrogen starvation. These data demonstrate that E. coli is well poised to detoxify •NO in the absence of useable nitrogen and suggest that the stringent response could be a useful target to potentiate the antibacterial activity of •NO.
Collapse
Affiliation(s)
- Xuanqing Wan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA
| | - Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
19
|
Verma R, Pradhan D, Nayek A, Singh H, Jain AK, Khan LA. Target-based drug repurposing against Candida albicans-A computational modeling, docking, and molecular dynamic simulations study. J Cell Biochem 2021; 123:289-305. [PMID: 34672012 DOI: 10.1002/jcb.30163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/21/2022]
Abstract
The emergence of multidrug-resistant strains of Candida albicans has become a global threat mostly due to co-infection with immune-compromised patients leading to invasive candidiasis. The life-threatening form of the disease can be managed quickly and effectively by drug repurposing. Thus, the study used in silico approaches to evaluate Food and Drug Administration (FDA) approved drugs against three drug targets-TRR1, TOM40, and YHB1. The tertiary structures of three drug targets were modeled, refined, and evaluated for their structural integrity based on PROCHECK, ERRAT, and PROSA. High-throughput virtual screening of FDA-approved drugs (8815), interaction analysis, and energy profiles had revealed that DB01102 (Arbutamine), DB01611 (Hydroxychloroquine), and DB09319 (Carindacillin) exhibited better binding affinity with TRR1, TOM40, and YHB1, respectively. Notably, the molecular dynamic simulation explored that Gln45, Thr119, and Asp288 of TRR1; Thr107 and Ser121 of TOM40; Arg193, Glu213, and Ser228 of YHB1 are crucial residues for stable drug-target interaction. Additionally, it also prioritized Arbutamine-TRR1 as the best drug-target complex based on MM-PBSA (-52.72 kcal/mol), RMSD (2.43 Å), and radius of gyration (-21.49 Å) analysis. In-depth, PCA results supported the findings of molecular dynamic simulations. Interestingly, the conserved region (>70%) among the TRR1 sequences from pathogenic Candida species indicated the effectiveness of Arbutamine against multiple species of Candida as well. Thus, the study dispenses new insight and enriches the understanding of developing an advanced technique to consider potential antifungals against C. albicans. Nonetheless, a detailed experimental validation is needed to investigate the efficacy of Arbutamin against life-threatening candidiasis.
Collapse
Affiliation(s)
- Rashi Verma
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India.,Biomedical Informatics Centre, Indian Council of Medical Research, New Delhi, India
| | - Dibyabhaba Pradhan
- Computational Genomics Centre, All India Institute of Medical Sciences, Indian Council of Medical Research, New Delhi, India
| | - Arnab Nayek
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Harpreet Singh
- Computational Genomics Centre, All India Institute of Medical Sciences, Indian Council of Medical Research, New Delhi, India
| | - Arun Kumar Jain
- Biomedical Informatics Centre, Indian Council of Medical Research, New Delhi, India
| | | |
Collapse
|
20
|
Das A, Rajeev A, Bhunia S, Arunkumar M, Chari N, Sankaralingam M. Synthesis, characterization and antimicrobial activity of nickel(II) complexes of tridentate N3 ligands. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
21
|
Saghaug CS, Klotz C, Kallio JP, Aebischer T, Langeland N, Hanevik K. Genetic Diversity of the Flavohemoprotein Gene of Giardia lamblia: Evidence for High Allelic Heterozygosity and Copy Number Variation. Infect Drug Resist 2020; 13:4531-4545. [PMID: 33376360 PMCID: PMC7755369 DOI: 10.2147/idr.s274543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose The flavohemoprotein (gFlHb) in Giardia plays an important role in managing nitrosative and oxidative stress, and potentially also in virulence and nitroimidazole drug tolerance. The aim of this study was to analyze the genetic diversity of gFlHb in Giardia assemblages A and B clinical isolates. Methods gFlHb genes from 20 cultured clinical Giardia isolates were subjected to PCR amplification and cloning, followed by Sanger sequencing. Sequences of all cloned PCR fragments from each isolate were analyzed for single nucleotide variants (SNVs) and compared to genomic Illumina sequence data. Identical clone sequences were sorted into alleles, and diversity was further analyzed. The number of gFlHb gene copies was assessed by mining PacBio de novo assembled genomes in eight isolates. Homology models for assessment of SNV's potential impact on protein function were created using Phyre2. Results A variable copy number of the gFlHb gene, between two and six copies, depending on isolate, was found. A total of 37 distinct sequences, representing different alleles of the gFlHb gene, were identified in AII isolates, and 41 were identified in B isolates. In some isolates, up to 12 different alleles were found. The total allelic diversity was high for both assemblages (>0.9) and was coupled with a nucleotide diversity of <0.01. The genetic variation (SNVs per CDS length) was 4.8% in sub-assemblage AII and 5.4% in assemblage B. The number of non-synonymous (ns) SNVs was high in gFIHb of both assemblages, 1.6% in A and 3.0% in B, respectively. Some of the identified nsSNV are predicted to alter protein structure and possibly function. Conclusion In this study, we present evidence that gFlHb, a putative protective enzyme against oxidative and nitrosative stress in Giardia, is a variable copy number gene with high allelic diversity. The genetic variability of gFlHb may contribute metabolic adaptability against metronidazole toxicity.
