Rapid Fluorescent-Based Detection of New Delhi ... - ACS Publications

Jun 21, 2019 - ABSTRACT: A method for rapid detection of metallo-β-lactamases NDM-5 and NDM-7 using conjugates of ... This is an open access article ...
0 downloads 0 Views 4MB Size
This is an open access article published under an ACS AuthorChoice License, which permits copying and redistribution of the article or any adaptations for non-commercial purposes.

Article Cite This: ACS Omega 2019, 4, 10891−10898

http://pubs.acs.org/journal/acsodf

Rapid Fluorescent-Based Detection of New Delhi Metallo-βLactamases by Photo-Cross-Linking Using Conjugates of Azidonaphthalimide and Zinc(II)-Chelating Motifs Monisha Singha,*,† Gaurav Kumar,*,‡ Diamond Jain,‡ Ganesh Kumar N,‡ Debashis Ray,† Anindya S. Ghosh,*,‡ and Amit Basak*,†,§ Department of Chemistry, ‡Department of Biotechnology, and §School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302 India

Downloaded via 79.110.31.70 on August 27, 2019 at 15:14:13 (UTC). See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.



S Supporting Information *

ABSTRACT: A method for rapid detection of metallo-β-lactamases NDM-5 and NDM-7 using conjugates of azidonaphthalimide and Zn(II) chelating motifs (like sulfonamides, hydroxamate, and terpyridine) is described. Incubation and irradiation, followed by gel electrophoresis, clearly show the presence of NDMs. The o-sulfonamide-based probe has the highest efficiency of detection for both the NDMs. The proteins are detectable at nM concentrations, and the method is also selective, works both in vitro and in vivo, as revealed by cellular imaging and also with clinical isolates.



positions 88 (Val → Leu) and 154 (Met → Leu),7 while NDM-7 differs at positions 130 (Asp → Asn) and 154 (Met → Leu).8 Although the crystal structure of NDM-1 (Figure 1) is known and not of NDM-5, it has been reported that the mutation at position 154 is proposed for inducing higher carbapenemase activity7,8 and/or stabilization,6d,9 which in

INTRODUCTION Antibiotic resistance has become a problem of immense concern in today’s world because of the appearance of so called “super bugs”.1 These super bacteria harboring more than two antibiotic-resistant genes have become multidrug resistant, and infections caused by them are difficult to treat.2 A major mechanism by which bacteria have acquired resistance is through production of metallo-β-lactamases (MBL)3 which uses an active site zinc to hydrolyze the β-lactam rings of widely used penicillins and cephalosporins thus making them inactive.4 As a result, carbapenems became one of the last lines of defense for treatment of such drug resistant infections. However, the emergence of new MBL-producing strains such as those producing New Delhi MBL (NDM BL) in recent years is conferring resistance5 to almost all β-lactam antibiotics including carbapenems. NDM-1 was first identified in 2008 in a Klebsiella pneumoniae isolate and has become the focus of worldwide attention because of the rapid dissemination of the corresponding blaNDM-type gene mainly amongst the Enterobacteriaceae and Acinetobacter spp. Because of the first description of NDM-1, multiple (>15) NDM variants have been reported, mostly arising from mutations causing substitutions of one or two residues at different locations of NDM-1.6 NDM-5 differs from NDM-1 by two substitutions at © 2019 American Chemical Society

Figure 1. Crystal structure of NDM-1. (A)NDM-1 monomer with highlighted motifs involved in protein dimerization and (B) proposed dimer form of NDM-1 with loop-10, loop-8, and alpha-3 residues involved in the interaction. Received: April 20, 2019 Accepted: June 6, 2019 Published: June 21, 2019 10891

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega

Article

Figure 2. Target photoaffinity probes 1−4.

Figure 3. (a) Reported24 NDM-1 inhibitor; docking poses of probe 2 (b) with NDM-1 and (c) with HCA II.

turn affects the discovery of new antibiotics.10 The mechanism of β-lactam resistance can be complex, involving, expression of β-lactamases and efflux pumps, porin loss, and alterations in penicillin-binding proteins (PBP). All these are associated with carbapenem resistance in gram negative bacteria.11 One of the reasons12,15,16 for NDM-harboring strains becoming a worldwide threat is certainly the lack of available antibiotics.13,14 However, the problem has become more complicated because of the absence of a standardized phenotypic test for MBL detection. Rapid and early detection of MBL strains including NDM producers is crucial not only for the right treatment but also to prevent their dissemination and associated nosocomial infections.



Recently, we have reported21 an efficient detection protocol for HCA II and PBP-5 by a simple gel electrophoresis technique using an azidonaphthalimide template with a built-in fluorophore and a linker. This has simplified the previously reported template-based strategies.22 Using a similar approach,21 molecules 1−4 having Zn(II)-chelating motifs attached to the template were designed (Figure 2) and synthesized in order to detect the presence of NDM-5 and NDM-7, in vitro and at a cellular level. Amongst all the probes, compound 2 with an o-sulphonamido moiety could detect the monomeric protein at ∼26 kDa with the most prominent visualizable fluorescent band under UV after photo-crosslinking followed by gel electrophoresis. The detection was also selective as demonstrated by preferential cross-linking of NDMs in the presence of other zinc enzymes like HCA II, carboxypeptidase, calf intestinal alkaline phosphatase, as well as nonmetallo enzymes like ovalbumin, class A serine-β-lactamase CTX-M-1523 and also in cell lysate (both overexpressed and clinical isolates). Herein, we describe the synthesis of the probes and the results of subsequent SDS-polyacrylamide gel electrophoresis and microscopic studies in detail. It may be pointed out that the design of the probes was inspired by a recent report by Cheng et al.24 on the screening of sulfonamides as NDM-1 inhibitors. The strongest inhibitor has been shown to be a sulfonamide with ortho-ligating imino nitrogen. Moreover, molecular docking studies using the pdb structure of NDM-1 indicated the possibility of cation πinteraction as well as agostic interaction between the zinc ion and the aryl ring bearing the o-sulfonamide moiety of probe 2. For the p-sulfonamide, being much further away from the active site, the binding interactions are less. Interestingly with HCA II, which is a Zn(II)-containing enzyme, the probe 2 fails

