1
|
Suppression of Grape White Rot Caused by Coniella vitis Using the Potential Biocontrol Agent Bacillus velezensis GSBZ09. Pathogens 2022; 11:pathogens11020248. [PMID: 35215191 PMCID: PMC8876275 DOI: 10.3390/pathogens11020248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 12/10/2022] Open
Abstract
Grape white rot caused by Coniella vitis is prevalent in almost all grapevines worldwide and results in a yield loss of 10–20% annually. Bacillus velezensis is a reputable plant growth-promoting bacterial. Strain GSBZ09 was isolated from grapevine cv. Red Globe (Vitis vinifera) and identified as B. velezensis according to morphological, physiological, biochemical characteristics and a multilocus gene sequence analysis (MLSA) based on six housekeeping genes (16S rRNA, gyrB, rpoD, atpD, rho and pgk). B. velezensis GSBZ09 was screened for antifungal activity against C. vitis under in vitro and in vivo conditions. GSBZ09 presented broad spectrum antifungal activity and produced many extracellular enzymes that remarkably inhibited the mycelial growth and spore germination of C. vitis. Furthermore, GSBZ09 had a high capacity for indole-3-acetic acid (IAA) production, siderophore production, and mineral phosphate solubilization. Pot experiments showed that the application of GSBZ09 significantly decreased the disease index of the grape white rot, directly promoted the growth of grapes, and upregulated defense-related enzymes. Overall, the features of B. velezensis GSBZ09 make it a potential strain for application as a biological control agent against C. vitis.
Collapse
|
2
|
Dyett BP, Yu H, Sarkar S, Strachan JB, Drummond CJ, Conn CE. Uptake Dynamics of Cubosome Nanocarriers at Bacterial Surfaces and the Routes for Cargo Internalization. ACS APPLIED MATERIALS & INTERFACES 2021; 13:53530-53540. [PMID: 34726885 DOI: 10.1021/acsami.1c09909] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Antibiotic-resistant bacteria pose a significant threat to humanity. Gram-negative strains have demonstrated resistance to last resort antibiotics, partially due to their outer membrane, which hinders transport of antimicrobials into the bacterium. Nanocarrier (NC)-mediated drug delivery is one proposed strategy for combating this emerging issue. Here, the uptake of self-assembled lipid nanocarriers of cubic symmetry (cubosomes) into bacteria revealed fundamental differences in the uptake mechanism between Gram-positive and Gram-negative bacteria. For Gram-positive bacteria, the NCs adhere to the outer peptidoglycan layers and slowly internalize to the bacterium. For Gram-negative bacteria, the NCs interact in two stages, fusion with the outer lipid membrane and then diffusion through the inner wall. The self-assembled nature of the cubosomes imparts a unique ability to transfer payloads via membrane fusion. Remarkably, the fusion uptake mechanism allowed rapid NC internalization by the Gram-negative bacteria, overcoming the outer membrane responsible for their heightened resilience. Here this is demonstrated by the marked reduction in the minimal inhibition concentration required for antibiotics against a pathogenic strain of Gram-negative bacteria, Escherichia coli. These results provide mechanistic insight for the development of lipid NCs as a new tool to combat bacteria.
Collapse
Affiliation(s)
- Brendan P Dyett
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Haitao Yu
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Department of Chemical Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Sampa Sarkar
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Jamie B Strachan
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Charlotte E Conn
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
3
|
Jia F, Wang J, Zhang L, Zhou J, He Y, Lu Y, Liu K, Yan W, Wang K. Multiple action mechanism and in vivo antimicrobial efficacy of antimicrobial peptide Jelleine-I. J Pept Sci 2020; 27:e3294. [PMID: 33283388 DOI: 10.1002/psc.3294] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Accepted: 11/19/2020] [Indexed: 12/28/2022]
Abstract
With the extensive use of antibiotics in medicine, agriculture and food chemistry, the emergence of multi-drug resistant bacteria become more and more frequent and posed great threats to human health and life. So novel antimicrobial agents were urgently needed to defend the resistant bacteria. Jelleine-I was a small antimicrobial peptide (AMP) with eight amino acids in its sequence. It was believed to be an ideal template for developing antimicrobial agents. In the present study, the possible action mode against both gram-negative bacteria and gram-positive bacteria and in vivo antimicrobial activity was explored. Our results showed that Jelleine-I exhibits its antimicrobial activity mainly by disrupting the integrity of the cell membrane, which would not be affected by the conventional resistant mechanism. It also aims at some intracellular targets such as genomic DNA to inhibit the growth of microbes. In addition, the result of in vivo antimicrobial activity experiment showed that Jelleine-I performed a good therapeutic effect toward the mice with Escherichia coli infected peritonitis. Notably, Jelleine-I has negligible cytotoxicity toward the tested mammalian cells, indicating excellent cell selectivity between prokaryotic cells and eurkayotic cells. In summary, our results showed that Jelleine-I would be a potential candidate to be developed as a novel antimicrobial agent.
Collapse
Affiliation(s)
- Fengjing Jia
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Jiayi Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Lishi Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Jingjing Zhou
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Yuhang He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Yaqi Lu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Kexin Liu
- School/Hospital of Stomatology, Lanzhou University, West Donggang Road 199, Lanzhou, 730000, China
| | - Wenjin Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| | - Kairong Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Research Unit of Peptide Science of Chinese Academy of Medical Sciences 2019RU066, Lanzhou University, West Donggang Road, 199, Lanzhou, 730000, China
| |
Collapse
|
4
|
Goneau LW, Delport J, Langlois L, Poutanen SM, Razvi H, Reid G, Burton JP. Issues beyond resistance: inadequate antibiotic therapy and bacterial hypervirulence. FEMS MICROBES 2020; 1:xtaa004. [PMID: 37333955 PMCID: PMC10117437 DOI: 10.1093/femsmc/xtaa004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/15/2020] [Indexed: 10/15/2023] Open
Abstract
The administration of antibiotics while critical for treatment, can be accompanied by potentially severe complications. These include toxicities associated with the drugs themselves, the selection of resistant organisms and depletion of endogenous host microbiota. In addition, antibiotics may be associated with less well-recognized complications arising through changes in the pathogens themselves. Growing evidence suggests that organisms exposed to antibiotics can respond by altering the expression of toxins, invasins and adhesins, as well as biofilm, resistance and persistence factors. The clinical significance of these changes continues to be explored; however, it is possible that treatment with antibiotics may inadvertently precipitate a worsening of the clinical course of disease. Efforts are needed to adjust or augment antibiotic therapy to prevent the transition of pathogens to hypervirulent states. Better understanding the role of antibiotic-microbe interactions and how these can influence disease course is critical given the implications on prescription guidelines and antimicrobial stewardship policies.
