Life Sciences and Agriculture

Polish Journal of Veterinary Sciences

Content

Polish Journal of Veterinary Sciences | 2024 | vol. 27 | No 1

Download PDF Download RIS Download Bibtex

Abstract

Coursing is a canine sport discipline invented for sighthounds. The dogs chase the mechanically operated lure on an open area at the track that consists of straight lines as well as turns. Thus, the dogs must rely mainly on their sight in order to chase the lure and to compete.
Whippets are the most popular sighthound breed in Poland and more and more dogs practice lure coursing on professional level. Several ocular disorders are known or presumed to be inherited in Whippets, so we decided to check how often they occur amongst competing dogs and if they have any impact on the results during competitions.
Forty-five regularly competing Whippets underwent complete ophthalmic examination, including evaluation of menace response, pupillary light reflex and dazzle reflex, biomicroscopic examination conducted before and after dilating pupils with topical tropicamide and fundus examination with indirect ophthalmoscopy. Refractive state of each eye was assessed via streak retinoscopy.
Ocular abnormalities were identified in 24 dogs (53.3%). The most common finding was vitreous degeneration identified in 9 dogs (20%), five dogs presented multiple lesions. There were no statistically significant differences regarding sex, age, and ranking points between the dogs with and without lesions.
However, some of the identified lesions are likely to progress, so we postulate that the ophthalmological examination should receive more attention in sport Whippets, to check if it has any impact on competing dogs at more advanced age.
Go to article

Bibliography

1. Act of 15 January 2015 on the protection of animals used for scientific or educational purposes, art 1.2 (5), Dz.U.2018.0.1207 https://isap.sejm.gov.pl/isap.nsf/download. xsp/WDU20150000266/U/D20150266Lj.pdf (Accessed on 3 February 2023)
2. .Act of 13 October 1995 on hunting law, Dz. U. 1995 Nr 147 poz. 713 https://isap.sejm.gov.pl/isap.nsf/download.xsp/WDU19951470713/U/D19950713Lj.pdf (Accessed on 3 February 2023)
3. Balicki I, Goleman M, Balicka A (2021) Ocular Abnormalities in Polish Hunting Dogs. Plos One 5; 16(11): e0258636
4. Bishop PN, Holmes DF, Kadler KE, Mcleod D, Bos KJ (2014) Age-related changes on the surface of vitreous collagen fibrils. Investig Oph-thalmol Vis Sci 45: 1041-1046
5. Coursing Polish Cup Results, 2021 https://coursing24.eu/ cupResultsPublic/2 (Accessed on 5 January 2023).
6. Crasta M, Arteaga K, Barachetti L, Guandalini A (2022) A multicenter retrospective evaluation of the prevalence of known and presumed heredi-tary eye diseases in Lagotto Romagnolo dog breed within a referral population in Italy (2012-2020). Vet Ophthalmol 25: 426-433
7. Day G, Powers M, Gyes N, Plummer L (2015) Visual Disorders in Agility Dogs with Jumping Problems. http://www. awe-somepaws.us/wp-content/uploads/2015/04/caninevision-project.pdf (Accessed on 3 February 2023)
8. de Oliveira JK, Bortolini M, Schaller M, Schuchmann RK, Moore BA, Montiani-Ferreira F (2020) The ophthalmic health and refractive state of working dogs in South Brazil. Open Vet J 10: 22-30. ECVO Manual: Breeds, 2021 https://www.ecvo.eu/media/ whippet.pdf (Accessed on 3 February 2023)
9. FCI International Guidelines for Lure Coursing Judges. www.fci.be (Accessed on 3 February 2023) Guandalini A, Di Girolamo N, Corvi R, Santillo D, Andreani V, Pinzo B (2017) Epidemiology of ocular disorders presumed to be inherited in three small Italian dog breeds in Italy. Vet Ophthalmol 21: 524-529. Hodowle – Whippety w Polsce (2022) http://www.whippety. topcharty.net/hodowle.php (Accessed on 11 November 2022)
10. Karamatic S, Goode R, Bageswaran N, Willet CE, Samaha G, Fergusion R, Mazrier H, Wade CM (2022) Genome-Wide Association Analysis for Chronic Superficial Keratitis in the Australian Racing Greyhound. Genes 13: 1328.
11. Kaukonen M, Lohi H (2018) Clinical and Genetic Characterization of Canine Vitreous Degeneration. ARVO Annual Meeting, Honolulu, Hawaii. Invest Ophthalmol Vis Sci 59: 6042.
12. Krishnan H, Diehl K, Stefanovski D, Aguirre GD (2019) Vitreous degeneration and associated ocular abnormalities in the dog. Vet Ophthalmol 23: 219-224.
13. Kubai MA, Bentley E, Miller PE, Mutti DO, Murphy CJ (2008) Refractive states of eyes and association between ametropia and breed in dogs. Amer J Vet Res 69: 946-951.
14. Lynch GL (2007) Ophtalmic examination findings in a group of retired racing Greyhounds. Vet Ophthalmol 10: 363-367.
15. Murphy CJ, Zodnik K, Mannis MJ (1997) Myopia and Refractive Error in Dogs. Investig Ophthalmol Vis Sci 33: 2459-2463.
16. Palmer SV, Gomes FE, McArt JA (2021) Ophtalmic disorders in a reffereal population of seven breeds of brachycephalic dogs: 970 cases (2008-2017). J Am Vet Med Assoc 259: 1318-1324.
17. Pont TR, Matas Riera M, Newton R, Donaldson D (2016) Corneal and anterior segment foreign body trauma in dogs: a review of 218 cases. Vet Ophthalmol 19: 386-397.
18. Somma AT, Moreno JC, Sato MT, Rodrigues BD, Bacellar-Galdino M, Occelli LM, Petersen-Jones SM, Montiani-Ferreira F (2017) Characteriza-tion of a novel form of progressive retinal atrophy in Whippet dogs: a clinical, electroretinographic, and breeding study. Vet Ophthalmol 20: 450-459.
19. Vitreous Degeneration in Whippets (2021) https://www.akcchf.org/research/research-portfolio/0011.html (Accessed on 9 January 2023)
Go to article