RESULTS AND DISCUSSION

We became interested in developing an early detection method for NDM-producing micro-organisms. At present, laboratory detection of MBL (including NDM) producers employs different techniques, all of which depend upon assaying the carbapenemase activity,13 like for example the modified Hodge test (MHT), UV spectrophotometry, MALDI TOF analysis, and Carba NP test. Amongst these, MHT is the only CLSIrecommended carbapenemase-screening method which, however, has a low sensitivity of ∼11% because of suppression of enzyme releases due to anchoring in the inner membrane.17 The sensitivity of MHT can be improved somewhat by the addition of a nonionic surfactant (Triton X-100).18 Other methods include PCR19 and loop-mediated isothermal amplification20 which, although more sensitive, put additional constrains like prior sequencing of the targeted gene and primer designing. 10892

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega

Article

Figure 4. Synthesis of target compounds.

Figure 5. SDS-PAGE experiment31 results show the binding efficiency of the synthesized probes with dimer and monomer forms of purified NDM5. NDM-5 (final concentration 10 μM) was used for cross-linking with compound concentration kept at 20 μM. The gel was run under nonreduced conditions. Gel image captured under ultraviolet light has been denoted as UV and the gel image after Coomassie blue staining has been denoted as CBB. M denotes protein molecular weight marker.

forms of the protein where the Zn ion acts as a bridge under low availability of Zn ions during protein folding. The cross-linking was initially studied with purified NDM530 under nonreducing conditions in which the protein exists both as monomeric and dimeric forms28,29 in solution as revealed by the appearance of two distinct bands under CBB at ∼26 and ∼50 kDa corresponding to the mono and dimeric forms, respectively. All the probes were incubated with both NDM-5 and NDM-7 under irradiation with UV. The appearance of fluorescent bands visible under UV demonstrated successful cross-linking, the cross-linking being stronger (thus more selective) with the monomeric form as revealed by the relative fluorescent intensities of the bands vis-à-vis Coomassie blue stained-bands (Figure 5). This observation is an indirect support to the hypothesis28 that the Zn ions are shared between two monomeric proteins in the dimer thus making the metal ion less exposed. Gratifyingly, the crosslinking also works in the case of irradiation with visible light, (the gel picture which is included in the Supporting Information). The same experiment was then repeated with a fixed concentration of protein under reducing conditions to evaluate the relative efficiency of cross-linking of various probes with the monomeric form of NDM. In this case also, all the probe molecules showed cross-linking as observed by the appearance of fluorescent bands when visualized under UV after gel electrophoresis. ImageJ software analysis32 revealed that amongst all the probes, compound 2 with the o-sulfonamido group showed the highest efficiency for visualization of NDM-

to show any binding, indicating possible selectivity of the probe (Figure 3). Backed up by the initial docking results, the synthesis started with the previously prepared GABA-derived azidonaphthalimide carboxylic acid 5. Like compound 1 whose synthesis has been reported earlier, the 2-sulphonamide25 based compound 2 was similarly prepared by treating 5 with bromoacetyl sulfanilamide 6 in dimethylformamide (DMF) in the presence of K2CO3. The synthesis of compound 3 was accomplished from the sodium salt of 5 by reacting with bromomethyl terpyridine 726 in dry DMF at room temperature for 10 h. Working up of the reaction mixture with brine followed by drying over Na2SO4 and evaporation afforded a yellow solid from which the product was purified by repeated precipitation from acetone−hexane (for 1 and 2) and ethyl acetate-hexane (for 3). The fluorescent template 5 was converted to the corresponding hydroxamic acid 4 (a well-known bidentate Zn(II)−chelator for HDAC27) by sequential reaction, first with ethyl chloroformate and N-methyl morpholine in dry DCM followed by reaction with hydroxylamine hydrochloride in the presence of KOH (Figure 4). MBLs such as B1 subclass NDM and BcII are present in monomeric and, may be, dimeric forms in the solution. From the crystal structure (Figure 1) of NDM-1, King and Strynadka28 predicted that the variable loops around the active site lead to broader substrate specificity and hence higher resistance. It is also predicted that loop-10, loop-8, and alpha-3 residues of two monomers of NDM-1 proteins interact to form the dimer. Selevsek et al.29 have shown that such dimers exist due to sharing of the same metal ions between two monomeric 10893

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega

Article

Figure 6. (A) Screening of binding efficiency for synthesized fluorescent probes with purified NDM-5; M = protein mol wt marker; C = NDM-5 in 2% DMSO. The concentration of each compound was maintained at 10 μM. (B) ImageJ analysis of Figure 6A; the X-axis denotes the compound numbers. SDS-PAGE was run under reduced conditions.

Figure 7. Protein concentration study with compound 2. Lane 1: 25 μM NDM-5 and DMSO (2%); lane 2: 25 μM NDM-5; lane 3: 20 μM NDM5; lane 4: 15 μM NDM-5; lane 5: 10 μM NDM-5; lane 6: 5 μM NDM-5; lane 7: 2 μM NDM-5; lane 8: 1 μM NDM-5; lane 9: 0.5 μM NDM-5; lane 10: protein marker. Final concentration of compound 2 was kept 20 μM in each lane from lane 2−9. SDS-PAGE was run under reduced conditions.