Collapse
Affiliation(s)
- Lee W Goneau
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto,1 King's College Cir, Toronto, ON M5S 1A8 Ontario, Canada
| | - Johannes Delport
- Department of Pathology, London Health Sciences Center - Victoria Hospital, 800 Commissioners Rd E, London, Ontario, Canada N6A 5W9
| | - Luana Langlois
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Susan M Poutanen
- Department of Laboratory Medicine and Pathobiology, University of Toronto,1 King's College Cir, Toronto, ON M5S 1A8 Ontario, Canada
- Department of Medicine, University of Toronto, 1 King's College Cir, Toronto, ON M5S 1A8 Toronto, Ontario, Canada
- Department of Microbiology, University Health Network and Sinai Health, 190 Elizabeth St. Toronto, ON M5G 2C4, Ontario, Canada
| | - Hassan Razvi
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Division of Urology, Department of Surgery, Western University, 1151 Richmond St, London, Ontario, N6A 3K7 Canada
| | - Gregor Reid
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Division of Urology, Department of Surgery, Western University, 1151 Richmond St, London, Ontario, N6A 3K7 Canada
| | - Jeremy P Burton
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, 268 Grosvenor St, London, Ontario, N6A 4V2 Canada
- Division of Urology, Department of Surgery, Western University, 1151 Richmond St, London, Ontario, N6A 3K7 Canada
| |
Collapse
|
5
|
Mühlen S, Dersch P. Treatment Strategies for Infections With Shiga Toxin-Producing Escherichia coli. Front Cell Infect Microbiol 2020; 10:169. [PMID: 32435624 PMCID: PMC7218068 DOI: 10.3389/fcimb.2020.00169] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/31/2020] [Indexed: 01/07/2023] Open
Abstract
Infections with Shiga toxin-producing Escherichia coli (STEC) cause outbreaks of severe diarrheal disease in children and the elderly around the world. The severe complications associated with toxin production and release range from bloody diarrhea and hemorrhagic colitis to hemolytic-uremic syndrome, kidney failure, and neurological issues. As the use of antibiotics for treatment of the infection has long been controversial due to reports that antibiotics may increase the production of Shiga toxin, the recommended therapy today is mainly supportive. In recent years, a variety of alternative treatment approaches such as monoclonal antibodies or antisera directed against Shiga toxin, toxin receptor analogs, and several vaccination strategies have been developed and evaluated in vitro and in animal models. A few strategies have progressed to the clinical trial phase. Here, we review the current understanding of and the progress made in the development of treatment options against STEC infections and discuss their potential.
Collapse
Affiliation(s)
- Sabrina Mühlen
- Institute for Infectiology, University of Münster, Münster, Germany.,German Center for Infection Research (DZIF), Associated Site University of Münster, Münster, Germany
| | - Petra Dersch
- Institute for Infectiology, University of Münster, Münster, Germany.,German Center for Infection Research (DZIF), Associated Site University of Münster, Münster, Germany
| |
Collapse
|
6
|
Identification of Antibiotics That Diminish Disease in a Murine Model of Enterohemorrhagic Escherichia coli Infection. Antimicrob Agents Chemother 2020; 64:AAC.02159-19. [PMID: 32015030 DOI: 10.1128/aac.02159-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 01/27/2020] [Indexed: 11/20/2022] Open
Abstract
Infections with enterohemorrhagic Escherichia coli (EHEC) cause disease ranging from mild diarrhea to hemolytic-uremic syndrome (HUS) and are the most common cause of renal failure in children in high-income countries. The severity of the disease derives from the release of Shiga toxins (Stx). The use of antibiotics to treat EHEC infections is generally avoided, as it can result in increased stx expression. Here, we systematically tested different classes of antibiotics and found that their influence on stx expression and release varies significantly. We assessed a selection of these antibiotics in vivo using the Citrobacter rodentium ϕstx 2dact mouse model and show that stx 2d-inducing antibiotics resulted in weight loss and kidney damage despite clearance of the infection. However, several non-Stx-inducing antibiotics cleared bacterial infection without causing Stx-mediated pathology. Our results suggest that these antibiotics might be useful in the treatment of EHEC-infected human patients and decrease the risk of HUS development.
Collapse
|
7
|
Topsakal A, Ekren N, Kilic O, Oktar FN, Mahirogullari M, Ozkan O, Sasmazel HT, Turk M, Bogdan IM, Stan GE, Gunduz O. Synthesis and characterization of antibacterial drug loaded β-tricalcium phosphate powders for bone engineering applications. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:16. [PMID: 31965360 DOI: 10.1007/s10856-019-6356-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 12/28/2019] [Indexed: 06/10/2023]
Abstract
Powders of β-tricalcium phosphate [β-TCP, β-Ca3(PO4)2] and composite powders of β-TCP and polyvinyl alcohol (PVA) were synthesized by using wet precipitation methods. First, the conditions for the preparation of single phase β-TCP have been delineated. In the co-precipitation procedure, calcium nitrate and diammonium hydrogen phosphate were used as calcium and phosphorous precursors, respectively. The pH of the system was varied in the range 7-11 by adding designed amounts of ammonia solution. The filtered cakes were desiccated at 80 °C and subsequently calcined at different temperatures in the range between 700-1100 °C. Later on, rifampicin form II was used to produce drug-loaded β-TCP and PVA/β-TCP powders. All the synthesized materials have been characterized from morphological (by scanning electron microscopy) and structural-chemical (by X-ray diffraction and Fourier transform infrared spectroscopy) point of view. The drug loading capacity of the selected pure β-TCP powder has been assessed. The biological performance (cytocompatibility in fibroblast cell culture and antibacterial efficacy against Escherichia coli and Staphylococcus aureus) has been tested with promising results. Application perspectives of the designed drug-bioceramic-polymer blends are advanced and discussed.
