Search results

Filters

  • Journals
  • Authors
  • Contributor
  • Keywords
  • Date
  • Type

Search results

Number of results: 15
items per page: 25 50 75
Sort by:
Download PDF Download RIS Download Bibtex

Abstract

We talk to Prof. Andrzej Górski from the Hirszfeld Institute of Immunology and Experimental Therapy about what is going wrong with antibiotics and whether they might one day be replaced with bacteriophages.

Go to article

Authors and Affiliations

Andrzej Górski
Download PDF Download RIS Download Bibtex

Abstract

22 representative antibiotics, including 8 quinolones (QNs), 9 sulfonamides (SAs), and 5 macrolides (MCs) were selected to investigate their occurrence and removal efficiencies in a Wastewater Treatment Plant (WWTP) and their distribution in the receiving water of the Chaobai River in Beijing, China. Water quality monitoring was performed in an integrated way at different selected points in the WWTP to explore the potential mechanism of antibiotics removal during wastewater treatment. Water quality of the Chaobai River was also analyzed to examine environmental distribution in a river ecosystem. The results showed that within all the 22 compounds examined, 10 antibiotics were quantified in wastewater influent, 10 in effluent, and 7 in river. Sulfadiazine (SDZ, 396 ng/L) and Sulfamethazine (SMZ, 382 ng/L) were the dominating antibiotics in the influent. Both the conventional treatment and advanced Biological Aerated Filter (BAF) system was important for the removal of antibiotics from the wastewater. And the concentrations of selected antibiotics were ranged from 0-41.8 ng/L in the effluent-receiving river. Despite the fact that the concentrations were reduced more than 50% compared to effluent concentrations, WWTP discharge was still regarded as a dominant point-source input of antibiotics into the Chaobai River.

Go to article

Authors and Affiliations

Zhang Chunhui
Wang Liangliang
Gao Xiangyu
He Xudan
Download PDF Download RIS Download Bibtex

Abstract

Vibrio species are common inhabitants of aquatic environments and have been described in connection with fish and human diseases.
Six Vibrio species were isolated from diseased freshwater and ornamental fish in Poland. The strains were identified based on morphological and biochemical characteristics and confirmed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) as V. albensis (n=3) from Gymnocephalus cernua, Sander lucioperca, Paracheirodon innesi, and Xiphophorus hellerii; V. mimicus (n=1) from Xiphophorus maculatus; and V. vulnificus (n=1) from Nematobrycon palmeri. This is the first time that Vibrio species have been isolated and described from ornamental fish in Poland. The isolates were resistant to ampicillin (83.3%), gentamicin (16.6%), ciprofloxacin (16.6%), sulfamethoxazole-trimethoprim (16.6%), and chloramphenicol (16.6%). The multiple antibiotic resistance (MAR) index was 0.00-0.08 for V. albensis, 0.17 for V. mimicus, and 0.33 for V. vulnificus.
Our study confirmed the presence of potentially pathogenic Vibrio species in freshwater and ornamental fish. Therefore, further monitoring of the presence of Vibrio species, mainly in ornamental fish, is necessary.
Go to article

Bibliography

1. Aksak-Wąs BJ, Ripa A, Szakoła P, Horbacka K, Niścigorska-Olsen J, Witak-Jędra M, Zając-Marczewska M, Karasińska-Cieślak M, Kot J, Parczewski M (2021) Septic shock induced by Vibrio vulnificus in Northen Poland, a case report. Infect Drug Resist 14: 5027-5033.
2. Al-Dulaimi MM, Mutalib SA, Ghani MA, Zaini NA, Ariffin AA (2019) Multiple antibiotic resistance (MAR), plasmid profiles, and DNA polymorphisms among Vibrio vulnificus isolates. Antibiotics (Basel) 8: 68.
3. Araj GF, Taleb R, El Beayni NK, Goksu E (2019) Vibrio albensis: An unusual urinary tract infection in a healthy male. J Infect Public Health 12: 712-713.
4. Austin B, Austin DA (2007) Vibrios. In: Austin B, Austin DA (eds) Bacterial fish pathogens. Diseases of farmed and wild fish. Praxis Publishing Ltd, Chichester, UK, pp 499-601.
5. Baker-Austin C, McArthur JV, Lindell AH, Wright MS, Tuckfield RC, Gooch J, Warner L, Oliver J, Stepanauskas R (2009) Multi-site analysis reveals widespread antibiotic resistance in the marine pathogen Vibrio vulnificus. Microb Ecol 57: 151-159.
6. Baker-Austin C, Oliver JD, Alam M, Ali A, Waldor MK, Qadri F, Martinez-Urtaza J (2018) Vibrio spp. infections. Nat Rev Dis Primers 4: 8.
7. Bauer AW, Kirby WM, Sherris JC, Turck M (1966) Antibiotic susceptibility testing by a standardized single disk method. Am J Clin Pathol 45: 493-496.
8. Bisharat N, Agmon V, Finkelstein R, Raz R, Ben-Dror G, Lerner L, Soboh S, Colodner R, Cameron DN, Wykstra DL, Swerdlow DL, Farmer JJ 3rd (1999) Clinical, epidemiological, and microbiological features of Vibrio vulnificus biogroup 3 causing outbreaks of wound infection and bacteraemia in Israel. Israel Vibrio Study Group. Lancet 354: 1421-1424.
9. Cassini A, Högberg LD, Plachouras D, Quattrocchi A, Hoxha A, Simonsen GS, Colomb-Cotinat M, Kretzchmar ME, Devleesschauwer B, Cecchini M, Ouakrim DA, Oliveira TC, Struelens MJ, Suetens C, Monnet DL, Burden of AMR Collaborative Group (2019) At-tributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Eco-nomic Area in 2015: a population-level modelling analysis. Lancet Infect Dis 19: 5-66.
10. Cheng WC, Jan IS, Chen JM, Teng SH, Teng LJ, Sheng WH, Ko WC, Hsueh PR (2015) Evaluation of the Bruker biotyper ma-trix-assisted laser desorption ionization-time of flight mass spectrometry for identification of blood isolates of Vibrio species. J Clin Mi-crobiol 53: 1741-1744.
11. Clinical and Laboratory Standards Institute – CLSI (2015) Methods for antimicrobial dilution and disk susceptibility testing of infre-quently isolated or fastidious bacteria. 3rd ed. CLSI guideline M45. Wayne, PA.
12. Da Silva LV, Ossai S, Chigbu P, Parveen S (2021) Antimicrobial and genetic profiles of Vibrio vulnificus and Vibrio parahaemolyticus isolated from the Maryland Coastal Bays, United States. Front Microbiol 12: 676249.
13. Dieckmann R, Strauch E, Alter T (2010) Rapid identification and characterization of Vibrio species using whole-cell MALDI-TOF mass spectrometry. J Appl Microbiol 109: 199-211.
14. Dong HT, Nguyen VV, Le HD, Sangsuriya P, Jitrakorn S, Saksmerprome V, Senapin S, Rodkhum C (2015) Naturally concurrent in-fections of bacterial and viral pathogens in disease outbreaks in cultured Nile tilapia (Oreochromis niloticus) farms. Aquaculture 448: 427-435.
15. El-Deen AG, Elkamel AA (2015) Clinical and experimental study on vibriosis in ornamental fish. Assiut Vet Med J 61: 147-153.
16. Erler M, Wichels A, Heinemayer EA, Hauk G, Hippelein M, Reyes NT, Gerdts G (2015) VibrioBase: A MALDI-TOF MS database for fast identification of Vibrio spp. that are potentially pathogenic in humans. Syst Appl Microbiol 38: 16-25.
17. Esteve C, Alcaide E, Herraiz S, Canals R, Merino S, Tomás JM (2007) First description of nonmotile Vibrio vulnificus strains virulent for eels. FEMS Microbiol Lett 266: 90-97.
18. Geng Y, Liu D, Han S, Zhou Y, Wang KY, Huang XL, Chen DF, Peng X, Lai WM (2014) Outbreaks of vibriosis associated with Vib-rio mimicus in freshwater catfish in China. Aquaculture 433: 82-84.
19. Huzmi H, Ina-Salvany MY, Natrah FM, Syukri F, Karim M (2019) Strategies of controlling vibriosis in fish. Asian J Appl Sci 7: 513-521.
20. Kiani S, Naghavi NS, Nazari A (2016) Detection of Vibrio species isolated from ornamental guppy fish in Kashan, Isfahan, Iran fish culturing ponds. Biol J Microorg 4: 43-48.
21. Kiiyukia C, Nakajima A, Nakai T, Muroga K, Kawakami H, Hashimoto H (1992) Vibrio cholera non-O1 isolated from ayu fish (Pleco-glossus altivelis) in Japan. Appl Environ Microbiol 58: 3078-3082.
22. Kolada A (ed.) (2022) Wstępny raport ds. sytuacji na rzece Odrze. IOŚ-PIB. https://ios.edu.pl/wp-content/uploads/2022/10/
Wstepny-raport-zespolu-ds.-sytuacji-na-rzece-Odrze.pdf
23. Krumperman PH (1983) Multiple antibiotic resistance indexing Escherichia coli to identify high-risk sources of fecal contamination of foods. Appl Environ Microbiol 46: 165-170.
24. Kumarage PM, De Silva LADS, Heo G-J (2022) Aquatic environments: A potential source of antimicrobial-resistant Vibrio spp. J Appl Microbiol 133: 2267-2279.
25. Kurpas M, Michalska M, Zakrzewski A, Zorena K (2021) First report of the presence of Vibrio vulnificus in the Gulf of Gdansk. Int Ma-rit Health 72: 247-251.
26. Lupiani B, Baya AM, Magariños B, Romalde JL, Li T, Roberson BS, Hetrick FM, Toranzo AE (1993) Vibrio mimicus and Vibrio chol-erae non-01 isolated from wild and hatchery-reared fish. Gyobyo Kenkyu 28: 15-26.
27. Manchanayake T, Salleh A, Amal MN, Yasin IS, Zamri-Saad M (2023) Pathology and pathogenesis of Vibrio infection in fish: A re-view. Aquac Rep 28: 101459.
28. Manfrin A, Friso S, Perin R, Qualtieri K, Bovo G, Rodgers CJ (2001) Tropical fish importation from third countries: the potential risk of introducing human and aquatic animal pathogens. Risk analysis in aquatic animal health. In: Proceedings of an International Conference, OIE Headquarters, Paris, France. 8–10 February 2000, pp 167–169.
29. Noorlis A, Ghazali FM, Cheah YK, Tuan Zainazor TC, Wong WC, Tunung R, Pui CF, Nishibuchi M, Nakaguchi Y, Son R (2011) An-tibiotic resistance and biosafety of Vibrio cholera and Vibrio parahaemolyticus from freshwater fish at retail level. Int Food Res J 18: 1523-1530.
30. Osunla CA, Okoh AI (2017) Vibrio Pathogens: A public health concern in rural water resources in Sub-Saharan Africa. Int J Environ Res Public Health 14: 1188.
31. Rehulka J, Petras P, Marejkova M, Aldova E (2015) Vibrio choleae non-O1/non-O139 infection in fish in the Czech Republic. Vet Med - Czech 60: 16-22.
32. Senderovich Y, Izhaki I, Halpern M (2010) Fish as reservoirs and vectors of Vibrio cholerae. PLoS One 5: e8607
33. Shaw KS, Goldstein RE, He X, Jacobs JM, Crump BC, Sapkota AR (2014) Antimicrobial susceptibility of Vibrio vulnificus and Vibrio parahaemolyticus recovered from recreational and commercial areas of Chesapeake Bay and Maryland Coastal Bays. PLoS One 9: e89616.
34. Stypułkowska-Misiurewicz H, Pancer K, Roszkowiak A (2006) Two unrelated cases of septicaemia due to Vibrio cholerae non-O1, non-O139 in Poland, July and August 2006. Euro Surveill 11: 3088.
35. Stypułkowska-Misiurewicz H, Stasiak J, Janczyk M, Tomaszewska E, Pancer K (1995) Vibrio cholerae non-01 isolated in Poland from the Bug river. Przegl Epidemiol 49: 237-243.
36. Tison DL, Nishibuchi M, Greenwood JD, Seidler RJ (1982) Vibrio vulnificus biogroup 2: new biogroup pathogenic for eels. Appl En-viron Microbiol 44: 640-646.
37. Vandenberghe J, Thompson FL, Gomez-Gil B, Swings J (2003) Phenotypic diversity amongst Vibrio isolates from marine aquaculture systems. Aquaculture 219: 9-20.
38. Warner E, Oliver JD (2008) Population structures of two genotypes of Vibrio vulnificus in oysters (Crassostrea virginica) and seawater. Appl Environ Microbiol 74: 80-85.
39. Zago V, Zambon M, Civettini M, Zaltum O, Manfrin A (2017) Virulence-associated factors in Vibrio cholerae non-O1/non-O139 and V. mimicus strains isolated in ornamental fish species. J Fish Dis 40: 1857-1868.
40. Zhang X, Li YW, Mo ZQ, Luo XC, Sun HY, Liu P, Li AX, Zhou SM, Dan XM (2014) Outbreak of a novel disease associated with Vibrio mimicus infection in fresh water cultured yellow catfish, Pelteobagrus fulvidraco. Aquaculture 432: 119-124.
Go to article