Figure 8. Results of cross-linking with different proteins and cell lysates. For (A) OV = ovalbumin (14 μM); M = protein mol wt marker; ND = purified NDM-5 (10 μM). For (B) HA = purified HCA II; M = protein mol wt marker. For (C) EC = E. coli cell lysate harboring CTX-M-15 overexpressed protein; C = E. coli cell lysate harboring NDM-5 overexpressed protein with 2% DMSO; ECN = E. coli cell lysate harboring NDM-5 overexpressed protein; M = protein mol wt marker; B = only 2% DMSO; CP = purified carboxypeptidase A (10 μM); CIP = purified calf intestine phosphatase (10 μM); ND = purified NDM-5 (2 μM); ECC = E. coli cell lysate from clinical isolate harboring NDM-5.34

Figure 9. Microscopic analysis of E. coli cells having overexpressed SHV-14 and NDM-5 proteins bound to compound 2.

picture under UV vis-a-vis Coomassie stain failed to show any significant cross-linking with all these enzymes (Figure 8). These results demonstrated the in vitro selectivity of compound 2 towards NDM-5. The probe 2 can also detect NDM-5 in cell lysate where the cells have been overexpressed with the NDM-5 gene. The method is also applicable to detect the presence of NDM-5 even in clinical isolates. The results were also very similar in the case of NDM-7. For example, probe 2, which selectively binds to the NDM-7 monomer, has higher efficiency compared to other probes and the detection limit in this case was 625 nM using 80 μM of 2 (figure in Supporting Information).

5 (Figure 6) and hence subsequent experiments regarding detection limit and cell lysate were carried out with probe 2. For a protein concentration of 20 μM, the lowest concentration of compound 2 that can detect NDM-5 was 5 μM (figure in Supporting Information). With a compound 2 concentration of 20 μM, the protein (NDM-5) is detectable at as low as 500 nM concentration (Figure 7).33 For checking the selectivity of the probe molecule 2, incubation followed by UV irradiation was performed with different additional proteins that included other Zn-dependent enzymes like HCA II, carboxypeptidase A, and CI phosphatase. The cross-linking efficiency of probe 2 was also compared with ovalbumin and a serine β-lactamase CTX-M-15. The gel 10894

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega

Article

General Experimental Procedure. Preparation of Carboxylic Acid Derivatives of 4-Azido-1,8-naphthalimide (5). Procedure for preparation of the GABA−carboxylic acid derivative is the same as reported earlier (Cai, Y.; Zhan, J.; Shen, H.; Mao, D.; Ji, S.; Liu, R.; Yang, B.; Kong, D.; Wang, L.; Yang, Z. Anal Chem. 2016, 88, 740). Preparation of Bromoacetyl Derivatives of 2-Aminobenzene Sulfonamide (6). To a solution of sulfanilamide 8 (0.35 g, 2.03 mmol) in dry THF (10 mL), K2CO3 (0.561 g, 4.06 mmol) was added and stirred for 30 min. Keeping the temperature at 0 °C, bromoacetyl chloride (0.2 mL, 2.44 mmol) was added dropwise to the reaction mixture which was stirred for a further period of 30 min at 0 °C. Water was added and the mixture was extracted with EtOAc (50 mL × 2), washed with brine, dried over Na2SO4, and concentrated in vacuo to get the product as a white crystalline solid (76% yield). Spectral data: 1H NMR (600 MHz, acetone-d6): δ 10.05 (s, 1 H), 8.40 (d, J = 8.4 Hz, 1 H), 8.03 (d, J = 8.0 Hz, 1 H), 7.69 (t, J = 7.9 Hz, 1 H), 7.39 (t, J = 7.7 Hz, 1 H), 6.94 (s, 2 H), 4.26 (s, 2 H). 13C NMR: δ (150 MHz, acetone-d6) 165.4, 135.7, 133.9, 132.4, 128.8, 125.1, 123.4, 30.4. HRMS: calcd for C8H9N2O3SBrNa (M + Na), 314.9415; found, 314.9424. Synthesis of Final Compounds. Synthesis of 2-Oxo-2((2-sulfamoylphenyl)amino)ethyl 4-(6-Azido-1,3-dioxo-1Hbenzo[de]isoquinolin-2(3H)-yl)butanoate (2). To a solution of GABA−carboxylic acid 5 (0.15 mmol) in dry DMF (5 mL) under N2, anhydrous K2CO3 (0.18 mmol) was added and stirred for 30 min at room temperature. A solution of bromoacetyl derivatives of sulphonamide 6 (0.18 mmol) in dry DMF (2 mL) was added and stirring was continued for 10 h at room temperature. The reaction was quenched by adding water (30 mL) and the aqueous layer was extracted with EtOAc (30 mL x 2). The combined organic layers were washed with brine, aq. NaHCO3 and water, dried over anhydrous Na2SO4, and concentrated in vacuo. The yellowishbrown gummy product was first precipitated from the acetone−hexane mixture and the precipitate was washed with hexane 2−3 times to furnish the target material as a yellow solid (70% yield). Spectral data: 1H NMR (400 MHz, DMSOd6): δ 12.14 (s, 1H), 8.49 (d, J = 7.2 Hz, 1H), 8.42 (d, J = 8.0 Hz, 1H), 8.38 (d, J = 8.4 Hz, 1H), 7.82 (m, 2H), 7.68 (t, J = 7.5 Hz, 2H), 7.47 (t, J = 7.6 Hz, 1H), 7.37 (d, J = 8.2 Hz, 1H), 4.89 (s, 2H), 4.10 (t, J = 6.5 Hz, 2H), 2.58 (t, J = 7.2 Hz, 2H), 2.03−1.91 (m, 2H). 13C NMR (150 MHz, DMSO-d6): δ 172.1, 163.5, 163.1, 155.1, 142.9, 134.7, 133.3, 131.6, 131.6, 128.5, 128.4, 127.4, 126.7, 123.6, 122.3, 121.6, 118.3, 117.7, 116.0, 62.2, 30.9, 22.8. IR (cm−1): 2942, 2122, 1745, 1695, 1652, 1612, 1586, 1542, 1478, 1439, 1391, 1353, 1276, 1158, 1130, 996, 765, 588, 502. HRMS: calcd for C24H20N6O7SNa (M + Na), 559.1012; found, 559.1011. Synthesis of 4-([2,2′:6′,2″-Terpyridin]-4′-yl)benzyl 4-(6Azido-1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)butanoate (3). GABA−carboxylic acid 5 (0.11 g, 0.35 mmol) was dissolved in methanol and NaHCO3 (29 mg, 0.35 mmol) dissolved in a minimum amount of water was added. The mixture was stirred for 2 h at room temperature and then lyophilized to obtain the sodium salt of 5 as a yellow solid. The solid (0.14 g, 0.35 mmol) was dissolved in dry DMF (20 mL) and compound 7 (0.12 g, 0.35 mmol) was added. The reaction mixture was stirred for 12 h under a N2 atmosphere at room temperature. The reaction mixture was extracted with ethyl acetate (2 × 50 mL) and the combined organic layers were