Collapse
Affiliation(s)
- Aysenur Topsakal
- Center for Nanotechnology and Biomaterials Application and Research (NBUAM), Marmara University, 34722, Istanbul, Turkey
- Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, 34722, Istanbul, Turkey
| | - Nazmi Ekren
- Center for Nanotechnology and Biomaterials Application and Research (NBUAM), Marmara University, 34722, Istanbul, Turkey
- Department of Electric and Electronic Engineering, Faculty of Technology, Marmara University, 34722, Istanbul, Turkey
| | - Osman Kilic
- Center for Nanotechnology and Biomaterials Application and Research (NBUAM), Marmara University, 34722, Istanbul, Turkey
- Department of Electric and Electronic Engineering, Faculty of Engineering, Marmara University, 34722, Istanbul, Turkey
| | - Faik N Oktar
- Center for Nanotechnology and Biomaterials Application and Research (NBUAM), Marmara University, 34722, Istanbul, Turkey
- Department of Bioengineering, Faculty of Engineering, Marmara University, 34722, Istanbul, Turkey
| | - Mahir Mahirogullari
- Center for Nanotechnology and Biomaterials Application and Research (NBUAM), Marmara University, 34722, Istanbul, Turkey
- Department of Orthopedics and Traumatology, Memorial Hospital, 34390, Istanbul, Turkey
| | - Ozan Ozkan
- Bioengineering Division, Graduate School of Science and Engineering, Hacettepe University, Beyte, 06800, Ankara, Turkey
| | - Hilal Turkoglu Sasmazel
- Metallurgical and Materials Engineering Department, Faculty of Engineering, Atilim University, Incek, 06830, Ankara, Turkey
| | - Mustafa Turk
- Bioengineering Division, Engineering Faculty, Kirikkale University, Yahsihan, 71450, Kirikkale, Turkey
| | - Iuliana M Bogdan
- National Institute of Materials Physics, 077125, Magurele, Ilfov, Romania
| | - George E Stan
- National Institute of Materials Physics, 077125, Magurele, Ilfov, Romania
| | - Oguzhan Gunduz
- Center for Nanotechnology and Biomaterials Application and Research (NBUAM), Marmara University, 34722, Istanbul, Turkey.
- Department of Metallurgical and Materials Engineering, Faculty of Technology, Marmara University, 34722, Istanbul, Turkey.
| |
Collapse
|
8
|
Yuan L, Li L, Zheng F, Shi Y, Xie X, Chai A, Li B. The complete genome sequence of Rahnella aquatilis ZF7 reveals potential beneficial properties and stress tolerance capabilities. Arch Microbiol 2019; 202:483-499. [PMID: 31707426 DOI: 10.1007/s00203-019-01758-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 01/28/2023]
Abstract
Rahnella aquatilis ZF7 is a plant beneficial strain isolated from Sakura tree soil with potential for biocontrol. Here, we present the complete genome sequence of R. aquatilis ZF7, which consists of one 4.49 Mb circular chromosome and a 54-kb plasmid named pRAZF7. Phylogenetic analyses revealed that R. aquatilis ZF7 is much similar to the strains Rahnella sp. Y9602 and R. aquatilis HX2 than others evaluated. In this study, multiple genes encoding functions that likely contribute to plant growth promotion, biocontrol and stress tolerance were identified by comparative genome analyses, including IAA production, phosphate solubilization, antibiotic resistance and formation of Se nanoparticles (SeNPs). In addition, these functions were also confirmed by in vitro experiments. Considering its ability to form SeNPs, strain R. aquatilis ZF7 will contribute to nano-agriculture. Overall, the features of R. aquatilis ZF7 make it a high potential and competitive strain in biocontrol, and the genome data will help further studies on the mechanisms of plant growth promotion and biocontrol.
Collapse
Affiliation(s)
- Lifang Yuan
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Lei Li
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Fei Zheng
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin, 300384, China
| | - Yanxia Shi
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Xuewen Xie
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Ali Chai
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Baoju Li
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
9
|
Hussein HM, Al-Khoury DK, Abdelnoor AM, Rahal EA. Atorvastatin increases the production of proinflammatory cytokines and decreases the survival of Escherichia coli-infected mice. Sci Rep 2019; 9:11717. [PMID: 31406240 PMCID: PMC6690901 DOI: 10.1038/s41598-019-48282-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
To assess whether the immunosuppressive effects of atorvastatin outweigh its antibacterial ones in an infection, mice were infected with Escherichia coli and administered atorvastatin; survival rates were then monitored. Mice treated with atorvastatin post-infection showed a remarkable decrease in their survival rate. On the other hand, the higher the level of serum IFN-γ in the infected mice treated with atorvastatin, the lower was the survival rate. Levels of IL-4 were markedly depressed in all groups infected with E. coli and treated with atorvastatin. Since atorvastatin inhibits IFN-γ expression in the absence of bacterial infection, we examined whether bacterial lipopolysaccharide (LPS) was the element capable of overriding this inhibition. Mouse peripheral blood mononuclear cells were treated with atorvastatin and lipopolysaccharide ex vivo then proinflammatory (IFN-γ, TNFα, IL-6) and prohumoral/regulatory (IL-4, IL-13, IL-10) cytokine levels were analyzed in culture supernatants. While proinflammatory cytokine levels were decreased upon treatment with atorvastatin alone, their levels were markedly elevated by treatment with LPS, bacterial lysate or bacterial culture supernatant. On the other hand, atorvastatin exerted an inhibitory effect on production of the prohumoral/regulatory cytokines. Our data indicates that any consideration for statins as antimicrobial treatment should assess the possible adverse outcomes.
