1
|
Ferreira A, Silva D, Almeida C, Rodrigues ME, Silva S, Castro J, Mil-Homens D, García-Meniño I, Mora A, Henriques M, Oliveira A. Effect of phage vB_EcoM_FJ1 on the reduction of ETEC O9:H9 infection in a neonatal pig cell line. Vet Res 2023; 54:26. [PMID: 36949480 PMCID: PMC10035155 DOI: 10.1186/s13567-023-01157-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/02/2023] [Indexed: 03/24/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) colonizes the intestine of young pigs causing severe diarrhoea and consequently bringing high production costs. The rise of antibiotic selective pressure together with ongoing limitations on their use, demands new strategies to tackle this pathology. The pertinence of using bacteriophages as an alternative is being explored, and in this work, the efficacy of phage vB_EcoM_FJ1 (FJ1) in reducing the load of ETEC EC43-Ph (serotype O9:H9 expressing the enterotoxin STa and two adhesins F5 and F41) was assessed. Foreseeing the oral application on piglets, FJ1 was encapsulated on calcium carbonate and alginate microparticles, thus preventing phage release under adverse conditions of the simulated gastric fluid (pH 3.0) and allowing phage availability in simulated intestinal fluid (pH 6.5). A single dose of encapsulated FJ1, provided to IPEC-1 cultured cells (from intestinal epithelium of piglets) previously infected by EC43, provided bacterial reductions of about 99.9% after 6 h. Although bacteriophage-insensitive mutants (BIMs) have emerged from treatment, the consequent fitness costs associated with this new phenotype were demonstrated, comparatively to the originating strain. The higher competence of the pig complement system to decrease BIMs' viability, the lower level of colonization of IPEC-1 cells observed with these mutants, and the increased survival rates and health index recorded in infected Galleria mellonella larvae supported this observation. Most of all, FJ1 established a proof-of-concept of the efficiency of phages to fight against ETEC in piglet intestinal cells.
Collapse
Affiliation(s)
- Alice Ferreira
- ALS ControlVet, Zona Industrial de Tondela ZIMII, Lote 6, 3460-605, Tondela, Portugal
| | - Daniela Silva
- ALS ControlVet, Zona Industrial de Tondela ZIMII, Lote 6, 3460-605, Tondela, Portugal
| | - Carina Almeida
- ALS ControlVet, Zona Industrial de Tondela ZIMII, Lote 6, 3460-605, Tondela, Portugal
| | - Maria Elisa Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS- Associate Laboratory, 4800-122, Guimarães, Portugal
| | - Sónia Silva
- I.P - National Institute for Agrarian and Veterinarian Research (INIAV), Rua Dos Lagidos, 4485-655, Vila Do Conde, Portugal
| | - Joana Castro
- I.P - National Institute for Agrarian and Veterinarian Research (INIAV), Rua Dos Lagidos, 4485-655, Vila Do Conde, Portugal
| | - Dalila Mil-Homens
- Institute for Bioengineering and Biosciences (IBB), Instituto Superior Técnico, 1049-001, Lisbon, Portugal
| | - Isidro García-Meniño
- Laboratorio de Referencia de Escherichia Coli (LREC), Departamento de Microbioloxía E Parasitoloxía, Facultade de Veterinaria, Universidade de Santiago de Compostela (USC), 27002, Lugo, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
- Department for Biological Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Azucena Mora
- Laboratorio de Referencia de Escherichia Coli (LREC), Departamento de Microbioloxía E Parasitoloxía, Facultade de Veterinaria, Universidade de Santiago de Compostela (USC), 27002, Lugo, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Mariana Henriques
- CEB - Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
- LABBELS- Associate Laboratory, 4800-122, Guimarães, Portugal
| | - Ana Oliveira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal.
- LABBELS- Associate Laboratory, 4800-122, Guimarães, Portugal.
