151
|
Muto A, Goto S, Kurita D, Ushida C, Himeno H. Involvement of GcvB small RNA in intrinsic resistance to multiple aminoglycoside antibiotics in Escherichia coli. J Biochem 2021; 169:485-489. [PMID: 33169170 DOI: 10.1093/jb/mvaa122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/23/2020] [Indexed: 11/14/2022] Open
Abstract
Deleting the gene for small RNA GcvB in Escherichia coli was found to increase the sensitivity to several aminoglycoside antibiotics, such as neomycin, streptomycin, kanamycin, kasugamycin and spectinomycin, at low concentrations. GcvB, conserved in gram-negative enteric bacteria, is known to negatively control the expression of many genes for amino acid incorporation systems, especially the periplasmic ABC-transporter proteins. Deletions of several amino acid transporter genes in ΔgcvB cells decreased the antibiotic sensitivity to the wild-type level, suggesting that those genes are involved in uptake of aminoglycosides into the cell. Since GcvB is constitutively synthesized in growing cells, repressing synthesis of amino acid transporters, it contributes to the intrinsic resistance to several aminoglycoside antibiotics.
Collapse
Affiliation(s)
- Akira Muto
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Bioscience, Hirosaki University, Bunkyo-cho 3, Hirosaki, Aomori 036-8561, Japan
| | - Simon Goto
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Bioscience, Hirosaki University, Bunkyo-cho 3, Hirosaki, Aomori 036-8561, Japan
| | - Daisuke Kurita
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Bioscience, Hirosaki University, Bunkyo-cho 3, Hirosaki, Aomori 036-8561, Japan
| | - Chisato Ushida
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Bioscience, Hirosaki University, Bunkyo-cho 3, Hirosaki, Aomori 036-8561, Japan
| | - Hyota Himeno
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Bioscience, Hirosaki University, Bunkyo-cho 3, Hirosaki, Aomori 036-8561, Japan
| |
Collapse
|
152
|
Zuo P, Yu P, Alvarez PJJ. Aminoglycosides Antagonize Bacteriophage Proliferation, Attenuating Phage Suppression of Bacterial Growth, Biofilm Formation, and Antibiotic Resistance. Appl Environ Microbiol 2021; 87:e0046821. [PMID: 34020940 PMCID: PMC8276799 DOI: 10.1128/aem.00468-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/17/2021] [Indexed: 01/21/2023] Open
Abstract
The common cooccurrence of antibiotics and phages in both natural and engineered environments underscores the need to understand their interactions and implications for bacterial control and antibiotic resistance propagation. Here, aminoglycoside antibiotics that inhibit protein synthesis (e.g., kanamycin and neomycin) impeded the replication of coliphage T3 and Bacillus phage BSP, reducing their infection efficiency and mitigating their hindrance of bacterial growth, biofilm formation, and tolerance to antibiotics. For example, treatment with phage T3 reduced subsequent biofilm formation by Escherichia coli liquid cultures to 53% ± 5% of that of the no-phage control, but a smaller reduction of biofilm formation (89% ± 10%) was observed for combined exposure to phage T3 and kanamycin. Despite sharing a similar mode of action with aminoglycosides (i.e., inhibiting protein synthesis) and antagonizing phage replication, albeit to a lesser degree, tetracyclines did not inhibit bacterial control by phages. Phage T3 combined with tetracycline showed higher suppression of biofilm formation than when combined with aminoglycosides (25% ± 6% of the no-phage control). The addition of phage T3 to E. coli suspensions with tetracycline also suppressed the development of tolerance to tetracycline. However, this suppression of antibiotic tolerance development disappeared when tetracycline was replaced with 3 mg/liter kanamycin, corroborating the greater antagonism with aminoglycosides. Overall, this study highlights this overlooked antagonistic effect on phage proliferation, which may attenuate phage suppression of bacterial growth, biofilm formation, antibiotic tolerance, and maintenance of antibiotic resistance genes. IMPORTANCE The coexistence of residual antibiotics and phages is common in many environments, which underscores the need to understand their interactive effects on bacteria and the implications for antibiotic resistance propagation. Here, aminoglycosides acting as bacterial protein synthesis inhibitors impeded the replication of various phages. This alleviated the suppressive effects of phages against bacterial growth and biofilm formation and diminished bacterial fitness costs that suppress the emergence of tolerance to antibiotics. We show that changes in bacteria caused by environmentally relevant concentrations of sublethal antibiotics can affect phage-host dynamics that are commonly overlooked in vitro but can result in unexpected environmental consequences.
Collapse
Affiliation(s)
- Pengxiao Zuo
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - Pingfeng Yu
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - Pedro J. J. Alvarez
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| |
Collapse
|
153
|
Leyton B, Ramos JN, Baio PVP, Veras JFC, Souza C, Burkovski A, Mattos-Guaraldi AL, Vieira VV, Abanto Marin M. Treat Me Well or Will Resist: Uptake of Mobile Genetic Elements Determine the Resistome of Corynebacterium striatum. Int J Mol Sci 2021; 22:7499. [PMID: 34299116 PMCID: PMC8304765 DOI: 10.3390/ijms22147499] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
Corynebacterium striatum, a bacterium that is part of the normal skin microbiota, is also an opportunistic pathogen. In recent years, reports of infections and in-hospital and nosocomial outbreaks caused by antimicrobial multidrug-resistant C. striatum strains have been increasing worldwide. However, there are no studies about the genomic determinants related to antimicrobial resistance in C. striatum. This review updates global information related to antimicrobial resistance found in C. striatum and highlights the essential genomic aspects in its persistence and dissemination. The resistome of C. striatum comprises chromosomal and acquired elements. Resistance to fluoroquinolones and daptomycin are due to mutations in chromosomal genes. Conversely, resistance to macrolides, tetracyclines, phenicols, beta-lactams, and aminoglycosides are associated with mobile genomic elements such as plasmids and transposons. The presence and diversity of insertion sequences suggest an essential role in the expression of antimicrobial resistance genes (ARGs) in genomic rearrangements and their potential to transfer these elements to other pathogens. The present study underlines that the resistome of C. striatum is dynamic; it is in evident expansion and could be acting as a reservoir for ARGs.
Collapse
Affiliation(s)
- Benjamin Leyton
- Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile;
- Carrera de Bioquímica, Departamento de Ciencias Químicas y Recursos Naturales, Universidad de La Frontera, Temuco 4811230, Chile
| | - Juliana Nunes Ramos
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-361, Brazil; (J.N.R.); (P.V.P.B.); (J.F.C.V.); (V.V.V.)
- Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro—LDCIC/FCM/UERJ, Rio de Janeiro 20550-170, Brazil; (C.S.); (A.L.M.-G.)
| | - Paulo Victor Pereira Baio
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-361, Brazil; (J.N.R.); (P.V.P.B.); (J.F.C.V.); (V.V.V.)
| | - João Flávio Carneiro Veras
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-361, Brazil; (J.N.R.); (P.V.P.B.); (J.F.C.V.); (V.V.V.)
| | - Cassius Souza
- Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro—LDCIC/FCM/UERJ, Rio de Janeiro 20550-170, Brazil; (C.S.); (A.L.M.-G.)
| | - Andreas Burkovski
- Department of Biology, Professur für Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstrasse 5, 91058 Erlangen, Germany;
| | - Ana Luíza Mattos-Guaraldi
- Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro—LDCIC/FCM/UERJ, Rio de Janeiro 20550-170, Brazil; (C.S.); (A.L.M.-G.)
| | - Verônica Viana Vieira
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-361, Brazil; (J.N.R.); (P.V.P.B.); (J.F.C.V.); (V.V.V.)
| | - Michel Abanto Marin
- Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4811230, Chile;
| |
Collapse
|
154
|
Yang X, Ye W, Qi Y, Ying Y, Xia Z. Overcoming Multidrug Resistance in Bacteria Through Antibiotics Delivery in Surface-Engineered Nano-Cargos: Recent Developments for Future Nano-Antibiotics. Front Bioeng Biotechnol 2021; 9:696514. [PMID: 34307323 PMCID: PMC8297506 DOI: 10.3389/fbioe.2021.696514] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
In the recent few decades, the increase in multidrug-resistant (MDR) bacteria has reached an alarming rate and caused serious health problems. The incidence of infections due to MDR bacteria has been accompanied by morbidity and mortality; therefore, tackling bacterial resistance has become an urgent and unmet challenge to be properly addressed. The field of nanomedicine has the potential to design and develop efficient antimicrobials for MDR bacteria using its innovative and alternative approaches. The uniquely constructed nano-sized antimicrobials have a predominance over traditional antibiotics because their small size helps them in better interaction with bacterial cells. Moreover, surface engineering of nanocarriers offers significant advantages of targeting and modulating various resistance mechanisms, thus owe superior qualities for overcoming bacterial resistance. This review covers different mechanisms of antibiotic resistance, application of nanocarrier systems in drug delivery, functionalization of nanocarriers, application of functionalized nanocarriers for overcoming bacterial resistance, possible limitations of nanocarrier-based approach for antibacterial delivery, and future of surface-functionalized antimicrobial delivery systems.
Collapse
Affiliation(s)
- Xinfu Yang
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wenxin Ye
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yajun Qi
- Department of Pharmacy, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yin Ying
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhongni Xia
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
155
|
Sharma V, Kaur R, Salwan R. Streptomyces: host for refactoring of diverse bioactive secondary metabolites. 3 Biotech 2021; 11:340. [PMID: 34221811 DOI: 10.1007/s13205-021-02872-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/31/2021] [Indexed: 12/22/2022] Open
Abstract
Microbial secondary metabolites are intensively explored due to their demands in pharmaceutical, agricultural and food industries. Streptomyces are one of the largest sources of secondary metabolites having diverse applications. In particular, the abundance of secondary metabolites encoding biosynthetic gene clusters and presence of wobble position in Streptomyces strains make it potential candidate as a native or heterologous host for secondary metabolite production including several cryptic gene clusters expression. Here, we have discussed the developments in Streptomyces strains genome mining, its exploration as a suitable host and application of synthetic biology for refactoring genetic systems for developing chassis for enhanced as well as novel secondary metabolites with reduced genome and cleaned background.
Collapse
Affiliation(s)
- Vivek Sharma
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| | - Randhir Kaur
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| | - Richa Salwan
- College of Horticulture and Forestry, Dr YS Parmar University of Horticulture and Forestry, Neri, Hamirpur, Himachal Pradesh 177001 India
| |
Collapse
|
156
|
Dutta D, Kaushik A, Kumar D, Bag S. Foodborne Pathogenic Vibrios: Antimicrobial Resistance. Front Microbiol 2021; 12:638331. [PMID: 34276582 PMCID: PMC8278402 DOI: 10.3389/fmicb.2021.638331] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Foodborne illness caused by pathogenic Vibrios is generally associated with the consumption of raw or undercooked seafood. Fish and other seafood can be contaminated with Vibrio species, natural inhabitants of the marine, estuarine, and freshwater environment. Pathogenic Vibrios of major public health concerns are Vibrio cholerae, Vibrio parahaemolyticus, and Vibrio vulnificus. Common symptoms of foodborne Vibrio infection include watery diarrhea, stomach cramping, nausea, vomiting, fever, and chills. Administration of oral or intravenous rehydration salts solution is the mainstay for the management of cholera, and antibiotics are also used to shorten the duration of diarrhea and to limit further transmission of the disease. Currently, doxycycline, azithromycin, or ciprofloxacin are commonly used for V. cholerae, and doxycycline or quinolone are administered for V. parahaemolyticus, whereas doxycycline and a third-generation cephalosporin are recommended for V. vulnificus as initial treatment regimen. The emergence of antimicrobial resistance (AMR) in Vibrios is increasingly common across the globe and a decrease in the effectiveness of commonly available antibiotics poses a global threat to public health. Recent progress in comparative genomic studies suggests that the genomes of the drug-resistant Vibrios harbor mobile genetic elements like plasmids, integrating conjugative elements, superintegron, transposable elements, and insertion sequences, which are the major carriers of genetic determinants encoding antimicrobial resistance. These mobile genetic elements are highly dynamic and could potentially propagate to other bacteria through horizontal gene transfer (HGT). To combat the serious threat of rising AMR, it is crucial to develop strategies for robust surveillance, use of new/novel pharmaceuticals, and prevention of antibiotic misuse.