Collapse
Affiliation(s)
- Christina S Saghaug
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Christian Klotz
- Department of Infectious Diseases, Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| | - Juha P Kallio
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Toni Aebischer
- Department of Infectious Diseases, Unit 16 Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institute, Berlin, Germany
| | - Nina Langeland
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Norwegian National Advisory Unit on Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
22
|
Gardner AM, Gardner PR. Allostery in the nitric oxide dioxygenase mechanism of flavohemoglobin. J Biol Chem 2020; 296:100186. [PMID: 33310705 PMCID: PMC7948479 DOI: 10.1074/jbc.ra120.016637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
The substrates O2 and NO cooperatively activate the NO dioxygenase function of Escherichia coli flavohemoglobin. Steady-state and transient kinetic measurements support a structure-based mechanistic model in which O2 and NO movements and conserved amino acids at the E11, G8, E2, E7, B10, and F7 positions within the globin domain control activation. In the cooperative and allosteric mechanism, O2 migrates to the catalytic heme site via a long hydrophobic tunnel and displaces LeuE11 away from the ferric iron, which forces open a short tunnel to the catalytic site gated by the ValG8/IleE15 pair and LeuE11. NO permeates this tunnel and leverages upon the gating side chains triggering the CD loop to furl, which moves the E and F-helices and switches an electron transfer gate formed by LysF7, GlnE7, and water. This allows FADH2 to reduce the ferric iron, which forms the stable ferric–superoxide–TyrB10/GlnE7 complex. This complex reacts with internalized NO with a bimolecular rate constant of 1010 M−1 s−1 forming nitrate, which migrates to the CD loop and unfurls the spring-like structure. To restart the cycle, LeuE11 toggles back to the ferric iron. Actuating electron transfer with O2 and NO movements averts irreversible NO poisoning and reductive inactivation of the enzyme. Together, structure snapshots and kinetic constants provide glimpses of intermediate conformational states, time scales for motion, and associated energies.
Collapse
Affiliation(s)
- Anne M Gardner
- Research and Development Division, Miami Valley Biotech, Dayton, Ohio, USA; Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Paul R Gardner
- Research and Development Division, Miami Valley Biotech, Dayton, Ohio, USA; Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Chemistry and Biochemistry Department, University of Dayton, Dayton, Ohio, USA.
| |
Collapse
|
23
|
Sari S, Avci A, Koçak E, Kart D, Sabuncuoğlu S, Doğan İS, Özdemir Z, Bozbey İ, Karakurt A, Saraç S, Dalkara S. Antibacterial azole derivatives: Antibacterial activity, cytotoxicity, and in silico mechanistic studies. Drug Dev Res 2020; 81:1026-1036. [PMID: 33216362 DOI: 10.1002/ddr.21721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/13/2020] [Accepted: 07/03/2020] [Indexed: 01/09/2023]
Abstract
Azole antifungal drugs are commonly used in antifungal chemotherapy. Antibacterial effects of some topical antifungals, such as miconazole and econazole, have lately been revealed, which suggests a promising venue in antimicrobial chemotherapy. In this study, we tested an in-house azole collection with antifungal properties for their antibacterial activity to identify dual-acting hits using the broth microdilution method. The in vitro screen yielded a number of potent derivatives against gram-positive bacteria, Enterococcus faecalis and Staphylococcus aureus. Compound 73's minimum inhibitory concentration (MIC) value less than 1 μg/ml against S. aureus; however, none of the compounds showed noteworthy activity against methicillin-resistant S. aureus (MRSA). All the active compounds were found safe at their MIC values against the healthy fibroblast cells in the in vitro cytotoxicity test. Molecular docking studies of the most active compounds using a set of docking programs with flavohemoglobin (flavoHb) structure, the proposed target of the azole antifungals with antibacterial activity, presented striking similarities regarding the binding modes and interactions between the tested compounds and the antifungal drugs with crystallographic data. In addition to being noncytotoxic, the library was predicted to be drug-like and free of pan-assay interference compounds (PAINS). As a result, the current study revealed several potential azole derivatives with both antifungal and antibacterial activities. Inhibition of bacterial flavoHb was suggested as a possible mechanism of action for the title compounds.
Collapse
Affiliation(s)
- Suat Sari
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Ahmet Avci
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Ebru Koçak
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Didem Kart
- Department of Pharmaceutical Microbiology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Suna Sabuncuoğlu
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - İnci Selin Doğan
- Department of Pharmaceutical Chemistry, Karadeniz Technical University Faculty of Pharmacy, Trabzon, Turkey
| | - Zeynep Özdemir
- Department of Pharmaceutical Chemistry, İnönü University Faculty of Pharmacy, Malatya, Turkey
| | - İrem Bozbey
- Department of Pharmaceutical Chemistry, Erzincan Binali Yıldırım University Faculty of Pharmacy, Erzincan, Turkey
| | - Arzu Karakurt
- Department of Pharmaceutical Chemistry, İnönü University Faculty of Pharmacy, Malatya, Turkey
| | - Selma Saraç
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Sevim Dalkara
- Department of Pharmaceutical Chemistry, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| |
Collapse
|
24
|
Chou WK, Vaikunthan M, Schröder HV, Link AJ, Kim H, Brynildsen MP. Synergy Screening Identifies a Compound That Selectively Enhances the Antibacterial Activity of Nitric Oxide. Front Bioeng Biotechnol 2020; 8:1001. [PMID: 32984281 PMCID: PMC7477088 DOI: 10.3389/fbioe.2020.01001] [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: 02/12/2020] [Accepted: 07/31/2020] [Indexed: 01/04/2023] Open
Abstract
Antibiotic resistance poses a serious threat to global health. To reinforce the anti-infective arsenal, many novel therapeutic strategies to fight bacterial infections are being explored. Among them, anti-virulence therapies, which target pathways important for virulence, have attracted much attention. Nitric oxide (NO) defense systems have been identified as critical for the pathogenesis of various bacteria, making them an appealing therapeutic target. In this study, we performed chemical screens to identify inhibitors of NO detoxification in Escherichia coli. We found that 2-mercaptobenzothiazole (2-MBT) can potently inhibit cellular detoxification of NO, achieving a level of inhibition that resembled the effect of genetically removing Hmp, the dominant detoxification enzyme under oxygenated conditions. Further analysis revealed that in the presence of NO, 2-MBT impaired the catalysis of Hmp and synthesis of Hmp and other proteins, whereas in its absence there were minimal perturbations to growth and protein synthesis. In addition, by studying the structure-activity relationship of 2-MBT, we found that both sulfur atoms in 2-MBT were vital for its inhibition of NO detoxification. Interestingly, when 2-mercaptothiazole (2-MT), which lacked the benzene ring, was used, differing biological activities were observed, although they too were NO dependent. Specifically, 2-MT could still prohibit NO detoxification, though it did not interfere with Hmp catalysis; rather, it was a stronger inhibitor of protein synthesis and it reduced the transcript levels of hmp, which was not observed with 2-MBT. Overall, these results provide a strong foundation for further exploration of 2-MBT and 2-MT for therapeutic applications.