| |
Collapse
|
2
|
Farfán J, Gonzalez JM, Vives M. The immunomodulatory potential of phage therapy to treat acne: a review on bacterial lysis and immunomodulation. PeerJ 2022; 10:e13553. [PMID: 35910763 PMCID: PMC9332329 DOI: 10.7717/peerj.13553] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/17/2022] [Indexed: 01/17/2023] Open
Abstract
Background Characterized by an inflammatory pathogenesis, acne is the most common skin disorder worldwide. Altered sebum production, abnormal proliferation of keratinocytes, and microbiota dysbiosis represented by disbalance in Cutibacterium acnes population structure, have a synergic effect on inflammation of acne-compromised skin. Although the role of C. acnes as a single factor in acne development is still under debate, it is known that skin and skin-resident immune cells recognize this bacterium and produce inflammatory markers as a result. Control of the inflammatory response is frequently the target for acne treatment, using diverse chemical or physical agents including antibiotics. However, some of these treatments have side effects that compromise patient adherence and drug safety and in the case of antibiotics, it has been reported C. acnes resistance to these molecules. Phage therapy is an alternative to treat antibiotic-resistant bacterial strains and have been recently proposed as an immunomodulatory therapy. Here, we explore this perspective about phage therapy for acne, considering the potential immunomodulatory role of phages. Methodology Literature review was performed using four different databases (Europe PubMed Central-ePMC, Google Scholar, PubMed, and ScienceDirect). Articles were ordered and selected according to their year of publication, number of citations, and quartile of the publishing journal. Results The use of lytic bacteriophages to control bacterial infections has proven its promising results, and anti-inflammatory effects have been found for some bacteriophages and phage therapy. These effects can be related to bacterial elimination or direct interaction with immune cells that result in the regulation of pro-inflammatory cytokines. Studies on C. acnes bacteriophages have investigated their lytic activity, genomic structure, and stability on different matrices. However, studies exploring the potential of immunomodulation of these bacteriophages are still scarce. Conclusions C. acnes bacteriophages, as well as other phages, may have direct immunomodulatory effects that are yet to be fully elucidated. To our knowledge, to the date that this review was written, there are only two studies that investigate anti-inflammatory properties for C. acnes bacteriophages. In those studies, it has been evidenced reduction of pro-inflammatory response to C. acnes inoculation in mice after bacteriophage application. Nevertheless, these studies were conducted in mice, and the interaction with the immune response was not described. Phage therapy to treat acne can be a suitable therapeutic alternative to C. acnes control, which in turn can aid to restore the skin's balance of microbiota. By controlling C. acnes colonization, C. acnes bacteriophages can reduce inflammatory reactions triggered by this bacterium.
Collapse
Affiliation(s)
- Juan Farfán
- Biological Sciences Department, Faculty of Science, Universidad de Los Andes, Bogotá, Bogotá D.C., Colombia
| | - John M. Gonzalez
- School of Medicine, Universidad de Los Andes, Bogotá, Bogotá D.C., Colombia
| | - Martha Vives
- Biological Sciences Department, Faculty of Science, Universidad de Los Andes, Bogotá, Bogotá D.C., Colombia
| |
Collapse
|
3
|
Bakuradze N, Merabishvili M, Makalatia K, Kakabadze E, Grdzelishvili N, Wagemans J, Lood C, Chachua I, Vaneechoutte M, Lavigne R, Pirnay JP, Abiatari I, Chanishvili N. In Vitro Evaluation of the Therapeutic Potential of Phage VA7 against Enterotoxigenic Bacteroides fragilis Infection. Viruses 2021; 13:2044. [PMID: 34696475 PMCID: PMC8538522 DOI: 10.3390/v13102044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022] Open
Abstract
Since the beginning of the 20th century, bacteriophages (phages), i.e., viruses that infect bacteria, have been used as antimicrobial agents for treating various infections. Phage preparations targeting a number of bacterial pathogens are still in use in the post-Soviet states and are experiencing a revival in the Western world. However, phages have never been used to treat diseases caused by Bacteroides fragilis, the leading agent cultured in anaerobic abscesses and postoperative peritonitis. Enterotoxin-producing strains of B. fragilis have been associated with the development of inflammatory diarrhea and colorectal carcinoma. In this study, we evaluated the molecular biosafety and antimicrobial properties of novel phage species vB_BfrS_VA7 (VA7) lysate, as well as its impact on cytokine IL-8 production in an enterotoxigenic B. fragilis (ETBF)-infected colonic epithelial cell (CEC) culture model. Compared to untreated infected cells, the addition of phage VA7 to ETBF-infected CECs led to significantly reduced bacterial counts and IL-8 levels. This in vitro study confirms the potential of phage VA7 as an antibacterial agent for use in prophylaxis or in the treatment of B. fragilis infections and associated colorectal carcinoma.
Collapse
Affiliation(s)
- Nata Bakuradze
- Research & Development Department, George Eliava Institute of Bacteriophage, Microbiology and Virology, Tbilisi 0160, Georgia; (M.M.); (K.M.); (E.K.); (N.G.); (N.C.)
- Department of Biology, Faculty of Exact and Natural Sciences, Javakhishvili Tbilisi State University, Tbilisi 0179, Georgia
| | - Maya Merabishvili
- Research & Development Department, George Eliava Institute of Bacteriophage, Microbiology and Virology, Tbilisi 0160, Georgia; (M.M.); (K.M.); (E.K.); (N.G.); (N.C.)
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, 1120 Brussels, Belgium;
- Laboratory Bacteriology Research, Ghent University, 9000 Ghent, Belgium;
| | - Khatuna Makalatia
- Research & Development Department, George Eliava Institute of Bacteriophage, Microbiology and Virology, Tbilisi 0160, Georgia; (M.M.); (K.M.); (E.K.); (N.G.); (N.C.)