Authors and Affiliations

K. Miazga
1 2
J Tomkowicz
3
J Wilczak
4
A. Cywińska
5

  1. Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159c, 02-776 Warsaw, Poland
  2. Municipal Zoological Garden in Warsaw, Ratuszowa 1/3, 03‑461 Warsaw, Poland
  3. Veterinary Ophtalmology Center “EyeVet”, Chałupnicza 67, 51-503 Wrocław, Poland
  4. Department of Physiology, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159c, 02-776 Warsaw, Poland
  5. Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, Lwowska 1, 87-100, Torun, Poland
Download PDF Download RIS Download Bibtex

Abstract

Listeria monocytogenes is a ubiquitous microorganism that is isolated from a variety of sources such as soil, water, decaying vegetation, sewage, animal feeds, silage, farm environments and food-processing environments. This study aimed to determine the prevalence, serogroups, biofilm formation, virulence factor genes, and genetic relationships of L. monocytogenes strains isolated from beef meat and meat contact surfaces obtained from a slaughterhouse in Burdur, Turkey. In this study, a total of 179 beef meat and meat contact surface samples were analyzed for the presence of L. monocytogenes by polymerase chain reaction (PCR). Out of a total of 179 beef meat and meat contact surface samples, 83 (46.37%) were found to be contaminated with L. monocytogenes, with the highest incidence (53.01%) occurring in beef meat. In the present study, most of the isolated strains belonged to serogroups IIB and IVB (lineage I). The L. monocytogenes strain also contained monoA-B, prfA, plcA, plcB, mpl, hlyA, actA, gtcA, dltA, Fri, flaA, InlA, InlC, InlJ, and iap genes. Biofilm formation was not determined in the tested samples at pH 5.5 and different temperatures (4°C, 10°C, 25°C, and 37°C). However, strong biofilm formation was observed in 6.45% (2/31) of the strains at pH 7.0 after 48 h incubation at 37°C, and in 3.22% (1/31) of the strains at pH 7.0 after 48 h incubation at 4°C and 10°C. Pulsed-field gel electrophoresis (PFGE) results showed that L. monocytogenes isolates were clonally related, and cross-contamination was present. In addition, PFGE results also revealed that AscI had more distinguishing power than the ApaI restriction enzyme. These results indicate that L. monocytogenes detected from meat and meat contact surfaces in the slaughterhouse pose a potential risk to public health.
Go to article