Collapse
Affiliation(s)
- Hadi M Hussein
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Diva Kalash Al-Khoury
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Alexander M Abdelnoor
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon
| | - Elias A Rahal
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon. .,Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
10
|
Kakoullis L, Papachristodoulou E, Chra P, Panos G. Shiga toxin-induced haemolytic uraemic syndrome and the role of antibiotics: a global overview. J Infect 2019; 79:75-94. [DOI: 10.1016/j.jinf.2019.05.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 05/21/2019] [Accepted: 05/25/2019] [Indexed: 11/17/2022]
|
11
|
El Hafi B, Rasheed SS, Abou Fayad AG, Araj GF, Matar GM. Evaluating the Efficacies of Carbapenem/β-Lactamase Inhibitors Against Carbapenem-Resistant Gram-Negative Bacteria in vitro and in vivo. Front Microbiol 2019; 10:933. [PMID: 31114565 PMCID: PMC6503214 DOI: 10.3389/fmicb.2019.00933] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Background Carbapenem-resistant Gram-negative bacteria are a major clinical concern as they cause virtually untreatable infections since carbapenems are among the last-resort antimicrobial agents. β-Lactamases implicated in carbapenem resistance include KPC, NDM, and OXA-type carbapenemases. Antimicrobial combination therapy is the current treatment approach against carbapenem resistance in order to limit the excessive use of colistin; however, its advantages over monotherapy remain debatable. An alternative treatment strategy would be the use of carbapenem/β-lactamase inhibitor (βLI) combinations. In this study, we assessed the in vitro and in vivo phenotypic and molecular efficacies of three βLIs when combined with different carbapenems against carbapenem-resistant Gram-negative clinical isolates. The chosen βLIs were (1) Avibactam, against OXA-type carbapenemases, (2) calcium-EDTA, against NDM-1, and (3) Relebactam, against KPC-2. Methods Six Acinetobacter baumannii clinical isolates were screened for bla OXA-23-like, bla OXA-24/40, bla OXA-51-like, bla OXA-58, and bla OXA-143-like, and eight Enterobacteriaceae clinical isolates were screened for bla OXA-48, bla NDM-1, and bla KPC-2. The minimal inhibitory concentrations of Imipenem (IPM), Ertapenem (ETP), and Meropenem (MEM) with corresponding βLIs for each isolate were determined. The efficacy of the most suitable in vitro treatment option against each of bla OXA-48, bla NDM-1, and bla KPC-2 was assessed via survival studies in a BALB/c murine infection model. Finally, RT-qPCR was performed to assess the molecular response of the genes of resistance to the carbapenem/βLI combinations used under both in vitro and in vivo settings. Results Combining MEM, IPM, and ETP with the corresponding βLIs restored the isolates' susceptibilities to those antimicrobial agents in 66.7%, 57.1%, and 30.8% of the samples, respectively. Survival studies in mice revealed 100% survival rates when MEM was combined with either Avibactam or Relebactam against bla OXA-48 and bla KPC-2, respectively. RT-qPCR demonstrated the consistent overexpression of bla OXA-48 upon treatment, without hindering Avibactam's activity, while bla NDM-1 and bla KPC-2 experienced variable expression levels upon treatment under in vitro and in vivo settings despite their effective phenotypic results. Conclusion New carbapenem/βLI combinations may be viable alternatives to antimicrobial combination therapy as they displayed high efficacy in vitro and in vivo. Meropenem/Avibactam and Meropenem/Relebactam should be tested on larger sample sizes with different carbapenemases before progressing further in its preclinical development.
Collapse
Affiliation(s)
- Bassam El Hafi
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Sari S Rasheed
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Antoine G Abou Fayad
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - George F Araj
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
12
|
Salloum NA, Kissoyan KAB, Fadlallah S, Cheaito K, Araj GF, Wakim R, Kanj S, Kanafani Z, Dbaibo G, Matar GM. Assessment of combination therapy in BALB/c mice injected with carbapenem-resistant Enterobacteriaceae strains. Front Microbiol 2015; 6:999. [PMID: 26441926 PMCID: PMC4585037 DOI: 10.3389/fmicb.2015.00999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/07/2015] [Indexed: 11/30/2022] Open
Abstract
Monotherapeutic options for carbapenem resistant infections are limited. Studies suggest that combination therapy may be associated with better outcomes than monotherapies. However, this is still controversial. This study assessed, the efficacy of combination therapy against carbapenem resistant Enterobacteriaceae harboring singly various extended spectrum beta lactamase or carbapenemase encoding genes. Thus, four isolates harboring either blaCTXM-15, blaCTXM-15 and blaOXA-48, blaNDM-1, or blaKPC-2 genes were selected for testing. Minimal inhibitory concentration was determined by broth dilution method. Gene transcript levels on single and combined treatments were done in vitro and in vivo by qRT-PCR. Assessment of treatments was done in BALB/c mice according to a specific protocol. As such, the qRT-PCR revealed a significant decrease of transcript levels in all isolates upon using rifampicin or tigecycline, singly or in combination with colistin. However, variable levels were obtained using colistin singly or in combination with meropenem or fosfomycin. In vivo assessment showed that all combinations used were effective against isolates harboring blaCTXM-15, blaOXA-48, and blaNDM-1. Conversely, the most significant combination against the isolate harboring blaKPC-2 gene was colistin with either carbapenem, fosfomycin, or kanamycin. As a conclusion, combination therapy selected based on the type of carbapenemase produced, appeared to be non-toxic and might be effective in BALB/c mice. Therefore, the use of a rationally optimized combination therapy might lead to better results than monotherapy, however, clinical trials are needed for human consumption.
Collapse
Affiliation(s)
- Noor A Salloum
- Department of Experimental Pathology, Immunology and Microbiology, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Kohar Annie B Kissoyan
- Department of Experimental Pathology, Immunology and Microbiology, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Sukayna Fadlallah
- Department of Experimental Pathology, Immunology and Microbiology, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Katia Cheaito
- Department of Experimental Pathology, Immunology and Microbiology, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - George F Araj
- Pathology and Laboratory Medicine, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Rima Wakim
- Pediatrics and Adolescent Medicine, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Souha Kanj
- Internal Medicine, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Zeina Kanafani
- Internal Medicine, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Ghassan Dbaibo
- Pediatrics and Adolescent Medicine, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology and Microbiology, Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| |
Collapse
|
13
|
Agger M, Scheutz F, Villumsen S, Mølbak K, Petersen AM. Antibiotic treatment of verocytotoxin-producingEscherichia coli(VTEC) infection: a systematic review and a proposal. J Antimicrob Chemother 2015; 70:2440-6. [DOI: 10.1093/jac/dkv162] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/22/2015] [Indexed: 11/12/2022] Open
|
14
|
Rahal EA, Fadlallah SM, Nassar FJ, Kazzi N, Matar GM. Approaches to treatment of emerging Shiga toxin-producing Escherichia coli infections highlighting the O104:H4 serotype. Front Cell Infect Microbiol 2015; 5:24. [PMID: 25853096 PMCID: PMC4364364 DOI: 10.3389/fcimb.2015.00024] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/04/2015] [Indexed: 11/13/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) are a group of diarrheagenic bacteria associated with foodborne outbreaks. Infection with these agents may result in grave sequelae that include fatality. A large number of STEC serotypes has been identified to date. E. coli serotype O104:H4 is an emerging pathogen responsible for a 2011 outbreak in Europe that resulted in over 4000 infections and 50 deaths. STEC pathogenicity is highly reliant on the production of one or more Shiga toxins that can inhibit protein synthesis in host cells resulting in a cytotoxicity that may affect various organ systems. Antimicrobials are usually avoided in the treatment of STEC infections since they are believed to induce bacterial cell lysis and the release of stored toxins. Some antimicrobials have also been reported to enhance toxin synthesis and production from these organisms. Various groups have attempted alternative treatment approaches including the administration of toxin-directed antibodies, toxin-adsorbing polymers, probiotic agents and natural remedies. The utility of antibiotics in treating STEC infections has also been reconsidered in recent years with certain modalities showing promise.