Collapse
Affiliation(s)
- Dipanjan Dutta
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Anupam Kaushik
- Department of Microbiology, National Centre for Disease Control, New Delhi, India
| | - Dhirendra Kumar
- Department of Microbiology, National Centre for Disease Control, New Delhi, India
| | | |
Collapse
|
157
|
Abstract
This article presents new data on Bifidobacterium longum MC-42—a strain that has been actively used for the preparation of commercial dairy products in Russia for almost 40 years. It was demonstrated that this strain possesses high activities of β-galactosidase, α-glucosidase, and leucine arylaminidase; inhibits the growth of pathogens such as Salmonella typhimurium, Staphylococcus aureus, and Escherichia coli; and can efficiently remove cholesterol from the cultural medium. The resistance of B. longum MC-42 determined for 15 commonly used antibiotics was in agreement with those previously reported for Bifidobacterium spp. The absence of frequently transmittable antibiotic resistance genes in the genome and the lack of undesirable activity of β-glucuronidase proved the safe use of B. longum МС-42 as a probiotic and starter culture. Additionally, the impact of two growth-promoting additives—yeast extract or milk protein hydrolysate containing supplementation—on the B. longum MC-42 fermentation profile was assessed. The introduction of these additives increases the maximum attainable viable cell count by orders of magnitude, significantly changed the profile of aminopeptidase activities in extracellular extracts, and influenced the antioxidant and antihypertensive properties of the obtained fermented products.
Collapse
|
158
|
Nafis A, Saad FE, Khalloufi FE, Kasrati A, Abbad A, Mezrioui N, Oudra B, Vasconcelos V, Hassani L. New insight into antimicrobial activities of Linaria ventricosa essential oil and its synergetic effect with conventional antibiotics. Arch Microbiol 2021; 203:4361-4366. [PMID: 34115157 DOI: 10.1007/s00203-021-02391-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
The purpose of the present study was to determine for the first time the volatile constituents, the antioxidant and antimicrobial activities of the essential oil (EO) of the endemic Moroccan Linaria ventricosa, alone or in combination with four known antibiotics. The major constituents were 2-methoxy-4-vinylphenol (17.4%), α-terpinene (13.64%) and 3,5-dimethylphenyl isocyanate (12.21%). The EO had moderate antioxidant potency, as measured by DPPH free radical scavenging (1.233 ± 0.031 mg/mL), ferric reducing antioxidant power assay (0.373 ± 0.019 mg/mL) and β-carotene/linoleic acid (0.922 ± 0.026 mg/mL). EO showed microbicidal activity against all microorganisms tested. The highest effectiveness was recorded against Candida albicans (IZ = 24 mm, MIC = 4.87 mg/mL and MMC = 9.75 mg/mL) and Candida glabrata (IZ = 22 mm, MIC = MMC = 4.87 mg/mL). Gram negative bacteria were the most resistant (MIC = MMC = 39 mg/mL). The combination of EO at sub-inhibitory concentrations with antibiotics showed a significant decrease in their individual MICs from 2 to 128 fold, being the best for ciprofloxacin and fluconazole against E. coli and C. albicans and C. glabrata, respectively.
Collapse
Affiliation(s)
- Ahmed Nafis
- Microbiology, Health and Environment Team, Faculty of Sciences, Chouaïb Doukkali University, El Jadida, Morocco.,Laboratory of Microbial Biotechnologies, Agrosciences and Environment, Faculty of Science, Semlalia University Cadi Ayyad, Marrakech, Morocco
| | - Fatima Ezzahraa Saad
- Laboratory of Water, Biodiversity and Climate Change, Faculty of Sciences Semlalia Marrakech, Cadi Ayyad University, Marrakech, Morocco
| | - Fatima El Khalloufi
- Laboratory of Chemistry, Modeling and Environmental Sciences, Polydisciplinary Faculty of Khouribga, Sultan Moulay Slimane University of Beni Mellal, BP. 145, 25000, Khouribga, Morocco
| | - Ayoub Kasrati
- Laboratory of Drug Science, Biomedical and Biotechnological Research, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Abdelaziz Abbad
- Laboratory of Microbial Biotechnologies, Agrosciences and Environment, Faculty of Science, Semlalia University Cadi Ayyad, Marrakech, Morocco
| | - Noureddine Mezrioui
- Laboratory of Microbial Biotechnologies, Agrosciences and Environment, Faculty of Science, Semlalia University Cadi Ayyad, Marrakech, Morocco
| | - Brahim Oudra
- Laboratory of Water, Biodiversity and Climate Change, Faculty of Sciences Semlalia Marrakech, Cadi Ayyad University, Marrakech, Morocco
| | - Vitor Vasconcelos
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, Terminal de Cruzeiros Do Porto de Leixões, Av. General Norton de Matos, S/n, 4450-208, Matosinhos, Portugal. .,Department of Biology, Faculty of Sciences, University of Porto, Rua Do Campo Alegre, 4169-007, Porto, Portugal.
| | - Lahcen Hassani
- Laboratory of Microbial Biotechnologies, Agrosciences and Environment, Faculty of Science, Semlalia University Cadi Ayyad, Marrakech, Morocco
| |
Collapse
|
159
|
Isgren CM. Improving clinical outcomes via responsible antimicrobial use in horses. EQUINE VET EDUC 2021. [DOI: 10.1111/eve.13502] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- C. M. Isgren
- Department of Equine Clinical Science Faculty of Health & Life Sciences Institute of Infection, Veterinary and Ecological Sciences University of Liverpool Leahurst UK
| |
Collapse
|
160
|
Chebotar' IV, Emelyanova MA, Bocharova JA, Mayansky NA, Kopantseva EE, Mikhailovich VM. The classification of bacterial survival strategies in the presence of antimicrobials. Microb Pathog 2021; 155:104901. [PMID: 33930413 DOI: 10.1016/j.micpath.2021.104901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 01/14/2023]
Abstract
The survival of bacteria under antibiotic therapy varies in nature and is based on the bacterial ability to employ a wide range of fundamentally different resistance mechanisms. This great diversity requires a disambiguation of the term 'resistance' and the development of a more precise classification of bacterial survival strategies during contact with antibiotics. The absence of a unified definition for the terms 'resistance', 'tolerance' and 'persistence' further aggravates the imperfections of the current classification system. This review suggests a number of original classification criteria that will take into account (1) the bacterial ability to replicate in the presence of antimicrobial agents, (2) existing evolutionary stability of a trait within a species, and (3) the presence or absence of specialized genes that determine the ability of a microorganism to decrease its own metabolism or switch it completely off. This review describes potential advantages of the suggested classification system, which include a better understanding of the relationship between bacterial survival in the presence of antibiotics and molecular mechanisms of cellular metabolism suppression, the opportunity to pinpoint targets to identify a true bacterial resistance profile. The true resistance profile in turn, could be used to develop effective diagnostic and antimicrobial therapy methods, while taking into consideration specific bacterial survival mechanisms.
Collapse
Affiliation(s)
- Igor V Chebotar'
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow, 119991, Russian Federation; Pirogov Russian National Research Medical University, 1 Ostrovitianov St., Moscow, 117997, Russian Federation
| | - Marina A Emelyanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow, 119991, Russian Federation
| | - Julia A Bocharova
- Pirogov Russian National Research Medical University, 1 Ostrovitianov St., Moscow, 117997, Russian Federation
| | - Nikolay A Mayansky
- Pirogov Russian National Research Medical University, 1 Ostrovitianov St., Moscow, 117997, Russian Federation
| | - Elena E Kopantseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow, 119991, Russian Federation
| | - Vladimir M Mikhailovich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., Moscow, 119991, Russian Federation.
| |
Collapse
|
161
|
The Influence of Permeability through Bacterial Porins in Whole-Cell Compound Accumulation. Antibiotics (Basel) 2021; 10:antibiotics10060635. [PMID: 34073313 PMCID: PMC8226570 DOI: 10.3390/antibiotics10060635] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 01/09/2023] Open
Abstract
The lack of new drugs for Gram-negative pathogens is a global threat to modern medicine. The complexity of their cell envelope, with an additional outer membrane, hinders internal accumulation and thus, the access of molecules to their targets. Our limited understanding of the molecular basis for compound influx and efflux from these pathogens is a major bottleneck for the discovery of effective antibacterial compounds. Here we analyse the correlation between the whole-cell compound accumulation of ~200 molecules and their predicted porin permeability coefficient (influx), using a recently developed scoring function. We found a strong linear relationship (74%) between the two, confirming porins key in compound uptake in Gram-negative bacteria. The analysis of this unique dataset aids to better understand the molecular descriptors behind whole-cell accumulation and molecular uptake in Gram-negative bacteria.
Collapse
|
162
|
Gil-Gil T, Ochoa-Sánchez LE, Baquero F, Martínez JL. Antibiotic resistance: Time of synthesis in a post-genomic age. Comput Struct Biotechnol J 2021; 19:3110-3124. [PMID: 34141134 PMCID: PMC8181582 DOI: 10.1016/j.csbj.2021.05.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
Antibiotic resistance has been highlighted by international organizations, including World Health Organization, World Bank and United Nations, as one of the most relevant global health problems. Classical approaches to study this problem have focused in infected humans, mainly at hospitals. Nevertheless, antibiotic resistance can expand through different ecosystems and geographical allocations, hence constituting a One-Health, Global-Health problem, requiring specific integrative analytic tools. Antibiotic resistance evolution and transmission are multilayer, hierarchically organized processes with several elements (from genes to the whole microbiome) involved. However, their study has been traditionally gene-centric, each element independently studied. The development of robust-economically affordable whole genome sequencing approaches, as well as other -omic techniques as transcriptomics and proteomics, is changing this panorama. These technologies allow the description of a system, either a cell or a microbiome as a whole, overcoming the problems associated with gene-centric approaches. We are currently at the time of combining the information derived from -omic studies to have a more holistic view of the evolution and spread of antibiotic resistance. This synthesis process requires the accurate integration of -omic information into computational models that serve to analyse the causes and the consequences of acquiring AR, fed by curated databases capable of identifying the elements involved in the acquisition of resistance. In this review, we analyse the capacities and drawbacks of the tools that are currently in use for the global analysis of AR, aiming to identify the more useful targets for effective corrective interventions.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Centro Nacional de Biotecnología, CSIC, Darwin 3, 28049 Madrid, Spain
| | | | - Fernando Baquero
- Department of Microbiology, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
- CIBER en Epidemiología y Salud Pública (CIBER-ESP), Madrid, Spain
| | | |
Collapse
|
163
|
Savage TJ, Rao S, Joerger J, Ozonoff A, McAdam AJ, Sandora TJ. Predictive Value of Direct Disk Diffusion Testing from Positive Blood Cultures in a Children's Hospital and Its Utility in Antimicrobial Stewardship. J Clin Microbiol 2021; 59:e02445-20. [PMID: 33692138 PMCID: PMC8316030 DOI: 10.1128/jcm.02445-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/06/2021] [Indexed: 12/19/2022] Open
Abstract
Accurate and early susceptibility results could reduce overuse of broad-spectrum antibiotics for empirical treatment of bacteremia. Direct disk diffusion testing (dDD) using nonstandardized inocula directly from blood cultures could facilitate earlier narrowing of antibiotics. To determine the predictive value of dDD compared with standardized antimicrobial susceptibility testing (AST), we performed a retrospective cohort study of 582 blood cultures from 495 pediatric patients with bacteremia. Positive and negative predictive value (PPV: number of isolates susceptible by both dDD and AST divided by the total number of isolates susceptible by dDD; NPV: number of isolates not susceptible [either intermediate or resistant] by both dDD and AST divided by the total number of isolates not susceptible by dDD), sensitivity, specificity, and 95% confidence interval were calculated for each bacterium-antibiotic combination. We evaluated the Antibiotic Spectrum Index of prescribed antibiotics to assess change in antibiotic prescribing after availability of Gram stain, dDD, and AST results. dDD results were available a median of 21 h before AST results. dDD had PPVs of ≥96% for most organism-antibiotic pairs, including 100% (CI 96 to 100%) for Staphylococcus aureus with oxacillin and 99% (CI 93 to 100%) for Enterobacterales with ceftriaxone. NPVs of dDD were variable and frequently lower than the PPV. Very major errors and major errors occurred in 31/5,454 (0.6%) and 231/5,454 (4.2%) organism-antibiotic combinations, respectively. Antibiotics were narrowed in 30% of cases after a dDD result and a further 25% of cases after AST result. dDD is highly predictive of susceptibility for many common organism-antibiotic combinations and provides actionable information one day earlier than standard susceptibility approaches. dDD has the potential to facilitate earlier deescalation to narrow-spectrum antibiotic treatment.