Bibliography

1. Abachin E, Poyart C, Pellegrini E, Milohanic E, Fiedler F, Berche P, Trieu-Cuot P (2002) Formation of D-alanyl-lipoteichoic acid is required for adhesion and virulence of Listeria monocytogenes. Mol Microbiol 43: 1-14.
2. Agostinho Davanzo EF, Dos Santos RL, Castro VH, Palma JM, Pribul BR, Dallago BS, Fuga B, Medeiros M, Titze de Almeida SS, da Costa HM, Rodrigues DD, Lincopan N, Perecmanis S, Santana AP (2021) Molecular characterization of Salmonella spp. and Listeria monocytogenes strains from biofilms in cattle and poultry slaughterhouses located in the federal District and State of Goia´s, Brazil. PLoS One 16: e0259687.
3. Andrade JC, João AL, Alonso CS, Barreto AS, Henriques AR (2020) Genetic subtyping, biofilm-forming ability and biocide suscepti-bility of Listeria monocytogenes strains isolated from a ready-to-eat food industry. Antibiotics (Basel) 9: 416.
4. Arslan S, Baytur S (2019) Prevalence and antimicrobial resistance of Listeria species and subtyping and virulence factors of Listeria monocytogenes from retail meat. J Food Saf 39: e12578.
5. Ayaz ND, Cufaoglu G (2016) Listeria monocytogenes as a foodborne pathogen: Biocontrol in foods using lytic bacteriophages. J Clin Microbiol Biochem Technol 2: 035-039.
6. Bubert A, Köhler S, Goebel W (1992) The homologous and heterologous regions within the iap gene allow genus- and species-specific identification of Listeria spp. by polymerase chain reaction. Appl Environ Microbiol 58: 2625-2632.
7. Bubert A, Riebe J, Schnitzler N, Schönberg A, Goebel W, Schubert P (1997) Isolation of catalase-negative Listeria monocytogenes strains from listeriosis patients and their rapid ıdentification by anti-p60 antibodies and/or PCR. J Clin Microbiol 35: 179-183.
8. Bubert A, Sokolovic Z, Chun SK, Papatheodorou L, Simm A, Goebel W (1999) Differential expression of Listeria monocytogenes virulence genes in mammalian host cells. Mol Gen Genet 261: 323-336.
9. Boukili M, Filali FR, Lafkih N, Bouymajane A, Sefiani M, Moumni M (2020) Prevalence, characterization and antimicrobial resistance of Listeria monocytogenes isolated from beef meat in Meknes city, Morocco. Germs 10: 74-80.
10. Çadırcı Ö, Gücükoğlu A, Terzi GG, Uyanık T, Alişarlı M (2018) The existence of Listeria monocytogenes in a cattle slaughterhouse and identification of serotypes by mPCR. Ankara Univ Vet Fak Derg 65: 305-311.
11. CDC (2017) Standard Operating Procedure for PulseNet PFGE of Listeria monocytogenes. https://www.cdc.gov/pulsenet/pdf/listeria-pfge-protocol-508c.pdf
12. Chavant P, Martinie B, Meylheuc T, Bellon-Fontaine MN, Hebraud M (2002) Listeria monocytogenes LO28: surface physicochemical properties and ability to form biofilms at different temperatures and growth phases. Appl Environ Microbiol 68: 728-737.
13. Chen M, Cheng J, Zhang J, Chen Y, Zeng H, Xue L, Lei T, Pang R, Wu S, Wu H, Zhang S, Wei X, Zhang Y, Ding Y, Wu Q (2019) Isolation, potential virulence, and population diversity of Listeria monocytogenes from meat and meat products in China. Front Microbiol 10: 946.
14. Cherifi T, Arsenault J, Pagotto F, Quessy S, Côté JC, Neira K, Fournaise S, Bekal S, Fravalo P (2020) Distribution, diversity and per-sistence of Listeria monocytogenes in swine slaughterhouses and their association with food and human listeriosis strains. PLoS One 15: e0236807.
15. Coban A, Pennone V, Sudagidan M, Molva C, Jordan K, Aydin A (2019) Prevalence, virulence characterization, and genetic related-ness of Listeria monocytogenes isolated from chicken retail points and poultry slaughterhouses in Turkey. Braz J Microbiol 50: 1063-1073.
16. Costa M, Pracca G, Sucari A, Galli L, Ibargoyen J, Gentiluomo J, Brusa V, Zugazua MM, Figueroa Y, Londero A, Roge A, Silva H, Der Ploeg CV, Signorini M, Oteiza JM, Leotta GA (2020) Comprehensive evaluation and implementation of improvement actions in bovine abattoirs to reduce pathogens exposure. Prev Vet Med 176: 104933.
17. Demaître N, Van Damme I, De Zutter L, Geeraerd AH, Rasschaert G, De Reu K (2020) Occurrence, distribution and diversity of Lis-teria monocytogenes contamination on beef and pig carcasses after slaughter. Meat Sci 169: 108177.
18. Doumith M, Buchrieser C, Glaser P, Jacquet C, Martin P (2004) Differentiation of the major Listeria monocytogenes serovars by multiplex PCR. J Clin Microbiol 42: 3819-3822.
19. Dussurget O, Dumas E, Archambaud C, Chafsey I, Chambon C, Hébraud M, Cossart P (2005) Listeria monocytogenes ferritin protects against multiple stresses and is required for virulence. FEMS Microbiol Lett 250: 253-261.