Collapse
Affiliation(s)
- Elias A Rahal
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Sukayna M Fadlallah
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Farah J Nassar
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Natalie Kazzi
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut Beirut, Lebanon
| |
Collapse
|
15
|
Delivery of rifampicin-chitin nanoparticles into the intracellular compartment of polymorphonuclear leukocytes. Int J Biol Macromol 2014; 74:36-43. [PMID: 25475841 DOI: 10.1016/j.ijbiomac.2014.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 10/24/2014] [Accepted: 11/01/2014] [Indexed: 12/29/2022]
Abstract
Polymorphonuclear leukocytes (PMNs) provide the primary host defence against invading pathogens by producing reactive oxygen species (ROS) and microbicidal products. However, few pathogens can survive for a prolonged period of time within the PMNs. Additionally their intracellular lifestyle within the PMNs protect themselves from the additional lethal action of host immune systems such as antibodies and complements. Antibiotic delivery into the intracellular compartments of PMNs is a major challenge in the field of infectious diseases. In order to deliver antibiotics within the PMNs and for the better treatment of intracellular bacterial infections we synthesized rifampicin (RIF) loaded amorphous chitin nanoparticles (RIF-ACNPs) of 350±50 nm in diameter. RIF-ACNPs nanoparticles are found to be non-hemolytic and non-toxic against a variety of host cells. The release of rifampicin from the prepared nanoparticles was ∼60% in 24 h, followed by a sustained pattern till 72 h. The RIF-ACNPs nanoparticles showed 5-6 fold enhanced delivery of RIF into the intracellular compartments of PMNs. The RIF-ACNPs showed anti-microbial activity against Escherichia coli, Staphylococcus aureus and a variety of other bacteria. In summary, our results suggest that RIF-ACNPs could be used to treat a variety of intracellular bacterial infections.
Collapse
|
16
|
Fadlallah SM, Rahal EA, Sabra A, Kissoyan KAB, Matar GM. Effect of rifampicin and gentamicin on Shiga toxin 2 expression level and the SOS response in Escherichia coli O104:H4. Foodborne Pathog Dis 2014; 12:47-55. [PMID: 25383748 DOI: 10.1089/fpd.2014.1824] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND A novel pathotype, Shiga toxin-producing Escherichia coli O104:H4, was the cause of a severe outbreak that affected European countries, mainly Germany, in 2011. The effect of different regimens of rifampicin and gentamicin were evaluated to determine possible treatment modes for the novel strain, and to evaluate the SOS response and its effect on toxin release. MATERIALS AND METHODS Pulsed-field gel electrophoresis (PFGE) was performed on the novel E. coli O104:H4 pathotype and two pre-outbreak E. coli O104:H4 CDC strains. Transcript levels of the stx2 and recA gene (SOS response inducer) were evaluated using quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR) in the novel E. coli O104:H4 samples subjected to different regimens of rifampicin and gentamicin. Consequently, reverse passive latex agglutination (RPLA) was used to determine the Stx2 titers in these samples. Western blot was performed to determine the LexA levels (SOS response repressor) in E. coli O104:H4. The efficacy of treatment with antimicrobial agents was assessed in BALB/c mice. RESULTS The outbreak and pre-outbreak strains are closely related as shown by PFGE, which demonstrated slight genomic differences between the three strains. The transcription level of the stx2 gene in the new pathotype was 1.41- and 1.75-fold that of the 2009 EL-2050 and 2009 EL-2071 pre-outbreak strains, respectively. Moreover, the transcription level of the stx2 gene in the new pathotype was substantially decreased as a result of treatment with the different concentrations of the antimicrobial agents, but was enhanced when the antibiotics were administered at two subinhibitory levels. RPLA data were in accordance with the qRT-PCR results. E. coli O104:H4 exposed to gentamicin at both sub-minimum inhibitory concentration (MIC) levels led to high transcription levels of the recA gene and lack of expression of the LexA protein, implying that the SOS response was activated. Rifampicin at both sub-MIC levels resulted in low transcript levels of the recA gene, indicating that the SOS response was not induced. In vivo, the highest survival rate in BALB/c mice was observed in the group that was treated with the minimum bactericidal concentration (MBC) of gentamicin. CONCLUSION The use of antimicrobial agents in E. coli O104:H4 infection seems to be effective at the MIC and MBC levels. This provides a promising ground for treatment of E. coli O104:H4.
Collapse
Affiliation(s)
- Sukayna M Fadlallah
- Department of Experimental Pathology, Immunology, & Microbiology, Faculty of Medicine, American University of Beirut , Beirut, Lebanon
| | | | | | | | | |
Collapse
|
17
|
Bernedo-Navarro RA, Miyachiro MM, da Silva MJ, Reis CF, Conceição RA, Gatti MSV, Yano T. Peptides derived from phage display libraries as potential neutralizers of Shiga toxin-induced cytotoxicity in vitro and in vivo. J Appl Microbiol 2014; 116:1322-33. [PMID: 24447276 DOI: 10.1111/jam.12451] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 12/21/2013] [Accepted: 01/10/2014] [Indexed: 11/30/2022]
Abstract
AIMS To use the phage display technique to develop peptides with the capability to neutralize the cytotoxicity induced by Stx1 and Stx2 toxins produced by Shiga toxin-producing Escherichia coli (STEC). METHODS AND RESULTS The phage display technique permitted the development of three peptides, named PC7-12, P12-26 and PC7-30, which bind to the globotriaosylceramide (Gb3) receptor for Shiga toxins produced by STEC. Moreover, these peptides were capable of competing efficiently with the Shiga toxins for binding to Gb3. The peptides described herein partially inhibited the Stx-induced cytotoxicity of cell-free filtrates of STEC O157 : H7 and purified Stx toxins in Vero cells. The inhibition of lethality induced by Stx toxins in mice indicated that peptide PC7-30 inhibited the lethality caused by Stx1 (2LD50) in mice. CONCLUSIONS The phage display technique permitted the development of peptides that inhibited the cytotoxicity induced by Stx toxins in vitro. Peptide PC7-30 inhibited the lethality of Stx1 in vivo; this molecule would be a promising candidate for the development of therapeutic agents for STEC-related diseases in humans. SIGNIFICANCE AND IMPACT OF THE STUDY The selection of Gb3, the common receptor for Stx1 and Stx2, may contribute to the development of efficient neutralizers for both toxins, and our approach would be an interesting alternative for the development of therapeutic molecules for the treatment of diseases caused by STEC strains.