- Faculty of Medicine, Teaching University Geomedi, Tbilisi 0114, Georgia
| | - Elene Kakabadze
- Research & Development Department, George Eliava Institute of Bacteriophage, Microbiology and Virology, Tbilisi 0160, Georgia; (M.M.); (K.M.); (E.K.); (N.G.); (N.C.)
| | - Nino Grdzelishvili
- Research & Development Department, George Eliava Institute of Bacteriophage, Microbiology and Virology, Tbilisi 0160, Georgia; (M.M.); (K.M.); (E.K.); (N.G.); (N.C.)
- Institute of Medical and Public Health Research, IIia State University, Tbilisi 0162, Georgia; (I.C.); (I.A.)
| | - Jeroen Wagemans
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, 3001 Leuven, Belgium; (J.W.); (C.L.); (R.L.)
| | - Cedric Lood
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, 3001 Leuven, Belgium; (J.W.); (C.L.); (R.L.)
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium
| | - Irakli Chachua
- Institute of Medical and Public Health Research, IIia State University, Tbilisi 0162, Georgia; (I.C.); (I.A.)
- School of Medicine, New Vision University, Tbilisi 0159, Georgia
| | - Mario Vaneechoutte
- Laboratory Bacteriology Research, Ghent University, 9000 Ghent, Belgium;
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, 3001 Leuven, Belgium; (J.W.); (C.L.); (R.L.)
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, 1120 Brussels, Belgium;
| | - Ivane Abiatari
- Institute of Medical and Public Health Research, IIia State University, Tbilisi 0162, Georgia; (I.C.); (I.A.)
| | - Nina Chanishvili
- Research & Development Department, George Eliava Institute of Bacteriophage, Microbiology and Virology, Tbilisi 0160, Georgia; (M.M.); (K.M.); (E.K.); (N.G.); (N.C.)
- School of Medicine, New Vision University, Tbilisi 0159, Georgia
| |
Collapse
|
4
|
Ramos-Vivas J, Superio J, Galindo-Villegas J, Acosta F. Phage Therapy as a Focused Management Strategy in Aquaculture. Int J Mol Sci 2021; 22:10436. [PMID: 34638776 PMCID: PMC8508683 DOI: 10.3390/ijms221910436] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Therapeutic bacteriophages, commonly called as phages, are a promising potential alternative to antibiotics in the management of bacterial infections of a wide range of organisms including cultured fish. Their natural immunogenicity often induces the modulation of a variated collection of immune responses within several types of immunocytes while promoting specific mechanisms of bacterial clearance. However, to achieve standardized treatments at the practical level and avoid possible side effects in cultivated fish, several improvements in the understanding of their biology and the associated genomes are required. Interestingly, a particular feature with therapeutic potential among all phages is the production of lytic enzymes. The use of such enzymes against human and livestock pathogens has already provided in vitro and in vivo promissory results. So far, the best-understood phages utilized to fight against either Gram-negative or Gram-positive bacterial species in fish culture are mainly restricted to the Myoviridae and Podoviridae, and the Siphoviridae, respectively. However, the current functional use of phages against bacterial pathogens of cultured fish is still in its infancy. Based on the available data, in this review, we summarize the current knowledge about phage, identify gaps, and provide insights into the possible bacterial control strategies they might represent for managing aquaculture-related bacterial diseases.
Collapse
Affiliation(s)
- José Ramos-Vivas
- Grupo de Investigación en Acuicultura, Universidad de Las Palmas de Gran Canaria, 35214 Las Palmas de Gran Canaria, Spain; (J.R.-V.); (F.A.)
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, 39011 Santander, Spain
- Department of Project Management, Universidad Internacional Iberoamericana, Campeche 24560, Mexico
| | - Joshua Superio
- Faculty of Biosciences and Aquaculture, Nord University, 8049 Bodø, Norway;
| | | | - Félix Acosta
- Grupo de Investigación en Acuicultura, Universidad de Las Palmas de Gran Canaria, 35214 Las Palmas de Gran Canaria, Spain; (J.R.-V.); (F.A.)