Collapse
Affiliation(s)
- Timothy J Savage
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Shun Rao
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jill Joerger
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander J McAdam
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas J Sandora
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
164
|
Arana L, Gallego L, Alkorta I. Incorporation of Antibiotics into Solid Lipid Nanoparticles: A Promising Approach to Reduce Antibiotic Resistance Emergence. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:nano11051251. [PMID: 34068834 PMCID: PMC8151913 DOI: 10.3390/nano11051251] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Antimicrobial resistance is one of the biggest threats to global health as current antibiotics are becoming useless against resistant infectious pathogens. Consequently, new antimicrobial strategies are urgently required. Drug delivery systems represent a potential solution to improve current antibiotic properties and reverse resistance mechanisms. Among different drug delivery systems, solid lipid nanoparticles represent a highly interesting option as they offer many advantages for nontoxic targeted drug delivery. Several publications have demonstrated the capacity of SLNs to significantly improve antibiotic characteristics increasing treatment efficiency. In this review article, antibiotic-loaded solid lipid nanoparticle-related works are analyzed to summarize all information associated with applying these new formulations to tackle the antibiotic resistance problem. The main antimicrobial resistance mechanisms and relevant solid lipid nanoparticle characteristics are presented to later discuss the potential of these nanoparticles to improve current antibiotic treatment characteristics and overcome antimicrobial resistance mechanisms. Moreover, solid lipid nanoparticles also offer new possibilities for other antimicrobial agents that cannot be administrated as free drugs. The advantages and disadvantages of these new formulations are also discussed in this review. Finally, given the progress of the studies carried out to date, future directions are discussed.
Collapse
Affiliation(s)
- Lide Arana
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Unibertsitateko Ibilbidea, 7, 01006 Vitoria-Gasteiz, Spain
- Correspondence:
| | - Lucia Gallego
- Department of Immunology, Microbiology and Parasitology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Sarriena Auzoa z/g, 48940 Leioa, Bizkaia, Spain;
| | - Itziar Alkorta
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Sarriena Auzoa z/g, 48940 Leioa, Bizkaia, Spain;
| |
Collapse
|
165
|
Godzieba M, Ciesielski S. Natural DNA Intercalators as Promising Therapeutics for Cancer and Infectious Diseases. Curr Cancer Drug Targets 2021; 20:19-32. [PMID: 31589125 DOI: 10.2174/1568009619666191007112516] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/11/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
Cancer and infectious diseases are one of the greatest challenges of modern medicine. An unhealthy lifestyle, the improper use of drugs, or their abuse are conducive to the increase of morbidity and mortality caused by these diseases. The imperfections of drugs currently used in therapy for these diseases and the increasing problem of drug resistance have forced a search for new substances with therapeutic potential. Throughout history, plants, animals, fungi and microorganisms have been rich sources of biologically active compounds. Even today, despite the development of chemistry and the introduction of many synthetic chemotherapeutics, a substantial part of the new compounds being tested for treatment are still of natural origin. Natural compounds exhibit a great diversity of chemical structures, and thus possess diverse mechanisms of action and molecular targets. Nucleic acids seem to be a good molecular target for substances with anticancer potential in particular, but they may also be a target for antimicrobial compounds. There are many types of interactions of small-molecule ligands with DNA. This publication focuses on the intercalation process. Intercalators are compounds that usually have planar aromatic moieties and can insert themselves between adjacent base pairs in the DNA helix. These types of interactions change the structure of DNA, leading to various types of disorders in the functioning of cells and the cell cycle. This article presents the most promising intercalators of natural origin, which have aroused interest in recent years due to their therapeutic potential.
Collapse
Affiliation(s)
- Martyna Godzieba
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Sloneczna 45 G, 10-917 Olsztyn, Poland
| | - Slawomir Ciesielski
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, ul. Sloneczna 45 G, 10-917 Olsztyn, Poland
| |
Collapse
|
166
|
Borelli TC, Lovate GL, Scaranello AFT, Ribeiro LF, Zaramela L, Pereira-dos-Santos FM, Silva-Rocha R, Guazzaroni ME. Combining Functional Genomics and Whole-Genome Sequencing to Detect Antibiotic Resistance Genes in Bacterial Strains Co-Occurring Simultaneously in a Brazilian Hospital. Antibiotics (Basel) 2021; 10:antibiotics10040419. [PMID: 33920372 PMCID: PMC8070361 DOI: 10.3390/antibiotics10040419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 01/01/2023] Open
Abstract
(1) Background: The rise of multi-antibiotic resistant bacteria represents an emergent threat to human health. Here, we investigate antibiotic resistance mechanisms in bacteria of several species isolated from an intensive care unit in Brazil. (2) Methods: We used whole-genome analysis to identify antibiotic resistance genes (ARGs) and plasmids in 34 strains of Gram-negative and Gram-positive bacteria, providing the first genomic description of Morganella morganii and Ralstonia mannitolilytica clinical isolates from South America. (3) Results: We identified a high abundance of beta-lactamase genes in resistant organisms, including seven extended-spectrum beta-lactamases (OXA-1, OXA-10, CTX-M-1, KPC, TEM, HYDRO, BLP) shared between organisms from different species. Additionally, we identified several ARG-carrying plasmids indicating the potential for a fast transmission of resistance mechanism between bacterial strains. Furthermore, we uncovered two pairs of (near) identical plasmids exhibiting multi-drug resistance. Finally, since many highly resistant strains carry several different ARGs, we used functional genomics to investigate which of them were indeed functional. In this sense, for three bacterial strains (Escherichia coli, Klebsiella pneumoniae, and M. morganii), we identified six beta-lactamase genes out of 15 predicted in silico as those mainly responsible for the resistance mechanisms observed, corroborating the existence of redundant resistance mechanisms in these organisms. (4) Conclusions: Systematic studies similar to the one presented here should help to prevent outbreaks of novel multidrug-resistant bacteria in healthcare facilities.
Collapse
Affiliation(s)
- Tiago Cabral Borelli
- Department of Biology, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-901, Brazil; (T.C.B.); (G.L.L.); (A.F.T.S.); (L.F.R.)
| | - Gabriel Lencioni Lovate
- Department of Biology, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-901, Brazil; (T.C.B.); (G.L.L.); (A.F.T.S.); (L.F.R.)
| | - Ana Flavia Tonelli Scaranello
- Department of Biology, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-901, Brazil; (T.C.B.); (G.L.L.); (A.F.T.S.); (L.F.R.)
| | - Lucas Ferreira Ribeiro
- Department of Biology, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-901, Brazil; (T.C.B.); (G.L.L.); (A.F.T.S.); (L.F.R.)
| | - Livia Zaramela
- Department of Pediatrics, University of California San Diego, San Diego, CA 92161, USA;
| | - Felipe Marcelo Pereira-dos-Santos
- Department of Cell and Molecular Biology, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil; (F.M.P.-d.-S.); (R.S.-R.)
| | - Rafael Silva-Rocha
- Department of Cell and Molecular Biology, Faculdade de Medicina de Ribeirão Preto, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-900, Brazil; (F.M.P.-d.-S.); (R.S.-R.)
| | - María-Eugenia Guazzaroni
- Department of Biology, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP 14049-901, Brazil; (T.C.B.); (G.L.L.); (A.F.T.S.); (L.F.R.)
- Correspondence:
| |
Collapse
|
167
|
Khan J, Tarar SM, Gul I, Nawaz U, Arshad M. Challenges of antibiotic resistance biofilms and potential combating strategies: a review. 3 Biotech 2021; 11:169. [PMID: 33816046 DOI: 10.1007/s13205-021-02707-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
In this modern era, medicine is facing many alarming challenges. Among different challenges, antibiotics are gaining importance. Recent years have seen unprecedented increase in knowledge and understanding of various factors that are root cause of the spread and development of resistance in microbes against antibiotics. The infection results in the formation of microbial colonies which are termed as biofilms. However, it has been found that a multiple factors contribute in the formation of antimicrobial resistance. Due to higher dose of Minimum Bactericidal Concentration (MBC) as well as of Minimum Inhibitory Concentration (MIC), a large batch of antibiotics available today are of no use as they are ineffective against infections. Therefore, to control infections, there is dire need to adopt alternative treatment for biofilm infection other than antibiotics. This review highlights the latest techniques that are being used to cure the menace of biofilm infections. A wide range of mechanisms has been examined with particular attention towards avenues which can be proved fruitful in the treatment of biofilms. Besides, newer strategies, i.e., matrix centered are also discussed as alternative therapeutic techniques including modulating microbial metabolism, matrix degrading enzyme, photodynamic therapy, natural compounds quorum sensing and nanotechnology which are being used to disrupt extra polymeric substances (EPS) matrix of desired bacterial biofilms.
Collapse
Affiliation(s)
- Javairia Khan
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Sumbal Mudassar Tarar
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Iram Gul
- Department of Earth and Environmental Sciences, Hazara University, Mansehra, Pakistan
| | - Uzam Nawaz
- Department of Statistics, The Women University Multan, Multan, Pakistan
| | - Muhammad Arshad
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| |
Collapse
|
168
|
Tarazi YH, Dwekat AFA, Ismail ZB. Molecular characterization of Salmonella spp. isolates from river and dam water, irrigated vegetables, livestock, and poultry manures in Jordan. Vet World 2021; 14:813-819. [PMID: 33935432 PMCID: PMC8076478 DOI: 10.14202/vetworld.2021.813-819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/16/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Aim Salmonellosis is an important food-borne and zoonotic disease with high morbidity and mortality rates. The objectives of this study were to isolate, serotype, and genetically characterize Salmonella spp. from Zarqa river and King Talal dam waters, vegetables irrigated by such waters, and manure of poultry and livestock farms located in the Zarqa river basin in Jordan. In addition, certain virulence factors and antimicrobial resistance patterns of isolated Salmonella strains were determined. Materials and Methods A total of 250 samples were cultured using routine microbiological methods. Suspected Salmonella spp. were identified based on colony morphology and confirmed using biochemical and molecular methods. Virulence genes including invA, stn, and pCT plasmid were detected using multiplex PCR. Phylogenetic analysis was performed using pulsed-field gel electrophoresis (PFGE). Results In total, 32/250 (12.8%) Salmonella spp. isolates were recovered from different sources. Of these, the most common serotype was Salmonella subspecies 1 (23 isolates), followed by Salmonella enterica serovar Typhimurium (4 isolates), Salmonella enterica serovar Typhi (3 isolates), and finally Salmonella enterica serovar Enteritidis (2 isolates). The PFGE indicated that Salmonella enterica serovar Typhimurium isolated from poultry manure and from parsley were closely related (84.6%). Salmonella enterica serovar Enteritidis isolated from the dam water was closely related to Salmonella enterica serovar Enteritidis isolated from spearmint (73.8%). Salmonella enterica serovar Typhi isolated from the river and dam water were 100% related to Salmonella enterica serovar Typhi isolated from lettuce. In the antimicrobial sensitivity test, 14 out of 32 (43.8%) isolated Salmonella strains were resistant to two or more of the major antimicrobial agent groups. However, the majority of isolates were sensitive to ceftriaxone, ciprofloxacin, cefuroxime, and gentamicin (97%, 93.8%, and 87.5%, 84.4%, respectively). All isolates were resistant to erythromycin and amoxicillin. Conclusion Results of this study indicate a serious potential threat to public health associated with consuming leafy green vegetables grown on the banks of Zarqa river and its dam because of widespread Salmonella spp. contamination. Appropriate monitoring of irrigation water must be applied to reduce the possibility of cross-contamination.
Collapse
Affiliation(s)
- Yaser H Tarazi
- Department of Veterinary Basic Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Abdallah F Al Dwekat
- Department of Veterinary Basic Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Zuhair Bani Ismail
- Department of Veterinary Clinical Sciences, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
169
|
Full pathogen characterisation: species identification including the detection of virulence factors and antibiotic resistance genes via multiplex DNA-assays. Sci Rep 2021; 11:6001. [PMID: 33727586 PMCID: PMC7966752 DOI: 10.1038/s41598-021-85438-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/19/2021] [Indexed: 11/08/2022] Open
Abstract
Antibiotic resistances progressively cause treatment failures, and their spreading dynamics reached an alarming level. Some strains have already been classified as highly critical, e.g. the ones summarised by the acronym ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.). To restrain this trend and enable effective medication, as much information as possible must be obtained in the least possible time. Here, we present a DNA microarray-based assay that screens for the most important sepsis-relevant 44 pathogenic species, 360 virulence factors (mediate pathogenicity in otherwise non-pathogenic strains), and 409 antibiotic resistance genes in parallel. The assay was evaluated with 14 multidrug resistant strains, including all ESKAPE pathogens, mainly obtained from clinical isolates. We used a cost-efficient ligation-based detection platform designed to emulate the highly specific multiplex detection of padlock probes. Results could be obtained within one day, requiring approximately 4 h for amplification, application to the microarray, and detection.