Collapse
Affiliation(s)
- R A Bernedo-Navarro
- Departamento de Genética, Evolução e Bioagentes, Instituto de Biologia-UNICAMP, Campinas, SP, Brazil
| | | | | | | | | | | | | |
Collapse
|
18
|
Kern WV. [Management of patients with EHEC/HUS. Lessons and perspectives from clinical infectious disease specialists]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2013; 56:87-94. [PMID: 23275960 DOI: 10.1007/s00103-012-1577-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 2011 EHEC/HUS outbreak created uncertainty regarding the previous recommendations of withholding antibiotic therapy and regarding rational measures for prevention of invasive meningococcal disease after treatment with the monoclonal antibody eculizumab. For both these areas, an expert panel of the German Society for Infectious Diseases in cooperation with representatives of other learned societies and health institutions has drafted and consented guidelines that were published online on 1 June 2011 (i.e., ~10 days after the peak of the epidemic) and 4 June 2011. A summary of the guidelines in English was made available online on 4 June 2011. The time until ad hoc guidelines are made publicly available should and can be shortened in similar scenarios in the future. To this end-among other things-scientific societies in Germany linked to infectious diseases and medical microbiology have established a permanent working group called AFIM. This working group will facilitate timely identification and appointment of experts and expert panels, and will make use of new ways of rapid and effective sharing and dissemination of knowledge and ad hoc guidelines in the medical community and public domain if needed. In the case of disease outbreaks, immediate telephone conferences among all professionals involved, close cooperation between institutions and expert groups, and avoidance of premature unconsented information and press releases will be critical. We expect that proceeding in this way will also have a major impact on proper planning, professional design, and adequate analysis of clinical studies and endpoints appropriate for the outbreak situation.
Collapse
Affiliation(s)
- W V Kern
- Zentrum Infektiologie und Reisemedizin und IFB-Zentrum für chronische Immundefizienz, Medizinische Universitätsklinik Freiburg, Hugstetterstr. 55, 79106, Freiburg, Deutschland.
| |
Collapse
|
19
|
Nassar FJ, Rahal EA, Sabra A, Matar GM. Effects of subinhibitory concentrations of antimicrobial agents on Escherichia coli O157:H7 Shiga toxin release and role of the SOS response. Foodborne Pathog Dis 2013; 10:805-12. [PMID: 23808851 DOI: 10.1089/fpd.2013.1510] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Treatment of Escherichia coli O157:H7 by certain antimicrobial agents often exacerbates the patient's condition by increasing either the release of preformed Shiga toxins (Stx) upon cell lysis or their production through the SOS response-triggered induction of Stx-producing prophages. Recommended subinhibitory concentrations (sub-MICs) of azithromycin (AZI), gentamicin (GEN), imipenem (IMI), and rifampicin (RIF) were evaluated in comparison to norfloxacin (NOR), an SOS-inducer, to assess the role of the SOS response in Stx release. Relative expression of recA (SOS-inducer), Q (late antitermination gene of Stx-producing prophage), stx1, and stx2 genes was assessed at two sub-MICs of the antimicrobials for two different strains of E. coli O157:H7 using reverse transcription-real-time polymerase chain reaction. Both strains at the two sub-MICs were also subjected to Western blotting for LexA protein expression and to reverse passive latex agglutination for Stx detection. For both strains at both sub-MICs, NOR and AZI caused SOS-induced Stx production (high recA, Q, and stx2 gene expression and high Stx2 production), so they should be avoided in E. coli O157:H7 treatment; however, sub-MICs of RIF and IMI induced Stx2 production in an SOS-independent manner except for one strain at the first twofold dilution below MIC of RIF where Stx2 production decreased. Moreover, GEN caused somewhat increased Stx2 production due to its mode of action rather than any effect on gene expression. The choice of antimicrobial therapy should rely on the antimicrobial mode of action, its concentration, and on the nature of the strain.
Collapse
Affiliation(s)
- Farah J Nassar
- Department of Experimental Pathology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | |
Collapse
|
20
|
Magwedere K, Dang HA, Mills EW, Cutter CN, Roberts EL, DebRoy C. Incidence of Shiga toxin–producing Escherichia coli strains in beef, pork, chicken, deer, boar, bison, and rabbit retail meat. J Vet Diagn Invest 2013; 25:254-8. [DOI: 10.1177/1040638713477407] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The objective of the current study was to determine the incidence of contamination by the top 7 Shiga toxin–producing Escherichia coli (STEC) O-groups, responsible for the majority of E. coli infections in human beings, in retail meat from different animal species. Samples from ground beef ( n = 51), ground pork ( n = 16), ground chicken ( n = 16), and game meat (deer, wild boar, bison, and rabbit; n = 55) were collected from retail vendors for the detection of 7 STEC O-groups (O26, O45, O103, O111, O121, O145, and O157). Meat samples were tested by using a multiplex polymerase chain reaction assay targeting the wzx gene of O antigen gene clusters of the 7 STEC O-groups. The positive samples were further tested for Shiga toxin genes ( stx1 and stx2). Out of a total of 83 ground beef, pork, and chicken samples, 17 (20%) carried O121, 9 (10%) carried O45, 8 (9%) carried O157, 3 (3%) carried O103, and 1 (1%) carried O145. None of the samples were positive for O26, O111, or the stx gene. All 3 white-tailed deer samples (100%) were positive for O45, O103, or both, 2 (10%) out of 20 red deer samples exhibited the presence of O103, and all 3 bison samples were contaminated with either O121, O145, or O157. One sample from ground deer, contaminated with E. coli O45, carried the stx1 gene. This preliminary investigation illustrates the importance of microbiological testing of pathogens in meat products, as well as the recognized need for increased surveillance and research on foodborne pathogens.