Collapse
Affiliation(s)
- Wen Kang Chou
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Mathini Vaikunthan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Hendrik V. Schröder
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| | - A. James Link
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
- Frick Chemistry Laboratory, Department of Chemistry, Princeton University, Princeton, NJ, United States
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Hahn Kim
- Frick Chemistry Laboratory, Department of Chemistry, Princeton University, Princeton, NJ, United States
- Small Molecule Screening Center, Princeton University, Princeton, NJ, United States
| | - Mark P. Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| |
Collapse
|
25
|
Khramtsov YV, Ulasov AV, Rosenkranz AA, Georgiev GP, Sobolev AS. Stabilization of Modular Nanotransporters by Embedding Hemin in Them in a New Strain with Heme Receptor Expression. DOKL BIOCHEM BIOPHYS 2020; 490:47-49. [PMID: 32342313 DOI: 10.1134/s1607672920010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 11/23/2022]
Abstract
It was found that the use of a new strain-producer Escherichia coli, expressing the heme receptor ChuA, enables obtaining a hemin-containing modular nanotransporter (MNT) for drug delivery into the nuclei of target cells. The hemin-containing MNT becomes stabilized, which leads to an increase in its thermal stability and prevents aggregation of this protein.
Collapse
Affiliation(s)
- Yu V Khramtsov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A V Ulasov
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A A Rosenkranz
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Moscow State University, Moscow, Russia
| | - G P Georgiev
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - A S Sobolev
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia. .,Moscow State University, Moscow, Russia.
| |
Collapse
|
26
|
Abstract
Flavohaemoglobins were first described in yeast as early as the 1970s but their functions were unclear. The surge in interest in nitric oxide biology and both serendipitous and hypothesis-driven discoveries in bacterial systems have transformed our understanding of this unusual two-domain globin into a comprehensive, yet undoubtedly incomplete, appreciation of its pre-eminent role in nitric oxide detoxification. Here, I focus on research on the flavohaemoglobins of microorganisms, especially of bacteria, and update several earlier and more comprehensive reviews, emphasising advances over the past 5 to 10 years and some controversies that have arisen. Inevitably, in light of space restrictions, details of nitric oxide metabolism and globins in higher organisms are brief.
Collapse
Affiliation(s)
- Robert K. Poole
- Department of Molecular Biology and Biotechnology, The University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| |
Collapse
|
27
|
Abstract
Nitric oxide (NO) is a radical that is used as an attack molecule by immune cells. NO can interact and damage a range of biomolecules, and the biological outcome for bacteria assaulted with NO will be governed by how the radical distributes within their biochemical reaction networks. Measurement of those NO fluxes is complicated by the low abundance and transience of many of its reaction products. To overcome this challenge, we use computational modeling to translate measurements of several biochemical species (e.g., NO, O2, NO2-) into NO flux distributions. In this chapter, we provide a detailed protocol, which includes experimental measurements and computational modeling, to estimate the NO flux distribution in an Escherichia coli culture. Those fluxes will have uncertainty associated with them and we also discuss how further experiments and modeling can be employed for flux refinement.
Collapse
Affiliation(s)
| | - Xuanqing Wan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
28
|
Butcher D, Moussaoui M, Baciou L, Miksovska J. Impact of azole drugs on energetics, kinetics, and ligand migration pathways of CO photo-dissociation in bacterial flavohemoglobins. RSC Adv 2020; 10:17930-17941. [PMID: 35515592 PMCID: PMC9053618 DOI: 10.1039/d0ra02529a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/21/2020] [Indexed: 01/18/2023] Open
Abstract
Flavohemoglobins (fHbs) are heme proteins found in prokaryotic and eukaryotic microbes. They are involved in NO detoxification through an NO˙ dioxygenase mechanism. The N-terminal heme globin domain allows for binding of gaseous ligands whereas a C-terminal NADH/FADH binding domain facilitates association of redox cofactors necessary for ligand reduction. The NO˙ dioxygenase function is important in facilitating immune resistance by protecting the cell from nitrosative stress brought about by a host organism; as a result, bacterial flavoHbs have recently been considered as targets for the development of new antibiotics. Here, photoacoustic calorimetry and transient absorption spectroscopy have been used to characterize energetics, structural dynamics, and kinetics of CO migration within bacterial flavoHbs from Ralstonia eutropha (FHP) and Staphylococcus aureus (HMPSa) in the presence and absence of antibiotic azole compounds. In FHP, the ligand photo-release is associated with ΔH = 26.2 ± 7.0 kcal mol−1 and ΔV = 25.0 ± 1.5 mL mol−1 while in HMPSa, ΔH = 34.7 ± 8.0 kcal mol−1 and ΔV = 28.6 ± 17 mL mol−1 were observed, suggesting distinct structural changes associated with ligand escape from FHP and HMPSa. In the presence of ketoconazole, the CO escape leads to a more negative enthalpy change and volume change whereas association of miconazole to FHP or HMPSa does not impact the reaction volume. These data are in agreement with the computational results that propose distinct binding sites for ketoconazole and miconazole on CO bound FHP. Miconazole or ketoconazole binding to either protein has only a negligible impact on the CO association rates, indicating that azole drugs do not impact flavoHbs interactions with gaseous ligands but may inhibit the NOD activity through preventing the electron transfer between FAD and heme cofactors. Impact of ketoconalzole and miconazole on structural dynamics of flavohemoglobin.![]()
Collapse
Affiliation(s)
- David Butcher
- Department of Chemistry and Biochemistry
- Florida International University
- Miami
- USA
| | - Myriam Moussaoui
- Laboratoire de Chimie Physique
- UMR8000
- Université Paris Sud
- CNRS
- Université Paris Saclay
| | - Laura Baciou
- Laboratoire de Chimie Physique
- UMR8000
- Université Paris Sud
- CNRS
- Université Paris Saclay
| | - Jaroslava Miksovska
- Department of Chemistry and Biochemistry
- Florida International University
- Miami
- USA
- Biomolecular Sciences Institute
| |
Collapse
|
29
|
Lindahl PA. A comprehensive mechanistic model of iron metabolism in Saccharomyces cerevisiae. Metallomics 2019; 11:1779-1799. [PMID: 31531508 DOI: 10.1039/c9mt00199a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ironome of budding yeast (circa 2019) consists of approximately 139 proteins and 5 nonproteinaceous species. These proteins were grouped according to location in the cell, type of iron center(s), and cellular function. The resulting 27 groups were used, along with an additional 13 nonprotein components, to develop a mesoscale mechanistic model that describes the import, trafficking, metallation, and regulation of iron within growing yeast cells. The model was designed to be simultaneously mutually autocatalytic and mutually autoinhibitory - a property called autocatinhibitory that should be most realistic for simulating cellular biochemical processes. The model was assessed at the systems' level. General conclusions are presented, including a new perspective on understanding regulatory mechanisms in cellular systems. Some unsettled issues are described. This model, once fully developed, has the potential to mimic the phenotype (at a coarse-grain level) of all iron-related genetic mutations in this simple and well-studied eukaryote.