| |
Collapse
|
5
|
Podlacha M, Grabowski Ł, Kosznik-Kawśnicka K, Zdrojewska K, Stasiłojć M, Węgrzyn G, Węgrzyn A. Interactions of Bacteriophages with Animal and Human Organisms-Safety Issues in the Light of Phage Therapy. Int J Mol Sci 2021; 22:8937. [PMID: 34445641 PMCID: PMC8396182 DOI: 10.3390/ijms22168937] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bacteriophages are viruses infecting bacterial cells. Since there is a lack of specific receptors for bacteriophages on eukaryotic cells, these viruses were for a long time considered to be neutral to animals and humans. However, studies of recent years provided clear evidence that bacteriophages can interact with eukaryotic cells, significantly influencing the functions of tissues, organs, and systems of mammals, including humans. In this review article, we summarize and discuss recent discoveries in the field of interactions of phages with animal and human organisms. Possibilities of penetration of bacteriophages into eukaryotic cells, tissues, and organs are discussed, and evidence of the effects of phages on functions of the immune system, respiratory system, central nervous system, gastrointestinal system, urinary tract, and reproductive system are presented and discussed. Modulations of cancer cells by bacteriophages are indicated. Direct and indirect effects of virulent and temperate phages are discussed. We conclude that interactions of bacteriophages with animal and human organisms are robust, and they must be taken under consideration when using these viruses in medicine, especially in phage therapy, and in biotechnological applications.
Collapse
Affiliation(s)
- Magdalena Podlacha
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Łukasz Grabowski
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| | - Katarzyna Kosznik-Kawśnicka
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| | - Karolina Zdrojewska
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Małgorzata Stasiłojć
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (K.Z.); (M.S.); (G.W.)
| | - Alicja Węgrzyn
- Laboratory of Phage Therapy, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Kładki 24, 80-822 Gdansk, Poland; (Ł.G.); (K.K.-K.)
| |
Collapse
|
6
|
Chechushkov A, Kozlova Y, Baykov I, Morozova V, Kravchuk B, Ushakova T, Bardasheva A, Zelentsova E, Allaf LA, Tikunov A, Vlassov V, Tikunova N. Influence of Caudovirales Phages on Humoral Immunity in Mice. Viruses 2021; 13:1241. [PMID: 34206836 PMCID: PMC8310086 DOI: 10.3390/v13071241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022] Open
Abstract
Bacteriophages are promising antibacterial agents. Although they have been recognized as bacterial viruses and are considered to be non-interacting with eukaryotic cells, there is growing evidence that phages may have a significant impact on the immune system via interactions with macrophages, neutrophils, and T-cell polarization. In this study, the influence of phages of podovirus, siphovirus, and myovirus morphotypes on humoral immunity of CD-1 mice was investigated. In addition, tissue distribution of the phages was tested in these mice. No common patterns were found either in the distribution of phages in mice or in changes in the levels of cytokines in the sera of mice once injected with phages. Importantly, pre-existing IgM-class antibodies directed against capsid proteins of phages with myovirus and siphovirus morphotypes were identified in mice before immunization. After triple immunization of CD1-mice with phages without any adjuvant, levels of anti-phage serum polyclonal IgG antibodies increased. Immunogenic phage proteins recognized by IgM and/or IgG antibodies were identified using Western blot analysis and mass spectrometry. In addition, mice serum collected after immunization demonstrated neutralizing properties, leading to a substantial decrease in infectivity of investigated phages with myovirus and siphovirus morphotypes. Moreover, serum samples collected before administration of these phages exhibited some ability to reduce the phage infectivity. Furthermore, Proteus phage PM16 with podovirus morphotype did not elicit IgM or IgG antibodies in immunized mice, and no neutralizing activities against PM16 were revealed in mouse serum samples before and after immunization.
Collapse
Affiliation(s)
- Anton Chechushkov
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Yuliya Kozlova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Ivan Baykov
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Vera Morozova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Bogdana Kravchuk
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Tatyana Ushakova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Alevtina Bardasheva
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Ekaterina Zelentsova
- International Tomography Center Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Lina Al Allaf
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Artem Tikunov
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Valentin Vlassov
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| | - Nina Tikunova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.C.); (Y.K.); (I.B.); (V.M.); (B.K.); (T.U.); (A.B.); (L.A.A.); (A.T.); (V.V.)
| |
Collapse
|
7
|
Aghaee BL, Mirzaei MK, Alikhani MY, Mojtahedi A. Sewage and sewage-contaminated environments are the most prominent sources to isolate phages against Pseudomonas aeruginosa. BMC Microbiol 2021; 21:132. [PMID: 33931013 PMCID: PMC8088035 DOI: 10.1186/s12866-021-02197-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 04/15/2021] [Indexed: 01/21/2023] Open
Abstract
Background P. aeruginosa is the primary source of hospital-acquired infections. Unfortunately, antibiotic resistance is growing to precariously high levels, making the infections by this pathogen life-threatening and hard to cure. One possible alternative to antibiotics is to use phages. However, the isolation of phages suitable for phage therapy— be lytic, be efficient, and have a broad host range —against some target bacteria has proven difficult. To identify the best places to look for these phages against P. aeruginosa we screened hospital sewages, soils, and rivers in two cities. Results We isolated eighteen different phages, determined their host range, infection property, and plaque morphology. We found that the sewage and sewage-contaminated environments are the most reliable sources for the isolation of Pseudomonas phages. In addition, phages isolated from hospital sewage showed the highest efficiency in lysing the bacteria used for host range determination. In contrast, phages from the river had larger plaque size and lysed bacteria with higher levels of antibiotic resistance. Conclusions Our findings provided additional support for the importance of sewage as the source of phage isolation. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02197-z.