20. Furrer B, Candrian U, Hoefelein C, Luethy J (1991) Detection and identification of Listeria monocytogenes in cooked sausage products and in milk by in vitro amplification of haemolysin gene fragments. J Appl Bacteriol 70: 372-379.
21. Graves LM, Hunter SB, Ong AR, Schoonmaker-Bopp D, Hise K, Kornstein L, DeWitt WE, Hayes PS, Dunne E, Mead P, Swaminathan B (2005) Microbiological aspects of the investigation that traced the 1998 outbreak of listeriosis in the United States to contaminated hot dogs and establishment of molecular subtyping-based surveillance for Listeria monocytogenes in the PulseNet Network. J Clin Microbiol 43: 2350-2355.
22. Hellström S (2011) Contamination routes and control of Listeria monocytogenes in food production. Academic dissertation. University of Helsinki, Faculty of Veterinary Medicine, Helsinki, Finland. SBN 978-952-10-7109-6 (PDF)
23. Hitchins AD, Jinneman K, Chen Y (2022) Chapter 10: Detection of Listeria monocytogenes in foods and environmental samples, and enumeration of Listeria monocytogenes in foods. In: Food and Drug Administraiton Bacteriological Analytical Manual (BAM), Food and Drug Administraiton. https://www.fda.gov/food/laboratory-methods-food/bamchapter-10-detection-listeria-monocytogenes-foods-andenvironmental-samples-and-enumeration
24. Iglesias MA, Kroning IS, Decol LT, de Melo Franco BD, da Silva WP (2017) Occurrence and phenotypic and molecular characteriza-tion of Listeria monocytogenes and Salmonella spp. in slaughterhouses in southern Brazil. Food Res Int 100: 96-101.
25. ISO (2017) International Organization for Standardization, EN ISO 11290-1:2017. Microbiology of the food chainHorizontal method for the detection and enumeration of Listeria monocytogenes and of Listeria spp. Part 1: Detection method. https://www.iso.org/obp/ui/#iso:std:iso: 11290:-1:ed-2:v1:en
26. Jang YS, Moon JS, Kang HJ, Bae D, Seo KH (2021) Prevalence, characterization, and antimicrobial susceptibility of Listeria monocytogenes from raw beef and slaughterhouse environments in Korea. Foodborne Pathog Dis 18: 419-425.
27. Jaradat ZW, Schutze GE, Bhunia AK (2002) Genetic homogeneity among Listeria monocytogenes strains from infected patients and meat products from two geographic locations determined by phenotyping, ribotyping and PCR analysis of virulence genes. Int J Food Microbiol 76: 1-10.
28. Jennison AV, Masson JJ, Fang NX, Graham RM, Bradbury MI, Fegan N, Gobius KS, Graham TM, Guglielmino CJ, Brown JL, Fox EM (2017) Analysis of the Listeria monocytogenes population structure among isolates from 1931 to 2015 in Australia. Front Microbiol 8: 603.
29. Kayode AJ, Igbinosa EO, Okoh AI (2019) Overview of listeriosis in the Southern African Hemisphere-Review. J Food Saf 40: e12732.
30. Kyoui D, Takahashi H, Miya S, Kuda T, Kimura B (2014) Comparison of the major virulence-related genes of Listeria monocytogenes in Internalin A truncated strain 36-25-1 and a clinical wild-type strain. BMC Microbiol 14: 15.
31. Leimeister-Wachter M, Domann E, Chakraborty T (1991) Detection of a gene encoding a phosphatidylinositolspecific phospholipase C that is co-ordinately expressed with listeriolysin in Listeria monocytogenes. Mol Microbiol 5: 361-366.
32. Li X, Shi X, Song Y, Yao S, Li K, Shi B, Sun J, Liu Z, Zhao W, Zhao C, Wang J (2022) Genetic diversity, antibiotic resistance, and virulence profiles of Listeria monocytogenes from retail meat and meat processing. Food Res Int 162: 112040.
33. Liu D, Ainsworth AJ, Austin FW, Lawrence ML (2004) Use of PCR primers derived from a putative transcriptional regulator gene for species-specific determination of Listeria monocytogenes. Int J Food Microbiol 91: 297-304.
34. Liu D, Lawrence ML, Ainsworth AJ, Austin FW (2008) Genotypic identification. In: Liu D (ed) Handbook of Listeria monocytogenes, 1st ed., Boca Raton: CRC Press, Taylor & Francis Group, pp 169-202.
35. Liu D, Lawrence ML, Austin FW, Ainsworth AJ (2007) A multiplex PCR for species- and virulence-specific determination of Listeria monocytogenes. J Microbiol Methods 71: 133-140.
36. Mazaheri T, Ripolles-Avila C, Hascoët AS, Rodríguez-Jerez JJ (2020) Effect of an enzymatic treatment on the removal of mature Listeria monocytogenes biofilms: A quantitative and qualitative study. Food Control 114: 107266.
37. Neves E, Lourenco A, Silva AC, Coutinho R, Brito L (2008) Pulsed-field gel electrophoresis (PFGE) analysis of Listeria monocytogenes isolates from different sources and geographical origins and representative of the twelve serovars. Syst Appl Microbiol 31: 387-392.