Collapse
Affiliation(s)
- Paul A Lindahl
- Departments of Chemistry and of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843-3255, USA.
| |
Collapse
|
30
|
Brynildsen MP. Nitric Oxide Stress as a Metabolic Flux. Adv Microb Physiol 2018; 73:63-76. [PMID: 30262110 DOI: 10.1016/bs.ampbs.2018.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nitric oxide (NO) is an antimicrobial metabolite produced by immune cells to prohibit infection. Due to its reactivity, NO has numerous reaction routes available to it in biological systems with some leading to cellular damage and others producing innocuous compounds. Pathogens have evolved resistance mechanisms toward NO, and many of these take the form of enzymes that chemically passivate the molecule. In essence, bacteria have channeled NO flux toward useful or harmless compounds, and away from pathways that damage cellular components. Pathogens devoid of detoxification enzymes have been found to have compromised survival in different infection models, which suggests that diverting flux away from NO defenses could be a viable antiinfective strategy. From this perspective, potentiation of NO stress mirrors challenges in metabolic engineering where researchers endeavor to divert flux away from endogenous pathways and toward those that produce desirable biomolecules. In this review, we cast NO stress as a metabolic flux and discuss how the tools and methodologies of metabolic engineering are well suited for analysis of this bacterial stress response. We provide examples of such interdisciplinary applications, discuss the benefits of considering NO stress from a flux perspective, as well as the pitfalls, and offer a vision for how metabolic engineering analyses can assist in deciphering the economics underlying bacterial responses to multistress conditions that are characteristic of the phagosomes of immune cells.
Collapse
Affiliation(s)
- Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States.
| |
Collapse
|
31
|
Moussaoui M, Misevičienė L, Anusevičius Ž, Marozienė A, Lederer F, Baciou L, Čėnas N. Quinones and nitroaromatic compounds as subversive substrates of Staphylococcus aureus flavohemoglobin. Free Radic Biol Med 2018; 123:107-115. [PMID: 29793040 DOI: 10.1016/j.freeradbiomed.2018.05.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/09/2018] [Accepted: 05/18/2018] [Indexed: 11/23/2022]
Abstract
In microorganisms, flavohemoglobins (FHbs) containing FAD and heme (Fe3+, metHb) convert NO. into nitrate at the expense of NADH and O2. FHbs contribute to bacterial resistance to nitrosative stress. Therefore, inhibition of FHbs functions may decrease the pathogen virulence. We report here a kinetic study of the reduction of quinones and nitroaromatic compounds by S. aureus FHb. We show that this enzyme rapidly reduces quinones and nitroaromatic compounds in a mixed single- and two-electron pathway. The reactivity of nitroaromatics increased upon an increase in their single-electron reduction potential (E17), whereas the reactivity of quinones poorly depended on their E17 with a strong preference for a 2-hydroxy-1,4-naphthoquinone structure. The reaction followed a 'ping-pong' mechanism. In general, the maximal reaction rates were found lower than the maximal presteady-state rate of FAD reduction by NADH and/or of oxyhemoglobin (HbFe2+O2) formation (~130 s-1, pH 7.0, 25 °C), indicating that the enzyme turnover is limited by the oxidative half-reaction. The turnover studies showed that quinones prefreqently accept electrons from reduced FAD, and not from HbFe2+O2. These results suggest that quinones and nitroaromatics act as 'subversive substrates' for FHb, and may enhance the cytotoxicity of NO. by formation of superoxide and by diverting the electron flux coming from reduced FAD. Because quinone reduction rate was increased by FHb inhibitors such as econazole, ketoconazole, and miconazole, their combined use may represent a novel chemotherapeutical approach.
Collapse
Affiliation(s)
- Myriam Moussaoui
- Laboratoire de Chimie Physique, Université Paris Sud, CNRS UMR 8000, 91405 Orsay Cedex France
| | - Lina Misevičienė
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Žilvinas Anusevičius
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Audronė Marozienė
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania
| | - Florence Lederer
- Laboratoire de Chimie Physique, Université Paris Sud, CNRS UMR 8000, 91405 Orsay Cedex France
| | - Laura Baciou
- Laboratoire de Chimie Physique, Université Paris Sud, CNRS UMR 8000, 91405 Orsay Cedex France
| | - Narimantas Čėnas
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania.