Authors and Affiliations

A. Pastuszka
1
L. Guz
1
K. Michalak
2
D. Pietras-Ożga
2
K. Puk
1

  1. Department of Biology and Fish Diseases, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, 20-950 Lublin, Poland
  2. Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences, Głęboka 30, 20-612 Lublin, Poland
Download PDF Download RIS Download Bibtex

Abstract

This article describes monitoring results of raw wastewater from one Polish municipal wastewater treatment plant (WWTP). The residues of 30 pharmaceutics belonging to particular drugs classes such as contrast media. antibiotics, lipids regulators, antiphologisties, psychiatric and anticpilcptic agents, drug's metabolites and 2 musk compounds have been investigated. The investigation showed occurrence of 20 out of 32 selected compounds above their limit of detection. Iopromide, a compound belonging to contrast media, was noticed at the highest concentration. The concentration of this compound in WWTP-influent was equaled to 27.0 μg/dm3• Other drugs, such as, like iopamidol, iomeprol, diatrizoat, iohexol, sulfomethoxazole, carbamazepine, ibuprofen, ibuprofen-OH, naproxen, diclofenac, bczafibrate, ketoprofen, and musk compound - galaxolide were detected at maximum concentration between I .O μg/dm3 (bezafibratc) and 13.0 μg/dm3 (iomcprol). The acidic compounds such as gemfibrozil and indomethacin were determined above their limit of detection, with concentration up to 0.22 μg/dm3 and 0.42 ug/dm', respectively. Based on the literature data, the above-mentioned drugs arc not completely removed from sewage during treatment processes and with effluent from WWTP they are introduced to receiving waters. Due to their chemical properties, residues of pharmaceutics may persist in the environment and the present knowledge about their ccotoxicological effects is insufficient.
Go to article

Authors and Affiliations

Ewa Felis
Korneliusz Miksch
Joanna Surmacz-Górska
Thomas Ternes
Download PDF Download RIS Download Bibtex

Abstract

This study mainly focused on the current situation of antibiotic pollution in coastal wetlands by screening for four common antibiotics (norfloxacin - NOR, ofloxacin - OFL, azithromycin - AZM, and roxithromycin - RXM) and two coastal wetland plants (Suaeda and Nelumbo nucifera), to determine the removal of antibiotic pollution by phytoremediation technology and its mechanism. We aimed to provide ideas for the remediation of antibiotics in coastal wetlands and their mechanisms of action in the context of intensive farming. The results showed that both plants had remediation effects on all four antibiotics, the phytoremediation of NOR and OFL was particularly significant, and the remediation effect of N. nucifera was better than that of Suaeda . The removal rates of the four antibiotics by Suaeda and N. nucifera at low antibiotic concentrations (10–25 μg/L) reached 48.9%–100% and 77.3%–100%, respectively. The removal rates of the four antibiotics at high antibiotic concentrations (50–200 μg/L) reached 7.5%–73.2% and 22%–84.6%, respectively. Moreover, AZM was only detected in trace amounts in the roots of N. nucifera, and RXM was not detected in either plant body.
Go to article

Bibliography

  1. Blasco, D. (1994). The Ramsar Convention manual: a guide to the Convention on Wetlands of International Importance especially as Waterfowl Habitat. Water 1994.
  2. Burken, J.G. & Schnoor, J.L. (1998). Predictive relationships for uptake of organic contaminants by hybrid poplar trees. Environ. Sci. Technol. 32 (21), 3379-3385. DOI:10.1021/es9706817.
  3. Calheiros, C., Rangel, A.& Castro, P. (2007). Constructed wetland systems vegetated with different plants applied to the treatment of tannery wastewater. Water Res. 41(8), pp. 1790-1798. DOI:10.1016/j.watres.2007.01.012.
  4. Chen, X.J., Li, F.Y. & He, Y.B. (2012). Remediation effect of two kinds of aquatic plants on water contaminated by antibiotics. Subtrop. Plant Sci. 41 (4), 1-7. (in Chinese).
  5. Chiou, C.T., Sheng, G. & Manes, M. (2001). A partition-limited model for the plant uptake of organic contaminants from soil and water. Environ. Sci. Technol. 35 (7), pp. 1437-1444. DOI:10.1021/es0017561.
  6. Dettenmaier, E.M., Doucette, W.J. & Bugbee, W.J. (2009). Chemical hydrophobicity and uptake by plant roots. Environ. Sci. Technol. 43 (2), pp. 324-329. DOI:https://doi.org/10.1021/es801751x.
  7. Ellis, J.B. (2006). Pharmaceutical and personal care products in urban receiving waters. Environ. Pollut. 144, pp. 184-189. DOI:10.1016/j.envpol.2005.12.018.
  8. Geng, J., Liu, X., Wang, J. & Li, S. (2022). Accumulation and risk assessment of antibiotics in edible plants grown in contaminated farmlands: A review. Sci. Total Environ. 853, 158616. DOI:10.1016/J.SCITOTENV.2022.158616.
  9. Grote, M., Schwake, A.C., Michel, R., Stevens, H., Heyser, W., Langenkamper, G., Betsche, T. & Freitag, M. (2007). Incorporation of veterinary antibiotics into crop-s from manured soil. Federal Res. Centre Agric. 1 (1), pp. 25-32.
  10. Hoang, T.T.T., Tu, L.T.C., Le, N.P. & Dao, Q.P. (2013). A preliminary study on the phytoremediation o-f antibiotic contaminated sediment. Int. J. Phytoremediat. 15 (1), 65-76. DOI:10.1080/15226514.2012.670316.
  11. Hu, D.F. & Coats, J.R. (2007). Aerobic degradation and photolysis of tylosin in water and soil. Environ. Tech. Chem. 26, pp. 884-889. DOI:10.1897/06-197R.1.
  12. Jiang, L., Hu, X., Yin, D., Zhang, H. & Yu, Z. (2011). Occurrence, distribution and seasonal variation of antibiotics in the Huangpu River, Shanghai, China. Chemosphere 82 (6), pp. 822-828. DOI:10.1016/j.chemosphere.2010.11.028.
  13. KasprZyk-Hordern, B. & Dinsdsle, R. (2008). The occurrence of pharmaceuticals, personal care products, endocrine disruptors and illicit drugs in surface water in South Wales, UK. Water Res. 42 (13), pp. 3498-3518. DOI:10.1016/j.watres.2008.04.026.
  14. Kay, P., Blackwell, P.A. & Boxall, A.B.A. (2005). A lysimeter experiment to investigate the leaching of veterinary antibiotics through a clay soil and comparison with field data. Environ. Pollut. 134 (2), pp. 333-341. DOI:10.1016/j.envpol.2004.07.021.
  15. Kolpin, D.W., Furlong, E.T., Meyer, M.T., Thurman, E.M., Zaugg, S.D., Barber, L.B. & Buxton, H.T. (2002). Pharmaceuticals, hormones, and other organic wastewater contaminants in U.S. stream, 1999-2000. A national reconnaissance. Environ. Sci. Technol. 36 (6), pp. 1202-1211. DOI:10.1021/ES011055J.
  16. Kumar, K., Gupta, S.C., Baidoo, S., Chander, Y. & Rosen, C.J. (2005). Antibiotic uptake by plants from soil fertilized with animal manure. J. Environ. Qual. 34 (6), pp. 2082-2085. DOI:10.2134/jeq2005.0026.
  17. Maier, M.L.V. & Tjeerdema, R.S. (2018). Azithromycin sorption and biodegradation in a simulated California river system. Chemosphere, 190, pp. 471-480. DOI:10.1016/j.chemosphere.2017.10.008.
  18. Managaki, S., Murata, A., Takada, H., Tuyen, B.C. & Chiem, N.H. (2007). Distribution of macrolides, sulfonamides, and trimethoprim in tropical waters: ubiquitous occurrence of veterinary antibiotics in the Mekong Delta. Environ. Sci. Technol. 41 (23), pp. 8004-8010. DOI:10.1021/es0709021.
  19. Mauricio, C.H. & Francis, J. (2017). Mangroves enhance local fisheries catches: a global meta-analysis. Fish. 18 (1), pp. 79-93. DOI:10.1111/faf.12168.
  20. Ostrowski, A., Connolly, R.M. & Sievers, M. (2021). Evaluating multiple stressor research in coastal wetlands: a systematic review. Mar. Environ. Res. 164, 105239. DOI:10.1016/j.marenvres.2020.105239.
  21. Peng, X.Z., Yu, Y.J., Tang, C.M., Tan, J.H., Huang, Q.X., Wang, Z.D. (2008). Occurrence of steroid estrogens, endocrine-disrupting phenols, and acid pharmaceutical residues in urban riverine water of the Pearl River Delta, South China. Sci. Total Environ. 397 (1-3), pp/ 158-166. DOI:10.1016/j.scitotenv.2008.02.059.
  22. Sun, Q.Y., Peng, Y.S., Liu, Y., Xu, J.R., Ren, K.J. & Fang, X.T. (2017). Residues and migration characteristics of antibiotics ciprofloxacin(CIP) in two mangrove wetlands. J. Environm. Sci. (China) 37 (03), pp. 1057-1064. DOI:10.13671/j.hjkxxb.2016.0327.
  23. Thuy, H.T.T. & Tu, T.C.L. (2014). Degradation of Selected Pharmaceuticals in Coastal Wetland Water and Sediments. Water Air Soil Poll. 225 (5), pp. 1-9. DOI: 10.1007/s11270-014-1940-y.
  24. Yan, C.X., Yang, Y., Zhou, J. L., Liu, M., Nie, M.H., Shi, H. & Gu, L.J. (2013). Antibiotics in the surface water of the Yangtze Estuary: Occurrence, distribution and risk assessment. Environ. Pollut. 175, pp. 22-29. DOI:10.1016/j.envpol.2012.12.008.
  25. Yao, L.I., Zhang, J.R., Yu-Huang, W.U., Cai, J. & Cui, Y.B. (2017). Review on Antibiotic Pollution and Phytoremediation in Coastal Wetland. DEStech Transac. Environ. Ener. Ear. Sci.(ese). DOI:10.12783/dteees/ese2017/14358.
Go to article