38. Nishibori T, Cooray K, Xiong H, Kawamura I, Fujita M, Mitsuyama M (1995) Correlation between the presence of virulence-associated genes as determined by PCR and actual virulence to mice in various strains of Listeria spp. Microbiol Immunol 39: 343-349.
39. Oevermann A, Zurbriggen, A Vandevelde M (2010) Rhombencephalitis caused by Listeria monocytogenes in humans and ruminants: a zoonosis on the rise? Interdiscip Perspect Infect Dis 2010: 632513.
40. Oh H, Kim S, Lee S, Lee H, Ha J, Lee J, Choi Y, Choi KH, Yoon Y (2018) Prevalence, serotype diversity, genotype and antibiotic resistance of Listeria monocytogenes isolated from carcasses and human in Korea. Korean J Food Sci Anim Resour 38: 851-865.
41. Olaimat AN, Al-Holy MA, Shahbaz HM, Al-Nabulsi AA, Abu Ghoush MH, Osaili TM, Ayyash MM, Holley RA (2018) Emergence of antibiotic resistance in Listeria monocytogenes isolated from food products: a comprehensive review. Compr Rev Food Sci Food Saf 17: 1277-1292.
42. Orsi RH, den Bakker HC, Wiedmann M (2011) Listeria monocytogenes lineages: Genomics, evolution, ecology, and phenotypic characteristics. Int J Med Microbiol 301: 79-96.
43. Orsi RH, Wiedmann M (2016) Characteristics and distribution of Listeria spp., including Listeria species newly described since 2009. Appl Microbiol Biotechnol 100: 5273-5287.
44. Papatzimos G, Kotzamanidis C, Kyritsi M, Malissiova E, Economou V, Giantzi V, Zdragas A, Hadjichristodoulou C, Sergelidis D (2022) Prevalence and characteristics of Listeria monocytogenes in meat, meat products, food handlers and the environment of the meat processing and the retail facilities of a company in Northern Greece. Lett Appl Microbiol 74: 367-376.
45. Penesyan A, Paulsen IT, Kjelleberg S, Gillings MR (2021) Three faces of biofilms: a microbial lifestyle, a nascent multicellular organism, and an incubator for diversity. NPJ Biofilms and Microbiomes, 7: 80.
46. Poimenidou SV, Dalmasso M, Papadimitriou K, Fox EM, Skandamis PN, Jordan K (2018) Virulence gene sequencing highlights similarities and differences in sequences in Listeria monocytogenes Serotype 1/2a and 4b strains of clinical and food origin from 3 different geographic locations. Front Microbiol 9: 1103.
47. Promadej N, Fiedler F, Cossart P, Dramsi S, Kathariou S (1999) Cell wall teichoic acid glycosylation in Listeria monocytogenes serotype 4b requires gtcA, a novel, serogroup-specific gene. J Bacteriol 181: 418-425.
48. Sahin S, Mogulkoç MN, Kalın R (2020) Prevalence and serotype distribution of Listeria monocytogenes isolated from retail raw meats. J Fac Vet Med Erciyes Univ 17: 22-27.
49. Slama RB, Miladi H, Chaieb K, Bakhrouf A (2013) Survival of Listeria monocytogenes cells and the effect of extended frozen storage (-20°C) on the expression of its virulence gene. Appl Biochem Biotechnol 170: 1174-1183.
50. Soni DK, Singh M, Singh DV, Dubey SK (2014) Virulence and genotypic characterization of Listeria monocytogenes isolated from vegetable and soil samples. BMC Microbiol 14: 241.
51. Sudagidan M, Cavusoglu C, Bacakoglu F (2008) Investigation of the virulence genes in methicillin-resistant Staphylococcus aureus strains isolated from biomaterial surfaces. Mikrobiyol Bul 42: 29-39.
52. Teixeira LA, Carvalho FT, Vallim DC, Pereira RC, Neto AC, Vieira BS, Carvalho RC, Figueiredo EE (2020) Listeria monocytogenes in export-approved beef from Mato Grosso, Brazil: prevalence, molecular characterization and resistance to antibiotics and disinfectants. Microorganisms 8: 18.
53. Vasquez-Boland JA, Kocks C, Dramsi S, Ohayon H, Geoffroy C, Mengaud J, Cossart P (1992) Nucleotide sequence of the lecithinase operon of Listeria monocytogenes and possible role of lecithinase in cell-to-cell spread. Infect Immun 60: 219-230.
54. Yan H, Neogi SB, Mo Z, Guan W, Shen Z, Zhang S, Li L, Yamasaki S, Shi L, Zhong N (2010) Prevalence and characterization of an-timicrobial resistance of foodborne Listeria monocytogenes isolates in Hebei province of Northern China, 2005-2007. Int J Food Mi-crobiol 144: 310-316.
55. Yucel N, Citak S, Onder M (2005) Prevalence and antibiotic resistance of Listeria species in meat products in Ankara, Turkey. Food Microbiol 22: 241-245.
56. Zhang H, Wang J, Chang Z, Liu X, Chen W, Yu Y, Wang X, Dong Q, Ye Y, Zhang X (2021) Listeria monocytogenes contamination characteristics in two ready-to-eat meat plants from 2019 to 2020 in Shanghai. Front Microbiol 12: 729114.
Go to article