| |
Collapse
|
32
|
Abstract
Urinary tract infection (UTI) is one of the most common bacterial infections in humans, and the majority are caused by uropathogenic Escherichia coli (UPEC). The rising antibiotic resistance among UPEC and the frequent failure of antibiotics to effectively treat recurrent UTI and catheter-associated UTI motivate research on alternative ways of managing UTI. Abundant evidence indicates that the toxic radical nitric oxide (NO), formed by activation of the inducible nitric oxide synthase, plays an important role in host defence to bacterial infections, including UTI. The major source of NO production during UTI is from inflammatory cells, especially neutrophils, and from the uroepithelial cells that are known to orchestrate the innate immune response during UTI. NO and reactive nitrogen species have a wide range of antibacterial targets, including DNA, heme proteins, iron-sulfur clusters, and protein thiol groups. However, UPEC have acquired a variety of defence mechanisms for protection against NO, such as the NO-detoxifying enzyme flavohemoglobin and the NO-tolerant cytochrome bd-I respiratory oxidase. The cytotoxicity of NO-derived intermediates is nonspecific and may be detrimental to host cells, and a balanced NO production is crucial to maintain the tissue integrity of the urinary tract. In this review, we will give an overview of how NO production from host cells in the urinary tract is activated and regulated, the effect of NO on UPEC growth and colonization, and the ability of UPEC to protect themselves against NO. We also discuss the attempts that have been made to develop NO-based therapeutics for UTI treatment.
Collapse
|
33
|
Tatara AM, Rozich AJ, Kontoyiannis PD, Watson E, Albert ND, Bennett GN, Mikos AG. Econazole-releasing porous space maintainers for fungal periprosthetic joint infection. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:70. [PMID: 29752591 PMCID: PMC6009980 DOI: 10.1007/s10856-018-6073-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/21/2018] [Indexed: 06/08/2023]
Abstract
While antibiotic-eluting polymethylmethacrylate space maintainers have shown efficacy in the treatment of bacterial periprosthetic joint infection and osteomyelitis, antifungal-eluting space maintainers are associated with greater limitations for treatment of fungal musculoskeletal infections including limited elution concentration and duration. In this study, we have designed a porous econazole-eluting space maintainer capable of greater inhibition of fungal growth than traditional solid space maintainers. The eluted econazole demonstrated bioactivity in a concentration-dependent manner against the most common species responsible for fungal periprosthetic joint infection as well as staphylococci. Lastly, these porous space maintainers retain compressive mechanical properties appropriate to maintain space before definitive repair of the joint or bony defect.
Collapse
Affiliation(s)
- Alexander M Tatara
- Department of Bioengineering, Rice University, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Emma Watson
- Department of Bioengineering, Rice University, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Nathaniel D Albert
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
34
|
Nitric Oxide-Mediated Induction of Dispersal in Pseudomonas aeruginosa Biofilms Is Inhibited by Flavohemoglobin Production and Is Enhanced by Imidazole. Antimicrob Agents Chemother 2018; 62:AAC.01832-17. [PMID: 29263060 DOI: 10.1128/aac.01832-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023] Open
Abstract
The biological signal molecule nitric oxide (NO) was found to induce biofilm dispersal across a range of bacterial species, which led to its consideration for therapeutic strategies to treat biofilms and biofilm-related infections. However, biofilms are often not completely dispersed after exposure to NO. To better understand this phenomenon, we investigated the response of Pseudomonas aeruginosa biofilm cells to successive NO treatments. When biofilms were first pretreated with a low, noneffective dose of NO, a second dose of the signal molecule at a concentration usually capable of inducing dispersal did not have any effect. Amperometric analysis revealed that pretreated P. aeruginosa cells had enhanced NO-scavenging activity, and this effect was associated with the production of the flavohemoglobin Fhp. Further, quantitative real-time reverse transcription-PCR (qRT-PCR) analysis showed that fhp expression increased by over 100-fold in NO-pretreated biofilms compared to untreated biofilms. Biofilms of mutant strains harboring mutations in fhp or fhpR, encoding a NO-responsive regulator of fhp, were not affected in their dispersal response after the initial pretreatment with NO. Overall, these results suggest that FhpR can sense NO to trigger production of the flavohemoglobin Fhp and inhibit subsequent dispersal responses to NO. Finally, the addition of imidazole, which can inhibit the NO dioxygenase activity of flavohemoglobin, attenuated the prevention of dispersal after NO pretreatment and improved the dispersal response in older, starved biofilms. This study clarifies the underlying mechanisms of impaired dispersal induced by repeated NO treatments and offers a new perspective for improving the use of NO in biofilm control strategies.
Collapse
|
35
|
Abstract
Nitric oxide (NO) is a cellular signalling molecule widely conserved among organisms, including microorganisms such as bacteria, yeasts, and fungi, and higher eukaryotes such as plants and mammals. NO is mainly produced by the activities of NO synthase (NOS) or nitrite reductase (NIR). There are several NO detoxification systems, including NO dioxygenase (NOD) and S-nitrosoglutathione reductase (GSNOR). NO homeostasis, based on the balance between NO synthesis and degradation, is important for regulating its physiological functions, since an excess of NO causes nitrosative stress due to the high reactivity of NO and NO-derived compounds. In yeast, NO may be involved in stress responses, but the role of NO and the mechanism underlying NO signalling are poorly understood due to the lack of mammalian NOS orthologs in the yeast genome. NOS and NIR activities have been observed in yeast cells, but the gene-encoding NOS and the mechanism by which NO production is catalysed by NIR remain unclear. On the other hand, yeast cells employ NOD and GSNOR to maintain intracellular redox balance following endogenous NO production, treatment with exogenous NO, or exposure to environmental stresses. This article reviews NO metabolism (synthesis, degradation) and its regulation in yeast. The physiological roles of NO in yeast, including the oxidative stress response, are also discussed. Such investigations into NO signalling are essential for understanding how NO modulates the genetics and physiology of yeast. In addition to being responsible for the pathology and pharmacology of various degenerative diseases, NO signalling may be a potential target for the construction and engineering of industrial yeast strains.
