1
|
Wali AF, Talath S, Sridhar SB, Shareef J, Goud M, Rangraze IR, Alaani NN, Mohamed OI. A Comprehensive Review on Bioactive Molecules and Advanced Microorganism Management Technologies. Curr Issues Mol Biol 2024; 46:13223-13251. [PMID: 39590383 PMCID: PMC11592628 DOI: 10.3390/cimb46110789] [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: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The advent of new strains of resistant microbes and the concomitant growth in multidrug resistance have made antimicrobial resistance an urgent public health concern. New antimicrobials are desperately needed to boost the success rates of treating infectious diseases and save lives. There are many intriguing biomolecules with antibacterial action, which are mostly unexplored in microorganisms. This review article describes the importance of natural compounds against microorganisms using advanced techniques to protect individuals from diseases. We have conducted an extensive literature review using databases such as SCOPUS, SCI, PUBMED, ScienceDirect, and Medline to gather relevant information. Our review covers various microorganism sources for antimicrobials, antifungal drugs, micro-culturing techniques, and microbial-based microsystems' applications. Every kind of higher trophic life depends on microorganisms for sustenance. The unseen majority is essential to understanding how humans and other living forms can survive anthropogenic climate change. The article discusses antimicrobial substances and the latest techniques and strategies for developing effective treatments. Novel model systems and cutting-edge biomolecular and computational methodologies could help researchers enhance antimicrobial resistance by completely capitalizing on lead antimicrobials.
Collapse
Affiliation(s)
- Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sathvik B. Sridhar
- Department of Clinical Pharmacy and Pharmacology, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates; (S.B.S.); (J.S.)
| | - Javedh Shareef
- Department of Clinical Pharmacy and Pharmacology, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates; (S.B.S.); (J.S.)
| | - Manjunatha Goud
- Department of Biochemistry, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates;
| | - Imran Rashid Rangraze
- Department of Internal Medicine, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates;
| | - Nowar Nizar Alaani
- Department of General Education, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates; (N.N.A.); (O.I.M.)
| | - Omnia Ibrahim Mohamed
- Department of General Education, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates; (N.N.A.); (O.I.M.)
| |
Collapse
|
2
|
Optimization of the Hemolysis Assay for the Assessment of Cytotoxicity. Int J Mol Sci 2023; 24:ijms24032914. [PMID: 36769243 PMCID: PMC9917735 DOI: 10.3390/ijms24032914] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/09/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
In vitro determination of hemolytic properties is a common and important method for preliminary evaluation of cytotoxicity of chemicals, drugs, or any blood-contacting medical device or material. The method itself is relatively straightforward, however, protocols used in the literature vary substantially. This leads to significant difficulties both in interpreting and in comparing the obtained values. Here, we examine how the different variables used under different experimental setups may affect the outcome of this assay. We find that certain key parameters affect the hemolysis measurements in a critical manner. The hemolytic effect of compounds tested here varied up to fourfold depending on the species of the blood source. The use of different types of detergents used for generating positive control samples (i.e., 100% hemolysis) produced up to 2.7-fold differences in the calculated hemolysis ratios. Furthermore, we find an expected, but substantial, increase in the number of hemolyzed erythrocytes with increasing erythrocyte concentration and with prolonged incubation time, which in turn affects the calculated hemolysis ratios. Based on our findings we propose an optimized protocol in an attempt to standardize future hemolysis studies.
Collapse
|
3
|
Jarkhi A, Lee AHC, Sun Z, Hu M, Neelakantan P, Li X, Zhang C. Antimicrobial Effects of L-Chg10-Teixobactin against Enterococcus faecalis In Vitro. Microorganisms 2022; 10:microorganisms10061099. [PMID: 35744616 PMCID: PMC9228388 DOI: 10.3390/microorganisms10061099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/22/2022] [Accepted: 05/22/2022] [Indexed: 11/18/2022] Open
Abstract
Objective: Teixobactin and its analogues are a new class of antibiotics that have no detectable bacterial resistance. This study was designed to determine the antibacterial and antibiofilm activities of a novel teixobactin analogue, L-Chg10-teixobactin, against two strains of Enterococcus faecalis (E. faecalis). Materials and Methods: The efficacy of L-Chg10-teixobactin against two strains of E. faecalis (ATCC 29212 and 47077) was determined using Clinical and Laboratory Standards Institute methods. L-Chg10-teixobactin was prepared at a stock concentration of 1 mg/mL in 5% DMSO. The minimum inhibitory concentration (MIC) was calculated using a two-fold serial broth dilution method, utilizing a 96-well plate. The minimum bactericidal concentration (MBC) was determined by plating the bacteria onto agar to define the concentration that resulted in 99.9% of bacterial death. Ampicillin was used as the control. The effect of L-Chg10-teixobactin on the inhibition of ATCC 47077 strain biofilm formation was determined by measuring the minimum biofilm inhibitory concentration (MBIC) using the safranin assay, while the eradication of the preformed biofilm was determined by measuring the minimum biofilm eradication concentration (MBEC) using the XTT assay. For nonlinear data, the log dose–response curve was plotted to calculate the optimum concentration using Excel (version 16.51, Microsoft® excel. 2021, Microsoft Corporation, Reymond, WA, USA). The data are presented as mean ± standard deviation (SD). Results: The MIC and MBC values of L-Chg10-teixobactin against both strains of E. faecalis were 0.8 μg/mL. The MIC of ampicillin was 1.25 μg/mL for ATCC 29212 and ranged from 1.25 to 5 μg/mL for ATCC 47077. The MBC of ampicillin for ATCC 29212 and ATCC 47077 was 10 and 20 μg/mL, respectively. The MIC and MBC of ampicillin were much higher compared with those of L-Chg10-teixobactin. The MBEC80 of L-Chg10-teixobactin was 4.60 μg/mL for ATCC 47077, which was much lower than that of ampicillin (20 μg/mL). Conclusions:L-Chg10-teixobactin demonstrated potent antibacterial and antibiofilm effects against E. faecalis, suggesting its potential role an effective antibacterial and antibiofilm agent in endodontic treatment.