Collapse
Affiliation(s)
- Bahareh Lashtoo Aghaee
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Center Munich and Technical University of Munich, 85764, Neuherberg, Bavaria, Germany
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Brucellosis research center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
8
|
Aghaee BL, Khan Mirzaei M, Alikhani MY, Mojtahedi A, Maurice CF. Improving the Inhibitory Effect of Phages against Pseudomonas aeruginosa Isolated from a Burn Patient Using a Combination of Phages and Antibiotics. Viruses 2021; 13:334. [PMID: 33670028 PMCID: PMC7926668 DOI: 10.3390/v13020334] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Antibiotic resistance causes around 700,000 deaths a year worldwide. Without immediate action, we are fast approaching a post-antibiotic era in which common infections can result in death. Pseudomonas aeruginosa is the leading cause of nosocomial infection and is also one of the three bacterial pathogens in the WHO list of priority bacteria for developing new antibiotics against. A viable alternative to antibiotics is to use phages, which are bacterial viruses. Yet, the isolation of phages that efficiently kill their target bacteria has proven difficult. Using a combination of phages and antibiotics might increase treatment efficacy and prevent the development of resistance against phages and/or antibiotics, as evidenced by previous studies. Here, in vitro populations of a Pseudomonas aeruginosa strain isolated from a burn patient were treated with a single phage, a mixture of two phages (used simultaneously and sequentially), and the combination of phages and antibiotics (at sub-minimum inhibitory concentration (MIC) and MIC levels). In addition, we tested the stability of these phages at different temperatures, pH values, and in two burn ointments. Our results show that the two-phages-one-antibiotic combination had the highest killing efficiency against the P. aeruginosa strain. The phages tested showed low stability at high temperatures, acidic pH values, and in the two ointments. This work provides additional support for the potential of using combinations of phage-antibiotic cocktails at sub-MIC levels for the treatment of multidrug-resistant P. aeruginosa infections.
Collapse
Affiliation(s)
- Bahareh Lashtoo Aghaee
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 65178-38678, Iran;
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Center Munich and Technical University of Munich, 85764 Neuherberg, Germany;
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 0B1, Canada
| | - Mohammad Yousef Alikhani
- Department of Microbiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan 65178-38678, Iran;
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht 41938-33697, Iran
| | - Corinne F. Maurice
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 0B1, Canada
| |
Collapse
|
9
|
Górski A, Borysowski J, Miȩdzybrodzki R. Bacteriophage Interactions With Epithelial Cells: Therapeutic Implications. Front Microbiol 2021; 11:631161. [PMID: 33537024 PMCID: PMC7848012 DOI: 10.3389/fmicb.2020.631161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Andrzej Górski
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland.,Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland.,Infant Jesus Hospital, The Medical University of Warsaw, Warsaw, Poland
| | - Jan Borysowski
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| | - Ryszard Miȩdzybrodzki
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland.,Phage Therapy Unit, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland.,Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Derome N, Filteau M. A continuously changing selective context on microbial communities associated with fish, from egg to fork. Evol Appl 2020; 13:1298-1319. [PMID: 32684960 PMCID: PMC7359827 DOI: 10.1111/eva.13027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Fast increase of fish aquaculture production to meet consumer demands is accompanied by important ecological concerns such as disease outbreaks. Meanwhile, food waste is an important concern with fish products since they are highly perishable. Recent aquaculture and fish product microbiology, and more recently, microbiota research, paved the way to a highly integrated approach to understand complex relationships between host fish, product and their associated microbial communities at health/disease and preservation/spoilage frontiers. Microbial manipulation strategies are increasingly validated as promising tools either to replace or to complement traditional veterinary and preservation methods. In this review, we consider evolutionary forces driving fish microbiota assembly, in particular the changes in the selective context along the production chain. We summarize the current knowledge concerning factors governing assembly and dynamics of fish hosts and food microbial communities. Then, we discuss the current microbial community manipulation strategies from an evolutionary standpoint to provide a perspective on the potential for risks, conflict and opportunities. Finally, we conclude that to harness evolutionary forces in the development of sustainable microbiota manipulation applications in the fish industry, an integrated knowledge of the controlling abiotic and especially biotic factors is required.