Authors and Affiliations

Junwen Ma
1 4
Yubo Cui
1
Peijing Kuang
1
Chengdong Ma
2
Mingyue Zhang
1
Zhaobo Chen
1
Ke Zhao
3

  1. College of Environment and Resources, Dalian Minzu University, Dalian, 116600, China
  2. Department of Marine Ecological Environment Information,National Marine Environmental Monitoring Center, Dalian, 116023, China
  3. Key Laboratory of Songliao Aquatic Environment, Ministry of Education,Jilin Jianzhu University, Changchun, 130118, China
  4. Product and Technology Development Center,Beijing Enterprises Water Group Limited, Beijing, 100102, China
Download PDF Download RIS Download Bibtex

Abstract

Antibiotics are used for postsurgical wound healing purposes but unfortunately, resistance against them demands some alternatives for quick recovery. Sepsis of wounds is a challenge for medical as well as veterinary professionals. Nanoparticles have significant advantages in wound treatment and drug resistance reversal. This study was conducted to appreciate emerging alternates of antibiotics like zinc oxide nanoparticles and plant extracts in topical application. Zinc oxide is considered a good wound healer and its nanoparticles are easy to access. So, the efficacies of zinc oxide nanoparticles and sweet flag plant extract ointments were tested to compare modern and traditional therapeutics as sweet flag is considered a pure medicinal plant. Rabbits were selected for this study due to the healing properties of their skin. Wounds were inflicted on the thoracolumbar region and treated for 29 days post-surgically daily with normal saline and the ointment of zinc oxide nanoparticles and sweet flag extract ointment, prepared in a hydrophilic solvent. Wound shrinkage was observed daily and histopathological analysis was made and results were compared. Zinc oxide nanoparticles ointment showed the most satisfactory results for every parameter included in the study. No side effects of its topical application were observed. Healing was normal without any complications. The preparations of zinc oxide nanoparticles may help in the era of antibiotic resistance as topical drugs in the future.
Go to article

Authors and Affiliations

A. Abbas
1 2
S.A. Muhammad
3
A. Ashar
4
S.A. Mehfooz
2
A. Rauf
3
M. Bakhsh
3
T. Nadeem
5
H. Fu
1

  1. Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
  2. Department of Clinical Medicine and Surgery, University of Agriculture, Faisalabad 38000, Pakistan
  3. University of Veterinary and Animal Sciences, Lahore, CVAS Jhang 35200, Pakistan
  4. Wilson College of Textiles, North Carolina State University, North Carolina, USA
  5. University of Veterinary and Animal Sciences, Lahore, Para Veterinary Institute (PVI), Karor 31100, Layyah, Pakistan
Download PDF Download RIS Download Bibtex

Abstract

Nontuberculous mycobacteria (NTM) have recently emerged as important bacterial pathogens of both animals and humans. In this study, we aimed to evaluate the effect of a combination of ten antibiotics with an inhibitor of efflux pumps (EPI), i.e. berberine (BER), against 6 strains of NTM. Our results showed that the BER potentiated the anti-mycobacterial activities of the antibiotics. Overall, our findings show the importance of BER in increasing the efficacy of antibiotics in NTM.
Go to article

Bibliography


Gaba S, Saini A, Singh G, Monga V (2021) An insight into the medicinal attributes of berberine derivatives: a review. Bioorg Med Chem 38: 116143.
Gentry EJ, Jampani HB, Keshavarz-Shokri A, Morton MD, Velde DV, Telikepalli H, Mitscher LA, Shawar R, Humble D, Baker W (1998) Antitubercular natural products: berberine from the roots of commercial hydrastis canadensis powder. Isolation of inactive 8-oxotetrahydrothalifendine, canadine, beta-hydrastine, and two new quinic acid esters, hycandinic acid esters-1 and -2. J Nat Prod 61: 1187-1193.
Guz L, Puk K (2022) Antibiotic susceptibility of mycobacteria isolated from ornamental fish. J Vet Res 66: 69-76.
Menichini M, Lari N, Rindi L (2020) Effect of efflux pump inhibitors on the susceptibility of Mycobacterium avium complex to clarithromycin. J Antibiot 73: 128-132.
Puk K, Guz L (2020) Occurrence of Mycobacterium spp. in ornamental fish. Ann Agric Environ Med 27: 535-539.
Song L, Wu X (2016) Development of efflux pump inhibitors in antituberculosis therapy. Int J Antimicrob Agents 47: 421-429.
Van Ingen J, Boeree MJ, Van Soolingen D, Mouton JW (2012) Resistance mechanisms and drug susceptibility testing of nontuberculous mycobacteria. Drug Resist Updat 15: 149-161.
Wang Y, Fu H, Li Y, Jiang J, Song D (2012) Synthesis and biological evaluation of 8-substituted berberine derivatives as novel an-ti-mycobacterial agents. Acta Pharm Sin B 2: 581-587.
Zhou XY, Ye XG, He LT, Zhang SR, Wang RL, Zhou J, He ZS (2016) In vitro characterization and inhibition of the interaction between ciprofloxacin and berberine against multidrug-resistant Klebsiella pneumoniae. J Antibiot 69: 741-746.
Go to article

Authors and Affiliations

K. Puk
1
L. Guz
1

  1. Department of Biology and Fish Diseases, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, 20-033 Lublin, Poland
Download PDF Download RIS Download Bibtex

Abstract

Since fluoroquinolone (FQ) antibiotics are extensively used both in human and veterinary medicine their accumulation in the environment is causing increasing concern. The aim of the study was to isolate a microbial consortium resistant to ofl oxacin and norfl oxacin and able to biodegrade both antibiotics. Green compost was used as a source of microorganisms. The biodegradation effi ciency was monitored by changes of antibiotics concentrations and toxicity. The microbial consortium was composed of two bacterial isolates: Klebsiella pneumoniae (K2) and Achromobacter sp. (K3) and two fungi Candida manassasensis (K1) and Trichosporon asahii (K4). All the isolates were characterized as highly resistant to both antibiotics – ofl oxacin and norfl oxacin. FQs were supplied individually into the culture medium in the presence of an easily degradable carbon source – glucose. Biodegradation of norfl oxacin was much faster than ofl oxacin biodegradation. During 20 days of the experiment, the norfl oxacin level decreased by more than 80%. Ofl oxacin was generally biodegraded thereafter at relatively slow biodegradation rate. After 28 days the ofl oxacin level decreased by 60%. Similarly, the toxicity of biodegraded antibiotics decreased 4-fold and 3.5-fold for norfl oxacin and ofl oxacin, respectively. The ability of the bacterial-fungal consortium to degrade antibiotics and reduce toxicity could help to reduce environmental pollution with these pharmaceutical.

Go to article

Authors and Affiliations

Łukasz Jałowiecki
Grażyna Płaza
Helene Ejhed
Monika Nawrotek
Download PDF Download RIS Download Bibtex

Abstract

Wild ruminants are an interesting topic for research because only limited information exists regarding their microbiota. They could also be an environmental reservoir of undesirable bacteria for other animals or humans. In this study faeces of the 21 free-living animals was sampled (9 Cervus elaphus-red deer, adult females, 12 Capreolus capreolus-roe deer, young females). They were culled by selective-reductive shooting during the winter season of 2014/2015 in the Strzałowo Forest District-Piska Primeval Forest (53° 36 min 43.56 sec N, 21° 30 min 58.68 sec E) in Poland. Buttiauxella sp. is a psychrotolerant, facultatively anaerobic, Gram-negative rod anaerobic bacte- rial species belonging to the Phylum Proteobacteria, Class Gammaproteobacteria, Order Entero- bacteriales, Family Enterobacteriacae and to Genus Buttiauxella. Buttiauxella sp. has never previ- ously been reported in wild ruminants. In this study, identification, antimicrobial profile and sensitivity to enterocins of Buttiauxella strains were studied as a contribution to the microbiota of wild animals, but also to extend knowledge regarding the antimicrobial spectrum of enterocins. Five strains were identified using the MALDI-TOF identification system (evaluation score value was up to 2.224) and allotted to the genus Buttiauxella including the species Buttiauxella gaviniae,

B. ferragutiae, B. agrestis. Strains were DNase negative, and they hydrolysed esculin; fermentation of L-arabinose, D-mannitol and D-mannose was positive. Dulcitol, inositol reaction, urea and indol were negative. Buttiauxella strains did not form biofilm. They were resistant to at least one of the 13 antibiotics tested. B. agrestis 2/109/1 was resistant to amdinocillin, clindamycin and pen- icillin. However, Buttiauxella strains were sensitive to the enterocins used (inhibition activity ranged from 100 to 25 600 AU/ml).

Go to article

Authors and Affiliations

A. Lauková
M. Pogány Simonová
I. Kubašová
R. Miltko
G. Bełżecki
V. Strompfová
Download PDF Download RIS Download Bibtex

Abstract

To analyze the composition of norfloxacin-resistant bacteria and norfloxacin-degrading bacteria in pond water and sediment in subtropical China, the composition of antibiotic resistant bacteria in pond water and sediment enriched with norfloxacin-containing medium was analyzed by high-throughput sequencing. Sediment and water samples were collected from 3 fish ponds in subtropical China, and domesticated with norfloxacin, subsequently norfloxacin-resistant bacteria through high-throughput sequencing of 16S rDNA, and isolated norfloxacin- degrading bacteria. Our results showed that the pond sediment and water contain a variety of norfloxacin-resistant bacteria, mainly from Proteobacteria, Bacteroidetes, Actinobacteria, Firmicutes, and Chloroflexi. Moreover, we isolated two norfloxacin-degrading bacteria (NorXu-2 and NorXu-3). The norfloxacin-degrading rate by NorXu-2 and NorXu-3 in the culture mediums with 200 μg/mL was the highest, which was up to 49.71% and 35.79%,respectively. When the norfloxacin concentration was 200 μg/mL, NorXu-2 and NorXu-3 had the best norfloxacin-degrading effect at pH of 6, and the degradation rates were 53.64% and 45.54%, respectively. Moreover, NorXu-3 exhibited a good tolerance to high NaCl concentration. These results not only provided basic data for the follow-up study of the molecular mechanism of antimicrobial microbial degradation, but also provided potential norfloxacin degrading bacteria for norfloxacin removal and bioremediation in aquaculture environment.
Go to article