Encouraged by the in vitro results, we then carried out in vivo studies using fluorescence cell imaging to detect NDM-5 and NDM-7. Upon expression of NDM-5 and NDM-7 in the Escherichia coli host (CS109) from an expressible plasmid (pBAD18-cam) under the control of an arabinose promoter and subsequent labeling of the cells with compound 2 revealed that only the CS109 cells expressing NDM-5 or NDM-7 were detectable under fluorescent microscopy (Figure 8); no fluorescence was observed in the control CS109. The CS109 cells expressing NDM-5 as well as NDM-7 when labeled with compound 2 have clearly shown the localization of the MBL at the cell periphery. This is most likely at the periplasmic space of the host because the expressed NDM-5 or NDM-7 gene has the constitutive signal peptide for transporting across the cytoplasmic membrane (see Figure 9, column 2 and 3 fluorescent images). The observation further expands the cell permeability and precision of compound 2 in the detection of NDM-5 and NDM-7 at the cellular level.



CONCLUSIONS In summary, the overall work provides an in-depth study on the utility of various protein binding fluorescent compounds in detection and localization of MBLs such as NDM-5 and NDM7 both at the cellular level and in vitro. Moreover, the compound 2 showed selective binding towards both the NDMs over other proteins used in the study as revealed by the combination of in vitro binding studies with cell lysates and whole cell microscopy images. The method has been validated for use against clinical isolates. Future studies will be aimed towards other carbapenemase resistant MBLs like VIM and IMP and in vitro screening of possible inhibitors triggered by visible light.



EXPERIMENTAL SECTION

General Experimental Details. All reactions were performed under a nitrogen atmosphere unless otherwise stated. Glassware used in reactions was thoroughly oven-dried. All commercial grade reagents were used without further purification, and solvents were dried prior to use following the standard protocol. Thin-layer chromatography was carried out on precoated plates (silica gel 60 F254), and the spots were visualized by exposure to UV light and/or by staining with iodine. All crude products were purified by silica gel flash column chromatography (230−400 mesh) with petroleum ether/ethyl acetate as the eluent and characterized by NMR and mass spectrometry unless otherwise mentioned. Melting points were determined in open capillary tubes and are reported as uncorrected. 1H and 13C NMR spectra for all the compounds were recorded at 400/500/600 and 100/125/150 MHz (Bruker UltrashieldTM 400, AscendTM 500, AscendTM 600), respectively. The spectra were recorded in deuterochloroform (CDCl3) and deuterated dimethyl sulfoxide (DMSO-d6) as solvents at room temperature. Chemical shifts are reported in ppm from tetramethylsilane with the solvent resonance as rgw internal standard (CDCl3: δH = 7.26, δC = 77.16 ppm). Data for 1H NMR are reported as follows: chemical shift (δ ppm), multiplicity (s = singlet, br s = broad singlet, d = doublet, t = triplet, q = quartet, m = multiplet, dd = doublet of doublet), coupling constant (Hz), and integration. Data for 13C NMR are reported as the chemical shift. HRMS spectra using ESI were recorded on an ESI−FTMS mass spectrometer. 10895