Collapse
|
4
|
Singh RB, Das S, Chodosh J, Sharma N, Zegans ME, Kowalski RP, Jhanji V. Paradox of complex diversity: Challenges in the diagnosis and management of bacterial keratitis. Prog Retin Eye Res 2021; 88:101028. [PMID: 34813978 DOI: 10.1016/j.preteyeres.2021.101028] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Bacterial keratitis continues to be one of the leading causes of corneal blindness in the developed as well as the developing world, despite swift progress since the dawn of the "anti-biotic era". Although, we are expeditiously developing our understanding about the different causative organisms and associated pathology leading to keratitis, extensive gaps in knowledge continue to dampen the efforts for early and accurate diagnosis, and management in these patients, resulting in poor clinical outcomes. The ability of the causative bacteria to subdue the therapeutic challenge stems from their large genome encoding complex regulatory networks, variety of unique virulence factors, and rapid secretion of tissue damaging proteases and toxins. In this review article, we have provided an overview of the established classical diagnostic techniques and therapeutics for keratitis caused by various bacteria. We have extensively reported our recent in-roads through novel tools for accurate diagnosis of mono- and poly-bacterial corneal infections. Furthermore, we outlined the recent progress by our group and others in understanding the sub-cellular genomic changes that lead to antibiotic resistance in these organisms. Finally, we discussed in detail, the novel therapies and drug delivery systems in development for the efficacious management of bacterial keratitis.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Leiden University Medical Center, 2333, ZA Leiden, the Netherlands
| | - Sujata Das
- Cornea and Anterior Segment Services, LV Prasad Eye Institute, Bhubaneshwar, India
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Namrata Sharma
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Michael E Zegans
- Department of Ophthalmology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Regis P Kowalski
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; The Charles T Campbell Ophthalmic Microbiology Laboratory, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Vishal Jhanji
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; The Charles T Campbell Ophthalmic Microbiology Laboratory, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Xu L, She P, Chen L, Li S, Zhou L, Hussain Z, Liu Y, Wu Y. Repurposing Candesartan Cilexetil as Antibacterial Agent for MRSA Infection. Front Microbiol 2021; 12:688772. [PMID: 34589063 PMCID: PMC8473943 DOI: 10.3389/fmicb.2021.688772] [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: 03/31/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is an important pathogen causing hospital-acquired infections. Methicillin-resistant S. aureus (MRSA), biofilms, and persisters are highly tolerant to traditional antibiotics and make it difficult to treat. Therefore, new antimicrobial agents are urgently needed to treat hard-to-eradicate diseases caused by this bacterium. In this study, candesartan cilexetil (CC), an angiotensin hypertension drug, had strong antimicrobial activity against S. aureus with minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) of 8-16 μg/ml and 16-32 μg/ml. CC exhibited limited cytotoxicity and low potential to induce drug resistance. In addition, it showed a synergistic antibacterial effect when combined with gentamicin and tobramycin. The effective concentrations to inhibit MRSA biofilm formation were 16-64 μg/ml, and intractable persisters were killed at 4-8 × MIC. Through the analysis of its mechanism of action, it was evident that the membrane permeability was disrupted as well as the cell structure was damaged. Furthermore, we demonstrated that CC had antibacterial effects in vivo in MRSA-infected murine skin abscess models. In conclusion, these results imply that CC might be a potential antibacterial agent for the treatment of S. aureus-associated infections.
Collapse
Affiliation(s)
- Lanlan Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shijia Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Linying Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zubair Hussain
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaqian Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Upert G, Luther A, Obrecht D, Ermert P. Emerging peptide antibiotics with therapeutic potential. MEDICINE IN DRUG DISCOVERY 2021; 9:100078. [PMID: 33398258 PMCID: PMC7773004 DOI: 10.1016/j.medidd.2020.100078] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/27/2020] [Indexed: 02/09/2023] Open
Abstract
This review covers some of the recent progress in the field of peptide antibiotics with a focus on compounds with novel or established mode of action and with demonstrated efficacy in animal infection models. Novel drug discovery approaches, linear and macrocyclic peptide antibiotics, lipopeptides like the polymyxins as well as peptides addressing targets located in the plasma membrane or in the outer membrane of bacterial cells are discussed.