Collapse
Affiliation(s)
- Nicolas Derome
- Institut de Biologie Intégrative et des Systèmes (IBIS)Université LavalQuébecQCCanada
- Département de BiologieUniversité LavalQuébecQCCanada
| | - Marie Filteau
- Département de BiologieUniversité LavalQuébecQCCanada
- Département des Sciences des alimentsInstitut sur la nutrition et les aliments fonctionnels (INAF)Université LavalQuébecQCCanada
| |
Collapse
|
11
|
Yıldızlı G, Coral G, Ayaz F. Immunostimulatory Activities of Coliphages on In Vitro Activated Mammalian Macrophages. Inflammation 2019; 43:595-604. [DOI: 10.1007/s10753-019-01140-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
12
|
Abstract
Clinical trial results of phage treatment of bacterial infections show a low to moderate efficacy, and the variation in infection clearance between subjects within studies is often large. Phage therapy is complicated and introduces many additional components of variance as compared to antibiotic treatment. A large part of the variation is due to in vivo pharmacokinetics and pharmacodynamics being virtually unknown, but also to a lack of standardisation. This is a consequence of the great variation of phages, bacteria, and infections, which results in different experiments or trials being impossible to compare, and difficulties in estimating important parameter values in a quantitative and reproducible way. The limitations of phage therapy will have to be recognised and future research focussed on optimising infection clearance rates by e.g. selecting phages, bacteria, and target bacterial infections where the prospects of high efficacy can be anticipated, and by combining information from new mathematical modelling of in vivo pharmacokinetic and pharmacodynamic processes and quantitatively assessed experiments.
Collapse
Affiliation(s)
- Anders S. Nilsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
13
|
Efficacy and safety assessment of two enterococci phages in an in vitro biofilm wound model. Sci Rep 2019; 9:6643. [PMID: 31040333 PMCID: PMC6491613 DOI: 10.1038/s41598-019-43115-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/18/2019] [Indexed: 01/22/2023] Open
Abstract
Chronic wounds affect thousands of people worldwide, causing pain and discomfort to patients and represent significant economical burdens to health care systems. The treatment of chronic wounds is very difficult and complex, particularly when wounds are colonized by bacterial biofilms which are highly tolerant to antibiotics. Enterococcus faecium and Enterococcus faecalis are within the most frequent bacteria present in chronic wounds. Bacteriophages (phages) have been proposed as an efficient and alternative against antibiotic-resistant infections, as those found in chronic wounds. We have isolated and characterized two novel enterococci phages, the siphovirus vB_EfaS-Zip (Zip) and the podovirus vB_EfaP-Max (Max) to be applied during wound treatment. Both phages demonstrated lytic behavior against E. faecalis and E. faecium. Genome analysis of both phages suggests the absence of genes associated with lysogeny. A phage cocktail containing both phages was tested against biofilms formed in wound simulated conditions at a multiplicity of infection of 1.0 and a 2.5 log CFU.mL−1 reduction in the bacterial load after at 3 h of treatment was observed. Phages were also tested in epithelial cells colonized by these bacterial species and a 3 log CFU.mL−1 reduction was observed using both phages. The high efficacy of these new isolated phages against multi-species biofilms, their stability at different temperatures and pH ranges, short latent periods and non-cytotoxicity to epithelial cells suggest their therapeutic use to control infectious biofilms present in chronic wounds.
Collapse
|
14
|
Pearce SC, Coia HG, Karl JP, Pantoja-Feliciano IG, Zachos NC, Racicot K. Intestinal in vitro and ex vivo Models to Study Host-Microbiome Interactions and Acute Stressors. Front Physiol 2018; 9:1584. [PMID: 30483150 PMCID: PMC6240795 DOI: 10.3389/fphys.2018.01584] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
The gut microbiome is extremely important for maintaining homeostasis with host intestinal epithelial, neuronal, and immune cells and this host-microbe interaction is critical during times of stress or disease. Environmental, nutritional, and cognitive stress are just a few factors known to influence the gut microbiota and are thought to induce microbial dysbiosis. Research on this bidirectional relationship as it pertains to health and disease is extensive and rapidly expanding in both in vivo and in vitro/ex vivo models. However, far less work has been devoted to studying effects of host-microbe interactions on acute stressors and performance, the underlying mechanisms, and the modulatory effects of different stressors on both the host and the microbiome. Additionally, the use of in vitro/ex vivo models to study the gut microbiome and human performance has not been researched extensively nor reviewed. Therefore, this review aims to examine current evidence concerning the current status of in vitro and ex vivo host models, the impact of acute stressors on gut physiology/microbiota as well as potential impacts on human performance and how we can parlay this information for DoD relevance as well as the broader scientific community. Models reviewed include widely utilized intestinal cell models from human and animal models that have been applied in the past for stress or microbiology research as well as ex vivo organ/tissue culture models and new innovative models including organ-on-a-chip and co-culture models.