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega

Article

37 °C. The protein expression was analyzed via 15% SDSPAGE after sonication. Cell lysates used at 49 μL of 0.9 OD600nm, compound used at 20 μM concentration. Overexpression of β-lactamase CTX-M-15 was studied in E. coli BL21 (DE3) under the control of a T7 promoter system. E. coli cell culture harboring recombinant CTX-M-15 was induced with 0.2 mM of IPTG at an OD600nm−0.6 and allowed to grow at 37 °C till OD600nm reached ∼1.0. The cells were then harvested and resuspended in 50 mM HEPES buffer and lysed by sonication. Prepared lysates were analyzed via 15% SDS-PAGE for expression. Cell lysates were used at a concentration of ∼1 μg/μL with compound used at 20 μM concentration. Similarly, clinical isolate-harboring NDM-5 was grown under antibiotic stress (Ampicillin 200 μg/mL) till OD600nm reached ∼1.0. The cells were harvested, resuspended in 50 mM HEPES buffer, and lysed by sonication. Reaction conditions were kept similar to previous experiments with a final cell lysate protein concentration of ∼1.0 μg/μL. Protein Purification and Compound Binding Study. The NDM-5, NDM-7, and CTX-M-15 proteins were overexpressed in the pET28a(+) vector under 0.5 mM IPTG induction, in BL21 (DE3) cells, at 20 °C overnight with constant shaking. The cells were sonicated and the cell lysate, thus obtained after removal of the cell debris, was loaded in the AKTA prime column (HisTrapTM HP, GE Healthcare, Uppsala, Sweden). The protein was washed initially with a low concentration of imidazole (25 mM) and subsequently eluted with 500 mM imidazole. Furthermore, excess imidazole was removed via dialysis and the concentration of purified protein was checked via the Bradford method. The SDS-PAGE analysis was done to assess the quality of purified proteins. The purity level of proteins after dialysis was more than 95%. Afterwards, the protein samples were used for further in vitro analysis. Preferential Binding of the Compound to the NDM Monomer form than the Dimer Form. To know the preferential binding of fluorescent compounds to NDM monomer and dimer forms, 15% SDS-PAGE was done under nonreducing conditions. The NDM protein was used at 10 μM, the compound was used at 20 μM. The cross-linking was done under UV light for 30 min on ice, followed by heat denaturation (at 95 °C for 5 min), and 15% SDS-PAGE analysis. Microscopic Analysis of Compound 2 Binding to Overexpressed NDM-5 and SHV-14 in E. coli Cells. Furthermore, to know the localization of the compound in E. coli cells harboring overexpressed NDM-5, NDM-7, and SHV-14 proteins, the bacterial cells were allowed to bind compound 2 (20 μM) for 15 min postinduction period of 16 h at 37 °C. Thereafter, the cells were allowed to stay under UV light for 15 min for cross-linking of compound 2 with overexpressed NDM-5, NDM-7, and SHV-14 within the E. coli cells. This step was followed by microscopy to record the images under both phase contrast and fluorescence (using DAPI filter). The microscopy slides were coated with 0.1% (w/v) poly-L-lysine prior to mounting of overexpressed bacterial cells. The experiment was also repeated with control E. coli cells devoid of NDM-5, NDM-7, and SHV-14.

washed with brine to remove DMF. Removal of ethyl acetate furnished a yellow solid which was further reprecipitated with ethyl acetate and hexane (75% yield). Spectral data: 1H NMR (400 MHz, chloroform-d): δ 8.75−8.58 (m,7 H), 8.55 (d, J = 8.0 Hz, 1 H), 8.40 (d, J = 8.4 Hz, 1 H), 7.87 (d, J = 6.0 Hz, 4 H), 7.75−7.67 (m, 1 H), 7.48−7.41 (m, 3 H), 7.35 (t, J = 6.2 Hz, 2 H), 5.16 (s, 2 H), 4.26 (t, J = 7.0 Hz, 2 H), 2.54 (t, J = 7.4 Hz, 2 H), 2.15 (p, J = 7.1 Hz, 2 H). 13C NMR (125 MHz, chloroform-d): δ 172.9, 164.2, 163.7, 156.3, 156.1, 149.9, 149.3, 143.6, 138.4, 137.0, 132.4, 131.9, 129.3, 129.0, 128.8, 127.6, 127.0, 124.5, 124.0, 122.7, 121.5, 119.0, 114.8, 66.0, 39.7, 32.1, 23.6. IR (cm−1): 3404, 2931, 2123, 1708, 1352, 1272, 1111, 784, 726, 612. HRMS: calcd for C38H27N7O4H (M + H), 646.2203; found, 646.2201. Synthesis of 4-(6-Azido-1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)-N-hydroxybutanamide (4). To a solution of the compound 5 (0.1 g, 0.31 mmol) in dichloromethane (20 mL) at 0 °C, ethyl chloroformate (35 μL, 0.37 mmol) and N-methyl morpholine (41 mg, 0.40 mmol) were added and the mixture was stirred for 10 min. Hydroxylamine (prepared by stirring a methanolic solution of hydroxylamine hydrochloride (32 mg, 0.47 mmol) with KOH in methanol, followed by filtration to remove KCl) was added and the mixture was stirred for 15 min. The solvent was removed under reduced pressure to obtain yellow gummy oil. After washing with dichloromethane, the hydroxamic acid 4 was obtained as a yellow solid (58% yield). Spectral data: 1H NMR (400 MHz, methanol-d4): δ 8.58 (d, J = 7.3 Hz, 1 H), 8.55 (d, J = 8.0 Hz, 1 H), 8.49 (d, J = 8.5 Hz, 1 H), 7.80 (t, J = 7.8 Hz, 1 H), 7.64 (d, J = 8.0 Hz, 1 H), 4.18 (t, J = 7.0 Hz, 2 H), 2.20 (t, J = 7.6 Hz, 2 H), 2.02 (p, J = 7.7 Hz, 2 H). 13C (125 MHz, methanol-d4): δ 172.2, 165.5, 165.1, 145.3, 133.1, 133.1, 130.4, 130.0, 128.1, 125.6, 123.7, 119.8, 116.3, 40.7, 31.5, 25.4. IR (cm−1): 3309, 2946, 2123, 1730, 1272, 1119, 733, 624. HRMS: calcd for, C16H13N5O4Na (M + Na) 362.0865; found, 362.0868. Capture Experiment Protocol. The capture experiment was studied with overexpressed proteins at the cellular level and with purified proteins (in vitro) under different conditions of cross-linking under (UV light) and reducing and nonreducing conditions. In the absence of a reducing environment, the NDM protein exists as dimer and monomer forms in the solution. Moreover, under reducing conditions (i.e., in the presence of DTT or BME in the loading dye of SDS-PAGE), a single band of NDM protein was observed. However, under nonreducing condition, two distinct bands were observed (26 and 50 kDa approximately). In all the experiments purified proteins are used in the range 5−20 μM depending on the experimental requirement in 50 mM HEPES buffer (pH 7.2), each compound was used at 20 μM concentration (except in compound concentration study). The whole reaction was set in 50 μL reaction volume and crossing-linking was done under the UV light for 30 min. The image was recorded under Typhoon FLA7000, UV light and white light conditions to confirm the compound binding and protein stability. For more details about the buffer used and the capture experiment protocol see Basak et al. Chem. Commun., 2017, 53, 13015. SDS-PAGE Gel Experimental Details. Cellular Expression and Selective Binding of Compound to β-Lactamase Overexpressed Proteins in Whole Cell Lysates. Cellular expression of NDM-5 and NDM-7 was studied in E. coli CS109 cells under the control of an araC promoter. The cells were induced with 0.2% arabinose and incubated overnight at 10896