Bibliography

  1. Caporaso, J.G., Kuczynski, J., Stombaugh, J., Bittinger, K., Bushman, F.D., Costello, E.K., Fierer, N., Peña, A.G., Goodrich, J.K., Gordon, J.I., Huttley, G.A., Kelley, S.T., Knights, D., Koenig, J.E., Ley, R.E., Lozupone, C.A., McDonald, D., Muegge, B.D., Pirrung, M., Reeder, J., Sevinsky, J.R., Turnbaugh, P.J., Walters, W.A., Widmann, J., Yatsunenko, T., Zaneveld, J. & Knight, R. (2010). QIIME allows analysis of high-throughput community sequencing data. Nature Methods, 7, 5, pp. 335-336. DOI:10.1038/nmeth.f.303
  2. Edgar, R.C. (2013). UPARSE: Highly accurate OTU sequences from microbial amplicon reads. Nature Methods, 10, 10, pp. 996-998. DOI:10.1038/nmeth.2604
  3. FAO. (2020). The State of World Fisheries and Aquaculture 2020. Sustainability in Action. FAO, Rome 2020. DOI:10.4060/ca9229en.
  4. Fu, B.M., Chen, L.W., Cai, T.M., Yang, Q. & Ding, D.H. (2017). Isolation and characterization of norfloxacin-degrading bacterium strain NOR-36. Acta Scientiae Circumstantiae, 37, 2, pp. 576-584. DOI:10.13671/j.hjkxxb.2016.0245
  5. Gamoń, F., Tomaszewski, M., Cema, G. & Ziembińska-Buczyńska, A. (2022). Adsorption of oxytetracycline and ciprofloxacin on carbon-based nanomaterials as affected by pH. Archives of Environmental Protection, 48, 2, pp. 34-41. DOI:10.24425/aep.2022.140764
  6. Gong, W., Gao, S., Zhu, Y., Wang, G., Zhang, K., Li, Z., Yu, E., Tian, J., Xia, Y., Xie, J. & Ni, J. (2021). Effect of the aerobic denitrifying bacterium Pseudomonas furukawaii ZS1 on microbiota compositions in grass carp culture water. Water, 13, pp. 1329, DOI:10.3390/w13101329
  7. Guo, J., Zhang, Y., Zhou, X. & Liu, Z. (2016). Occurrence and removal of fluoroquinolones in municipal sewage: a review. Environmental Pollution and Control, 38, 2, pp. 75-80. DOI:10.15985/j.cnki.1001-3865.2016.02.015
  8. Han, Y., Wang, J., Zhao, Z., Chen, J., Lu, H. & Liu, G. (2017). Fishmeal application induces antibiotic resistance gene propagation in mariculture sediment. Environmental Science & Technology, 51, 18, pp. 10850-10860. DOI:10.1021/acs.est.7b02875
  9. Hao, Q., Xu, X., Chen, H., Liu, S., Chen, J., Liu, S. & Ying, G. (2017). Residual antibiotics in the Nansha aquaculture area of Guangzhou. Journal of Tropical Oceanography, 36, 1, pp. 106-113. DOI:10.11978/2016001
  10. Jałowiecki, Ł., Płaza, G., Ejhed, H. & Nawrotek, M. (2019). Aerobic biodegradation of norfloxacin and ofloxacin by a microbial consortium. Archives of Environmental Protection, 45, 4, pp. 40-47. DOI:10.24425/aep.2019.130240
  11. Laxminarayan, R., Duse, A., Wattal, C., Zaidi, A.K.M., Wertheim, H.F.L., Sumpradit, N., Vlieghe, E., Hara, G.L., Gould, I.M., Goossens, H., Greko, C., So, A.D., Bigdeli, M., Tomson, G., Woodhouse, W., Ombaka, E., Peralta, A.Q., Qamar, F.N., Mir, F., Kariuki, S., Bhutta, Z.A., Coates, A., Bergstrom, R., Wright, G.D., Borwn, E.D. & Cars, O. (2013). Antibiotic resistance - the need for global solution. The Lancet Infectious Diseases, 13, 12, pp. 1057-1098. DOI:10.1016/S1473-3099(13)70318-9
  12. Lemańska, N., Felis, E., Poraj-Kobielska, M., Gajda-Meissner, Z. & Hofrichter, M. (2021). Comparison of sulphonamides decomposition efficiency in ozonation and enzymatic oxidation processes. Archives of Environmental Protection, 47, 1, pp. 10-18. DOI:10.24425/aep.2021.13643
  13. Liang, X., Shi, Z. & Huang, X. (2013). Occurrence of antibiotics in typical aquaculture of the Pearl River Estuary. Ecology and Environmental Sciences, 22, 2, pp. 304-310. DOI: 10.3969/j.issn.1674-5906.2013.02.022
  14. Lin, B.K., Lyu, J., Lyu, X.J., Yu, H.Q., Hu, Z., Lam, J.C.W. & Lam, P.K.S. (2015). Characterization of cephalexin degradation capabilities of two Pseudomonas strains isolated from activated sludge. Journal of Hazardous Materials, 282, pp. 158-164. DOI:10.1016/j.jhazmat.2014.06.080
  15. Liu, H., Yang, Y.K., Ge, Y.H., Zhao, L., Long, S. & Zhang, R. (2016). Interaction between common antibiotics and a Shewanella strain isolated from an enhanced biological phosphorus removal activated sludge system. Bioresource Technology, 222, pp. 114-122. DOI:10.1016/j.biortech.2016.09.096
  16. Liu, X. & Lu, S. (2018). Occurrence and ecological risk of typical antibiotics in surface water of the Datong Lake, China. China Environmental Science, 28, 1, pp. 320-329. DOI:10.3969/j.issn.1000-6923.2018.01.036
  17. Magoc, T. & Salzberg, S.L. (2011). FLASH: Fast length adjustment of short reads to improve genome assemblies. Bioinformatics, 27, 21, pp. 2957-2963. DOI:10.1093/bioinformatics/btr507
  18. Mao, L.T., Huang, J., Chen, Z.G., Ma, X.L. & Liu, H.R. (2019). Norfloxacin resistant bacterial compositions in sediments of Chinese subtropical fish pond. Applied Ecology and Environmental Research, 17, 1, pp. 1039-1048. DOI:10.15666/aeer/1701_10391048
  19. Monteiro, S.H., Garcia, F., Gozi, K.S., Romera, D.M., Francisco, J.G., Roura-Andrade, G.C.R. & Tornisielo, V.L. (2016). Relationship between antibiotic residues and occurrence of resistant bacteria in Nile tilapia (Oreochromis niloticus) cultured in cage-farm. Journal of Environmental Science and Health, Part B, Pesticides, Food Contaminants, and Agricultural Wastes, 51, 12, pp. 817-823. DOI:10.1080/03601234.2016.12008457
  20. Mühling, M., Woolven-Allen, J., Murrell, J.C. & Joint, I. (2018). Improved group-specific PCR primers for denaturing gradient gel electrophoresis analysis of the genetic diversity of complex microbial communities. The ISME Journal, 2, pp. 379-392. DOI:10.1038/ismej.2007.97
  21. Mulla, S.I., Hu, A., Sun, Q., Li, J., Suanon, F., Ashfaq, M. & Yu, C.-P. (2018). Biodegradation of sulfamethoxazole in bacteria from three different origins. Journal of Environmental Management, 206, pp. 93-102. DOI:10.1016/j.jenvman.2017.10.029
  22. Ni, J., Li, X., He, Z. & Xu, M. (2017). A novel method to determine the minimum number of sequences required for reliable microbial community analysis. Journal of Microbiological Methods, 139, pp. 196-201. DOI:10.1016/j.mimet.2017.06.006
  23. Shao, S.C., Hu, Y.Y., Cheng, J.H. & Chen, Y. (2018). Degradation of oxytetracycline (OTC) and nitrogen conversion characteristics using a novel strain. Chemical Engineering Journal, 354, pp. 758-766. DOI:10.1016/j.cej.2018.08.032
  24. Wang, M., Yu, S., Hong, Y. & Sun, D. (2011). Residual characterization of multi-categorized antibiotics in five typical aquaculture waters. Ecology and Environmental Sciences, 20, 5, pp. 934-939. DOI:10.3969/j.issn.1674-5906.2011.05.026
  25. Wu, Y., Feng, P.Y., Li, R., Chen, X., Li, X., Sumpradit, T. & Liu, P. (2019). Progress in microbial remediation of antibiotic-residue contaminated environment. Chinese Journal of Biotechnology, 35, 11, pp. 2133-2150. DOI:10.13345/j.cjb.190164
  26. Xiang, J., He, T., Wang, P., Xie, M., Xiang, J. & Ni, J. (2018). Opportunistic pathogens are abundant in the gut of cultured giant spiny frog (Paa spinosa). Aquaculture Research, 49, 5, pp. 2033-2041. DOI:10.1111/are.13660
  27. Yang, J., Ying, G., Liu, S., Zhou, L., Zhao, J., Tao, R. & Peng, P. (2012). Biological degradation and microbial function effect of norfloxacin in a soil under different conditions. Journal of Environmental Science and Health Part B, Pesticides, Food Contaminants, and Agricultural Wastes, 47, 4, pp. 288-295. DOI:10.1080/03601234.2012.638886
  28. Yang, Z., Fan, T.-J. & Xu, B. (2020). Norfloxacin induces apoptosis and necroptosis in human corneal epithelial cells. Toxicology in Vitro, 66, pp. 104868. DOI:10.1016/j.tiv.2020.104868
  29. Zhang, G., Xue, Y., Wang, Q., Wang, P., Yao, H., Zhang, W., Zhao, J. & Li, Y. (2019). Photocatalytic oxidation of norfloxacin by Zn0.9Fe0.1S supported on Ni-foam under visible light irradiation. Chemosphere, 230, pp. 406-415. DOI:10.1016/j.chemosphere.2019.05.015
  30. Zhang, J.Y., Peng, X.X. & Jia, X.S. (2019). Isolation and characterization of high efficiency sulfamethazine-degrading bacterium strain J2. Acta Scientiae Circumstantiae, 39, 9, pp. 2919-2927. DOI:10.13671/j.hjkxxb.2019.0096
  31. Zhang, X., Cui, L., Li, S., Liu, X., Han, X., Jiang, K., Yu, X., Xu, L., Wu, F., Song, D. & Gao H. 2020. China Fishery Statistical Yearbook 2020. China Agriculture Press, Beijing 2020.
  32. Zhao, T., Chen, Y., Han, W. & He, Y. (2016). The contamination characteristics and ecological risk assessment of typical antibiotics in the upper reaches of the Dongjiang River. Ecology and Environment Sciences, 25, 10, pp. 1707-1713. DOI:10.16258/j.cnki.1674-5906.2016.10.016
Go to article

Authors and Affiliations

Lutian Mao
1
Lifen Chen
1
Xirui Wang
1
Zhongbao Xu
1
Hui Ouyang
1
Biyou Huang
1
Libin Zhou
1

  1. Huizhou University, Huizhou City, China
Download PDF Download RIS Download Bibtex

Abstract

Staphylococcus aureus (S. aureus) has been recognized as one of the important zoonotic pathogens. However, it was limited about the epidemiology and genetic characteristics of S. aureus isolated from pigs in Hunan province, china. The aim of this study was to determine the characteristics of 163 S. aureus isolated from 590 pigs in Hunan Province, China. All isolates were characterized by agr typing, detection of virulence genes and antibiotic resistance genes, lethal test of mice and antibiotic susceptibility tests. The results showed that 30 strains of the 163 isolates were divided into agrI (18.40%), agrII (36/163, 22.09%), agrIII (20/163, 12.27%,), agrIV (20/163,12.27%) and the remaining 57 isolates were amplified negative by agr primers. In the 163 isolates, the detection rate of the virulence genes hlb, hld, hla, icaA, seb, fnbA, eta, etb, sea, tst and pvl ranged from 2.45% to 100%. The 43 isolates that were lethal to the mice, had β-hemolytic activity, the number of virulence genes of which was 7.8% higher than that of the remaining 120 non-fatal strains. The resistance rates of the 163 isolates to the 15 antibiotics were 0% (0/163) - 100% (163/163). All isolates were susceptible to Vancomycin and only 7 isolates were methicillin - resistant S. aureus (MRSA). The detection rates of the 11 resistance genes was 0% (0/163) - 100% (163/163). This study first to describes the epidemiology and characteristics of S. aureus from pigs in Hunan Province, which will help in tracking the evolution of epidemic strains and preventing pig-human transmission events.