Collapse
Key Words
- ADMET, absorption, distribution, metabolism and excretion – toxicity in pharmacokinetics
- AMP, antimicrobial peptide
- AMR, antimicrobial resistance
- ATCC, ATCC cell collection
- Antibiotic
- BAM, β-barrel assembly machinery
- CC50, cytotoxic concentration to kill 50% of cells
- CD, circular dichroism
- CFU, colony forming unit
- CLSI, clinical and laboratory standards institute
- CMS, colistin methane sulfonate
- DMPC, 1,2-dimyristoyl-sn-glycero-3-phosphocholine
- ESKAPE, acronym encompassing six bacterial pathogens (often carrying antibiotic resistance): Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumonia, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp
- FDA, U. S. Food and Drug Administration
- HABP, hospital acquired bacterial pneumonia
- HDP, host-defense peptide
- HEK293, human embryonic kidney 293 cells
- HK-2, human kidney 2 cells (proximal tubular cell line)
- HepG2, human hepatocellular carcinoma cell line
- Hpg, 4-hydroxy-phenyl glycine
- ITC, isothermal titration calorimetry
- KPC, Klebsiella pneumoniae metallo-β-lactamase C resistant
- LPS, lipopolysaccharide
- LptA, lipopolysaccharide transport protein A
- LptC, lipopolysaccharide transport protein C
- LptD, lipopolysaccharide transport protein D
- MDR, multidrug-resistant
- MH-I, Müller-Hinton broth I
- MH-II, Müller-Hinton broth II (cation adjusted)
- MIC, minimal inhibitory concentration
- MRSA, methicilline-resistant S. aureus
- MSSA, methicilline-sensitive S. aureus
- MoA, mechanism (mode) of action
- NDM-1, New Delhi metallo-β-lactamase resistant
- NOAEL, no adverse effect level
- ODL, odilorhabdin
- OMPTA (outer membrane targeting antibiotic)
- OMPTA, outer membrane targeting antibiotic
- Omp, outer membrane protein
- PBMC, peripheral mononuclear blood cell
- PBP, penicillin-binding protein
- PBS, phosphate-buffered saline
- PK, pharmacokinetics
- POPC, 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine
- POPG, 2-oleoyl-1-palmitoyl-sn-glycero-3-phospho-(1-glycerol)
- PrAMPs, polyproline antimicrobial peptides
- RBC, red blood cell
- SAR, structure-activity relationship
- SPR, surface plasmon resonance
- SPase I, signal peptidase I
- VABP, ventilator associated bacterial pneumonia
- VIM-1, beta-lactamase 2 (K. pneumoniae)
- VISA, vancomycin-intermediate S. aureus
- VRE, vancomycin-resistant enterococcus
- WHO, World Health Organization
- WT, wild type
- WTA, wall teichoic acid
- XDR, extremely drug-resistant
- antimicrobial peptide
- antimicrobial resistance
- bid, bis in die (two times a day)
- i.p., intraperitoneal
- i.v., intravenous
- lipopeptide
- mITT population, minimal intend-to-treat population
- peptide antibiotic
- s.c., subcutaneous
Collapse
Affiliation(s)
- Gregory Upert
- Polyphor Ltd, Hegenheimermattweg 125, 4123 Allschwil, Switzerland
| | - Anatol Luther
- Bachem AG, Hauptstrasse 114, 4416 Bubendorf, Switzerland
| | - Daniel Obrecht
- Polyphor Ltd, Hegenheimermattweg 125, 4123 Allschwil, Switzerland
| | - Philipp Ermert
- Polyphor Ltd, Hegenheimermattweg 125, 4123 Allschwil, Switzerland
| |
Collapse
|
7
|
Cele Z, Somboro AM, Amoako DG, Ndlandla LF, Balogun MO. Fluorinated Quaternary Chitosan Derivatives: Synthesis, Characterization, Antibacterial Activity, and Killing Kinetics. ACS OMEGA 2020; 5:29657-29666. [PMID: 33251401 PMCID: PMC7689678 DOI: 10.1021/acsomega.0c01355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/16/2020] [Indexed: 06/12/2023]
Abstract
Chitosan has become an established platform biopolymer with applications in biomedical engineering, nanomedicine, and the development of new materials with improved solubility, antimicrobial activity, and low toxicity. In this study, a series of chitosan derivatives were synthesized by conjugating various perfluorocarbon chains to chitosan via Schiff base formation or nucleophilic substitution, followed by quaternization with glycidyl trimethylammonium chloride to confer non-pH-dependent permanent positive charges. Synthesized fluorinated N-(2-hydroxypropyl)-3-trimethylammonium chitosan chloride polymers were characterized and investigated for their antibacterial efficacies against multidrug-resistant bacteria including clinical isolates. The polymers showed activity against both Gram-positive and Gram-negative bacteria (MIC = 64-512 μg/mL) but with greater potency against the former. They displayed rapid bactericidal properties, based on the MBC/MIC ratio, which were further confirmed by the time-kill kinetic assays. Given the properties presented here, fluorinated quaternary chitosan derivatives can serve as great candidates to be investigated as environmentally more benign, nontherapeutic antimicrobial agents that could serve as alternatives to the heavy reliance on antibiotics, which are currently in a very precarious state due to increasing occurrence of drug resistance.