Collapse
|
36
|
Gell DA. Structure and function of haemoglobins. Blood Cells Mol Dis 2017; 70:13-42. [PMID: 29126700 DOI: 10.1016/j.bcmd.2017.10.006] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/29/2017] [Accepted: 10/30/2017] [Indexed: 12/18/2022]
Abstract
Haemoglobin (Hb) is widely known as the iron-containing protein in blood that is essential for O2 transport in mammals. Less widely recognised is that erythrocyte Hb belongs to a large family of Hb proteins with members distributed across all three domains of life-bacteria, archaea and eukaryotes. This review, aimed chiefly at researchers new to the field, attempts a broad overview of the diversity, and common features, in Hb structure and function. Topics include structural and functional classification of Hbs; principles of O2 binding affinity and selectivity between O2/NO/CO and other small ligands; hexacoordinate (containing bis-imidazole coordinated haem) Hbs; bacterial truncated Hbs; flavohaemoglobins; enzymatic reactions of Hbs with bioactive gases, particularly NO, and protection from nitrosative stress; and, sensor Hbs. A final section sketches the evolution of work on the structural basis for allosteric O2 binding by mammalian RBC Hb, including the development of newer kinetic models. Where possible, reference to historical works is included, in order to provide context for current advances in Hb research.
Collapse
Affiliation(s)
- David A Gell
- School of Medicine, University of Tasmania, TAS 7000, Australia.
| |
Collapse
|
37
|
Brengel C, Thomann A, Schifrin A, Allegretta G, Kamal AAM, Haupenthal J, Schnorr I, Cho SH, Franzblau SG, Empting M, Eberhard J, Hartmann RW. Biophysical Screening of a Focused Library for the Discovery of CYP121 Inhibitors as Novel Antimycobacterials. ChemMedChem 2017; 12:1616-1626. [DOI: 10.1002/cmdc.201700363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/04/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Christian Brengel
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Andreas Thomann
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Alexander Schifrin
- Department of Biochemistry; Saarland University; Campus B2.2 66123 Saarbrücken Germany
| | - Giuseppe Allegretta
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Ahmed A. M. Kamal
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Isabell Schnorr
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Sang Hyun Cho
- Institute for Tuberculosis Research; College of Pharmacy; University of Illinois at Chicago; 833 S. Wood Street Chicago IL 60612-7231 USA
| | - Scott G. Franzblau
- Institute for Tuberculosis Research; College of Pharmacy; University of Illinois at Chicago; 833 S. Wood Street Chicago IL 60612-7231 USA
| | - Martin Empting
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Jens Eberhard
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
| | - Rolf W. Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland, HIPS; Department for Drug Design and Optimization; Campus E8.1 66123 Saarbrücken Germany
- Department of Pharmacy; Pharmaceutical and Medicinal Chemistry; Saarland University; Campus C2.3 66123 Saarbrücken Germany
| |
Collapse
|
38
|
Nenoff P, Koch D, Krüger C, Drechsel C, Mayser P. New insights on the antibacterial efficacy of miconazole in vitro. Mycoses 2017; 60:552-557. [PMID: 28370366 DOI: 10.1111/myc.12620] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/25/2017] [Accepted: 03/03/2017] [Indexed: 11/29/2022]
Abstract
Miconazole is a broad-spectrum antifungal used in topical preparations. In the present investigation the minimal inhibitory concentration (MIC) of miconazole for eighty wild type strains of gram-positive and gram-negative bacteria isolated from infected skin lesions was assessed using a modified agar dilution test (adapted to CLSI, Clinical Laboratory Standards Institute). 14 ATCC reference strains served as controls. Miconazole was found efficacious against gram-positive aerobic bacteria (n=62 species), the MICs against Staphylococcus (S.) aureus, S. spp., Streptococcus spp. und Enterococcus spp. ranged between 0.78 and 6.25 μg/mL. Interestingly, there were no differences in susceptibility between methicillin-susceptible (MSSA, 3) methicillin-resistant (MRSA, 6) and fusidic acid-resistant (FRSA, 2) S. aureus isolates. Strains of Streptococcus pyogenes (A-streptococci) (8) were found to be slightly more sensitive (0.78-1.563 μg/mL), while for gram-negative bacteria, no efficacy was found within the concentrations tested (MIC >200 μg/mL). In conclusion, for the gram-positive aerobic bacteria the MICs of miconazole were found within a range which is much lower than the concentration of miconazole used in topical preparations (2%). Thus topically applied miconazole might be a therapeutic option in skin infections especially caused by gram-positive bacteria even by those strains which are resistant to antibiotics.
Collapse
Affiliation(s)
- P Nenoff
- Labor für Medizinische Mikrobiologie, Rötha/OT Mölbis, Germany
| | - D Koch
- Labor für Medizinische Mikrobiologie, Rötha/OT Mölbis, Germany
| | - C Krüger
- Labor für Medizinische Mikrobiologie, Rötha/OT Mölbis, Germany
| | | | | |
Collapse
|
39
|
Ansell BRE, Baker L, Emery SJ, McConville MJ, Svärd SG, Gasser RB, Jex AR. Transcriptomics Indicates Active and Passive Metronidazole Resistance Mechanisms in Three Seminal Giardia Lines. Front Microbiol 2017; 8:398. [PMID: 28367140 PMCID: PMC5355454 DOI: 10.3389/fmicb.2017.00398] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/27/2017] [Indexed: 12/13/2022] Open
Abstract
Giardia duodenalis is an intestinal parasite that causes 200-300 million episodes of diarrhoea annually. Metronidazole (Mtz) is a front-line anti-giardial, but treatment failure is common and clinical resistance has been demonstrated. Mtz is thought to be activated within the parasite by oxidoreductase enzymes, and to kill by causing oxidative damage. In G. duodenalis, Mtz resistance involves active and passive mechanisms. Relatively low activity of iron-sulfur binding proteins, namely pyruvate:ferredoxin oxidoreductase (PFOR), ferredoxins, and nitroreductase-1, enable resistant cells to passively avoid Mtz activation. Additionally, low expression of oxygen-detoxification enzymes can allow passive (non-enzymatic) Mtz detoxification via futile redox cycling. In contrast, active resistance mechanisms include complete enzymatic detoxification of the pro-drug by nitroreductase-2 and enhanced repair of oxidized biomolecules via thioredoxin-dependent antioxidant enzymes. Molecular resistance mechanisms may be largely founded on reversible transcriptional changes, as some resistant lines revert to drug sensitivity during drug-free culture in vitro, or passage through the life cycle. To comprehensively characterize these changes, we undertook strand-specific RNA sequencing of three laboratory-derived Mtz-resistant lines, 106-2ID10, 713-M3, and WB-M3, and compared transcription relative to their susceptible parents. Common up-regulated genes encoded variant-specific surface proteins (VSPs), a high cysteine membrane protein, calcium and zinc channels, a Mad-2 cell cycle regulator and a putative fatty acid α-oxidase. Down-regulated genes included nitroreductase-1, putative chromate and quinone reductases, and numerous genes that act proximal to PFOR. Transcriptional changes in 106-2ID10 diverged from those in 713-r and WB-r (r ≤ 0.2), which were more similar to each other (r = 0.47). In 106-2ID10, a nonsense mutation in nitroreductase-1 transcripts could enhance passive resistance whereas increased transcription of nitroreductase-2, and a MATE transmembrane pump system, suggest active drug detoxification and efflux, respectively. By contrast, transcriptional changes in 713-M3 and WB-M3 indicated a higher oxidative stress load, attributed to Mtz- and oxygen-derived radicals, respectively. Quantitative comparisons of orthologous gene transcription between Mtz-resistant G. duodenalis and Trichomonas vaginalis, a closely related parasite, revealed changes in transcripts encoding peroxidases, heat shock proteins, and FMN-binding oxidoreductases, as prominent correlates of resistance. This work provides deep insight into Mtz-resistant G. duodenalis, and illuminates resistance-associated features across parasitic species.