Go to article

Authors and Affiliations

X. Zhang
G. Wang
C. Yin
Download PDF Download RIS Download Bibtex

Abstract

Mastitis is one of the most crucial diseases of dairy animals. Especially subclinical mastitis (SCM) has negative impacts on of dairy economy in term of reducing milk quality and quantity also premature culling and cost of therapy. Staphylococci are important etiological agents in SCM. The aim of the study was to investigate the biofilm production and antibiotic resistance profiles of Staphylococcus spp. other than S. aureus isolated from milks of Anatolian water buffalo with subclinical mastitis. Twenty-two coagulase negative staphylococci (CNS) identified phenotypically were also identified with PCR as Staphylococcus spp. other than S. aureus. Biofilm productions were investigated both by Congo Red Agar Method and PCR. The antibiotic resistance profiles of the isolates were determined by Disc Diffusion Method and they were antibiotyped. Only three (13.6%) isolates were biofilm positive both phenotypically and genotypically. All isolates except for two were resistant against at least two antibiotics. Multidrug-resistance among the isolates was low (13.6%). Antibiotyping results showed that the similarities among the strains were between 30-100%. Genotyping of the strains revealed that a genetic heterogeneity was found among CNS isolates and their similarities were between 43% and 93%. In conclusion, CNS isolates identified as subclinical mastitis agents in buffaloes showed a high antibiotic resistance profile especially against oxacillin and vancomycin. Further studies should be conducted to investigate new mechanisms and/or genes responsible for antibiotic resistance in buffaloes.
Go to article

Bibliography

Aslantaş Ö, Yılmaz MA, Yılmaz EŞ, Kurekci C (2014) Antimicrobial susceptibility pattern and SCCmec types of methicillin-resistant coagulase-negative staphylococci from subclinical bovine mastitis in Hatay, Turkey. Bull Vet Inst Pulawy 58: 563-566.
Athar M (2006) Preparation and evaluation of inactivated polyvalent vaccines for the control of mastitis in dairy buffaloes. PhD Dissertation Dept. Vet. Clinical Medicine and Surgery, Univ. Agri., Faisalabad, Pakistan.
Ba X, Harrison EM, Edwards GF, Holden MT, Larsen, AR, Petersen A, Skov RL, Peacock SJ, Parkhill J, Paterson GK, Holmes MA (2014) Novel mutations in penicillin-binding protein genes in clinical Staphylococcus aureus isolates that are methicillin resistant on susceptibility testing, but lack the mec gene. J Antimicrob Chemother 69: 594-597.
Becker K, Heilmann C, Peters G (2014) Coagulase-Negative Staphylococci. Clin 246 Microbiol Rev 27: 870-926.
Borghese A, Mazzi M (2005) Buffalo population and strategies in the world. In: Borghese A (ed) Buffalo production and research. Food and Agriculture Organization, Rome, Italy pp 1-41.
Boye K, Bartels MD, Andersen IS, Moller JA, Westh H (2007) A new multiplex PCR for easy screening of methicillin-resistant Staphylococcus aureus SCCmec types I-V. Clin Microbiol Infect 13: 725-727.
Bradley A (2002) Bovine mastitis: an evolving disease. Vet J 164: 116-128.
Chen XP, Li WG, Zheng H, Du HY, Zhang L, Zhang L, Che J, Wu Y, Liu SM, Lu JX (2017) Extreme diversity and multiple SCCmec elements in coagulase-negative Staphylococcus found in the Clinic and Community in Beijing, China. Ann Clin Microbiol Antimicrob 16: 57.
Ciftci A, Findik A, Onuk EE, Savasan S (2009) Detection of methicillin resistance and slime factor production of Staphylococcus aureus in bovine mastitis. Braz J Microbiol 40: 254-261.
Clinical and Laboratory Standards Institute (2019) Performance standards for antimicrobial susceptibility testing, 29th ed. CLSI document M100. Clinical and Laboratory Standards Institute, Wayne, PA. https://community.clsi.org/media/2663/m100ed29_sample.pdf
Dezfulian A, Aslani MM, Oskoui M, Farrokh P, Azimirad M, Dabiri H, Salehian MT, Zali MR (2012) Identification and characterization of a high vancomycin-resistant Staphylococcus aureus harboring VanA gene cluster isolated from diabetic foot ulcer. Iran J Basic Med Sci 15: 803-806 Ergun Y, Aslantas O, Doğruer G, Kirecci E, Sarıbay MK, Ates CT, Ulku A, Demir C (2009) Prevalence and etiology of subclinical mastitis in awassi dairy ewes in southern Turkey. Turk J Vet Anim Sci 33: 477-483.
Findik A, Akan N, Onuk EE, Çakıroğlu D, Ciftci A (2009) Methicillin resistance profile and molecular typing of Staphylococcus aureus strains isolated from noses of the healthy dogs. Kafkas Univ Vet Fak Derg 15: 925-930.
Fitzgerald JR, Hartigan PJ, Meaney WJ, Smyth CJ (2000) Molecular population and virulence factor analysis of Staphylococcus aureus from bovine intramammary infection. J Appl Microbiol 88: 1028-1037.
Gentilini E, Denamiel G, Betancor A, Rebuelto M, Fermepin RM, De Torres RA (2002) Antimicrobial susceptibility of coagulase-negative staphylococci isolated from bovine mastitis in Argentina. J Dairy Sci 85: 1913-1917.
Gulhan T, Boynukara B, Ciftci A, Sogut MU, Findik A (2015) Characterization of Enterococcus faecalis isolates originating from different sources for their virulence factors and genes, antibiotic resistance patterns, genotypes and biofilm production. Iran J Vet Res 16: 261-266.
Hanssen AM, Sollid JU (2006) SCCmecin staphylococci: genes on the move. FEMS Immunol Med Microbiol 46: 8-20.
Harrison EM, Paterson GK, Holden MT, Ba X, Rolo J, Morgan FJ, Pichon B, Kearns A, Zadoks RN, Peacock SJ, Parkhill J, Holmes MA (2014) A novel hybrid SCCmec- -mecC region in Staphylococcus sciuri. J Antimicrob Chemother 69: 911-918.
İnegol E, Türkyılmaz S (2012) Determination of SCCmec types in methicillin resistant staphylococci isolated from cows and farm workers. Ankara Univ Vet Fak Derg 59: 89-93.
Hiramatsu K, Kihara H, Yokota T (1992) Analysis of borderlineresistant strains of methicillin-resistant Staphylococcus aureus using polymerase chain reaction. Microbiol Immunol 36: 445-453.
Morandi S, Brasca M, Lodi R, Brusetti L, Andrighetto C, Lombardi A (2010) Biochemical profiles, restriction fragment length polymorphism (RFLP), random amplified polymorphic DNA (RAPD) and multilocus variable number tandem repeat analysis (MLVA) for typing Staphylococcus aureus isolated from dairy products. Res Vet Sci 88: 427-435.
Özenç E, Vural MR, Seker E, Uçar M (2008) An evaluation of subclinical mastitis during lactation in Anatolian buffaloes. Turk J Vet Anim Sci 32: 359-368.
Palazzo IC, Araujo ML, Darini AL (2005) First Report of Vancomycin-Resistant Staphylococci Isolated from Healthy Carriers in Brazil. J Clin Microbiol 43: 179-185.
Pamuk Ş, Şeker E, Yıldırım Y (2010) Antibiotic resistance of coagulase negative Staphylococci isolated from buffalo milk and some milk products. Kocatepe Vet J 3: 7-12.
Partridge SR, Kwong SM, Firth N, Jensen SO (2018) Mobile Genetic Elements Associated with Antimicrobial Resistance. Clin Microbiol Rev 31: e00088-17.
Petinaki E, Arvaniti A, Bartzavali C, Dimitracopoulos G, Spiliopoulou I (2002) Presence of mec Genes and Overproduction of Beta-Lactamase in the Expression of Low-Level Methicillin Resistance among Staphylococci. Chemotherapy 48: 174-181.
Pyörala S, Taponen S (2009) Coagulase-negative staphylococci-Emerging mastitis pathogens. Vet Microbiol 134: 3-8.
Qu Y, Zhao H, Nobrega DB, Cobo ER, Han B, Zhao Z, Li S, Li M, Barkema HW, Gao J (2018) Molecular epidemiology and distribution of antimicrobial resistance genes of Staphylococcus species isolated from Chinese dairy cows with clinical mastitis. J Dairy Sci 102: 1571-1583.
Raza A, Muhammad G, Sharif S, Atta A (2013) Biofilm producing Staphylococcus aureus and bovine mastitis: a review. Mol Microbiol Res 33: 1-8.
Reinoso E, Bettera S, Ferigerio C, DiRenzo M, Calozari A, Bongi C (2004) RAPD-PCR analysis of Staphylococcus aureus strains isolated from bovine and human hosts. Microbiol Res 159: 245-255.
Ruppe E, Barbier F, Mesli Y, Maiga A, Cojocaru R, Benkhalfat M, Benchouk S, Hassaine H, Maiga I, Diallo A, Koumare AK, Ouattara K, Soumare S, Dufourcq JB, Nareth C, Sarthou JL, Andremont A, Ruimy R (2009) Diversity of Staphylococcal cassette chromosome mec structures in methicillin- resistant Staphylococcus epidermidis and Staphylococcus haemolyticus strains among outpatients from four countries. Antimicrob Agents Chemother 53: 442-449.
Saber H, Jasni AS, Jamaluddin TZ, Ibrahim R (2017) A review of staphylococcal cassette chromosome mec (SCCmec) types in coagulase-negative staphylococci (CoNS) species. Malays J Med Sci 24: 7-18
Sawant AA, Gillespie BE, Oliver SP (2009) Antimicrobial susceptibility of coagulase-negative Staphylococcus species isolated from bovine milk. Vet Microbiol 134: 73-81.
Schalm OW, Carroll EJ, Jain NC (1971) Bovine mastitis. Bovine mastitis. LeaFebiger, Philadelphia USA. Siebert WT, Moreland N, Williams TW (1979) Synergy of vancomycin plus cefazolin or cephalothin against methicillin-resistance Staphylococcus epidermidis. J Infect Dis 139: 452-457.
Sudhan NA, Sharma N (2010) Mastitis: An important production disease of dairy animals. Smvs’ Dairy Year Book pp 72-88. Sujatha S, Praharaj I (2012) Glycopeptide resistance in Gram-positive cocci: a review. Interdiscip Perspect Infect Dis 2012: 781679
Taponen S, Pyörälä S (2009) Coagulase-negative staphylococci as cause of bovine mastitis-Not so different from Staphylococcus aureus? Vet Microbiol 134: 29-36.
Taponen S, Simojoki H, Haveri M, Larsen HD, Pyörälä S (2006) Clinical characteristics and persistence of bovine mastitis caused by different species of coagulase-negative staphylococci identified with API or AFLP. Vet Microbiol 115: 199-207.
Thorberg B (2008) Coagulase-Negative Staphylococci in Bovine Sub-Clinical Mastitis. Licentiate Thesis Department of Biomedical Sciences and Veterinary Public Health Swedish University of Agricultural Sciences, Report no. 2.
Turutoğlu H, Ercelik S, Ozturk D (2006) Antibiotic resistance of Staphylococcus aureus and coagulase-negative staphylococci isolated from bovine mastitis. Bull Vet Ins Pulawy 50: 41-45.
Versalovic J, Lupski JR (2002) Molecular detection and genotyping of pathogens: more accurate and rapid answers. Trends Microbiol 10: S15-21. Vurucu N, Savaşan S, Sezener MG (2019) Determination of Virulence Genes and Genetic Similarities of Mastitic Milk Originated Escherichia coli Isolates. J Agri Life Sci 2: 31-35.
Wielders CL, Vriens MR, Brisse S, De Graaf-Miltenburg LA, Troelstra A, Fleer A, Schmitz FJ, Verhoef J, Fluit AC (2001) Evidence for in-vivo transfer of mecA DNA between strains of Staphylococcus aureus. Lancet 357: 1674-1675.
Xu Z, Shah HN, Misra R, Chen J, Zhang W, Liu Y, Cutler RR, Mkrtchyan HV (2018) The prevalence, antibiotic resistance and mecA characterization of coagulase negative staphylococci recovered from non-healthcare settings in London, UK. Antimicrob Resist Infect Control 7: 73.
Yazdani R, Oshaghi M, Havayi A, Pishva E, Salehi R, Sadeghizadeh M, Foroohesh H (2006) Detection of icaAD gene and biofilm formation in Staphylococcus aureus isolates from wound infections. Iranian J Publ Health 35: 25-28.
Zare S, Derakhshandeh A, Haghkhah M, Naziri Z, Broujeni AM (2019) Molecular typing of Staphylococcus aureus from different sources by RAPD-PCR analysis. Heliyon 5: e02231.
Go to article