Collapse
|
170
|
Zhu N, Zhong C, Liu T, Zhu Y, Gou S, Bao H, Yao J, Ni J. Newly designed antimicrobial peptides with potent bioactivity and enhanced cell selectivity prevent and reverse rifampin resistance in Gram-negative bacteria. Eur J Pharm Sci 2021; 158:105665. [DOI: 10.1016/j.ejps.2020.105665] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/18/2020] [Accepted: 11/26/2020] [Indexed: 01/10/2023]
|
171
|
Sulaiman JE, Lam H. Evolution of Bacterial Tolerance Under Antibiotic Treatment and Its Implications on the Development of Resistance. Front Microbiol 2021; 12:617412. [PMID: 33717007 PMCID: PMC7952611 DOI: 10.3389/fmicb.2021.617412] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/12/2021] [Indexed: 01/08/2023] Open
Abstract
Recent laboratory evolution studies have shown that upon repetitive antibiotic treatments, bacterial populations will adapt and eventually became tolerant and resistant to the drug. Drug tolerance rapidly evolves upon frequent, intermittent antibiotic treatments, and such emerging drug tolerance seems to be specific to the treatment conditions, complicating clinical practice. Moreover, it has been shown that tolerance often promotes the development of resistance, which further reinforces the need of clinical diagnostics for antibiotic tolerance to reduce the occurrence of acquired resistance. Here, we discuss the laboratory evolution studies that were performed to track the development of tolerance in bacterial populations, and highlight the urgency of developing a comprehensive knowledge base of various tolerance phenotypes and their detection in clinics. Finally, we propose future directions for basic research in this growing field.
Collapse
Affiliation(s)
- Jordy Evan Sulaiman
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Henry Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| |
Collapse
|
172
|
Hobson C, Chan AN, Wright GD. The Antibiotic Resistome: A Guide for the Discovery of Natural Products as Antimicrobial Agents. Chem Rev 2021; 121:3464-3494. [PMID: 33606500 DOI: 10.1021/acs.chemrev.0c01214] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The use of life-saving antibiotics has long been plagued by the ability of pathogenic bacteria to acquire and develop an array of antibiotic resistance mechanisms. The sum of these resistance mechanisms, the antibiotic resistome, is a formidable threat to antibiotic discovery, development, and use. The study and understanding of the molecular mechanisms in the resistome provide the basis for traditional approaches to combat resistance, including semisynthetic modification of naturally occurring antibiotic scaffolds, the development of adjuvant therapies that overcome resistance mechanisms, and the total synthesis of new antibiotics and their analogues. Using two major classes of antibiotics, the aminoglycosides and tetracyclines as case studies, we review the success and limitations of these strategies when used to combat the many forms of resistance that have emerged toward natural product-based antibiotics specifically. Furthermore, we discuss the use of the resistome as a guide for the genomics-driven discovery of novel antimicrobials, which are essential to combat the growing number of emerging pathogens that are resistant to even the newest approved therapies.
Collapse
Affiliation(s)
- Christian Hobson
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Andrew N Chan
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Gerard D Wright
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
173
|
Isgren CM, Williams NJ, Fletcher OD, Timofte D, Newton RJ, Maddox TW, Clegg PD, Pinchbeck GL. Antimicrobial resistance in clinical bacterial isolates from horses in the UK. Equine Vet J 2021; 54:390-414. [PMID: 33566383 DOI: 10.1111/evj.13437] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Surveillance of antimicrobial resistance (AMR) in horses is important to aid empirical treatment decisions and highlight emerging AMR threats. OBJECTIVE To describe the AMR patterns of common groups of bacteria from clinical submissions from horses in the UK during 2018, and to determine how this varies by sample site and type of submitting veterinary practice. STUDY DESIGN Prospective observational study. METHODS All data on bacterial culture and subsequent antimicrobial susceptibility testing (AST) collected in 2018 from six large equine diagnostic laboratories were included. Resistance patterns were analysed including resistance to 1 or 2 antimicrobial classes, multidrug resistance (MDR), extensively drug resistant (XDR), resistance to highest priority critically important antimicrobials and isolates where there was no readily available treatment for adult horses in the UK. Submitting practices were classified according to whether they treated referral cases or not (first opinion). Comparisons between proportions and resistance for each bacterial group and sample site was performed using Chi-squared (or Fisher's exact test). RESULTS A total of 6,018 bacterial isolates from 4,038 diagnostic submissions were included from respiratory (n = 1555), urogenital (n = 1,010), skin/hair/wound/abscess (n = 753), surgical site infection (SSI) /catheter-related-infection (CRI) /orthopaedic infections (n = 347) and unknown/'other' submissions (n = 373). There were 2,711 Gram-negative isolates and 3,307 Gram-positive isolates. Prevalence of MDR for E. coli was 31.7%, Staphylococcus spp. 25.3% and > 25% for the majority of bacterial isolates from SSI/CRI/orthopaedic submissions. For Enterococcus spp. there was no readily available treatment for adult horses in the UK in 30.2% of positive submissions. MDR was significantly higher from referral hospital than first opinion submissions for the majority of pathogens (except Actinobacillus spp. and Pasteurella spp. and β-haemolytic Streptococcus spp.). MAIN LIMITATIONS Since culture and susceptibility results are not systematic analyses based on harmonised methods, selection bias could impact the findings. CONCLUSIONS Ongoing surveillance is essential to understand emerging patterns of resistance. MDR is high in SSI/CRI/orthopaedic infections, which is important for hospital biosecurity and guiding treatment decisions. Harmonisation of diagnostic procedures and interpretation of results amongst veterinary laboratories will improve AMR surveillance and data comparison among laboratories.
Collapse
Affiliation(s)
- Cajsa M Isgren
- Department of Equine Clinical Science, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, UK
| | - Nicola J Williams
- Department of Livestock and One Health, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, UK
| | - Owen D Fletcher
- Department of Equine Clinical Science, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, UK
| | - Dorina Timofte
- Department of Veterinary Anatomy, Physiology & Pathology, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, UK
| | | | - Thomas W Maddox
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, Cheshire, UK
| | - Peter D Clegg
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, Cheshire, UK
| | - Gina L Pinchbeck
- Department of Livestock and One Health, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health & Life Sciences, University of Liverpool, Neston, UK
| |
Collapse
|
174
|
Li Y, Xu Z, Han W, Cao H, Umarov R, Yan A, Fan M, Chen H, Duarte CM, Li L, Ho PL, Gao X. HMD-ARG: hierarchical multi-task deep learning for annotating antibiotic resistance genes. MICROBIOME 2021; 9:40. [PMID: 33557954 PMCID: PMC7871585 DOI: 10.1186/s40168-021-01002-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/08/2021] [Indexed: 05/07/2023]
Abstract
BACKGROUND The spread of antibiotic resistance has become one of the most urgent threats to global health, which is estimated to cause 700,000 deaths each year globally. Its surrogates, antibiotic resistance genes (ARGs), are highly transmittable between food, water, animal, and human to mitigate the efficacy of antibiotics. Accurately identifying ARGs is thus an indispensable step to understanding the ecology, and transmission of ARGs between environmental and human-associated reservoirs. Unfortunately, the previous computational methods for identifying ARGs are mostly based on sequence alignment, which cannot identify novel ARGs, and their applications are limited by currently incomplete knowledge about ARGs. RESULTS Here, we propose an end-to-end Hierarchical Multi-task Deep learning framework for ARG annotation (HMD-ARG). Taking raw sequence encoding as input, HMD-ARG can identify, without querying against existing sequence databases, multiple ARG properties simultaneously, including if the input protein sequence is an ARG, and if so, what antibiotic family it is resistant to, what resistant mechanism the ARG takes, and if the ARG is an intrinsic one or acquired one. In addition, if the predicted antibiotic family is beta-lactamase, HMD-ARG further predicts the subclass of beta-lactamase that the ARG is resistant to. Comprehensive experiments, including cross-fold validation, third-party dataset validation in human gut microbiota, wet-experimental functional validation, and structural investigation of predicted conserved sites, demonstrate not only the superior performance of our method over the state-of-art methods, but also the effectiveness and robustness of the proposed method. CONCLUSIONS We propose a hierarchical multi-task method, HMD-ARG, which is based on deep learning and can provide detailed annotations of ARGs from three important aspects: resistant antibiotic class, resistant mechanism, and gene mobility. We believe that HMD-ARG can serve as a powerful tool to identify antibiotic resistance genes and, therefore mitigate their global threat. Our method and the constructed database are available at http://www.cbrc.kaust.edu.sa/HMDARG/ . Video abstract (MP4 50984 kb).
Collapse
Affiliation(s)
- Yu Li
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
- Department of Computer Science and Engineering (CSE), The Chinese University of Hong Kong (CUHK), Hong Kong, People's Republic of China
| | - Zeling Xu
- School of Biological Sciences, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Wenkai Han
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Huiluo Cao
- Carol Yu Center for Infection and Department of Microbiology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Ramzan Umarov
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Aixin Yan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Huan Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, People's Republic of China
| | - Carlos M Duarte
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
- Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Pak-Leung Ho
- Carol Yu Center for Infection and Department of Microbiology, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia.
| |
Collapse
|
175
|
Hamida RS, Ali MA, Goda DA, Al-Zaban MI. Lethal Mechanisms of Nostoc-Synthesized Silver Nanoparticles Against Different Pathogenic Bacteria. Int J Nanomedicine 2020; 15:10499-10517. [PMID: 33402822 PMCID: PMC7778443 DOI: 10.2147/ijn.s289243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/05/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increasing antibiotic resistance and the emergence of multidrug-resistant (MDR) pathogens have led to the need to develop new therapeutic agents to tackle microbial infections. Nano-antibiotics are a novel generation of nanomaterials with significant antimicrobial activities that target bacterial defense systems including biofilm formation, membrane permeability, and virulence activity. PURPOSE In addition to AgNO3, the current study aimed to explore for first time the antibacterial potential of silver nanoparticles synthesized by Nostoc sp. Bahar_M (N-SNPs) and their killing mechanisms against Streptococcus mutans, methicillin-resistant Staphylococcus aureus, Escherichia coli, and Salmonella typhimurium. METHODS Potential mechanisms of action of both silver species against bacteria were systematically explored using agar well diffusion, enzyme (lactate dehydrogenase (LDH) and ATPase) and antioxidant (glutathione peroxidase and catalase) assays, and morphological examinations. qRT-PCR and SDS-PAGE were employed to investigate the effect of both treatments on mfD, flu, and hly gene expression and protein patterns, respectively. RESULTS N-SNPs exhibited greater biocidal activity than AgNO3 against the four tested bacteria. E. coli treated with N-SNPs showed significant surges in LDH levels, imbalances in other antioxidant and enzyme activities, and marked morphological changes, including cell membrane disruption and cytoplasmic dissolution. N-SNPs caused more significant upregulation of mfD expression and downregulation of both flu and hly expression and increased protein denaturation compared with AgNO3. CONCLUSION N-SNPs exhibited significant inhibitory potential against E. coli by direct interfering with bacterial cellular structures and/or enhancing oxidative stress, indicating their potential for use as an alternative antimicrobial agent. However, the potential of N-SNPs to be usable and biocompatible antibacterial drug will evaluate by their toxicity against normal cells.
Collapse
Affiliation(s)
- Reham Samir Hamida
- Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohamed Abdelaal Ali
- Biotechnology Unit, Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Doaa A Goda
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), Alexandria, Egypt
| | - Mayasar Ibrahim Al-Zaban
- Department of Biology, College of Science Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
176
|
Raw Cow Milk Bacterial Consortium as Bioindicator of Circulating Anti-Microbial Resistance (AMR). Animals (Basel) 2020; 10:ani10122378. [PMID: 33322611 PMCID: PMC7763537 DOI: 10.3390/ani10122378] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
The environment, including animals and animal products, is colonized by bacterial species that are typical and specific of every different ecological niche. Natural and human-related ecological pressure promotes the selection and expression of genes related to antimicrobial resistance (AMR). These genes might be present in a bacterial consortium but might not necessarily be expressed. Their expression could be induced by the presence of antimicrobial compounds that could originate from a given ecological niche or from human activity. In this work, we applied (meta)proteomics analysis of bacterial compartment of raw milk in order to obtain a method that provides a measurement of circulating AMR involved proteins and gathers information about the whole bacterial composition. Results from milk analysis revealed the presence of 29 proteins/proteoforms linked to AMR. The detection of mainly β-lactamases suggests the possibility of using the milk microbiome as a bioindicator for the investigation of AMR. Moreover, it was possible to achieve a culture-free qualitative and functional analysis of raw milk bacterial consortia.