Collapse
Affiliation(s)
- Brendan R. E. Ansell
- Faculty of Veterinary and Agricultural Sciences, The University of MelbourneMelbourne, VIC, Australia
| | - Louise Baker
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
| | - Samantha J. Emery
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
| | - Malcolm J. McConville
- Bio21 Molecular Science and Biotechnology Institute, The University of MelbourneMelbourne, VIC, Australia
| | - Staffan G. Svärd
- Department of Cell and Molecular Biology, Uppsala UniversityUppsala, Sweden
| | - Robin B. Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of MelbourneMelbourne, VIC, Australia
| | - Aaron R. Jex
- Faculty of Veterinary and Agricultural Sciences, The University of MelbourneMelbourne, VIC, Australia
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
| |
Collapse
|
40
|
Cavoue T, Bounou Abassi H, Vayssade M, Nguyen Van Nhien A, Kang IK, Kwon GW, Pourceau G, Dubot P, Abbad Andaloussi S, Versace DL. Imidazolium-based titanium substrates against bacterial colonization. Biomater Sci 2017; 5:561-569. [DOI: 10.1039/c6bm00715e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Photografting of a silane-derived imidazole compound on titanium substrates against bacterial colonization.
Collapse
|
41
|
Chou WK, Brynildsen MP. A biochemical engineering view of the quest for immune-potentiating anti-infectives. Curr Opin Chem Eng 2016. [DOI: 10.1016/j.coche.2016.08.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
42
|
Nitric oxide signaling in yeast. Appl Microbiol Biotechnol 2016; 100:9483-9497. [DOI: 10.1007/s00253-016-7827-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/17/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022]
|
43
|
Endogenous nitric oxide accumulation is involved in the antifungal activity of Shikonin against Candida albicans. Emerg Microbes Infect 2016; 5:e88. [PMID: 27530748 PMCID: PMC5034102 DOI: 10.1038/emi.2016.87] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 12/28/2022]
Abstract
The aim of the present study was to investigate the role of nitric oxide (NO) in the antifungal activity of Shikonin (SK) against Candida albicans (C. albicans) and to clarify the underlying mechanism. The results showed that the NO donors S-nitrosoglutathione (GSNO) and L-arginine could enhance the antifungal activity of SK, whereas the NO production inhibitor Nω-nitro-L-arginine methyl ester (L-NAME) attenuated antifungal action. Using the fluorescent dye 3-amino,4-aminomethyl-2′, 7-difluorescein, diacetate (DAF-FM DA), we found that the accumulation of NO in C. albicans was increased markedly by SK in a time- and dose-dependent manner. In addition, the results of real-time reverse transcription-PCR (RT-PCR) demonstrated that the transcription level of YHB1 in C. albicans was greatly increased upon incubation of SK. Consistently, the YHB1-null mutant (yhb1Δ/Δ) exhibited a higher susceptibility to SK than wild-type cells. In addition, although the transcription level of CTA4 in C. albicans was not significantly changed when exposed to SK, the CTA4-null mutant (cta4Δ/Δ) was more susceptible to SK. Collectively, SK is the agent found to execute its antifungal activity directly via endogenous NO accumulation, and NO-mediated damage is related to the suppression of YHB1 and the function of CTA4.
Collapse
|
44
|
Devarajan N, Suresh P. Framework-Copper-Catalyzed C−N Cross-Coupling of Arylboronic Acids with Imidazole: Convenient and Ligand-Free Synthesis of N-Arylimidazoles. ChemCatChem 2016. [DOI: 10.1002/cctc.201600480] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Nainamalai Devarajan
- Department of Natural Products Chemistry; School of Chemistry; Madurai Kamaraj University; Madurai- 625021 India
| | - Palaniswamy Suresh
- Department of Natural Products Chemistry; School of Chemistry; Madurai Kamaraj University; Madurai- 625021 India
| |
Collapse
|
45
|
Abstract
If life without heme-Fe were at all possible, it would definitely be different. Indeed this complex and versatile iron-porphyrin macrocycle upon binding to different “globins” yields hemeproteins crucial to sustain a variety of vital functions, generally classified, for convenience, in a limited number of functional families. Over-and-above the array of functions briefly outlined below, the spectacular progress in molecular genetics seen over the last 30 years led to the discovery of many hitherto unknown novel hemeproteins in prokaryotes and eukaryotes. Here, we highlight a few basic aspects of the chemistry of the hemeprotein universe, in particular those that are relevant to the control of heme-Fe reactivity and specialization, as sculpted by a variety of interactions with the protein moiety.