Collapse
Affiliation(s)
- Zamani
E.D. Cele
- Biopolymer
Modification & Therapeutics Laboratory, Chemicals Cluster, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria 0001, South Africa
| | - Anou M. Somboro
- Biomedical
Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Daniel G. Amoako
- Biomedical
Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4000, South Africa
| | - Lindokuhle F. Ndlandla
- Biopolymer
Modification & Therapeutics Laboratory, Chemicals Cluster, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria 0001, South Africa
| | - Mohammed O. Balogun
- Biopolymer
Modification & Therapeutics Laboratory, Chemicals Cluster, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria 0001, South Africa
| |
Collapse
|
8
|
Shen T, Chen L, Liu Y, Shi S, Liu Z, Cai K, Liao C, Wang C. Decanoic acid modification enhances the antibacterial activity of PMAP-23RI-Dec. Eur J Pharm Sci 2020; 157:105609. [PMID: 33141035 DOI: 10.1016/j.ejps.2020.105609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/26/2020] [Accepted: 10/19/2020] [Indexed: 01/12/2023]
Abstract
Antimicrobial peptides are a new type of antibacterial drugs with a broad antibacterial spectrum. Based on our previous research, PMAP-23RI-Dec was designed by modifying the C-terminal of PMAP-23RI with decanoic acid. In this study, we measured the antibacterial activity, stability, hemolysis, and cytotoxicity of PMAP-23RI-Dec. The mechanism of PMAP-23RI-Dec on biofilm and cell membranes were also studied. The results show that PMAP-23RI-Dec exhibited high antibacterial activity and stability, but the hemolytic activity and cytotoxicity of PMAP-23RI-Dec were not enhanced. Moreover, PMAP-23RI-Dec could inhibit biofilm formation at low concentrations, and enhance the killing effect on bacteria by changing the permeability of their cell membranes. Finally, PMAP-23RI-Dec reduced Pseudomonas aeruginosa GIM1.551 and Staphylococcus aureus ATCC25923 damage to organs, and showed superior efficacy against peritonitis. PMAP-23RI-Dec also reduced the scope of abscess and alleviated wound infections. Our research indicated that PMAP-23RI-Dec is a new antibacterial agent with potential clinical application.
Collapse
Affiliation(s)
- Tengfei Shen
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Liangliang Chen
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Yongqing Liu
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Shuaibing Shi
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Zhixin Liu
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Kairui Cai
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Chengshui Liao
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China; Henan Provincial Open Laboratory of Key Disciplines in Environmental and Animal Products Safety, Henan University of Science and Technology, Luoyang, Henan 471000, China.
| | - Chen Wang
- The Key Lab of Veterinary Biological Products, Henan University of Science and Technology, Luoyang, Henan 471000, China; Henan Provincial Open Laboratory of Key Disciplines in Environmental and Animal Products Safety, Henan University of Science and Technology, Luoyang, Henan 471000, China.
| |
Collapse
|
9
|
Srivastava S, Dashora K, Ameta KL, Singh NP, El-Enshasy HA, Pagano MC, Hesham AEL, Sharma GD, Sharma M, Bhargava A. Cysteine-rich antimicrobial peptides from plants: The future of antimicrobial therapy. Phytother Res 2020; 35:256-277. [PMID: 32940412 DOI: 10.1002/ptr.6823] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/26/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022]
Abstract
There has been a spurt in the spread of microbial resistance to antibiotics due to indiscriminate use of antimicrobial agents in human medicine, agriculture, and animal husbandry. It has been realized that conventional antibiotic therapy would be less effective in the coming decades and more emphasis should be given for the development of novel antiinfective therapies. Cysteine rich peptides (CRPs) are broad-spectrum antimicrobial agents that modulate the innate immune system of different life forms such as bacteria, protozoans, fungi, plants, insects, and animals. These are also expressed in several plant tissues in response to invasion by pathogens, and play a crucial role in the regulation of plant growth and development. The present work explores the importance of CRPs as potent antimicrobial agents, which can supplement and/or replace the conventional antibiotics. Different plant parts of diverse plant species showed the presence of antimicrobial peptides (AMPs), which had significant structural and functional diversity. The plant-derived AMPs exhibited potent activity toward a range of plant and animal pathogens, protozoans, insects, and even against cancer cells. The cysteine-rich AMPs have opened new avenues for the use of plants as biofactories for the production of antimicrobials and can be considered as promising antimicrobial drugs in biotherapeutics.