Collapse
|
177
|
Cyanobacterial blooms contribute to the diversity of antibiotic-resistance genes in aquatic ecosystems. Commun Biol 2020; 3:737. [PMID: 33277584 PMCID: PMC7718256 DOI: 10.1038/s42003-020-01468-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022] Open
Abstract
Cyanobacterial blooms are a global ecological problem that directly threatens human health and crop safety. Cyanobacteria have toxic effects on aquatic microorganisms, which could drive the selection for resistance genes. The effect of cyanobacterial blooms on the dispersal and abundance of antibiotic-resistance genes (ARGs) of concern to human health remains poorly known. We herein investigated the effect of cyanobacterial blooms on ARG composition in Lake Taihu, China. The numbers and relative abundances of total ARGs increased obviously during a Planktothrix bloom. More pathogenic microorganisms were present during this bloom than during a Planktothrix bloom or during the non-bloom period. Microcosmic experiments using additional aquatic ecosystems (an urban river and Lake West) found that a coculture of Microcystis aeruginosa and Planktothrix agardhii increased the richness of the bacterial community, because its phycosphere provided a richer microniche for bacterial colonization and growth. Antibiotic-resistance bacteria were naturally in a rich position, successfully increasing the momentum for the emergence and spread of ARGs. These results demonstrate that cyanobacterial blooms are a crucial driver of ARG diffusion and enrichment in freshwater, thus providing a reference for the ecology and evolution of ARGs and ARBs and for better assessing and managing water quality.
Collapse
|
178
|
Chernova OA, Chernov VM, Mouzykantov AA, Baranova NB, Edelstein IA, Aminov RI. Antimicrobial drug resistance mechanisms among Mollicutes. Int J Antimicrob Agents 2020; 57:106253. [PMID: 33264670 DOI: 10.1016/j.ijantimicag.2020.106253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 07/08/2020] [Accepted: 11/22/2020] [Indexed: 12/11/2022]
Abstract
Representatives of the Mollicutes class are the smallest, wall-less bacteria capable of independent reproduction. They are widespread in nature, most are commensals, and some are pathogens of humans, animals and plants. They are also the main contaminants of cell cultures and vaccine preparations. Despite limited biosynthetic capabilities, they are highly adaptable and capable of surviving under various stress and extreme conditions, including antimicrobial selective pressure. This review describes current understanding of antibiotic resistance (ABR) mechanisms in Mollicutes. Protective mechanisms in these bacteria include point mutations, which may include non-target genes, and unique gene exchange mechanisms, contributing to transfer of ABR genes. Better understanding of the mechanisms of emergence and dissemination of ABR in Mollicutes is crucial to control these hypermutable bacteria and prevent the occurrence of highly ABR strains.
Collapse
Affiliation(s)
- Olga A Chernova
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Centre of RAS, Kazan, Russian Federation
| | - Vladislav M Chernov
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Centre of RAS, Kazan, Russian Federation
| | - Alexey A Mouzykantov
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Centre of RAS, Kazan, Russian Federation
| | - Natalya B Baranova
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Centre of RAS, Kazan, Russian Federation
| | - Inna A Edelstein
- Smolensk State Medical University, Ministry of Health of Russian Federation, Smolensk, Russian Federation
| | - Rustam I Aminov
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation.
| |
Collapse
|
179
|
Deciphering Streptococcal Biofilms. Microorganisms 2020; 8:microorganisms8111835. [PMID: 33233415 PMCID: PMC7700319 DOI: 10.3390/microorganisms8111835] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Streptococci are a diverse group of bacteria, which are mostly commensals but also cause a considerable proportion of life-threatening infections. They colonize many different host niches such as the oral cavity, the respiratory, gastrointestinal, and urogenital tract. While these host compartments impose different environmental conditions, many streptococci form biofilms on mucosal membranes facilitating their prolonged survival. In response to environmental conditions or stimuli, bacteria experience profound physiologic and metabolic changes during biofilm formation. While investigating bacterial cells under planktonic and biofilm conditions, various genes have been identified that are important for the initial step of biofilm formation. Expression patterns of these genes during the transition from planktonic to biofilm growth suggest a highly regulated and complex process. Biofilms as a bacterial survival strategy allow evasion of host immunity and protection against antibiotic therapy. However, the exact mechanisms by which biofilm-associated bacteria cause disease are poorly understood. Therefore, advanced molecular techniques are employed to identify gene(s) or protein(s) as targets for the development of antibiofilm therapeutic approaches. We review our current understanding of biofilm formation in different streptococci and how biofilm production may alter virulence-associated characteristics of these species. In addition, we have summarized the role of surface proteins especially pili proteins in biofilm formation. This review will provide an overview of strategies which may be exploited for developing novel approaches against biofilm-related streptococcal infections.
Collapse
|
180
|
Ogunrinu OJ, Norman KN, Vinasco J, Levent G, Lawhon SD, Fajt VR, Volkova VV, Gaire T, Poole TL, Genovese KJ, Wittum TE, Scott HM. Can the use of older-generation beta-lactam antibiotics in livestock production over-select for beta-lactamases of greatest consequence for human medicine? An in vitro experimental model. PLoS One 2020; 15:e0242195. [PMID: 33196662 PMCID: PMC7668573 DOI: 10.1371/journal.pone.0242195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022] Open
Abstract
Though carbapenems are not licensed for use in food animals in the U.S., carbapenem resistance among Enterobacteriaceae has been identified in farm animals and their environments. The objective of our study was to determine the extent to which older-generation β-lactam antibiotics approved for use in food animals in the U.S. might differentially select for resistance to antibiotics of critical importance to human health, such as carbapenems. Escherichia coli (E. coli) strains from humans, food animals, or the environment bearing a single β-lactamase gene (n = 20 each) for blaTEM-1, blaCMY-2, and blaCTX-M-* or else blaKPC/IMP/NDM (due to limited availability, often in combination with other bla genes), were identified, along with 20 E. coli strains lacking any known beta-lactamase genes. Baseline estimates of intrinsic bacterial fitness were derived from the population growth curves. Effects of ampicillin (32 μg/mL), ceftriaxone (4 μg/mL) and meropenem (4 μg/mL) on each strain and resistance-group also were assessed. Further, in vitro batch cultures were prepared by mixing equal concentrations of 10 representative E. coli strains (two from each resistance gene group), and each mixture was incubated at 37°C for 24 hours in non-antibiotic cation-adjusted Mueller-Hinton II (CAMH-2) broth, ampicillin + CAMH-2 broth (at 2, 4, 8, 16, and 32 μg/mL) and ceftiofur + CAMH-2 broth (at 0.5, 1, 2, 4, and 8μg/mL). Relative and absolute abundance of resistance-groups were estimated phenotypically. Line plots of the raw data were generated, and non-linear Gompertz models and multilevel mixed-effect linear regression models were fitted to the data. The observed strain growth rate distributions were significantly different across the groups. AmpC strains (i.e., blaCMY-2) had distinctly less robust (p < 0.05) growth in ceftriaxone (4 μg/mL) compared to extended-spectrum beta-lactamase (ESBL) producers harboring blaCTX-M-*variants. With increasing beta-lactam antibiotic concentrations, relative proportions of ESBLs and CREs were over-represented in the mixed bacterial communities; importantly, this was more pronounced with ceftiofur than with ampicillin. These results indicate that aminopenicillins and extended-spectrum cephalosporins would be expected to propagate carbapenemase-producing Enterobacteriaceae in food animals if and when Enterobacteriaceae from human health care settings enter the food animal environment.
Collapse
Affiliation(s)
- Olanrewaju J. Ogunrinu
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Keri N. Norman
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas, United States of America
| | - Javier Vinasco
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Gizem Levent
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Sara D. Lawhon
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Virginia R. Fajt
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, Texas, United States of America
| | - Victoria V. Volkova
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Tara Gaire
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Toni L. Poole
- Southern Plains Agricultural Research Center, United States Department of Agriculture, College Station, Texas, United States of America
| | - Kenneth J. Genovese
- Southern Plains Agricultural Research Center, United States Department of Agriculture, College Station, Texas, United States of America
| | - Thomas E. Wittum
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - H. Morgan Scott
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
181
|
Qiu H, Si Z, Luo Y, Feng P, Wu X, Hou W, Zhu Y, Chan-Park MB, Xu L, Huang D. The Mechanisms and the Applications of Antibacterial Polymers in Surface Modification on Medical Devices. Front Bioeng Biotechnol 2020; 8:910. [PMID: 33262975 PMCID: PMC7686044 DOI: 10.3389/fbioe.2020.00910] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/15/2020] [Indexed: 01/04/2023] Open
Abstract
Medical device contamination caused by microbial pathogens such as bacteria and fungi has posed a severe threat to the patients' health in hospitals. Due to the increasing resistance of pathogens to antibiotics, the efficacy of traditional antibiotics treatment is gradually decreasing for the infection treatment. Therefore, it is urgent to develop new antibacterial drugs to meet clinical or civilian needs. Antibacterial polymers have attracted the interests of researchers due to their unique bactericidal mechanism and excellent antibacterial effect. This article reviews the mechanism and advantages of antimicrobial polymers and the consideration for their translation. Their applications and advances in medical device surface coating were also reviewed. The information will provide a valuable reference to design and develop antibacterial devices that are resistant to pathogenic infections.
Collapse
Affiliation(s)
- Haofeng Qiu
- School of Medicine, Ningbo University, Ningbo, China
| | - Zhangyong Si
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Yang Luo
- School of Medicine, Ningbo University, Ningbo, China
| | - Peipei Feng
- School of Medicine, Ningbo University, Ningbo, China
| | - Xujin Wu
- School of Medicine, Ningbo University, Ningbo, China
| | - Wenjia Hou
- School of Medicine, Ningbo University, Ningbo, China
| | - Yabin Zhu
- School of Medicine, Ningbo University, Ningbo, China
| | - Mary B. Chan-Park
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Long Xu
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Dongmei Huang
- Ningbo Baoting Biotechnology Co., Ltd., Ningbo, China
| |
Collapse
|
182
|
The Household Resistome: Frequency of β-Lactamases, Class 1 Integrons, and Antibiotic-Resistant Bacteria in the Domestic Environment and Their Reduction during Automated Dishwashing and Laundering. Appl Environ Microbiol 2020; 86:AEM.02062-20. [PMID: 32978137 PMCID: PMC7657631 DOI: 10.1128/aem.02062-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/20/2020] [Indexed: 11/20/2022] Open
Abstract
Households provide a habitat for bacteria originating from humans, animals, foods, contaminated clothes, or other sources. Thus, bacteria carrying antibiotic resistance genes (ARGs) may be introduced via household members, animals, or the water supply from external habitats into private households and vice versa. Since data on antibiotic resistance (ABR) in the domestic environment are limited, this study aimed to determine the abundance of β-lactamase, mobile colistin resistance, and class 1 integron genes and the correlation of their presence and to characterize phenotypically resistant strains in 54 private households in Germany. Additionally, the persistence of antibiotic-resistant bacteria during automated dishwashing compared to that during laundering was assessed. Shower drains, washing machines, and dishwashers were sampled and analyzed using quantitative real-time PCR. Resistant strains were isolated, followed by identification and antibiotic susceptibility testing using a Vitek 2 system. The results showed a significantly higher relative ARG abundance of 0.2367 ARG copies/16S rRNA gene copies in shower drains than in dishwashers (0.1329 ARG copies/16S rRNA gene copies) and washing machines (0.0006 ARG copies/16S rRNA gene copies). bla CMY-2, bla ACT/MIR, and bla OXA-48 were the most prevalent ARG, and intI1 occurred in 96.3% of the households, while no mcr genes were detected. Several β-lactamase genes co-occurred, and the resistance of bacterial isolates correlated positively with genotypic resistance, with carbapenemase genes dominating across isolates. Antibiotic-resistant bacteria were significantly reduced during automated dishwashing as well as laundering tests and did not differ from susceptible strains. Overall, the domestic environment may represent a potential reservoir of β-lactamase genes and β-lactam-resistant bacteria, with shower drains being the dominant source of ABR.IMPORTANCE The abundance of antibiotic-resistant bacteria and ARGs is steadily increasing and has been comprehensively analyzed in natural environments, animals, foods, and wastewater treatment plants. In this respect, β-lactams and colistin are of particular interest due to the emergence of multidrug-resistant Gram-negative bacteria. Despite the connection of private households to these environments, only a few studies have focused on the domestic environment so far. Therefore, the present study further investigated the occurrence of ARGs and antibiotic-resistant bacteria in shower drains, washing machines, and dishwashers. The analysis of the domestic environment as a potential reservoir of resistant bacteria is crucial to determine whether households contribute to the spread of ABR or may be a habitat where resistant bacteria from the natural environment, humans, food, or water are selected due to the use of detergents, antimicrobial products, and antibiotics. Furthermore, ABR could limit the options for the treatment of infections arising in the domestic environment.