Collapse
Affiliation(s)
- Kudakwashe Magwedere
- Departments of Veterinary and Biomedical Sciences (Dang, Roberts, DebRoy), Pennsylvania State University, University Park, PA
- Animal Science (Magwedere, Mills), Pennsylvania State University, University Park, PA
- Food Science (Cutter), Pennsylvania State University, University Park, PA
| | - Huu Anh Dang
- Departments of Veterinary and Biomedical Sciences (Dang, Roberts, DebRoy), Pennsylvania State University, University Park, PA
- Animal Science (Magwedere, Mills), Pennsylvania State University, University Park, PA
- Food Science (Cutter), Pennsylvania State University, University Park, PA
| | - Edward W. Mills
- Departments of Veterinary and Biomedical Sciences (Dang, Roberts, DebRoy), Pennsylvania State University, University Park, PA
- Animal Science (Magwedere, Mills), Pennsylvania State University, University Park, PA
- Food Science (Cutter), Pennsylvania State University, University Park, PA
| | - Catherine N. Cutter
- Departments of Veterinary and Biomedical Sciences (Dang, Roberts, DebRoy), Pennsylvania State University, University Park, PA
- Animal Science (Magwedere, Mills), Pennsylvania State University, University Park, PA
- Food Science (Cutter), Pennsylvania State University, University Park, PA
| | - Elisabeth L. Roberts
- Departments of Veterinary and Biomedical Sciences (Dang, Roberts, DebRoy), Pennsylvania State University, University Park, PA
- Animal Science (Magwedere, Mills), Pennsylvania State University, University Park, PA
- Food Science (Cutter), Pennsylvania State University, University Park, PA
| | - Chitrita DebRoy
- Departments of Veterinary and Biomedical Sciences (Dang, Roberts, DebRoy), Pennsylvania State University, University Park, PA
- Animal Science (Magwedere, Mills), Pennsylvania State University, University Park, PA
- Food Science (Cutter), Pennsylvania State University, University Park, PA
| |
Collapse
|
21
|
Rahal EA, Kazzi N, Nassar FJ, Matar GM. Escherichia coli O157:H7-Clinical aspects and novel treatment approaches. Front Cell Infect Microbiol 2012; 2:138. [PMID: 23162800 PMCID: PMC3498739 DOI: 10.3389/fcimb.2012.00138] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 10/19/2012] [Indexed: 12/25/2022] Open
Abstract
Escherichia coli O157:H7 is a notorious pathogen often contracted by intake of contaminated water or food. Infection with this agent is associated with a broad spectrum of illness ranging from mild diarrhea and hemorrhagic colitis to the potentially fatal hemolytic uremic syndrome (HUS). Treating E. coli O157:H7 infection with antimicrobial agents is associated with an increased risk of severe sequelae such as HUS. The difficulty in treating this bacterium using conventional modalities of antimicrobial agent administration has sparked an interest in investigating new therapeutic approaches to this bacterium. These approaches have included the use of probiotic agents and natural products with variable success rates. In addition, novel modalities and regimen of antimicrobial agent administration have been assessed in an attempt at decreasing their association with aggravating infection outcomes.
Collapse
Affiliation(s)
- Elias A Rahal
- Faculty of Medicine, Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut Beirut, Lebanon
| | | | | | | |
Collapse
|
22
|
Corogeanu D, Willmes R, Wolke M, Plum G, Utermöhlen O, Krönke M. Therapeutic concentrations of antibiotics inhibit Shiga toxin release from enterohemorrhagic E. coli O104:H4 from the 2011 German outbreak. BMC Microbiol 2012; 12:160. [PMID: 22853739 PMCID: PMC3438081 DOI: 10.1186/1471-2180-12-160] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 07/02/2012] [Indexed: 11/25/2022] Open
Abstract
Background The shiga toxin-producing E. coli (STEC) O104:H4 caused a major outbreak in Germany in spring 2011. STEC are usually susceptible to common antibiotics. However, antibiotic treatment of STEC-infected patients is not recommended because STEC may enhance production and release of shiga toxins (STX) in response to antibiotics, which eventually enhances the frequency and severity of clinical symptoms, including haemolytic uraemic syndrome (HUS) and fatalities. Results We characterized the response to antibiotics of STEC O104:H4 isolates from two HUS patients during the German STEC outbreak in spring 2011 in comparison to the common STEC O157:H7. Liquid cultures of STEC O157:H7 and O104:H4 were incubated with graded dilutions of the antibiotics ciprofloxacin, meropenem, fosfomycin, gentamicin, rifampicin, and chloramphenicol. At defined times of antibiotic treatment, transcriptional activation of the STX2 gene, contents of STX and STX-activity in the culture supernatants were quantified. Unlike the common serotype O157:H7, STEC O104:H4 does not release STX in response to therapeutic concentrations of ciprofloxacin, meropenem, fosfomycin, and chloramphenicol. Conclusions In future outbreaks, the response of the respective epidemiologic STEC strain to antibiotics should be rapidly characterized in order to identify antibiotics that do not enhance the release of STX. This will eventually allow clinical studies tackling the question whether antibiotic treatment impacts on the eradication of STEC, clinical course of disease, and frequency of carriers.