Collapse
Affiliation(s)
- Shilpi Srivastava
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, India
| | - Kavya Dashora
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi, India
| | - Keshav Lalit Ameta
- Department of Chemistry, School of Liberal Arts and Sciences, Mody University of Science and Technology, Lakshmangarh, Rajasthan, India
| | | | - Hesham Ali El-Enshasy
- Institute of Bioproduct Development (IBD), School of Chemical and Energy Engineering, Universiti Teknologi Malaysia (UTM), Skudai, Johor Bahru, Malaysia.,City of Scientific Research and Technology Applications (SRTA), New Burg Al Arab, Alexandria, Egypt
| | | | - Abd El-Latif Hesham
- Genetics Department, Faculty of Agriculture, Beni-Suef University, Beni-Suef, Egypt
| | | | - Minaxi Sharma
- Department of Food Technology, Akal College of Agriculture, Eternal University, Baru Sahib, India
| | - Atul Bhargava
- Department of Botany, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
10
|
Tsotetsi N, Amoako DG, Somboro AM, Khumalo HM, Khan RB. Molecular mechanisms underlying the renoprotective effects of 1,4,7-triazacyclononane: a βeta-lactamase inhibitor. Cytotechnology 2020; 72:785-796. [PMID: 32920746 DOI: 10.1007/s10616-020-00422-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Broad-spectrum β-lactam antibiotics such as penicillin are routinely used against both Gram-negative and Gram-positive bacteria. However, bacteria that produce β-lactamase have developed resistance against these antibiotics by cleaving the β-lactam ring and rendering the antibiotic inactive. To combat this effect, 1,4,7- Triazacyclononane (TACN), a cyclic organic compound derived from cyclononanes has been shown to preserve the activity of β-lactam antibiotics by inhibiting β-lactamase. However, its cytotoxic effects require elucidation. Given that the cytotoxic target for many therapeutics is the kidney, this study investigated the effects of TACN on human embryonic kidney cells (Hek293) cells. Hek293 cells were treated with TACN (0-500 µM) for 24 h and the cytotoxicity was assessed (MTT and LDH assay). Apoptosis was luminometrically detected by measuring phosphatidylserine externalisation and caspase activity and fluorescently detecting necrosis. DNA fragmentation was visualised using fluorescent microscopy. Expression of the apoptosis-related protein were determined by western blot. The results generated indicate that TACN does not initiate necrosis as LDH was decreased. Likewise, decreased apoptosis was supported by the decreased phosphatidylserine, caspases, Bax, cleaved PARP, IAP and NF-kB. However, increased DNA fragmentation was associated with increased p53. Therefore, effects of TACN at the nucleus, produced a p53 response to initiate DNA repair and did not culminate in cell death. The findings show that TACN is not cytotoxic to Hek293 cells via the apoptotic route. Since TACN did not induce cell death, its potential as a metallo-β-lactamase inhibitor (MBLI) may be exploited to counteract the effect of MBL-producing bacteria. Restoring β-lactam activity will curb the global menace of antibiotic resistance.
Collapse
Affiliation(s)
- Nrateng Tsotetsi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Daniel G Amoako
- Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Anou M Somboro
- Biomedical Resource Unit, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hezekiel M Khumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Rene B Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
11
|
Gunjal VB, Thakare R, Chopra S, Reddy DS. Teixobactin: A Paving Stone toward a New Class of Antibiotics? J Med Chem 2020; 63:12171-12195. [PMID: 32520557 DOI: 10.1021/acs.jmedchem.0c00173] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Antimicrobial resistance is a serious threat to human health worldwide, prompting research efforts on a massive scale in search of novel antibiotics to fill an urgent need for a remedy. Teixobactin, a macrocyclic depsipeptide natural product, isolated from uncultured bacteria (Eleftheria terrae), displayed potent activity against several Gram-positive pathogenic bacteria. The distinct pharmacological profile and interesting structural features of teixobactin with nonstandard amino acid (three d-amino acids and l-allo-enduracididine) residues attracted several research groups to work on this target molecule in search of novel antibiotics with new mechanism. Herein, we present a comprehensive and critical perspective on immense possibilities offered by teixobactin in the domain of drug discovery. Efforts made by various research groups since its isolation are discussed, highlighting the molecule's considerable potential with special emphasis on replacement of amino acids. Critical analysis of synthetic efforts, SAR studies, and the way forward are provided hereunder.
Collapse
Affiliation(s)
- Vidya B Gunjal
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ritesh Thakare
- CSIR-Central Drug Research Institute, Sector 10, Janakipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - Sidharth Chopra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.,CSIR-Central Drug Research Institute, Sector 10, Janakipuram Extension, Sitapur Road, Lucknow 226031, Uttar Pradesh, India
| | - D Srinivasa Reddy
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
12
|
Mode of action of teixobactins in cellular membranes. Nat Commun 2020; 11:2848. [PMID: 32503964 PMCID: PMC7275090 DOI: 10.1038/s41467-020-16600-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/12/2020] [Indexed: 12/16/2022] Open
Abstract
The natural antibiotic teixobactin kills pathogenic bacteria without detectable resistance. The difficult synthesis and unfavourable solubility of teixobactin require modifications, yet insufficient knowledge on its binding mode impedes the hunt for superior analogues. Thus far, teixobactins are assumed to kill bacteria by binding to cognate cell wall precursors (Lipid II and III). Here we present the binding mode of teixobactins in cellular membranes using solid-state NMR, microscopy, and affinity assays. We solve the structure of the complex formed by an improved teixobactin-analogue and Lipid II and reveal how teixobactins recognize a broad spectrum of targets. Unexpectedly, we find that teixobactins only weakly bind to Lipid II in cellular membranes, implying the direct interaction with cell wall precursors is not the sole killing mechanism. Our data suggest an additional mechanism affords the excellent activity of teixobactins, which can block the cell wall biosynthesis by capturing precursors in massive clusters on membranes. The natural antibiotic teixobactin kills bacteria by direct binding to their cognate cell wall precursors (Lipid II and III). Here authors use solid-state NMR to reveal the native binding mode of teixobactins and show that teixobactins only weakly bind to Lipid II in anionic cellular membranes.