Collapse
Affiliation(s)
- Paolo Ascenzi
- Dipartimento di Scienze, Università Roma Tre, Viale Marconi 446, I-00146 Roma, Italy
| | - Maurizio Brunori
- Dipartimento di Scienze Biochimiche “Alessandro Rossi Fanelli” and Istituto Pasteur — Fondazione Cenci, Bolognetti, Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185 Roma, Italy
| |
Collapse
|
46
|
Gardner PR, Gardner DP, Gardner AP. Globins Scavenge Sulfur Trioxide Anion Radical. J Biol Chem 2015; 290:27204-27214. [PMID: 26381408 DOI: 10.1074/jbc.m115.679621] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Indexed: 01/16/2023] Open
Abstract
Ferrous myoglobin was oxidized by sulfur trioxide anion radical (STAR) during the free radical chain oxidation of sulfite. Oxidation was inhibited by the STAR scavenger GSH and by the heme ligand CO. Bimolecular rate constants for the reaction of STAR with several ferrous globins and biomolecules were determined by kinetic competition. Reaction rate constants for myoglobin, hemoglobin, neuroglobin, and flavohemoglobin are large at 38, 120, 2,600, and ≥ 7,500 × 10(6) m(-1) s(-1), respectively, and correlate with redox potentials. Measured rate constants for O2, GSH, ascorbate, and NAD(P)H are also large at ∼100, 10, 130, and 30 × 10(6) m(-1) s(-1), respectively, but nevertheless allow for favorable competition by globins and a capacity for STAR scavenging in vivo. Saccharomyces cerevisiae lacking sulfite oxidase and deleted of flavohemoglobin showed an O2-dependent growth impairment with nonfermentable substrates that was exacerbated by sulfide, a precursor to mitochondrial sulfite formation. Higher O2 exposures inactivated the superoxide-sensitive mitochondrial aconitase in cells, and hypoxia elicited both aconitase and NADP(+)-isocitrate dehydrogenase activity losses. Roles for STAR-derived peroxysulfate radical, superoxide radical, and sulfo-NAD(P) in the mechanism of STAR toxicity and flavohemoglobin protection in yeast are suggested.
Collapse
|
47
|
An ensemble-guided approach identifies ClpP as a major regulator of transcript levels in nitric oxide-stressed Escherichia coli. Metab Eng 2015; 31:22-34. [DOI: 10.1016/j.ymben.2015.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/21/2015] [Accepted: 06/15/2015] [Indexed: 11/23/2022]
|
48
|
Robinson JL, Adolfsen KJ, Brynildsen MP. Deciphering nitric oxide stress in bacteria with quantitative modeling. Curr Opin Microbiol 2014; 19:16-24. [PMID: 24983704 DOI: 10.1016/j.mib.2014.05.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 05/02/2014] [Accepted: 05/26/2014] [Indexed: 12/22/2022]
Abstract
Many pathogens depend on nitric oxide (NO•) detoxification and repair to establish an infection, and inhibitors of these systems are under investigation as next-generation antibiotics. Because of the broad reactivity of NO• and its derivatives with biomolecules, a deep understanding of how pathogens sense and respond to NO•, as an integrated system, has been elusive. Quantitative kinetic modeling has been proposed as a method to enhance analysis and understanding of NO• stress at the systems-level. Here we review the motivation for, current state of, and future prospects of quantitative modeling of NO• stress in bacteria, and suggest that such mathematical approaches would prove equally useful in the study of other broadly reactive antimicrobials, such as hydrogen peroxide (H2O2).
Collapse
Affiliation(s)
- Jonathan L Robinson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Kristin J Adolfsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
49
|
Antimicrobial Agents Act Differently on Staphyloccocus aureus and Ralstonia eutropha Flavohemoglobins. Appl Biochem Biotechnol 2014; 173:1023-37. [DOI: 10.1007/s12010-014-0938-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/21/2014] [Indexed: 11/25/2022]
|
50
|
Bang CS, Kinnunen A, Karlsson M, Önnberg A, Söderquist B, Persson K. The antibacterial effect of nitric oxide against ESBL-producing uropathogenic E. coli is improved by combination with miconazole and polymyxin B nonapeptide. BMC Microbiol 2014; 14:65. [PMID: 24629000 PMCID: PMC3984681 DOI: 10.1186/1471-2180-14-65] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/07/2014] [Indexed: 12/29/2022] Open
Abstract
Background Nitric oxide (NO) is produced as part of the host immune response to bacterial infections, including urinary tract infections. The enzyme flavohemoglobin, coded by the hmp gene, is involved in protecting bacterial cells from the toxic effects of NO and represents a potentially interesting target for development of novel treatment concepts against resistant uropathogenic bacteria. The aim of the present study was to investigate if the in vitro antibacterial effects of NO can be enhanced by pharmacological modulation of the enzyme flavohemoglobin. Results Four clinical isolates of multidrug-resistant extended-spectrum β-lactamase (ESBL)-producing uropathogenic E. coli were included in the study. It was shown that the NO-donor substance DETA/NO, but not inactivated DETA/NO, caused an initial growth inhibition with regrowth noted after 8 h of exposure. An hmp-deficient strain showed a prolonged growth inhibition in response to DETA/NO compared to the wild type. The imidazole antibiotic miconazole, that has been shown to inhibit bacterial flavohemoglobin activity, prolonged the DETA/NO-evoked growth inhibition. When miconazole was combined with polymyxin B nonapeptide (PMBN), in order to increase the bacterial wall permeability, DETA/NO caused a prolonged bacteriostatic response that lasted for up to 24 h. Conclusion An NO-donor in combination with miconazole and PMBN showed enhanced antimicrobial effects and proved effective against multidrug-resistant ESBL-producing uropathogenic E. coli.
Collapse
Affiliation(s)
| | | | | | | | | | - Katarina Persson
- Faculty of Medicine and Health, iRiSC - Inflammatory Response and Infection Susceptibility Centre, Örebro University, SE- 701 82 Örebro, Sweden.
| |
Collapse
|