Collapse
Affiliation(s)
- Sarah C Pearce
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Heidi G Coia
- National Research Council, The National Academies of Sciences, Engineering, and Medicine, Washington, DC, United States.,711th Human Performance Wing, Airforce Research Laboratory, Airman Systems Directorate, Human-Centered ISR Division, Molecular Mechanisms Branch, Wright-Patterson Air Force Base, Dayton, OH, United States
| | - J P Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Ida G Pantoja-Feliciano
- Soldier Protection and Optimization Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| | - Nicholas C Zachos
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kenneth Racicot
- Performance Nutrition Team, Combat Feeding Directorate, Natick Soldier Research, Development and Engineering Center, Natick, MA, United States
| |
Collapse
|
15
|
Effects of Staphylococcus aureus Bacteriophage K on Expression of Cytokines and Activation Markers by Human Dendritic Cells In Vitro. Viruses 2018; 10:v10110617. [PMID: 30413044 PMCID: PMC6266804 DOI: 10.3390/v10110617] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/03/2018] [Accepted: 11/06/2018] [Indexed: 01/21/2023] Open
Abstract
A potential concern with bacteriophage (phage) therapeutics is a host-versus-phage response in which the immune system may neutralize or destroy phage particles and thus impair therapeutic efficacy, or a strong inflammatory response to repeated phage exposure might endanger the patient. Current literature is discrepant with regard to the nature and magnitude of innate and adaptive immune response to phages. The purpose of this work was to study the potential effects of Staphylococcus aureus phage K on the activation of human monocyte-derived dendritic cells. Since phage K acquired from ATCC was isolated around 90 years ago, we first tested its activity against a panel of 36 diverse S. aureus clinical isolates from military patients and found that it was lytic against 30/36 (83%) of strains. Human monocyte-derived dendritic cells were used to test for an in vitro phage-specific inflammatory response. Repeated experiments demonstrated that phage K had little impact on the expression of pro- and anti-inflammatory cytokines, or on MHC-I/II and CD80/CD86 protein expression. Given that dendritic cells are potent antigen-presenting cells and messengers between the innate and the adaptive immune systems, our results suggest that phage K does not independently affect cellular immunity or has a very limited impact on it.
Collapse
|
16
|
Shan J, Ramachandran A, Thanki AM, Vukusic FBI, Barylski J, Clokie MRJ. Bacteriophages are more virulent to bacteria with human cells than they are in bacterial culture; insights from HT-29 cells. Sci Rep 2018; 8:5091. [PMID: 29572482 PMCID: PMC5865146 DOI: 10.1038/s41598-018-23418-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
Bacteriophage therapeutic development will clearly benefit from understanding the fundamental dynamics of in vivo phage-bacteria interactions. Such information can inform animal and human trials, and much can be ascertained from human cell-line work. We have developed a human cell-based system using Clostridium difficile, a pernicious hospital pathogen with limited treatment options, and the phage phiCDHS1 that effectively kills this bacterium in liquid culture. The human colon tumorigenic cell line HT-29 was used because it simulates the colon environment where C. difficile infection occurs. Studies on the dynamics of phage-bacteria interactions revealed novel facets of phage biology, showing that phage can reduce C. difficile numbers more effectively in the presence of HT-29 cells than in vitro. Both planktonic and adhered Clostridial cell numbers were successfully reduced. We hypothesise and demonstrate that this observation is due to strong phage adsorption to the HT-29 cells, which likely promotes phage-bacteria interactions. The data also showed that the phage phiCDHS1 was not toxic to HT-29 cells, and phage-mediated bacterial lysis did not cause toxin release and cytotoxic effects. The use of human cell lines to understand phage-bacterial dynamics offers valuable insights into phage biology in vivo, and can provide informative data for human trials.
Collapse
Affiliation(s)
- Jinyu Shan
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK.
| | - Ananthi Ramachandran
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Anisha M Thanki
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Fatima B I Vukusic
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Jakub Barylski
- Department of Molecular Virology, Faculty of Biology, Adam Mickiewicz University, 61-614, Poznan, Poland
| | - Martha R J Clokie
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
17
|
Abedon ST. Bacteriophage Clinical Use as Antibacterial "Drugs": Utility and Precedent. Microbiol Spectr 2017; 5:10.1128/microbiolspec.bad-0003-2016. [PMID: 28840811 PMCID: PMC11687515 DOI: 10.1128/microbiolspec.bad-0003-2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Indexed: 12/25/2022] Open
Abstract
For phage therapy-the treatment of bacterial infections using bacterial viruses-a key issue is the conflict between apparent ease of clinical application, on the one hand, and on the other hand, numerous difficulties that can be associated with undertaking preclinical development. These conflicts between achieving efficacy in the real world versus rigorously understanding that efficacy should not be surprising because equivalent conflicts have been observed in applied biology for millennia: exploiting the inherent, holistic tendencies of useful systems, e.g., of dairy cows, inevitably is easier than modeling those systems or maintaining effectiveness while reducing such systems to isolated parts. Trial and error alone, in other words, can be a powerful means toward technological development. Undertaking trial and error-based programs, especially in the clinic, nonetheless is highly dependent on those technologies possessing both inherent safety and intrinsic tendencies toward effectiveness, but in this modern era we tend to forget that ideally there would exist antibacterials which could be thus developed, that is, with tendencies toward both safety and effectiveness, and which are even relatively inexpensive. Consequently, we tend to demand rigor as well as expense of development even to the point of potentially squandering such utility, were it to exist. In this review I lay out evidence that in phage therapy such potential, in fact, does exist. Advancement of phage therapy unquestionably requires effective regulation as well as rigorous demonstration of efficacy, but after nearly 100 years of clinical practice, perhaps not as much emphasis on strictly laboratory-based proof of principle.