Collapse
|
13
|
Koulenti D, Xu E, Song A, Sum Mok IY, Karageorgopoulos DE, Armaganidis A, Tsiodras S, Lipman J. Emerging Treatment Options for Infections by Multidrug-Resistant Gram-Positive Microorganisms. Microorganisms 2020; 8:E191. [PMID: 32019171 PMCID: PMC7074912 DOI: 10.3390/microorganisms8020191] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
Antimicrobial agents are currently the mainstay of treatment for bacterial infections worldwide. However, due to the increased use of antimicrobials in both human and animal medicine, pathogens have now evolved to possess high levels of multi-drug resistance, leading to the persistence and spread of difficult-to-treat infections. Several current antibacterial agents active against Gram-positive bacteria will be rendered useless in the face of increasing resistance rates. There are several emerging antibiotics under development, some of which have been shown to be more effective with an improved safety profile than current treatment regimens against Gram-positive bacteria. We will extensively discuss these antibiotics under clinical development (phase I-III clinical trials) to combat Gram-positive bacteria, such as Staphylococcus aureus, Enterococcus faecium and Streptococcus pneumoniae. We will delve into the mechanism of actions, microbiological spectrum, and, where available, the pharmacokinetics, safety profile, and efficacy of these drugs, aiming to provide a comprehensive review to the involved stakeholders.
Collapse
Affiliation(s)
- Despoina Koulenti
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
- 2nd Critical Care Department, Attikon University Hospital, 12462 Athens, Greece;
| | - Elena Xu
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
| | - Andrew Song
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
| | - Isaac Yin Sum Mok
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
| | - Drosos E. Karageorgopoulos
- 4th Department of Internal Medicine, Attikon University Hospital, 12462 Athens, Greece; (D.E.K.); (S.T.)
| | | | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Attikon University Hospital, 12462 Athens, Greece; (D.E.K.); (S.T.)
| | - Jeffrey Lipman
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia; (E.X.); (A.S.); (I.Y.S.M.); (J.L.)
- Department of Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
- Anesthesiology and Critical Care, Centre Hospitalier Universitaire De Nîmes (CHU), University of Montpellier, 30029 Nîmes, France
| |
Collapse
|
14
|
Karas JA, Chen F, Schneider-Futschik EK, Kang Z, Hussein M, Swarbrick J, Hoyer D, Giltrap AM, Payne RJ, Li J, Velkov T. Synthesis and structure-activity relationships of teixobactin. Ann N Y Acad Sci 2019; 1459:86-105. [PMID: 31792983 DOI: 10.1111/nyas.14282] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/15/2022]
Abstract
The discovery of antibiotics has led to the effective treatment of bacterial infections that were otherwise fatal and has had a transformative effect on modern medicine. Teixobactin is an unusual depsipeptide natural product that was recently discovered from a previously unculturable soil bacterium and found to possess potent antibacterial activity against several Gram positive pathogens, including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococci. One of the key features of teixobactin as an antibiotic lead is that resistance could not be generated in a laboratory setting. This is proposed to be a result of a mechanism of action that involves binding to essential cell wall synthesis building blocks, lipid II and lipid III. Since the initial isolation report in 2015, significant efforts have been made to understand its unique mechanism of action, develop efficient synthetic routes for its production, and thus enable the generation of analogues for structure-activity relationship studies and optimization of its pharmacological properties. Our review provides a comprehensive treatise on the progress in understanding teixobactin chemistry, structure-activity relationships, and mechanisms of antibacterial activity. Teixobactin represents an exciting starting point for the development of new antibiotics that can be used to combat multidrug-resistant bacterial ("superbug") infections.
Collapse
Affiliation(s)
- John A Karas
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Fan Chen
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Elena K Schneider-Futschik
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia.,Lung Health Research Centre, Department of Pharmacology & Therapeutics, the University of Melbourne, Parkville, Victoria, Australia
| | - Zhisen Kang
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Maytham Hussein
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - James Swarbrick
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Hoyer
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia.,The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia.,Department of Molecular Medicine, the Scripps Research Institute, La Jolla, California
| | - Andrew M Giltrap
- School of Chemistry, the University of Sydney, Sydney, New South Wales, Australia
| | - Richard J Payne
- School of Chemistry, the University of Sydney, Sydney, New South Wales, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
15
|
Chiorean S, Antwi I, Carney DW, Kotsogianni I, Giltrap AM, Alexander FM, Cochrane SA, Payne RJ, Martin NI, Henninot A, Vederas JC. Dissecting the Binding Interactions of Teixobactin with the Bacterial Cell-Wall Precursor Lipid II. Chembiochem 2019; 21:789-792. [PMID: 31552694 DOI: 10.1002/cbic.201900504] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Indexed: 12/11/2022]
Abstract
The prevalence of life-threatening, drug-resistant microbial infections has challenged researchers to consider alternatives to currently available antibiotics. Teixobactin is a recently discovered "resistance-proof" antimicrobial peptide that targets the bacterial cell wall precursor lipid II. In doing so, teixobactin exhibits potent antimicrobial activity against a wide range of Gram-positive organisms. Herein we demonstrate that teixobactin and several structural analogues are capable of binding lipid II from both Gram-positive and Gram-negative bacteria. Furthermore, we show that when combined with known outer membrane-disrupting peptides, teixobactin is active against Gram-negative organisms.