Collapse
|
183
|
Wallace MJ, Fishbein SRS, Dantas G. Antimicrobial resistance in enteric bacteria: current state and next-generation solutions. Gut Microbes 2020; 12:1799654. [PMID: 32772817 PMCID: PMC7524338 DOI: 10.1080/19490976.2020.1799654] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Antimicrobial resistance is one of the largest threats to global health and imposes substantial burdens in terms of morbidity, mortality, and economic costs. The gut is a key conduit for the genesis and spread of antimicrobial resistance in enteric bacterial pathogens. Distinct bacterial species that cause enteric disease can exist as invasive enteropathogens that immediately evoke gastrointestinal distress, or pathobionts that can arise from established bacterial commensals to inflict dysbiosis and disease. Furthermore, various environmental reservoirs and stressors facilitate the evolution and transmission of resistance. In this review, we present a comprehensive discussion on circulating resistance profiles and gene mobilization strategies of the most problematic species of enteric bacterial pathogens. Importantly, we present emerging approaches toward surveillance of pathogens and their resistance elements as well as promising treatment strategies that can circumvent common resistance mechanisms.
Collapse
Affiliation(s)
- M. J. Wallace
- Department of Pathology & Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - S. R. S. Fishbein
- Department of Pathology & Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - G. Dantas
- Department of Pathology & Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA,The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA,Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA,CONTACT G. Dantas Department of Pathology & Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| |
Collapse
|
184
|
Qiao J, Purro M, Liu Z, Xiong MP. Effects of Polyethyelene Glycol-Desferrioxamine:Gallium Conjugates on Pseudomonas aeruginosa Outer Membrane Permeability and Vancomycin Potentiation. Mol Pharm 2020; 18:735-742. [PMID: 33147036 DOI: 10.1021/acs.molpharmaceut.0c00820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pseudomonas aeruginosa exhibits a broad spectrum of intrinsic antibiotic resistance because of the limited permeability of its outer membrane. Given this situation, molecules that could make Gram-negative bacteria more permeable and more susceptible to large-scaffold Gram-positive antibiotics may be advantageous. Herein, we evaluate the antimicrobial activity of a series of targeted poly(ethylene glycol)-desferrioxamine/gallium (PEG-DG) conjugates that can improve the sensitivity of P. aeruginosa to the glycopeptide vancomycin (VAN). We observed that single-ended mPEG-DG and double-ended PEG-DG2 conjugates characterized by PEG MW ≥2000 synergistically enhanced the sensitivity of VAN against P. aeruginosa reference strains PAO1 and ATCC 27853 and three clinically isolated carbapenem-resistant strains, but not Escherichia coli strain ATCC 25922. Although the exact mechanism of this phenomenon is currently under investigation, PEG-DG conjugates enhanced nitrocefin (NCF), hexidium iodide (HI), and VAN permeability only when PEG and DG were directly conjugated. The two most important physicochemical factors contributing to the synergistic activity observed with VAN relate to (1) the final concentration of DG ligands conjugated to the polymer and (2) the polymer length, wherein MW ≥2000 yielded a similar fractional inhibitory concentration.
Collapse
Affiliation(s)
- Jing Qiao
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Max Purro
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Zhi Liu
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - May P Xiong
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
185
|
Gonçalves J, Luís Â, Gradillas A, García A, Restolho J, Fernández N, Domingues F, Gallardo E, Duarte AP. Ayahuasca Beverages: Phytochemical Analysis and Biological Properties. Antibiotics (Basel) 2020; 9:antibiotics9110731. [PMID: 33114334 PMCID: PMC7690887 DOI: 10.3390/antibiotics9110731] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
Ayahuasca is a psychoactive beverage, originally consumed by indigenous Amazon tribes, of which consumption has been increasing worldwide. The aim of this study was to evaluate the phytochemical profile, as well as the antioxidant, anti-inflammatory and antimicrobial properties of decoctions of four individual plants, a commercial mixture and four mixtures of two individual plants used in the Ayahuasca preparation. For this purpose, a phytochemical characterization was performed, determining the content of flavonoids, total phenolic compounds, and analyzing the phenolic profile. Besides, 48 secondary metabolites were investigated by ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q/TOF-MS) and their concentration estimated with real standards when present. The antioxidant activity was evaluated by both the β-carotene bleaching test and DPPH free radical scavenging assay, and the anti-inflammatory activity was determined by a protein denaturation method. Finally, the antimicrobial properties were evaluated using the disc diffusion assay, resazurin microtiter method, anti-quorum sensing and anti-biofilm activity assays. The obtained results showed that, in general, the samples have a high content of phenolic compounds and flavonoids with noticeable differences, reflecting on remarkable antioxidant and anti-inflammatory activities. Significant antimicrobial properties were also observed, with emphasis on the effect of B. caapi and P. harmala on planktonic and biofilm cells of A. baumannii, inhibiting both the biofilm formation and the production of violacein pigment.
Collapse
Affiliation(s)
- Joana Gonçalves
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.G.); (J.R.); (F.D.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBI Medical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
| | - Ângelo Luís
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.G.); (J.R.); (F.D.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBI Medical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
- Correspondence: (Â.L.); (E.G.); Tel.: +351-275-329-002/3 (Â.L.); +351-275-329-002/3 (E.G.)
| | - Ana Gradillas
- CEMBIO, Center for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain; (A.G.); (A.G.)
| | - Antonia García
- CEMBIO, Center for Metabolomics and Bioanalysis, Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain; (A.G.); (A.G.)
| | - José Restolho
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.G.); (J.R.); (F.D.); (A.P.D.)
| | - Nicolás Fernández
- Cátedra de Toxicología y Química Legal, Laboratorio de Asesoramiento Toxicológico Analítico (CENATOXA), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113AAD Buenos Aires, Argentina;
| | - Fernanda Domingues
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.G.); (J.R.); (F.D.); (A.P.D.)
| | - Eugenia Gallardo
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.G.); (J.R.); (F.D.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBI Medical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
- Correspondence: (Â.L.); (E.G.); Tel.: +351-275-329-002/3 (Â.L.); +351-275-329-002/3 (E.G.)
| | - Ana Paula Duarte
- Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (J.G.); (J.R.); (F.D.); (A.P.D.)
- Laboratório de Fármaco-Toxicologia, UBI Medical, Universidade da Beira Interior, Estrada Municipal 506, 6200-284 Covilhã, Portugal
| |
Collapse
|
186
|
Insights into bacterial cell division from a structure of EnvC bound to the FtsX periplasmic domain. Proc Natl Acad Sci U S A 2020; 117:28355-28365. [PMID: 33097670 PMCID: PMC7668044 DOI: 10.1073/pnas.2017134117] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The peptidoglycan layer is a core component of the bacterial cell envelope that provides a barrier to the environment and protection from osmotic shock. During division, bacteria must break and rebuild the peptidoglycan layer to enable separation of daughter cells. In E. coli, two of the three amidases responsible (AmiA and AmiB) are regulated by a single periplasmic activator (EnvC) that is, itself, controlled by an atypical ABC transporter (FtsEX) tethered to the cytoplasmic septal Z-ring. Here we define the structural basis for the interaction of FtsEX with EnvC and suggest a molecular mechanism for amidase activation where EnvC autoinhibition is relieved by ATP-driven conformational changes transmitted through the FtsEX-EnvC complex. FtsEX is a bacterial ABC transporter that regulates the activity of periplasmic peptidoglycan amidases via its interaction with the murein hydrolase activator, EnvC. In Escherichia coli, FtsEX is required to separate daughter cells after cell division and for viability in low-osmolarity media. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for amidase activation, but the process itself is poorly understood. Here we present the 2.1 Å structure of the FtsX periplasmic domain in complex with its periplasmic partner, EnvC. The EnvC-FtsX periplasmic domain complex has a 1-to-2 stoichiometry with two distinct FtsX-binding sites located within an antiparallel coiled coil domain of EnvC. Residues involved in amidase activation map to a previously identified groove in the EnvC LytM domain that is here found to be occluded by a “restraining arm” suggesting a self-inhibition mechanism. Mutational analysis, combined with bacterial two-hybrid screens and in vivo functional assays, verifies the FtsEX residues required for EnvC binding and experimentally test a proposed mechanism for amidase activation. We also define a predicted link between FtsEX and integrity of the outer membrane. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for resistance to membrane-attacking antibiotics and detergents to which E. coli would usually be considered intrinsically resistant. These structural and functional data provide compelling mechanistic insight into FtsEX-mediated regulation of EnvC and its downstream control of periplasmic peptidoglycan amidases.
Collapse
|
187
|
Teelucksingh T, Thompson LK, Cox G. The Evolutionary Conservation of Escherichia coli Drug Efflux Pumps Supports Physiological Functions. J Bacteriol 2020; 202:e00367-20. [PMID: 32839176 PMCID: PMC7585057 DOI: 10.1128/jb.00367-20] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bacteria harness an impressive repertoire of resistance mechanisms to evade the inhibitory action of antibiotics. One such mechanism involves efflux pump-mediated extrusion of drugs from the bacterial cell, which significantly contributes to multidrug resistance. Intriguingly, most drug efflux pumps are chromosomally encoded components of the intrinsic antibiotic resistome. In addition, in terms of xenobiotic detoxification, bacterial efflux systems often exhibit significant levels of functional redundancy. Efflux pumps are also considered to be highly conserved; however, the extent of conservation in many bacterial species has not been reported and the majority of genes that encode efflux pumps appear to be dispensable for growth. These observations, in combination with an increasing body of experimental evidence, imply alternative roles in bacterial physiology. Indeed, the ability of efflux pumps to facilitate antibiotic resistance could be a fortuitous by-product of ancient physiological functions. Using Escherichia coli as a model organism, we here evaluated the evolutionary conservation of drug efflux pumps and we provide phylogenetic analysis of the major efflux families. We show the E. coli drug efflux system has remained relatively stable and the majority (∼80%) of pumps are encoded in the core genome. This analysis further supports the importance of drug efflux pumps in E. coli physiology. In this review, we also provide an update on the roles of drug efflux pumps in the detoxification of endogenously synthesized substrates and pH homeostasis. Overall, gaining insight into drug efflux pump conservation, common evolutionary ancestors, and physiological functions could enable strategies to combat these intrinsic and ancient elements.
Collapse
Affiliation(s)
- Tanisha Teelucksingh
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Laura K Thompson
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Georgina Cox
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
188
|
Kongkham B, Prabakaran D, Puttaswamy H. Opportunities and challenges in managing antibiotic resistance in bacteria using plant secondary metabolites. Fitoterapia 2020; 147:104762. [PMID: 33069839 DOI: 10.1016/j.fitote.2020.104762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022]
Abstract
Development of antibiotic resistance (ABR) in bacteria and its multidimensional spread is an emerging global threat that needs immediate attention. Extensive antibiotics (AB) usage results in development of ABR in bacteria by target modification, production of AB degrading enzymes, porin modifications, efflux pumps overexpression, etc. To counter this, apart from strict regulation of AB use and behavioural changes, research and development (R&D) of newer antimicrobials are in place. One such emerging approach to combat ABR is the use of structurally and functionally diverse plant secondary metabolites (PSMs) in combination with the conventional AB. Either the PSMs are themselves antimicrobial or they potentiate the activity of the AB through a range of mechanisms. However, their use is lagging due to poor knowledge of mode of action, structure-activity relationships, pharmacokinetics, etc. This review paper discussed the opportunities and challenges in managing ABR using PSMs. Mechanisms of ABR development in bacteria and current strategies to counter them were studied and the areas where PSMs can play an important role were highlighted. The use of PSMs, both as an anti-resistance and anti-virulence agent in combination therapy to counter multi-drug resistance along with their mechanisms of action, has been discussed in detail. The difficulties in the commercialisation of PSMs and strategies to overcome them along with future priority areas of research have also been given. Following the given R&D path will definitely help in better understanding and utilising the full potential of PSMs in solving the problem of antimicrobial resistance (AMR).