Authors and Affiliations

H. Gurler
1
A. Findik
2
M.G. Sezener
2

  1. Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
  2. Department of Microbiology, Faculty of Veterinary Medicine, University of Ondokuz Mayis, Samsun, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The aim was to explore the feasibility of using bamboo vinegar powder as an antibiotics substitute in weaning piglets. Forty-five healthy Duroc × Landrance × Yorshire piglets (weight 6.74 ± 0.17 kg; age 31 days) were randomly divided into the control group (basic diet), ANT group (basic diet + 0.12% compound antibiotics), BV1 group (basic diet + 0.1% bamboo vinegar powder), BV5 group (basic diet + 0.5% bamboo vinegar powder) and BV10 group (basic diet + 1% bamboo vinegar powder). MyD88 and CD14 expression in immune tissues was examined using real-time PCR. MyD88 expression in the control group were significantly lower than that in other groups in all tissues (p<0.05), while CD14 expression showed the opposite trend. MyD88 expression was significantly higher in the BV10 group than in other groups in lung tissue (P<0.05), significantly higher in the ANT group than in the BV1 group in the kidneys (P<0.05), significantly higher in the BV10 group than in the BV1 group in the thymus (P<0.05), and signifi- cantly higher in the BV1 group than in the BV10 group in the lymphatic tissue (P<0.05). These differences between experimental groups were not observed for the CD14 gene (P>0.05). Thus, adding bamboo vinegar powder to the basic diet of weaning piglets had immune effects similar to antibiotics and the effect was dose-dependent. Moreover, the MyD88 and CD14 genes appear to play a role in these immune effects

Go to article

Authors and Affiliations

W.Y. Qin
L.N. Gan
L. Dong
L.H. Yu
S.L. Wu
W.B. Bao
Download PDF Download RIS Download Bibtex

Abstract

Mycoplasma bovis is a highly contagious pathogen that causes clinical or subclinical mastitis. The present study was aimed for the isolation, molecular characterization and antibiogram determination of M. bovis from raw milk samples. Milk samples were collected randomly from lactating cows and buffaloes from different tehsils of district Faisalabad, Pakistan. Samples were inoculated on modified Hayflick medium and biochemical tests were performed for further confirmation of isolated M. bovis. Out of total 400 milk samples, 184 (46%) samples were found positive for culture method. The 16S-rRNA gene polymerase chain reaction was performed for molecular characterization of isolated M. bovis strains. Out of total 400 milk samples, 240 (60%) positive for M. bovis through PCR method were examined. The 16S-rRNA gene PCR positive isolated M. bovis strains were sequenced and results were compared using Maximum-likelihood method and sequenced strains of M. bovis were aligned and analyzed by Clustal W software. Antibiogram of isolated M. bovis strains was analyzed by disc diffusion assay against eight commonly used antibiotics. Tylosin (30μg) and Tilmicosin (15ug) showed inhibition zones of 32.34 ± 1.10 mm and 17.12 ± 0.93 mm respectively against isolated M. bovis which were found sensitive. Isolated M. bovis was found resistant to other commonly used antibiotics. Statistical analysis revealed that p-value was < 0.05 and the odds ratio was >1.0 at 95% CI. This study complemented the lack of epidemiological knowledge of molecular characterization, comparative effectiveness and resistance trends of isolated M. bovis strains against commonly used antibiotics.
Go to article

Bibliography

  1. Adorno BM, Salina A, Joaquim S, Guimarães FF, Lopes BC, Menozzi B, Langoni H (2021) Presence of Mollicutes and Mycoplasma bovis in nasal swabs from calves and in milk from cows with clinical mastitis. Vet Zootec 28: 001-009.
  2. Ahmad Z, Babar S, Abbas F, Awan MA, Abubakar M, Attique MA, Hassan Y, Rashid N, Ali M (2011) Identification and molecular characterization of Mycoplasma species from bovine lungs samples collected from slaughter house, Quetta, Balochistan, Pakistan. Pak J Life Soc Sci 9: 91-97.
  3. Ahmad Z, Babar S, Abbas F, Awan MA, Shafee M, Tariq MM, Mengal MA, Rashid N, Amin S, Taj K, Ali M (2014) Prevalence of Mycoplasma bovis in respiratory tract of cattle slaughtered in Balochistan, Pakistan. Pak Vet J 34: 46-49.
  4. Alhussen MA, Kirpichenko VV, Yatsentyuk SP, Nesterov AA, Byadovskaya OP, Zhbanovat TV, Sprygin AV (2021) Mycoplasma bovis, M. bovigenitalium and M. dispar as Bovine Pathogens: Brief characteristics of the pathogens (review). Agric Biol 56: 245-260.
  5. Kumar A, Verma AK, Gangwar NK, Rahal A (2012) Isolation, characterization and antibiogram of Mycoplasma bovis in sheep pneumonia. Asian J Anim Vet Adv 7: 149-157.
  6. Behera S, Rana R, Gupta PK, Kumar D, Sonal, Rekha V, Arun TR, Jena D (2018) Development of real- time PCR assay for the detection of Mycoplasma bovis. Trop Anim Health Prod 50: 875-882.
  7. Bokma J, Vereecke N, De Bleecker K, Callens J, Ribbens S, Nauwynck H, Haesebrouck F, Theuns S, Boyen F, Pardon B (2020) Phylogenomic analysis of Mycoplasma bovis from Belgian veal, dairy and beef herds. Vet Res 51: 121.
  8. Bokma J, Vereecke N, Nauwynck H, Haesebrouck F, Theuns S, Pardon B, Boyen F (2021) Genome-wide association study reveals genetic markers for antimicrobial resistance in Mycoplasma bovis. Microbiol Spectr 9: e0026221.
  9. Buller H, Blokhuis H, Jensen P, Keeling L (2018) Towards farm animal welfare and sustainability. Animals 2018, 8: 81.
  10. Caria M, Boselli C, Murgia L, Rosati R, Pazzona A (2013) Influence of low vacuum levels on milking characteristics of sheep, goat and buffalo. J Agr Eng 44: 217- 220.
  11. Cheng WN, Han SG (2020) Bovine mastitis: risk factors, therapeutic strategies, and alternative treatments – A review. Asian-Australas J Anim Sci . 33: 1699-1713.
  12. Deeney A S, Collins R, Ridley AM (2021) Identification of Mycoplasma species and related organisms from ruminants in England and Wales during 2005-2019. BMC Vet Res 17: 325.
  13. Farid MA, Abo-Shosha AA, Belal ES, Hassan MM (2018) Genotyping of pathogenic Mycoplasma bovis isolated from cattle in Kafr El-Sheikh Province, Egypt. J Pure Appl Microbiol. 12: 2103-2109.
  14. Ghafar A, Mcgill D, Stevenson MA, Badar M, Kumbher A, Warriach MH, Gasser RB, Jabbar A (2020) A participatory investigation of bovine health and production issues in Pakistan. Front Vet Sci 7: 248.
  15. Hata E, Harada T, Itoh M (2019) Relationship between antimicrobial susceptibility and multilocus sequence type of Mycoplasma bovis isolates and development of a method for rapid detection of point mutations involved in decreased susceptibility to macrolides, lincosamides, tetracyclines, and spectinomycin. Appl Environ Microbiol 85: e0057519.
  16. Hudzicki J (2009) Kirby-Bauer disk diffusion susceptibility test protocol. Am J Mol Biol 8: 2009.
  17. Ilyas F, Gillani DQ, Yasin M, Iqbal MA, Javed I, Ahmad S, Nabi I (2022) Impact of Livestock and Fisheries on Economic Growth: An Empirical Analysis from Pakistan. Sarhad J Agric 38: 160-169.
  18. Imandar M, Pourbakhsh SA, Jamshidian M, Salehi TZ (2018) Isolation, identification and molecular characterization of Mycoplasma bovis in mastitic dairy cattle by PCR and culture methods. J Hell Vet Med Soc 69: 815-822.
  19. Imran M, Rehman I, Sulehria AQ, Butt YM, Khan AM, Ziauddin A (2021) Profile of Antimicrobial Susceptibility from Cattles’s Milk Isolates Suffering from Mastitis in District Lahore. J Biores Manag 8: 6-14.
  20. Khan ZU (2022) Laws, Issues, Challenges, Analysis of Livestock Sector and International Best Practices. J Dev Soc Sci 3: 271-283.
  21. Klein U, de Jong A, Moyaert H, El Garch F, Leon R, Richard-Mazet A, Rose M, Maes D, Pridmore A, Thomson JR, Ayling RD (2017) Antimicrobial susceptibility monitoring of Mycoplasma hyopneumoniae and Mycoplasma bovis isolated in Europe. Vet Microbiol 204: 188-193.
  22. Konigsson MH, Bolske G, Johansson KE (2002) Intraspecific variation in the 16S- rRNA gene sequences of Mycoplasma agalactiae and Mycoplasma bovis strains. Vet Microbiol 85: 209-220.
  23. Mahmood F, Khan A, Hussain R, Khan IA, Abbas RZ, Ali HM, Younus M (2017) Patho-bacteriological investigation of an outbreak of Mycoplasma bovis infection in calves-Emerging stealth assault. Microb Pathog 107: 404-408.
  24. Maunsell FP, Donovan GA, (2009) Mycoplasma bovis infections in young calves. Vet Clin North Am Food Anim Pract 25: 139-177.
  25. Maunsell FP, Woolums AR, Francoz D, Rosenbusch RF, Step DL, Wilson DJ, Janzen ED (2011) Mycoplasma bovis infections in cattle. J Vet Inter Med 25: 772-783.
  26. Mojsoska B, Ghoul M, Perron GG, Jenssen H, Alatraktchi FA (2021) Changes in toxin production of environmental Pseudomonas aeruginosa isolates exposed to sub- inhibitory concentrations of three common antibiotics. PloS One 16: e0248014.
  27. Nicholas RA, Fox LK, Lysnyansky I (2016) Mycoplasma mastitis in cattle: To cull or not to cull. Vet J 216: 142-147.
  28. Niu J, Wang D, Yan M, Chang Z, Xu Y, Sizhu S, Li Z, Hu S, Bi D (2021) Isolation, identification and biological characteristics of Mycoplasma bovis in yaks. Microb Pathog 150: 104691.
  29. Pal A, Chakravarty AK (2020) Disease resistance for different livestock species. Genet Breed Dis Resist Livest 2020: 271-296.
  30. Passchyn P, Piepers S, De Meulemeester L, Boyen F, Haesebrouck F, De Vliegher S (2012) Between-herd prevalence of Mycoplasma bovis in bulk milk in Flanders, Belgium Res Vet Sci 92: 219-220.
  31. Perez-Casal J, Prysliak T, Maina T, Suleman M, Jimbo S (2017) Status of the development of a vaccine against Mycoplasma bovis. Vaccine 35: 2902-2907.
  32. Romero J, Benavides E, Meza C (2018) Assessing financial impacts of subclinical mastitis on Colombian dairy farms. Front Vet Sci 5: 273
  33. Rossetti BC, Frey J, Pilo P (2010) Direct detection of Mycoplasma bovis in milk and tissue samples by real-time PCR. Molar Cell Pro. 24: 321-323.
  34. Salina A, Timenetsky J, Barbosa MS, Azevedo CM, Langoni H (2020) Microbiological and molecular detection of Mycoplasma bovis in milk samples from bovine clinical mastitis. Pesqui Vet Bras 40: 82-87.
  35. Shao Y, Wang Y, Yuan Y, Xie Y (2021) A systematic review on antibiotics misuse in livestock and aquaculture and regulation implications in China. Sci Total Environ 798: 149205.
  36. Abadi AT, Rizvanov AA, Haertlé T, Blatt NL (2019) World Health Organization report: current crisis of antibiotic resistance. BioNanoScience 9: 778-788.
  37. Gogoi-Tiwari J, Tiwari HK, Wawegama NK, Premachandra C, Robertson ID, Fisher AD, Waichigio FK, Irons P, Aleri JW (2022) Prevalence of Mycoplasma bovis Infection in Calves and Dairy Cows in Western Australia. Vet Sci 9: 351-358.
  38. Vereecke N, Bokma J, Haesebrouck F, Nauwynck H, Boyen F, Pardon B, Theuns S (2020) High quality genome assemblies of Mycoplasma bovis using a taxon- specific Bonito basecaller for MinION and Flongle long-read nanopore sequencing. BMC Bioinform 21: 517.
  39. Verraes C, Claeys W, Cardoen S, Daube G, De Zutter L, Imberechts H, Dierick K, Herman L (2014) A review of the microbiological hazards of raw milk from animal species other than cows. Inter Dairy J 39: 121-130.
  40. Wisselink HJ, Smid B, Plater J, Ridley A, Andersson AM, Aspan A, Pohjanvirta T, Vahanikkila N, Larsen H, Hogberg J, Colin A, Tardy F (2019) A European interlaboratory trial to evaluate the performance of different PCR methods for Mycoplasma bovis diagnosis. BMC Vet Res 15: 86.
Go to article