Collapse
Affiliation(s)
- Sorina Chiorean
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta, T6G 2G2, Canada
| | - Isaac Antwi
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta, T6G 2G2, Canada
| | - Daniel W Carney
- Ferring Research Institute, Inc., 4245 Sorrento Valley Boulevard, San Diego, CA, 92121, USA
| | - Ioli Kotsogianni
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE, Leiden, NL
| | - Andrew M Giltrap
- School of Chemistry, University of Sydney, Chemistry Building (F11) Eastern Avenue, Sydney, NSW, 2006, Australia
| | - Francesca M Alexander
- School of Chemistry and Chemical Engineering, Queen's University, 39 Stranmillis Road, Belfast, BT9 5AG, UK
| | - Stephen A Cochrane
- School of Chemistry and Chemical Engineering, Queen's University, 39 Stranmillis Road, Belfast, BT9 5AG, UK
| | - Richard J Payne
- School of Chemistry, University of Sydney, Chemistry Building (F11) Eastern Avenue, Sydney, NSW, 2006, Australia
| | - Nathaniel I Martin
- Biological Chemistry Group, Institute of Biology Leiden, Leiden University, Sylviusweg 72, 2333 BE, Leiden, NL
| | - Antoine Henninot
- Ferring Research Institute, Inc., 4245 Sorrento Valley Boulevard, San Diego, CA, 92121, USA
| | - John C Vederas
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta, T6G 2G2, Canada
| |
Collapse
|
16
|
Zhou J, Liu Y, Shen T, Chen L, Zhang C, Cai K, Liu Z, Meng X, Zhang L, Liao C, Wang C. Enhancing the antibacterial activity of PMAP-37 by increasing its hydrophobicity. Chem Biol Drug Des 2019; 94:1986-1999. [PMID: 31437351 DOI: 10.1111/cbdd.13601] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/09/2019] [Accepted: 07/27/2019] [Indexed: 12/15/2022]
Abstract
With increasing resistance against conventional antibiotics, there is an urgent need to discover novel substances to replace antibiotics. This need provides an opportunity for the development of antimicrobial peptides (AMPs). To develop new AMPs with effective and safe therapeutic effects, two PMAP-37 analogs called PMAP-37(R13-I) and PMAP-37(K20/27-I) were designed to increase hydrophobicity. Antimicrobial susceptibility testing and animal infection models were used to assess their antibacterial activity. The results showed that the minimal inhibitory concentrations of PMAP-37(R13-I) were lower than those of PMAP-37 for two gram-negative strains. Compared with PMAP-37, PMAP-37(K20/27-I) not only inhibited the growth of most bacterial strains, but also exhibited antibacterial activity against Shigella flexneri CICC21534. In addition, PMAP-37(K20/27-I) exhibited pH and thermal stability. PMAP-37(R13-I) had a therapeutic effect only in mice infected with Salmonella typhimurium SL1344. However, PMAP-37(K20/27-I) exhibited the therapeutic effects, whether in the clinical symptoms, the tissue lesions, or the tissue bacterial loads and the survival rates in mice infected with Staphylococcus aureus ATCC25923 or S. typhimurium SL1344. Therefore, PMAP-37(K20/27-I) can be used as a substitute for antibiotics against infection with bacterial strains.
Collapse
Affiliation(s)
- Jiangfei Zhou
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yongqing Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Tengfei Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Liangliang Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Cong Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Kairui Cai
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Zhixin Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Xiangmiao Meng
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Ling Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Chengshui Liao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Chen Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
17
|
Gomes A, Bessa LJ, Fernandes I, Ferraz R, Mateus N, Gameiro P, Teixeira C, Gomes P. Turning a Collagenesis-Inducing Peptide Into a Potent Antibacterial and Antibiofilm Agent Against Multidrug-Resistant Gram-Negative Bacteria. Front Microbiol 2019; 10:1915. [PMID: 31481944 PMCID: PMC6710338 DOI: 10.3389/fmicb.2019.01915] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Antimicrobial resistance is becoming one the most serious health threats worldwide, as it not only hampers effective treatment of infectious diseases using current antibiotics, but also increases the risks of medical procedures like surgery, transplantation, bone and dental implantation, chemotherapy, or chronic wound management. To date, there are no effective measures to tackle life-threatening nosocomial infections caused by multidrug resistant bacterial species, of which Gram-negative species within the so-called "ESKAPE" pathogens are the most worrisome. Many such bacteria are frequently isolated from severely infected skin lesions such as diabetic foot ulcers (DFU). In this connection, we are pursuing new peptide constructs encompassing antimicrobial and collagenesis-inducing motifs, to tackle skin and soft tissue infections by exerting a dual effect: antimicrobial protection and faster healing of the wound. This produced peptide 3.1-PP4 showed MIC values as low as 1.0 and 2.1 μM against Escherichia coli and Pseudomonas aeruginosa, respectively, and low toxicity to HFF-1 human fibroblasts. Remarkably, the peptide was also potent against multidrug-resistant isolates of Klebsiella pneumoniae, E. coli, and P. aeruginosa (MIC values between 0.5 and 4.1 μM), and hampered the formation of/disaggregated K. pneumoniae biofilms of resistant clinical isolates. Moreover, this notable hybrid peptide retained the collagenesis-inducing behavior of the reference cosmeceutical peptide C16-PP4 ("Matrixyl"). In conclusion, 3.1-PP4 is a highly promising lead toward development of a topical treatment for severely infected skin injuries.