Collapse
Affiliation(s)
- Bhani Kongkham
- Environmental Biotechnology Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Delhi 110016, India
| | - Duraivadivel Prabakaran
- Environmental Biotechnology Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Delhi 110016, India
| | - Hariprasad Puttaswamy
- Environmental Biotechnology Laboratory, Centre for Rural Development and Technology, Indian Institute of Technology Delhi, Delhi 110016, India.
| |
Collapse
|
189
|
Triggiano F, Calia C, Diella G, Montagna MT, De Giglio O, Caggiano G. The Role of Urban Wastewater in the Environmental Transmission of Antimicrobial Resistance: The Current Situation in Italy (2010-2019). Microorganisms 2020; 8:E1567. [PMID: 33053645 PMCID: PMC7600224 DOI: 10.3390/microorganisms8101567] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 12/12/2022] Open
Abstract
Scientific studies show that urban wastewater treatment plants (UWWTP) are among the main sources of release of antibiotics, antibiotic resistance genes (ARG) and antibiotic-resistant bacteria (ARB) into the environment, representing a risk to human health. This review summarizes selected publications from 1 January 2010 to 31 December 2019, with particular attention to the presence and treatment of ARG and ARB in UWWTPs in Italy. Following a brief introduction, the review is divided into three sections: (i) phenotypic assessment (ARB) and (ii) genotypic assessment (ARG) of resistant microorganisms, and (iii) wastewater treatment processes. Each article was read entirely to extract the year of publication, the geographical area of the UWWTP, the ARB and ARG found, and the type of disinfection treatment used. Among the ARB, we focused on the antibiotic resistance of Escherichia coli, Klebsiella pneumoniae, and Enterococci in UWWTP. The results show that the information presented in the literature to date is not exhaustive; therefore, future scientific studies at the national level are needed to better understand the spread of ARB and ARG, and also to develop new treatment methods to reduce this spread.
Collapse
Affiliation(s)
| | | | | | | | - Osvalda De Giglio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Piazza G. Cesare 11, 70124 Bari, Italy; (F.T.); (C.C.); (G.D.); (M.T.M.); (G.C.)
| | | |
Collapse
|
190
|
Memar MY, Yekani M, Celenza G, Poortahmasebi V, Naghili B, Bellio P, Baghi HB. The central role of the SOS DNA repair system in antibiotics resistance: A new target for a new infectious treatment strategy. Life Sci 2020; 262:118562. [PMID: 33038378 DOI: 10.1016/j.lfs.2020.118562] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/15/2020] [Accepted: 10/01/2020] [Indexed: 01/19/2023]
Abstract
Bacteria have a considerable ability and potential to acquire resistance against antimicrobial agents by acting diverse mechanisms such as target modification or overexpression, multidrug transporter systems, and acquisition of drug hydrolyzing enzymes. Studying the mechanisms of bacterial cell physiology is mandatory for the development of novel strategies to control the antimicrobial resistance phenomenon, as well as for the control of infections in clinics. The SOS response is a cellular DNA repair mechanism that has an essential role in the bacterial biologic process involved in resistance to antibiotics. The activation of the SOS network increases the resistance and tolerance of bacteria to stress and, as a consequence, to antimicrobial agents. Therefore, SOS can be an applicable target for the discovery of new antimicrobial drugs. In the present review, we focus on the central role of SOS response in bacterial resistance mechanisms and its potential as a new target for control of resistant pathogens.
Collapse
Affiliation(s)
- Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Giuseppe Celenza
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Vahdat Poortahmasebi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrooz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pierangelo Bellio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
191
|
Alves-Barroco C, Rivas-García L, Fernandes AR, Baptista PV. Tackling Multidrug Resistance in Streptococci - From Novel Biotherapeutic Strategies to Nanomedicines. Front Microbiol 2020; 11:579916. [PMID: 33123110 PMCID: PMC7573253 DOI: 10.3389/fmicb.2020.579916] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
The pyogenic streptococci group includes pathogenic species for humans and other animals and has been associated with enduring morbidity and high mortality. The main reason for the treatment failure of streptococcal infections is the increased resistance to antibiotics. In recent years, infectious diseases caused by pyogenic streptococci resistant to multiple antibiotics have been raising with a significant impact to public health and veterinary industry. The rise of antibiotic-resistant streptococci has been associated to diverse mechanisms, such as efflux pumps and modifications of the antimicrobial target. Among streptococci, antibiotic resistance emerges from previously sensitive populations as result of horizontal gene transfer or chromosomal point mutations due to excessive use of antimicrobials. Streptococci strains are also recognized as biofilm producers. The increased resistance of biofilms to antibiotics among streptococci promote persistent infection, which comprise circa 80% of microbial infections in humans. Therefore, to overcome drug resistance, new strategies, including new antibacterial and antibiofilm agents, have been studied. Interestingly, the use of systems based on nanoparticles have been applied to tackle infection and reduce the emergence of drug resistance. Herein, we present a synopsis of mechanisms associated to drug resistance in (pyogenic) streptococci and discuss some innovative strategies as alternative to conventional antibiotics, such as bacteriocins, bacteriophage, and phage lysins, and metal nanoparticles. We shall provide focused discussion on the advantages and limitations of agents considering application, efficacy and safety in the context of impact to the host and evolution of bacterial resistance.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Lorenzo Rivas-García
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal.,Biomedical Research Centre, University of Granada, Granada, Spain
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro Viana Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
192
|
Shanmugakani RK, Sugawara Y, Akeda Y, Hagiya H, Sakamoto N, Aye MM, Myint T, Hamada S, Tomono K. bla OXA -731 , a new chromosome-encoded bla OXA -48 -like variant in Shewanella sp. from the aquatic environment in Myanmar. ENVIRONMENTAL MICROBIOLOGY REPORTS 2020; 12:548-554. [PMID: 32776437 DOI: 10.1111/1758-2229.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/06/2020] [Indexed: 06/11/2023]
Abstract
Shewanella sp., the progenitors of blaOXA-48 -like genes are increasingly reported with the possession of different blaOXA-48 -like variants. This study aims to characterize blaOXA-731 , a new variant of a blaOXA-48 -like gene identified in Shewanella sp. isolated from the aquatic environment in Myanmar. Phylogenetic analysis of the blaOXA-731 sequence with other blaOXA-48 -like variants showed that it has the highest nucleotide identity of 86.09% with blaOXA-48 . However, the active site motifs in OXA-731 were 100% identical to that in OXA-48. Whole-genome sequencing analysis showed that blaOXA-731 is not surrounded by any mobile genetic elements. The genetic context of blaOXA-731 was found as similar to other blaOXA-48 -like genes previously identified in Shewanella sp. S1 nuclease pulsed-field gel electrophoresis followed by Southern blotting confirmed the location of blaOXA-731 in the chromosome of the Shewanella genome. Cloning and expression studies showed that OXA-731 has β-lactamase activity similar to OXA-48 and OXA-181, but it has no significant carbapenemase activity. Our results showed the significance of blaOXA-48 -like-carrying Shewanella sp. in the spreading of blaOXA-48 -like genes in the community.
Collapse
Affiliation(s)
- Rathina Kumar Shanmugakani
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
- Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, 565-0871, Japan
| | - Yo Sugawara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yukihiro Akeda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
- Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, 565-0871, Japan
| | - Hideharu Hagiya
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
- Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, 565-0871, Japan
| | - Noriko Sakamoto
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Mya Mya Aye
- Bacteriology Research Division, Department of Medical Research, Dagon Township, Yangon, 11191, Myanmar
| | - Thuzar Myint
- Bacteriology Research Division, Department of Medical Research, Dagon Township, Yangon, 11191, Myanmar
| | - Shigeyuki Hamada
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazunori Tomono
- Department of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
- Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
193
|
Do 1,8-naphthyridine sulfonamides possess an inhibitory action against Tet(K) and MsrA efflux pumps in multiresistant Staphylococcus aureus strains? Microb Pathog 2020; 147:104268. [DOI: 10.1016/j.micpath.2020.104268] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/14/2020] [Indexed: 11/17/2022]
|
194
|
Abstract
Antimicrobial resistance is developing rapidly and threatens to outstrip the rate at which new antimicrobials are introduced. Genetic recombination allows bacteria to rapidly disseminate genes encoding for antimicrobial resistance within and across species. Antimicrobial use creates a selective evolutionary pressure, which leads to further resistance. Antimicrobial stewardship, best use, and infection prevention are the most effective ways to slow the spread and development of antimicrobial resistance.
Collapse
Affiliation(s)
- Lindsay Morrison
- Division of Infectious Disease, McGaw Medical Center, Northwestern University Feinberg School of Medicine, 645 North Michigan Avenue, Suite 900, Chicago, IL 60611, USA.
| | - Teresa R Zembower
- Division of Infectious Disease, McGaw Medical Center, Northwestern University Feinberg School of Medicine, 645 North Michigan Avenue, Suite 900, Chicago, IL 60611, USA
| |
Collapse
|
195
|
Galindo-Méndez M. Antimicrobial Resistance in Escherichia coli. E. COLI INFECTIONS - IMPORTANCE OF EARLY DIAGNOSIS AND EFFICIENT TREATMENT 2020. [DOI: 10.5772/intechopen.93115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
196
|
Garzón V, Bustos RH, G. Pinacho D. Personalized Medicine for Antibiotics: The Role of Nanobiosensors in Therapeutic Drug Monitoring. J Pers Med 2020; 10:E147. [PMID: 32993004 PMCID: PMC7712907 DOI: 10.3390/jpm10040147] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 01/01/2023] Open
Abstract
Due to the high bacterial resistance to antibiotics (AB), it has become necessary to adjust the dose aimed at personalized medicine by means of therapeutic drug monitoring (TDM). TDM is a fundamental tool for measuring the concentration of drugs that have a limited or highly toxic dose in different body fluids, such as blood, plasma, serum, and urine, among others. Using different techniques that allow for the pharmacokinetic (PK) and pharmacodynamic (PD) analysis of the drug, TDM can reduce the risks inherent in treatment. Among these techniques, nanotechnology focused on biosensors, which are relevant due to their versatility, sensitivity, specificity, and low cost. They provide results in real time, using an element for biological recognition coupled to a signal transducer. This review describes recent advances in the quantification of AB using biosensors with a focus on TDM as a fundamental aspect of personalized medicine.
Collapse
Affiliation(s)
- Vivian Garzón
- PhD Biosciences Program, Universidad de La Sabana, Chía 140013, Colombia;
| | - Rosa-Helena Bustos
- Therapeutical Evidence Group, Clinical Pharmacology, Universidad de La Sabana, Chía 140013, Colombia;
| | - Daniel G. Pinacho
- Therapeutical Evidence Group, Clinical Pharmacology, Universidad de La Sabana, Chía 140013, Colombia;
| |
Collapse
|
197
|
Nadeem SF, Gohar UF, Tahir SF, Mukhtar H, Pornpukdeewattana S, Nukthamna P, Moula Ali AM, Bavisetty SCB, Massa S. Antimicrobial resistance: more than 70 years of war between humans and bacteria. Crit Rev Microbiol 2020; 46:578-599. [PMID: 32954887 DOI: 10.1080/1040841x.2020.1813687] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Development of antibiotic resistance in bacteria is one of the major issues in the present world and one of the greatest threats faced by mankind. Resistance is spread through both vertical gene transfer (parent to offspring) as well as by horizontal gene transfer like transformation, transduction and conjugation. The main mechanisms of resistance are limiting uptake of a drug, modification of a drug target, inactivation of a drug, and active efflux of a drug. The highest quantities of antibiotic concentrations are usually found in areas with strong anthropogenic pressures, for example medical source (e.g., hospitals) effluents, pharmaceutical industries, wastewater influents, soils treated with manure, animal husbandry and aquaculture (where antibiotics are generally used as in-feed preparations). Hence, the strong selective pressure applied by antimicrobial use has forced microorganisms to evolve for survival. The guts of animals and humans, wastewater treatment plants, hospital and community effluents, animal husbandry and aquaculture runoffs have been designated as "hotspots for AMR genes" because the high density of bacteria, phages, and plasmids in these settings allows significant genetic exchange and recombination. Evidence from the literature suggests that the knowledge of antibiotic resistance in the population is still scarce. Tackling antimicrobial resistance requires a wide range of strategies, for example, more research in antibiotic production, the need of educating patients and the general public, as well as developing alternatives to antibiotics (briefly discussed in the conclusions of this article).