Authors and Affiliations

A. Jabbar
1
M. Ashraf
1
S.U. Rahman
1
M.S. Sajid
2

  1. Institute of Microbiology, University of Agriculture, Jail Road, Faisalabad, Punjab 38000, Pakistan
  2. Department of parasitology, University of Agriculture, Jail Road, Faisalabad, Punjab 38000, Pakistan
Download PDF Download RIS Download Bibtex

Abstract

Antimicrobial resistance in Salmonella has been associated with the presence of integrons and many other resistance mechanisms contributing to the spread of antimicrobial-resistant genes within and between livestock and human populations. In this study, the presence of Salmonella serovars from broiler and cattle samples and their antimicrobial resistance, integrons, tet resistance, ESBL and resistance genes carriage were investigated. Total of 209 litter (broiler farms) and fecal samples (cattle farms) were examined by bacteriological procedures, susceptibilities against 18 antimicrobials and genes carriages were detected by singleplex and multiplex PCR. A total of 46/209 (22 %) Salmonella strains were isolated. Six different Salmonella serotypes from 46 Salmonella isolates were identified and the most common serotype was S. Infantis 38 (82.6%) from broiler litter; followed by S. Kitenge 3 (6.5 %) from fecal sample. The highest occurrence of resistance observed for penicilline (46/46, %100), lincomycin (43/46, 93.5%) and 42 isolates (43/46, 93.5%) exhibited MDR. The overall occurrence of class 1, 2 and 3 integrons carrying Salmonella in tested samples were 63.04% (29/46), 43.5% (20/46) and 84.8% (39/46) respectively. Out of the 27 isolates produced an ESBL, mostly CTX and TEM. On 46 Salmonella isolates, in 16 (34.8%) Tcr’ genes were determined. Genotypic and phenotipic detection of ESBL genes found within integrons from Salmonella isolates from different sources (broiler and cattle) can provide powerful information about health and economic risk associated with transferable multidrug resistance.
Go to article