Collapse
Affiliation(s)
- Stephen T Abedon
- Department of Microbiology, The Ohio State University, Mansfield, OH 44906
| |
Collapse
|
18
|
Drilling AJ, Ooi ML, Miljkovic D, James C, Speck P, Vreugde S, Clark J, Wormald PJ. Long-Term Safety of Topical Bacteriophage Application to the Frontal Sinus Region. Front Cell Infect Microbiol 2017; 7:49. [PMID: 28286740 PMCID: PMC5323412 DOI: 10.3389/fcimb.2017.00049] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/08/2017] [Indexed: 12/13/2022] Open
Abstract
Background:Staphylococcus aureus biofilms contribute negatively to a number of chronic conditions, including chronic rhinosinusitis (CRS). With the inherent tolerance of biofilm-bound bacteria to antibiotics and the global problem of bacterial antibiotic resistance, the need to develop novel therapeutics is paramount. Phage therapy has previously shown promise in treating sinonasal S. aureus biofilms. Methods: This study investigates the long term (20 days) safety of topical sinonasal flushes with bacteriophage suspensions. The bacteriophage cocktail NOV012 against S. aureus selected for this work contains two highly characterized and different phages, P68 and K710. Host range was assessed against S. aureus strains isolated from CRS patients using agar spot tests. NOV012 was applied topically to the frontal sinus region of sheep, twice daily for 20 days. General sheep wellbeing, mucosal structural changes and inflammatory load were assessed to determine safety of NOV012 application. Results: NOV012 could lyse 52/61 (85%) of a panel of locally derived CRS clinical isolates. Application of NOV012 to the frontal sinuses of sheep for 20 days was found to be safe, with no observed inflammatory infiltration or tissue damage within the sinus mucosa. Conclusion: NOV012 cocktail appears safe to apply for extended periods to sheep sinuses and it could infect and lyse a wide range of S. aureus CRS clinical isolates. This indicates that phage therapy has strong potential as a treatment for chronic bacterial rhinosinusitis.
Collapse
Affiliation(s)
- Amanda J Drilling
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide Adelaide, SA, Australia
| | - Mian L Ooi
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide Adelaide, SA, Australia
| | - Dijana Miljkovic
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide Adelaide, SA, Australia
| | - Craig James
- Adelaide Pathology Partners Adelaide, SA, Australia
| | - Peter Speck
- School of Biological Sciences, Flinders University Bedford Park, SA, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide Adelaide, SA, Australia
| | | | - Peter-John Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide Adelaide, SA, Australia
| |
Collapse
|
19
|
Dufour N, Henry M, Ricard JD, Debarbieux L. Commentary: Morphologically Distinct Escherichia coli Bacteriophages Differ in Their Efficacy and Ability to Stimulate Cytokine Release In Vitro. Front Microbiol 2016; 7:1029. [PMID: 27443304 PMCID: PMC4923138 DOI: 10.3389/fmicb.2016.01029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/17/2016] [Indexed: 11/28/2022] Open
Affiliation(s)
- Nicolas Dufour
- AP-HP, Hôpital Louis Mourier, Service de Réanimation Médico-ChirurgicaleColombes, France; INSERM, Infection Antimicrobials Modelling Evolution, UMR 1137Paris, France; Department of Microbiology, Molecular Biology of Gene in Extremophiles, Institut PasteurParis, France
| | - Marine Henry
- Department of Microbiology, Molecular Biology of Gene in Extremophiles, Institut Pasteur Paris, France
| | - Jean-Damien Ricard
- AP-HP, Hôpital Louis Mourier, Service de Réanimation Médico-ChirurgicaleColombes, France; INSERM, Infection Antimicrobials Modelling Evolution, UMR 1137Paris, France; Université Paris Diderot, Infection Antimicrobials Modelling Evolution, UMR 1137Paris, France
| | - Laurent Debarbieux
- Department of Microbiology, Molecular Biology of Gene in Extremophiles, Institut Pasteur Paris, France
| |
Collapse
|