Collapse
Affiliation(s)
- Ana Gomes
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Lucinda J. Bessa
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Iva Fernandes
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Ricardo Ferraz
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Porto, Portugal
| | - Nuno Mateus
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Paula Gameiro
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Cátia Teixeira
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Paula Gomes
- Laboratório Associado para a Química Verde da Rede de Química e Tecnologia (LAQV-REQUIMTE), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| |
Collapse
|
18
|
|
19
|
Sosibo SC, Somboro AM, Amoako DG, Osei Sekyere J, Bester LA, Ngila JC, Sun DD, Kumalo HM. Impact of Pyridyl Moieties on the Inhibitory Properties of Prominent Acyclic Metal Chelators Against Metallo-β-Lactamase-Producing Enterobacteriaceae: Investigating the Molecular Basis of Acyclic Metal Chelators' Activity. Microb Drug Resist 2019; 25:439-449. [DOI: 10.1089/mdr.2018.0272] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Sphelele C. Sosibo
- Department of Applied Chemistry, University of Johannesburg, Doornfontein, Johannesburg, South Africa
| | - Anou M. Somboro
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Daniel G. Amoako
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - John Osei Sekyere
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Durban, South Africa
| | - Linda A. Bester
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Jane C. Ngila
- Department of Applied Chemistry, University of Johannesburg, Doornfontein, Johannesburg, South Africa
| | - Darren D. Sun
- Department of Applied Chemistry, University of Johannesburg, Doornfontein, Johannesburg, South Africa
- School of Civil and Environmental Engineering, Nanyang Technological University, Singapore, Singapore
| | - Hezekiel M. Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
20
|
Somboro AM, Amoako DG, Osei Sekyere J, Kumalo HM, Khan R, Bester LA, Essack SY. 1,4,7-Triazacyclononane Restores the Activity of β-Lactam Antibiotics against Metallo-β-Lactamase-Producing Enterobacteriaceae: Exploration of Potential Metallo-β-Lactamase Inhibitors. Appl Environ Microbiol 2019; 85:e02077-18. [PMID: 30478231 PMCID: PMC6344617 DOI: 10.1128/aem.02077-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 10/24/2018] [Indexed: 01/07/2023] Open
Abstract
Metallo-β-lactamase (MBL)-producing Enterobacteriaceae are of grave clinical concern, particularly as there are no metallo-β-lactamase inhibitors approved for clinical use. The discovery and development of MBL inhibitors to restore the efficacy of available β-lactams are thus imperative. We investigated a zinc-chelating moiety, 1,4,7-triazacyclononane (TACN), for its inhibitory activity against clinical carbapenem-resistant Enterobacteriaceae MICs, minimum bactericidal concentrations (MBCs), the serum effect, fractional inhibitory concentration indexes, and time-kill kinetics were determined using broth microdilution techniques according to Clinical and Laboratory Standards Institute (CSLI) guidelines. Enzyme kinetic parameters and the cytotoxic effects of TACN were determined using spectrophotometric assays. The interactions of the enzyme-TACN complex were investigated by computational studies. Meropenem regained its activity against carbapenemase-producing Enterobacteriaceae, with the MIC decreasing from between 8 and 64 mg/liter to 0.03 mg/liter in the presence of TACN. The TACN-meropenem combination showed bactericidal effects with an MBC/MIC ratio of ≤4, and synergistic activity was observed. Human serum effects on the MICs were insignificant, and TACN was found to be noncytotoxic at concentrations above the MIC values. Computational studies predicted that TACN inhibits MBLs by targeting their catalytic active-site pockets. This was supported by its inhibition constant (Ki ), which was 0.044 μM, and its inactivation constant (Kinact), which was 0.0406 min-1, demonstrating that TACN inhibits MBLs efficiently and holds promise as a potential inhibitor.IMPORTANCE Carbapenem-resistant Enterobacteriaceae (CRE)-mediated infections remain a significant public health concern and have been reported to be critical in the World Health Organization's priority pathogens list for the research and development of new antibiotics. CRE produce enzymes, such as metallo-β-lactamases (MBLs), which inactivate β-lactam antibiotics. Combination therapies involving a β-lactam antibiotic and a β-lactamase inhibitor remain a major treatment option for infections caused by β-lactamase-producing organisms. Currently, no MBL inhibitor-β-lactam combination therapy is clinically available for MBL-positive bacterial infections. Hence, developing efficient molecules capable of inhibiting these enzymes could be a promising way to overcome this phenomenon. TACN played a significant role in the inhibitory activity of the tested molecules against CREs by potentiating the activity of carbapenem. This study demonstrates that TACN inhibits MBLs efficiently and holds promises as a potential MBL inhibitor to help curb the global health threat posed by MBL-producing CREs.
Collapse
Affiliation(s)
- Anou M Somboro
- Antimicrobial Research Unit, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Daniel G Amoako
- Antimicrobial Research Unit, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - John Osei Sekyere
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Hezekiel M Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - René Khan
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Linda A Bester
- Biomedical Resource Unit, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sabiha Y Essack
- Antimicrobial Research Unit, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
21
|
Affiliation(s)
- Matthew W McCarthy
- a Weill Cornell Medical College, Division of General Internal Medicine , New York-Presbyterian Hospital , New York , NY , USA
| |
Collapse
|