Collapse
Affiliation(s)
- Diana Corogeanu
- Institute for Medical Microbiology, Immunology and Hygiene, Medical Center, University of Cologne, Cologne, Germany
| | | | | | | | | | | |
Collapse
|
23
|
Menne J, Nitschke M, Stingele R, Abu-Tair M, Beneke J, Bramstedt J, Bremer JP, Brunkhorst R, Busch V, Dengler R, Deuschl G, Fellermann K, Fickenscher H, Gerigk C, Goettsche A, Greeve J, Hafer C, Hagenmüller F, Haller H, Herget-Rosenthal S, Hertenstein B, Hofmann C, Lang M, Kielstein JT, Klostermeier UC, Knobloch J, Kuehbacher M, Kunzendorf U, Lehnert H, Manns MP, Menne TF, Meyer TN, Michael C, Münte T, Neumann-Grutzeck C, Nuernberger J, Pavenstaedt H, Ramazan L, Renders L, Repenthin J, Ries W, Rohr A, Rump LC, Samuelsson O, Sayk F, Schmidt BMW, Schnatter S, Schöcklmann H, Schreiber S, von Seydewitz CU, Steinhoff J, Stracke S, Suerbaum S, van de Loo A, Vischedyk M, Weissenborn K, Wellhöner P, Wiesner M, Zeissig S, Büning J, Schiffer M, Kuehbacher T. Validation of treatment strategies for enterohaemorrhagic Escherichia coli O104:H4 induced haemolytic uraemic syndrome: case-control study. BMJ 2012; 345:e4565. [PMID: 22815429 PMCID: PMC3400392 DOI: 10.1136/bmj.e4565] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate the effect of different treatment strategies on enterohaemorrhagic Escherichia coli O104:H4 induced haemolytic uraemic syndrome. DESIGN Multicentre retrospective case-control study. SETTING 23 hospitals in northern Germany. PARTICIPANTS 298 adults with enterohaemorrhagic E coli induced haemolytic uraemic syndrome. MAIN OUTCOME MEASURES Dialysis, seizures, mechanical ventilation, abdominal surgery owing to perforation of the bowel or bowel necrosis, and death. RESULTS 160 of the 298 patients (54%) temporarily required dialysis, with only three needing treatment long term. 37 patients (12%) had seizures, 54 (18%) required mechanical ventilation, and 12 (4%) died. No clear benefit was found from use of plasmapheresis or plasmapheresis with glucocorticoids. 67 of the patients were treated with eculizumab, a monoclonal antibody directed against the complement cascade. No short term benefit was detected that could be attributed to this treatment. 52 patients in one centre that used a strategy of aggressive treatment with combined antibiotics had fewer seizures (2% v 15%, P = 0.03), fewer deaths (0% v 5%, p = 0.029), required no abdominal surgery, and excreted E coli for a shorter duration. CONCLUSIONS Enterohaemorrhagic E coli induced haemolytic uraemic syndrome is a severe self limiting acute condition. Our findings question the benefit of eculizumab and of plasmapheresis with or without glucocorticoids. Patients with established haemolytic uraemic syndrome seemed to benefit from antibiotic treatment and this should be investigated in a controlled trial.
Collapse
Affiliation(s)
- Jan Menne
- Medical School Hannover, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bielaszewska M, Idelevich EA, Zhang W, Bauwens A, Schaumburg F, Mellmann A, Peters G, Karch H. Effects of antibiotics on Shiga toxin 2 production and bacteriophage induction by epidemic Escherichia coli O104:H4 strain. Antimicrob Agents Chemother 2012; 56:3277-82. [PMID: 22391549 PMCID: PMC3370775 DOI: 10.1128/aac.06315-11] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The role of antibiotics in treatment of enterohemorrhagic Escherichia coli (EHEC) infections is controversial because of concerns about triggering hemolytic-uremic syndrome (HUS) by increasing Shiga toxin (Stx) production. During the recent large EHEC O104:H4 outbreak, antibiotic therapy was indicated for some patients. We tested a diverse panel of antibiotics to which the outbreak strain is susceptible to interrogate the effects of subinhibitory antibiotic concentrations on induction of stx(2)-harboring bacteriophages, stx(2) transcription, and Stx2 production in this emerging pathogen. Ciprofloxacin significantly increased stx(2)-harboring phage induction and Stx2 production in outbreak isolates (P values of <0.001 to <0.05), while fosfomycin, gentamicin, and kanamycin insignificantly influenced them (P > 0.1) and chloramphenicol, meropenem, azithromycin, rifaximin, and tigecycline significantly decreased them (P ≤ 0.05). Ciprofloxacin and chloramphenicol significantly upregulated and downregulated stx(2) transcription, respectively (P < 0.01); the other antibiotics had insignificant effects (P > 0.1). Meropenem, azithromycin, and rifaximin, which were used for necessary therapeutic or prophylactic interventions during the EHEC O104:H4 outbreak, as well as tigecycline, neither induced stx(2)-harboring phages nor increased stx(2) transcription or Stx2 production in the outbreak strain. These antibiotics might represent therapeutic options for patients with EHEC O104:H4 infection if antibiotic treatment is inevitable. We await further analysis of the epidemic to determine if usage of these agents was associated with an altered risk of developing HUS.
Collapse
Affiliation(s)
- Martina Bielaszewska
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, Münster, Germany
| | | | - Wenlan Zhang
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, Münster, Germany
| | - Andreas Bauwens
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, Münster, Germany
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, Münster, Germany
- Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany
| | - Georg Peters
- Institute of Medical Microbiology, University of Münster, Münster, Germany
- Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany
| | - Helge Karch
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, Münster, Germany
- Interdisciplinary Center of Clinical Research (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
25
|
Rahal EA, Kazzi N, Sabra A, Abdelnoor AM, Matar GM. Decrease in Shiga toxin expression using a minimal inhibitory concentration of rifampicin followed by bactericidal gentamicin treatment enhances survival of Escherichia coli O157:H7-infected BALB/c mice. Ann Clin Microbiol Antimicrob 2011; 10:34. [PMID: 21906403 PMCID: PMC3180354 DOI: 10.1186/1476-0711-10-34] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 09/12/2011] [Indexed: 12/30/2022] Open
Abstract
Background Treatment of Escherichia coli O157:H7 infections with antimicrobial agents is controversial due to an association with potentially fatal sequelae. The production of Shiga toxins is believed to be central to the pathogenesis of this organism. Therefore, decreasing the expression of these toxins prior to bacterial eradication may provide a safer course of therapy. Methods The utility of decreasing Shiga toxin gene expression in E. coli O157:H7 with rifampicin prior to bacterial eradication with gentamicin was evaluated in vitro using real-time reverse-transcription polymerase chain reaction. Toxin release from treated bacterial cells was assayed for with reverse passive latex agglutination. The effect of this treatment on the survival of E. coli O157:H7-infected BALB/c mice was also monitored. Results Transcription of Shiga toxin-encoding genes was considerably decreased as an effect of treating E. coli O157:H7 in vitro with the minimum inhibitory concentration (MIC) of rifampicin followed by the minimum bactericidal concentration (MBC) of gentamicin (> 99% decrease) compared to treatment with gentamicin alone (50-75% decrease). The release of Shiga toxins from E. coli O157:H7 incubated with the MIC of rifampicin followed by addition of the MBC of gentamicin was decreased as well. On the other hand, the highest survival rate in BALB/c mice infected with E. coli O157:H7 was observed in those treated with the in vivo MIC equivalent dose of rifampicin followed by the in vivo MBC equivalent dose of gentamicin compared to mice treated with gentamicin or rifampicin alone. Conclusions The use of non-lethal expression-inhibitory doses of antimicrobial agents prior to bactericidal ones in treating E. coli O157:H7 infection is effective and may be potentially useful in human infections with this agent in addition to other Shiga toxin producing E. coli strains.
Collapse
Affiliation(s)
- Elias A Rahal
- Department of Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | | | |
Collapse
|