Collapse
Affiliation(s)
- Syeda Fatima Nadeem
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Umar Farooq Gohar
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Syed Fahad Tahir
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | - Hamid Mukhtar
- Institute of Industrial Biotechnology, Government College University, Lahore, Pakistan
| | | | - Pikunthong Nukthamna
- Faculty of Food Industry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand.,College of Research Methodology and Cognitive Science, Burapha University, Chonburi, Thailand
| | - Ali Muhammed Moula Ali
- Faculty of Food Industry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | | | - Salvatore Massa
- Faculty of Food Industry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand.,Department of Agricultural, Food and Environmental Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
198
|
Gudda FO, Waigi MG, Odinga ES, Yang B, Carter L, Gao Y. Antibiotic-contaminated wastewater irrigated vegetables pose resistance selection risks to the gut microbiome. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 264:114752. [PMID: 32417582 DOI: 10.1016/j.envpol.2020.114752] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 06/11/2023]
Abstract
Wastewater reuse in food crop irrigation has led to agroecosystem pollution concerns and human health risks. However, there is limited attention on the relationship of sub-lethal antibiotic levels in vegetables and resistance selection. Most risk assessment studies show non-significant toxicity, but overlook the link between antibiotics in crops and propagation of gut microbiome resistance selection. The review highlights the risk of antibiotics in treated water used for irrigation, uptake, and accumulation in edible vegetable parts. Moreover, it elucidates the risks to the adaptive resistance selection of the gut microbiome from sub-lethal antibiotic levels, as a result of dietary contaminated vegetables. Experiments have reported that bacterial resistance selection is possible at concentrations that are several hundred-folds lower than lethal effect levels on susceptible cells. Consequently, mutants selected at low antibiotic levels, such as those from vegetables, are fitter and more resistant compared to those selected at high concentrations. Necessary standardization, such as the development of minimum acceptable antibiotic limits allowable in food crop irrigation water, with a focus on minimum selection concentration, and not only toxicity, has been proposed. Wastewater irrigation offers environmental benefits and can contribute to food security, but it has non-addressed risks. Research gaps, future perspectives, and frameworks of mitigating the potential risks are discussed.
Collapse
Affiliation(s)
- Fredrick Owino Gudda
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China; Faculty of Environment and Resource Development, Department of Environmental Sciences, Egerton University, Box 536, Egerton, 20115, Kenya
| | - Michael Gatheru Waigi
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Emmanuel Stephen Odinga
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Bing Yang
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Laura Carter
- School of Geography, University of Leeds, Leeds, LS2 9JT, UK
| | - Yanzheng Gao
- Institute of Organic Contaminant Control and Soil Remediation, College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
199
|
Taitt CR, Leski TA, Prouty MG, Ford GW, Heang V, House BL, Levin SY, Curry JA, Mansour A, Mohammady HE, Wasfy M, Tilley DH, Gregory MJ, Kasper MR, Regeimbal J, Rios P, Pimentel G, Danboise BA, Hulseberg CE, Odundo EA, Ombogo AN, Cheruiyot EK, Philip CO, Vora GJ. Tracking Antimicrobial Resistance Determinants in Diarrheal Pathogens: A Cross-Institutional Pilot Study. Int J Mol Sci 2020; 21:ijms21165928. [PMID: 32824772 PMCID: PMC7460656 DOI: 10.3390/ijms21165928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Infectious diarrhea affects over four billion individuals annually and causes over a million deaths each year. Though not typically prescribed for treatment of uncomplicated diarrheal disease, antimicrobials serve as a critical part of the armamentarium used to treat severe or persistent cases. Due to widespread over- and misuse of antimicrobials, there has been an alarming increase in global resistance, for which a standardized methodology for geographic surveillance would be highly beneficial. To demonstrate that a standardized methodology could be used to provide molecular surveillance of antimicrobial resistance (AMR) genes, we initiated a pilot study to test 130 diarrheal pathogens (Campylobacter spp., Escherichia coli, Salmonella, and Shigella spp.) from the USA, Peru, Egypt, Cambodia, and Kenya for the presence/absence of over 200 AMR determinants. We detected a total of 55 different determinants conferring resistance to ten different categories of antimicrobials: genes detected in ≥ 25 samples included blaTEM, tet(A), tet(B), mac(A), mac(B), aadA1/A2, strA, strB, sul1, sul2, qacEΔ1, cmr, and dfrA1. The number of determinants per strain ranged from none (several Campylobacter spp. strains) to sixteen, with isolates from Egypt harboring a wider variety and greater number of genes per isolate than other sites. Two samples harbored carbapenemase genes, blaOXA-48 or blaNDM. Genes conferring resistance to azithromycin (ere(A), mph(A)/mph(K), erm(B)), a first-line therapeutic for severe diarrhea, were detected in over 10% of all Enterobacteriaceae tested: these included >25% of the Enterobacteriaceae from Egypt and Kenya. Forty-six percent of the Egyptian Enterobacteriaceae harbored genes encoding CTX-M-1 or CTX-M-9 families of extended-spectrum β-lactamases. Overall, the data provide cross-comparable resistome information to establish regional trends in support of international surveillance activities and potentially guide geospatially informed medical care.
Collapse
Affiliation(s)
- Chris R. Taitt
- US Naval Research Laboratory, Center for Biomolecular Science & Engineering, Washington, DC 20375, USA; (T.A.L.); (G.J.V.)
- Correspondence: ; Tel.: +1-011-202-404-4208
| | - Tomasz A. Leski
- US Naval Research Laboratory, Center for Biomolecular Science & Engineering, Washington, DC 20375, USA; (T.A.L.); (G.J.V.)
| | - Michael G. Prouty
- US Naval Medical Research Unit No. 2-Phnom Penh, Blvd Kim Il Sung, Khan Toul Kork, Phnom Penh, Cambodia; (M.G.P.); (G.W.F.); (V.H.)
| | - Gavin W. Ford
- US Naval Medical Research Unit No. 2-Phnom Penh, Blvd Kim Il Sung, Khan Toul Kork, Phnom Penh, Cambodia; (M.G.P.); (G.W.F.); (V.H.)
| | - Vireak Heang
- US Naval Medical Research Unit No. 2-Phnom Penh, Blvd Kim Il Sung, Khan Toul Kork, Phnom Penh, Cambodia; (M.G.P.); (G.W.F.); (V.H.)
| | - Brent L. House
- US Naval Medical Research Unit No. 3, Naval Air Station Sigonella, 95030 Sigonella, Italy; (B.L.H.); (S.Y.L.); (J.A.C.); (A.M.); (H.E.M.); (M.W.)
| | - Samuel Y. Levin
- US Naval Medical Research Unit No. 3, Naval Air Station Sigonella, 95030 Sigonella, Italy; (B.L.H.); (S.Y.L.); (J.A.C.); (A.M.); (H.E.M.); (M.W.)
| | - Jennifer A. Curry
- US Naval Medical Research Unit No. 3, Naval Air Station Sigonella, 95030 Sigonella, Italy; (B.L.H.); (S.Y.L.); (J.A.C.); (A.M.); (H.E.M.); (M.W.)
| | - Adel Mansour
- US Naval Medical Research Unit No. 3, Naval Air Station Sigonella, 95030 Sigonella, Italy; (B.L.H.); (S.Y.L.); (J.A.C.); (A.M.); (H.E.M.); (M.W.)
| | - Hanan El Mohammady
- US Naval Medical Research Unit No. 3, Naval Air Station Sigonella, 95030 Sigonella, Italy; (B.L.H.); (S.Y.L.); (J.A.C.); (A.M.); (H.E.M.); (M.W.)
| | - Momtaz Wasfy
- US Naval Medical Research Unit No. 3, Naval Air Station Sigonella, 95030 Sigonella, Italy; (B.L.H.); (S.Y.L.); (J.A.C.); (A.M.); (H.E.M.); (M.W.)
| | - Drake Hamilton Tilley
- US Naval Medical Research Unit No. 6 Peru, Lima 07001, Peru; (D.H.T.); (M.J.G.); (M.R.K.); (J.R.); (P.R.); (G.P.)
| | - Michael J. Gregory
- US Naval Medical Research Unit No. 6 Peru, Lima 07001, Peru; (D.H.T.); (M.J.G.); (M.R.K.); (J.R.); (P.R.); (G.P.)
| | - Matthew R. Kasper
- US Naval Medical Research Unit No. 6 Peru, Lima 07001, Peru; (D.H.T.); (M.J.G.); (M.R.K.); (J.R.); (P.R.); (G.P.)
| | - James Regeimbal
- US Naval Medical Research Unit No. 6 Peru, Lima 07001, Peru; (D.H.T.); (M.J.G.); (M.R.K.); (J.R.); (P.R.); (G.P.)
| | - Paul Rios
- US Naval Medical Research Unit No. 6 Peru, Lima 07001, Peru; (D.H.T.); (M.J.G.); (M.R.K.); (J.R.); (P.R.); (G.P.)
| | - Guillermo Pimentel
- US Naval Medical Research Unit No. 6 Peru, Lima 07001, Peru; (D.H.T.); (M.J.G.); (M.R.K.); (J.R.); (P.R.); (G.P.)
| | - Brook A. Danboise
- US Army Medical Research Directorate-Africa/Kenya, Kericho 20200, Kenya; (B.A.D.); (C.E.H.); (E.A.O.); (A.N.O.); (E.K.C.); (C.O.P.)
| | - Christine E. Hulseberg
- US Army Medical Research Directorate-Africa/Kenya, Kericho 20200, Kenya; (B.A.D.); (C.E.H.); (E.A.O.); (A.N.O.); (E.K.C.); (C.O.P.)
| | - Elizabeth A. Odundo
- US Army Medical Research Directorate-Africa/Kenya, Kericho 20200, Kenya; (B.A.D.); (C.E.H.); (E.A.O.); (A.N.O.); (E.K.C.); (C.O.P.)
| | - Abigael N. Ombogo
- US Army Medical Research Directorate-Africa/Kenya, Kericho 20200, Kenya; (B.A.D.); (C.E.H.); (E.A.O.); (A.N.O.); (E.K.C.); (C.O.P.)
| | - Erick K. Cheruiyot
- US Army Medical Research Directorate-Africa/Kenya, Kericho 20200, Kenya; (B.A.D.); (C.E.H.); (E.A.O.); (A.N.O.); (E.K.C.); (C.O.P.)
| | - Cliff O. Philip
- US Army Medical Research Directorate-Africa/Kenya, Kericho 20200, Kenya; (B.A.D.); (C.E.H.); (E.A.O.); (A.N.O.); (E.K.C.); (C.O.P.)
| | - Gary J. Vora
- US Naval Research Laboratory, Center for Biomolecular Science & Engineering, Washington, DC 20375, USA; (T.A.L.); (G.J.V.)
| |
Collapse
|
200
|
da Cruz Nizer WS, Inkovskiy V, Overhage J. Surviving Reactive Chlorine Stress: Responses of Gram-Negative Bacteria to Hypochlorous Acid. Microorganisms 2020; 8:E1220. [PMID: 32796669 PMCID: PMC7464077 DOI: 10.3390/microorganisms8081220] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/30/2020] [Accepted: 08/09/2020] [Indexed: 01/29/2023] Open
Abstract
Sodium hypochlorite (NaOCl) and its active ingredient, hypochlorous acid (HOCl), are the most commonly used chlorine-based disinfectants. HOCl is a fast-acting and potent antimicrobial agent that interacts with several biomolecules, such as sulfur-containing amino acids, lipids, nucleic acids, and membrane components, causing severe cellular damage. It is also produced by the immune system as a first-line of defense against invading pathogens. In this review, we summarize the adaptive responses of Gram-negative bacteria to HOCl-induced stress and highlight the role of chaperone holdases (Hsp33, RidA, Cnox, and polyP) as an immediate response to HOCl stress. We also describe the three identified transcriptional regulators (HypT, RclR, and NemR) that specifically respond to HOCl. Besides the activation of chaperones and transcriptional regulators, the formation of biofilms has been described as an important adaptive response to several stressors, including HOCl. Although the knowledge on the molecular mechanisms involved in HOCl biofilm stimulation is limited, studies have shown that HOCl induces the formation of biofilms by causing conformational changes in membrane properties, overproducing the extracellular polymeric substance (EPS) matrix, and increasing the intracellular concentration of cyclic-di-GMP. In addition, acquisition and expression of antibiotic resistance genes, secretion of virulence factors and induction of the viable but nonculturable (VBNC) state has also been described as an adaptive response to HOCl. In general, the knowledge of how bacteria respond to HOCl stress has increased over time; however, the molecular mechanisms involved in this stress response is still in its infancy. A better understanding of these mechanisms could help understand host-pathogen interactions and target specific genes and molecules to control bacterial spread and colonization.
Collapse
Affiliation(s)
| | | | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada; (W.S.d.C.N.); (V.I.)
| |
Collapse
|