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega



Article

(8) Gottig, S.; Hamprecht, A. G.; Christ, S.; Kempf, V. A. J.; Wichelhaus, T. A. Detection of NDM-7 in Germany, a new variant of the New Delhi metallo-β-lactamase with increased carbapenemase activity. J. Antimicrob. Chemother. 2013, 68, 1737−1740. (9) Bahr, G.; Vitor-Horen, L.; Bethel, C. R.; Bonomo, R. A.; González, L. J.; Vila, A. J. Clinical Evolution of New Delhi Metallo-βLactamase (NDM) Optimizes Resistance under Zn(II) Deprivation. Antimicrob. Agents Chemother. 2017, 62, No. e01849. (10) Gould, I. M.; Bal, A. M. New antibiotic agents in the pipeline and how they can help overcome microbial resistance. Virulence 2013, 4, 185−191. (11) Karthikeyan, K.; Thirunarayan, M. A.; Krishnan, P. Coexistence of blaOXA-23 with blaNDM-1 and armA in clinical isolates of Acinetobacter baumannii from India. J. Antimicrob. Chemother. 2010, 65, 2253−2254. (12) Gilbert, P.; McBain, A. J. Potential impact of increased use of biocides in consumer products on prevalence of antibiotic resistance. Clin. Microbiol. Rev. 2003, 16, 189−208. (13) Kumarasamy, K. K.; Toleman, M. A.; Walsh, T. R.; Bagaria, J.; Butt, F.; Balakrishnan, R.; Chaudhary, U.; Doumith, M.; Giske, C. G.; Irfan, S.; Krishnan, P.; Kumar, A. V.; Maharjan, S.; Mushtaq, S.; Noorie, T.; Paterson, D. L.; Pearson, A.; Perry, C.; Pike, R.; Rao, B.; Ray, U.; Sarma, J. B.; Sharma, M.; Sheridan, E.; Thirunarayan, M. A.; Turton, J.; Upadhyay, S.; Warner, M.; Welfare, W.; Livermore, D. M.; Woodford, N. Emergence of a new antibiotic resistance mechanism in India, Pakistan, and the UK: a molecular, biological, and epidemiological study. Lancet Infect. Dis. 2010, 10, 597−602. (14) Arpin, C.; Noury, P.; Boraud, D.; Coulange, L.; Manetti, A.; André, C.; M’Zali, F.; Quentin, C. NDM-1-Producing Klebsiella pneumoniae Resistant to Colistin in a French Community Patient without History of Foreign Travel. Antimicrob. Agents Chemother. 2012, 56, 3432−3434. (15) Walsh, T. R.; Weeks, J.; Livermore, D. M.; Toleman, M. A. Dissemination of NDM-1 positive bacteria in the New Delhi environment and its implications for human health: an environmental point prevalence study. Lancet Infect. Dis. 2011, 11, 355−362. (16) Wang, B.; Sun, D. Detection of NDM-1 carbapenemaseproducing Acinetobacter calcoaceticus and Acinetobacter junii in environmental samples from livestock farms. J. Antimicrob. Chemother. 2015, 70, 611−613. (17) (a) Birgy, A.; Bidet, P.; Genel, N.; Doit, C.; Decre, D.; Arlet, G.; Bingen, E. Phenotypic Screening of Carbapenemases and Associated β-Lactamases in Carbapenem-Resistant Enterobacteriaceae. J. Clin. Microbiol. 2012, 50, 1295−1302. (b) Tan, K. E.; Ellis, B. C.; Lee, R.; Stamper, P. D.; Zhang, S. X.; Carroll, K. C. Prospective Evaluation of a Matrix-Assisted Laser Desorption Ionization−Time of Flight Mass Spectrometry System in a Hospital Clinical Microbiology Laboratory for Identification of Bacteria and Yeasts: a Bench-byBench Study for Assessing the Impact on Time to Identification and Cost-Effectiveness. J. Clin. Microbiol. 2012, 50, 1419−1421. (18) Pasteran, F.; Gonzalez, L. J.; Albornoz, E.; Bahr, G.; Vila, A. J.; Corso, A. Triton Hodge Test: Improved Protocol for Modified Hodge Test for Enhanced Detection of NDM and Other Carbapenemase Producers. J. Clin. Microbiol. 2016, 54, 640−649. (19) Liu, W.; Zou, D.; Li, Y.; Wang, X.; He, X.; Wei, X.; Shao, C.; Li, X.; Shang, W.; Yu, K.; Liu, D.; Guo, J.; Yin, Z.; Yuan, J. Sensitive and rapid detection of the new Delhi metallo-β-lactamase gene by loopmediated isothermal amplification. J. Clin. Microbiol. 2012, 50, 1580− 1585. (20) Ota, Y.; Furuhashi, K.; Nanba, T.; Yamanaka, K.; Ishikawa, J.; Nagura, O.; Hamada, E.; Maekawa, M. A rapid and simple detection method for phenotypic antimicrobial resistance in Escherichia coli by loop-mediated isothermal amplification. J. Med. Microbiol. 2019, 68, 169−177. (21) Singha, M.; Roy, S.; Pandey, S. D.; Bag, S. S.; Bhattacharya, P.; Das, A.; Ghosh, S.; Ray, D.; Basak, A. Use of azidonaphthalimide carboxylic acids as fluorescent templates with a built-in photoreactive group and a flexible linker simplifies protein labeling studies:

ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsomega.9b01145. Other related gel experiments and NMR spectra of all new compounds (PDF)



AUTHOR INFORMATION

Corresponding Authors

*E-mail: *E-mail: *E-mail: *E-mail:

[email protected] (M.S.). [email protected] (G.K.). [email protected] (A.S.G.). [email protected] (A.M.).

ORCID

Debashis Ray: 0000-0002-4174-6445 Amit Basak: 0000-0003-4041-2846 Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS DST is acknowledged for an SERB grant (SB/S1/OC-94/ 2013) and for the JC Bose Fellowship to A.B. M.S. and G.K.N. are grateful to IIT Kharagpur, and G.K. acknowledges ICMR for providing fellowships.



REFERENCES

(1) Review on Antimicrobial resistance Final Report and Recommendations O’Neill J. May, 2016. (2) Khan, A. U.; Maryam, L.; Zarrilli, R. Structure, Genetics and Worldwide Spread of New Delhi Metallo-β-lactamase (NDM): a threat to public health. BMC Microbiol. 2017, 17, 101. (3) King, D. T.; Worrall, L. J.; Gruninger, R.; Strynadka, N. C. J. New Delhi Metallo-β-Lactamase: Structural Insights into β-Lactam Recognition and Inhibition. J. Am. Chem. Soc. 2012, 134, 11362− 11365. (4) Pitout, J. D. D.; Gregson, D. B.; Poirel, L.; McClure, J.-A.; Le, P.; Church, D. L. Detection of Pseudomonas aeruginosa Producing Metallo-β-Lactamases in a Large Centralized Laboratory. J. Clin. Microbiol. 2005, 43, 3129−3135. (5) Rasheed, J. K.; Kitchel, B.; Zhu, W.; Anderson, K. F.; Clark, N. C.; Ferraro, M. J.; Savard, P.; Humphries, R. M.; Kallen, A. J.; Limbago, B. M. New Delhi Metallo-β-Lactamase−producing Enterobacteriaceae, United States. Emerg. Infect. Dis. 2013, 19, 870−878. (6) (a) Kaase, M.; Nordmann, P.; Wichelhaus, T. A.; Gatermann, S. G.; Bonnin, R. A.; Poirel, L. J. NDM-2 carbapenemase in Acinetobacter baumannii from Egypt. J. Antimicrob. Chemother. 2011, 66, 1260−1262. (b) Khan, A. U.; Nordmann, P. Spread of carbapenemase NDM-1 producers: the situation in India and what may be proposed. Scand. J. Infect. Dis. 2012, 44, 531−535. (c) Tada, T.; Miyoshi-Akiyama, T.; Dahal, R. K.; Sah, M. K.; Ohara, H.; Kirikae, T.; Pokhrel, B. M. NDM-8 Metallo-β-Lactamase in a MultidrugResistant Escherichia coli Strain Isolated in Nepal. Antimicrob. Agents Chemother. 2013, 57, 2394−2396. (d) Makena, A.; Brem, J.; Pfeffer, I.; Geffen, R. E. J.; Wilkins, S. E.; Tarhonskaya, H.; Flashman, E.; Phee, L. M.; Wareham, D. W.; Schofield, C. J. Biochemical characterization of New Delhi metallo-β-lactamase variants reveals differences in protein stability. J. Antimicrob. Chemother. 2015, 70, 463−469. (e) Khan, A. U.; Parvez, S. Detection of bla(NDM-4) in Escherichia coli from hospital sewage. J. Med. Microbiol. 2014, 63, 1404−1406. (7) Hornsey, M.; Phee, L.; Wareham, D. W. A novel variant, NDM5, of the New Delhi metallo-β-lactamase in a multidrug-resistant Escherichia coli ST648 isolate recovered from a patient in the United Kingdom. Antimicrob. Agents Chemother. 2011, 55, 5952−5954. 10897

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898

ACS Omega

Article

(33) Use of higher concentration of probe (80 μM) improved the detection limit down to ∼150 nM. Attempts will be made to optimize the structural features of the probe to improve the detection limit further. (34) In Figure 8A, purified NDM-5 was without signal peptides and C-terminal anchoring amino acids, and the cell lysate from the clinical isolate (Figure 8C) has been used at 1μg/μl of ∼1.0 OD at 600 nm.