Bibliography


Aslam M, Checkley S, Avery B, Chalmers G, Bohaychuk V, Gensler G, Reid-Smith R, Boerlin P (2012) Phenotypic and genetic characteri-zation of antimicrobial resistance in Salmonella serovars isolated from retail meats in Alberta, Canada. Food Microbiol 32: 110-117.
Ahmed AM, Shimamoto T (2014) Characterization of integrons and resistance genes in multidrug-resistant Salmonella enterica isolated from meat and dairy products in Egypt. Int J Food Microbiol 189: 39-44.
Akiba M, Kusumoto M, Iwata T (2010) Rapid identification of Salmonella enterica serovars, Typhimurium, Choleraesuis, Infantis, Hadar, Enteritidis, Dublin and Gallinarum, by multiplex PCR. J Microbiol Methods 85: 9-15.
Antunes P, Machado J, Peixe L (2006) Characterization of antimicrobial resistance and class 1 and 2 integrons in Salmonella enterica isolates from different sources in Portugal. J Antimicrob Chemother 58: 297-304.
Asgharpour F, Mahmoud S, Marashi A, Moulana Z (2018) Molecular detection of class 1, 2 and 3 integrons and some antimicrobial resistance genes in Salmonella Infantis isolates. Iran J Microbiol 10: 104-110.
Barlow RS, Fegan N, Gobius KS (2009) Integron-containing bacteria in faeces of cattle from different production systems at slaughter. J Appl Microbiol 107: 540-545.
Bennett PM (1999) Integrons and gene cassettes: a genetic construction kit for bacteria. J Antimicrob Chemother 43: 1-4.
Bush K, Jacoby GA (2010) Updated functional classifica tion of beta-lactamases. Antimicrob Agents Chemother 54: 969-976.
Carfora V, Alba P, Leekitcharoenphon P, Ballarò D, Cordaro G, Di Matteo P, Donati V, Ianzano A, Iurescia M, Stravino F, Tagliaferri T, Battisti A, Franco A (2018) Colistin resistance mediated by mcr-1 in ESBL-producing, multidrug resistant Salmonella Infantis in broiler chicken industry, Italy (2016-2017). Front Microbiol 9:1880.
Castro-Vargas RE, Herrera-Sánchez MP, Rodríguez -Hernández R, Rondón-Barragán IS (2020) Antibiotic resistance in Salmonella spp. isolated from poultry: A global overview. Vet World 13: 2070-2084.
Chang Q, Wang W, Regev-Yochay G, Lipsitch M, Hanage WP (2015) Antibiotics in agriculture and the risk to human health: how worried should we be? Evol Appl 8: 240-247.
Chuanchuen R, Padungtod P (2009) Antimicrobial resistance genes in Salmonella enterica isolates from poultry and swine in Thai-land. J Vet Med Sci 71: 1349-1355.
CLSI (2020) Performans standards for antimicrobial disk and dilution susceptibility tests for bacteria isolated from animals, 5 th ed., Clinical and Laboratory Standarts Institute document Vet01S. Clinical and Laboratory Standards Institute, Wayne, Pennsylvania.
de Jong A, Smet A, Ludwig C, Stephan B, de Graef E, Vanrobaeys M, Haesebrouck F (2014) Antimicrobial susceptibility of Salmonella isolates from healthy pigs and chickens (2008-2011). Vet Microbiol 171: 298-306.
Dessie HK, Bae DH, Lee YJ (2013) Characterization of integrons and their cassettes in Escherichia coli and Salmonella isolates from poultry in Korea. Poult Sci 92: 3036-3043.
Duc VM, Nakamoto Y, Fujiwara A, Toyofuku H, Obi T, Chuma T (2019) Prevalence of Salmonella in broiler chickens in Kagoshi-ma, Japan in 2009 to 2012 and the relationship between serovars changing and antimicrobial resistance. BMC Vet Res 15: 108.
EFSA (2021) European Food Safety Authority, European Centre for Disease Prevention Control. The European Union One Health 2019 Zoonoses Report. EFSA J 19: e06406.
Firoozeh F, Shahcheraghi F, Salehi TZ, Karimi V, Aslani MM (2011) Antimicrobial resistance profile and presence of class I integrons among Salmonella enterica serovars isolated from human clinical specimens in Tehran, Iran. Iran J Microbiol 3: 112-117.
Franco A, Leekitcharoenphon P, Feltrin F, Alba P, Cordaro G, Iurescia M, Tolli R, D’Incau M, Staffolani M, Di Giannatale E, Hendriksen RS, Battisti A (2015) Emergence of a clonal lineage of multidrug-resistant ESBL-producing Salmonella Infantis transmitted from broilers and broiler meat to humans in Italy between 2011 and 2014. PLoS One 10: e0144802.
Frech G, Schwarz S (2000) Molecular analysis of tetracycline resistance in Salmonella enterica subsp. enterica serovars Typhimurium, Enteritidis, Dublin, Choleraesuis, Hadar and Saintpaul: construction and application of specific gene probes. J Appl Microbiol 89: 633-641.
Gal-Mor O, Valinsky L, Weinberger M, Guy S, Jaffe J, Schorr YI, Raisfeld A, Agmon V, Nissan I (2010) Multidrug -resistant Salmonella enterica serovar Infantis, Israel. Emerg Infect Dis 16: 1754-1757.
García-Soto S, Abdel-Glil MY, Tomaso H, Linde J, Methner U (2020) Emergence of multidrug-resistant Salmonella enterica subspecies enterica serovar Infantis of multilocus sequence type 2283 in German broiler farms. Front Microbiol 11: 1741.
Ghoddusi A, Fasaei BN, Salehi TZ, Akbarein H (2019) Serotype distribution and antimicrobial resistance of Salmonella isolates in human, chicken, and cattle in Iran. Arch Razi Inst 74: 259-266.
Goldstein C, Lee MD, Sanchez S, Hudson C, Phillips B, Register B, Grady M, Liebert C, Summers AO, White DG, Maurer JJ (2001) Inci-dence of class 1 and 2 integrases in clinical and commensal bacteria from livestock, companion animals, and exotics. Antimicrob Agents Chemother 45: 723-726.
Gutema FD, Agga GE, Abdi RD, De Zutter L, Duchateau L, Gabriël S (2019) Prevalence and serotype diversity of Salmonella in apparently healthy cattle: systematic review and meta-analysis of published studies, 2000-2017. Front Vet Sci 6: 102.
Hall RM (2010) Salmonella genomic islands and antibiotic resistance in Salmonella enterica. Future Microbiol 5: 1525-1538.
Hammuel C, Jatau ED, Whong CMZ (2014) Prevalence and antibiogram pattern of some nosocomial pathogens isolated from Hospital environment in Zaria, Nigeria. Aceh Int J Sci Technol 3: 131-139.
Hasman H, Mevius D, Veldman K, Olesen I, Aarestrup FM (2005) Beta-Lactamases among extended-spectrum beta-lactamase (ESBL)-resistant Salmonella from poultry, poultry products and human patients in the Netherlands. J Antimicrob Chemother 56: 115-121.
Hindermann D, Gopinath G, Chase H, Negrete F, Althaus D, Zurfluh K, Tall BD, Stephan R, Nüesch-Inderbinen M (2017) Salmonella enterica serovar Infantis from food and human infections, Switzerland, 2010-2015: poultry-related multidrug resistant clones and an emerging ESBL producing clonal lineage. Front Microbiol 8: 1322.
ISO (2017) ISO 6579-1, Microbiology of the food chain-horizontal method for the detection, enumeration and serotyping of Salmonella – Part 1: detection of Salmonella spp.; International Organization for Standardization, Geneva, Switzerland.
Issenhuth-JeanJean S, Roggentin P, Mikoleit M, Guibourdenche M, de Pinna E, Nair S, Fields IP, Weill FX (2014) Supplement 2008-2010 (no. 48) to the White-Kauffmann Scheme. Res Microbiol 165: 526-530.
Krumperman PH (1983) Multiple antibiotic resistance indexing of Escherichia coli to identify high-risk sources of fecal contamination of foods. Appl Environ Microbiol 46: 165-170.
Le Minor L (1992) The Genus Salmonella. In: Balows A, Truper HG, Dworkin M, Harder W, Schleifer KH A (eds) Handbook on the biology of bacteria: ecophysiology, isolation, ıdentification, Application. Springer-Verlag, New York, Berlin, Heidelberg, pp 2760-2774.
Leverstein-van Hall MA, Blok HE, Donders AR, Paauw A, Fluit AC, Verhoef J (2003) Multidrug resistance among Enterobacteriaceae is strongly associated with the presence of integrons and is independent of species or isolate origin. J Infect Dis 187: 251-259.
Livermore DM, Canton R, Gniadkowski M, Nordmann P, Rossolini GM, Arlet G, Ayala J, Coque TM, Kern -Zdanowicz I, Luzzaro F, Poirel L, Woodford N (2006) CTX-M: changing the face of ESBLs in Europe. J Antimicrob Chemother 59: 165-174.
Lu Y, Wu CM, Wu GJ, Zhao HY, He T, Cao XY, Dai L, Xia LN, Qin SS, Shen JZ (2011) Prevalence of antimicrobial resistance among Salmonella isolates from chicken in China. Foodborne Pathog Dis 8: 45-53.
Machado E, Ferreira J, Novais A, Peixe L, Canton R, Baquero F, Coque TM (2007) Preservation of integron types among Enterobacteriaceae producing extended-spectrum beta-lactamases in a Spanish hospital over a 15-year period (1988 to 2003). Antimicrob Agents Chemother 51: 2201-2204.
Mazel D (2006) Integrons: agents of bacterial evolution. Nat Rev Microbiol 4: 608-620.
McDermott PF, Zhao S, Tate H (2018) Antimicrobial resistance in non typhoidal Salmonella. Microbiol Spectr 6: 6.4.16.
Michalova E, Novotna P, Schlegelova J (2004) Tetracyclines in veterinary medicine and bacterial resistance to them. Vet Med 49: 79-100.
Morshed R, Peighambari SM (2010) Drug resistance, plasmid profile and random amplified polymorphic DNA analysis of Iranian isolates of Salmonella Enteritidis. New Microbiol 33: 47-56.
Ng LK, Martin I, Alfa M, Mulvey M (2001) Multiplex PCR for the detection of tetracycline resistant genes. Mol Cell Probes 15: 209-215.
Nógrády N, Király M, Davies R, Nagy B (2012) Multidrug resistant clones of Salmonella Infantis of broiler origin in Europe. Int J Food Microbiol 157:108-112.
Pan H, Zhou X, Chai W, Paudyal N, Li S, Zhou X, Zhou K, Wu Q, Wu B, Li G, Rajkovic A, Fang W, Rankin SC, Li Y, Xu X, Schifferli DM, Yue M (2019) Diversified sources for human infections by Salmonella enterica serovar Newport. Transbound Emerg Dis 66: 1044-1048.
Pate M, Micunovic J, Golob M, Vestby LK, Ocepek M (2019) Salmonella Infantis in broiler flocks in Slovenia: the prevalence of multidrug resistant strains with high genetic homogeneity and low biofilm-forming ability. Biomed Res Int 2019: 4981463.
Paterson DL, Bonomo RA (2005) Extended-spectrum beta-lactamases: a clinical update. Clin Microbiol Rev 18: 657-686.
Paudyal N, Pan H, Elbediwi M, Zhou X, Peng X, Li X, Fang W, Yue M (2019) Characterization of Salmonella Dublin isolated from bovine and human hosts. BMC Microbiol 19: 226.
Proietti PC, Stefanetti V, Musa L, Zicavo A, Dionisi AM, Bellucci S, Mensa AL, Menchetti L, Branciari R, Ortenzi R, Franciosini MP (2020) Genetic profiles and antimicrobial resistance patterns of Salmonella Infantis strains isolated in Italy in the food chain of broiler meat production. Antibiotics 9: 814
Rahmani M, Peighambari SM, Svendsen CA, Cavaco LM, Agersø Y, Hendriksen RS (2013) Molecular clonality and antimicrobial resistance in Salmonella enterica serovars Enteritidis and Infantis from broilers in three Northern regions of Iran. BMC Vet Res 9: 66.
Rao S, Maddox CW, Hoien-Dalen P, Lanka S, Weigel RM (2008) Diagnostic accuracy of Class I integron PCR method in detection of antibi-otic resistance in Salmonella isolates from swine production systems. J Clin Microbiol 46: 916-920.
Revathi G, Shannon KP, Stapleton PD, Jain BK, French GL (1998) An outbreak of extended-spectrum, beta-lactamase producing Salmonella Senftenberg in a burns ward. J Hosp Infect 40: 295-302.
Rhouma M, Letellier A (2017) Extended-spectrum beta-lactamases, carbapenemases and the mcr-1 gene: is there a historical link? Int J Anti-microb Agents 49: 269-271.
Roca I, Akova M, Baquero F, Carlet J, Cavaleri M, Coenen S, Cohen J, Findlay D, Gyssens I, Heure OE, Kahlmeter G, Kruse H, Laxmina-rayan R, Liébana E, López-Cerero L, MacGowan A, Martins M, Rodríguez-Baño J, Rolain J-M, Segovia C, Siqauque B, Tacconelli E, Wel-lington E, Vila J (2015) The global threat of antimicrobial resistance: science for intervention. New Microbes New Infect 6: 22-29.
Sefton AM (2002) Mechanisms of antimicrobial resistance: their clinical relevance in the new millennium. Drugs 62: 557-566.
Thomas KM, de Glanville WA, Barker CG, Benschop J, Buza JJ, Cleaveland S, Davis MA, French NP, Mmbaga BT, Prinsen G, Swai ES, Zadoks RN, Crump JA (2020) Prevalence of Campylobacter and Salmonella in African food animals and meat: a systematic review and meta-analysis. Int J Food Microbiol 315: 108382.
Threlfall EJ (2000) Epidemic Salmonella Typhimurium DT 104 - a truly international multiresistant clone. J Antimicrob Chemother 46: 7-10.
Threlfall EJ (2002) Antimicrobial drug resistance in Salmonella: problems and perspectives in food-and water-borne infections. FEMS Microbiol Rev 26: 141-148.
Trongjit S, Angkititrakul S, Tuttle RE, Poungseree J, Padungtod P, Chuanchuen R (2017) Prevalence and antimicrobial resistance in Salmonella enterica isolated from broiler chickens, pigs and meat products in Thailand -Cambodia border provinces. Microbiol Immu-nol 61: 23-33.
Yan SS, Pendrak ML, Abela-Ridder B, Punderson JW, Fedorko DP, Foley SL (2003) An overview of Salmonella typing: Public health perspectives. Clin Applied Immunol Rev 4: 189-204.
Yusuf E, Bax HI, Verkaik NJ, van Westreenen M (2021) An update on eight “New” antibiotics against multidrug -resistant Gram-negative bacteria. J Clin Med 10: 1068.
Zhao X, Hu M, Zhang Q, Zhao C, Zhang Y, Li L, Qi J, Luo Y, Zhou D, Liu Y (2020) Characterization of integrons and antimicrobial re-sistance in Salmonella from broilers in Shandong, China. Poult Sci 99: 7046-7054.
Zhao X, Ye C, Chang W, Sun S (2017a) Serotype distribution, antimicrobial resistance, and class 1 integrons profiles of Salmonella from animals in slaughterhouses in Shandong Province, China. Front Microbiol 8: 1049.
Zhao X, Yang J, Zhang B, Sun S, Chang W (2017b) Characterization of integrons and resistance genes in Salmonella isolates from farm animals in Shandong Province, China. Front Microbiol 8: 1300.
Zwe YH, Yen-Tang VC, Aung KT, Gutiérrez RA, Ng LC, Yuk HG (2018) Prevalence, sequence types, antibiotic resistance and, gyrA muta-tions of Salmonella isolated from retail fresh chicken meat in Singapore. Food Control 90: 233-240.
Go to article

Authors and Affiliations

O. Sahan Yapicier
1
D. Ozturk
2

  1. Republic of Turkey Ministry of Agriculture and Forestry Veterinary Control Central Research Institute, Bacteriological Diagnostic Laboratory, 06020, Ankara, Turkey
  2. Mehmet Akif Ersoy University, Faculty of Veterinary Medicine, Department of Microbiology, 15030, Burdur, Turkey

This page uses 'cookies'. Learn more