applications in selective tagging of HCAII and penicillin binding proteins. Chem. Commun. 2017, 53, 13015−13018. (22) (a) Hosoya, T.; Inoue, A.; Hiramatsu, T.; Aoyama, H.; Ikemoto, T.; Suzuki, M. Facile synthesis of diazido-functionalized biaryl compounds as radioisotope-free photoaffinity probes by Suzuki−Miyaura coupling. Bioorg. Med. Chem. 2009, 17, 2490. (b) Kiyonaka, S.; Kato, K.; Nishida, M.; Mio, K.; Numaga, T.; Sawaguchi, Y.; Yoshida, T.; Wakamori, M.; Mori, E.; Numata, T.; Ishii, M.; Takemoto, H.; Ojida, A.; Watanabe, K.; Uemura, A.; Kurose, H.; Morii, T.; Kobayashi, T.; Sato, Y.; Sato, C.; Hamachi, I.; Mori, Y. Selective and direct inhibition of TRPC3 channels underlies biological activities of a pyrazole compound. Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 5400. (c) Addy, P. S.; Saha, B.; Singh, N. D. P.; Das, A. K.; Bush, J. T.; Lejeune, C.; Schofield, C. J.; Basak, A. 1,3,5Trisubstituted benzenes as fluorescent photoaffinity probes for human carbonic anhydrase II capture. Chem. Commun. 2013, 49, 1930. (d) Lang, K.; Chin, J. W. Bioorthogonal Reactions for Labeling Proteins. ACS Chem. Biol. 2014, 9, 16. (e) Li, X.; Cao, J.-H.; Li, Y.; Rondard, P.; Zhang, Y.; Yi, P.; Liu, J.-F.; Nan, F.-J. Activity-Based Probe for Specific Photoaffinity Labeling γ-Aminobutyric Acid B (GABAB) Receptors on Living Cells: Design, Synthesis, and Biological Evaluation. J. Med. Chem. 2008, 51, 3057 and references therein . (f) Dülsner, E.; Lenz, T.; Jurinke, C. Bioorthogonal Reactions for Labeling Proteins. Nat. Methods 2008, 5, an10. (23) Ogbolu, D. O.; Terry Alli, O. A.; Webber, M. A.; Oluremi, A. S.; Oloyede, O. M. CTX-M-15 is Established in Most MultidrugResistant Uropathogenic Enterobacteriaceae and Pseudomonaceae from Hospitals in Nigeria. Eur J Microbiol Immunol. 2018, 8, 20−24. (24) Wang, X.; Lu, M.; Shi, Y.; Ou, Y.; Cheng, X. Discovery of novel new delhi metallo-β-lactamases-1 inhibitors by multistep virtual screening. PLoS One 2015, 10 (3), No. e0118290. (25) There are few literature reports on the use of sulphonamides as MBL inhibitors:. (a) Brem, J.; Rydjik, A.; Mcdonough, M.; Schofield, C. J.; Morrison, A.; Hewitt, J.; Pannifer, A.; Jones, P. Inhibitors of metallo-beta-lactamases. WO 2017093727 A1, 2017. (b) Eidam, O.; Romagnoli, C.; Caselli, E.; Babaoglu, K.; Pohlhaus, D. T.; Karpiak, J.; Bonnet, R.; Shoichet, B. K.; Prati, F. Design, Synthesis, Crystal Structures, and Antimicrobial Activity of Sulfonamide Boronic Acids as β-Lactamase Inhibitors. J. Med. Chem. 2010, 53, 7852−7863. (26) Mandal, A.; Maity, A.; Bag, S. S.; Bhattacharya, P.; Das, A. K.; Basak, A. Design and synthesis of dual probes for detection of metal ions by LALDI MS and fluorescence: application in Zn(II) imaging in cells. RSC Adv. 2017, 7, 7163−7169. (27) Bottomley, M. J.; Lo Surdo, P.; Di Giovine, P. D.; Cirillo, R.; Ferrigno, F.; Jones, P.; Neddermann, P.; De Francesco, R.; Steinkühler, C.; Gallinari, P.; Carfí, A. Structural and Functional Analysis of the Human HDAC4 Catalytic Domain Reveals a Regulatory Structural Zinc-binding Domain. J. Biol. Chem. 2008, 283, 26694−26704. (28) King, D.; Strynadka, N. Crystal structure of New Delhi metalloβ-lactamase reveals molecular basis for antibiotic resistance. Prot. Sci. 2011, 20, 1484−1491. (29) Selevsek, N.; Rival, S.; Tholey, A.; Heinzle, E.; Heinz, U.; Hemmingsen, L.; Adolph, W. H. Zinc Ion-induced Domain Organization in Metallo-β-lactamases A FLEXIBLE “ZINC ARM” For Rapid Metal Ion Transfer? J. Biol. Chem. 2009, 284, 16419− 16431. (30) Kumar, G.; Issa, B.; Kar, D.; Biswal, S.; Ghosh, A. S. E152A substitution drastically affects NDM-5 activity. FEMS Microbiology Letters 2017, 364, 1−6. (31) It may be mentioned that the electrophoresis were done under denatured conditions in the presence of SDS after incubating followed by photoirradiating the probes with protein under native conditions. The latter ensured attachment of the fluorophore probe to the protein even if the zinc is washed away during denaturation. (32) (a) Schneider, C. A.; Rasband, W. S.; Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis Nat. Methods 2012, 9, 671−675. (b) Collins, T. ImageJ for microscopy. Biotechniques 2007, 43, S25− S30. 10898

DOI: 10.1021/acsomega.9b01145 ACS Omega 2019, 4, 10891−10898