Authors and Affiliations

F. Tasci
1
M. Sudagidan
2
O. Yavuz
2
A. Soyucok
3
A. Aydin
4

  1. Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, 15030, Istiklal Campus, Burdur, Turkey
  2. Scientific and Technology Application and Research Center, Burdur Mehmet Akif Ersoy University, Burdur, 15030, Istiklal Campus, Burdur, Turkey
  3. Department of Food Processing, Food Agriculture and Livestock Vocational School, Burdur Mehmet Akif Ersoy University, 15030, Istiklal Campus, Burdur, Turkey
  4. Department of Food Hygiene and Technology, Faculty of Veterinary Medicine,Istanbul University-Cerrahpasa, 34320, Avcilar, Istanbul, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Candida albicans, a polymorphic yeast, is a physiological component of the human and animal commensal microbiome. It is an etiological factor of candidiasis, which is treated by azole antifungals. Growing resistance to azoles is a reason to look for other alternative treatment options. The pharmacotherapeutic use of plant extracts and essential oils has become increasingly important. In our experiment, C. albicans showed susceptibility to four observed plant extracts and essential oils from peppermint ( Mentha piperita), thyme ( Thymus vulgaris), sage ( Salvia officinalis), and oregano ( Origanum vulgare). Oregano plant extract and essential oil showed the highest antifungal activity, at MIC values of 4.9 mg/mL and 0.4 mg/mL respectively. Therefore, it was subjected to further research on the influence of virulence factors – biofilm formation, extracellular phospholipase production and germ tube formation. Oregano plant extract and essential oil showed an inhibitory effect on the observed C. albicans virulence factors at relatively low concentrations. The extract inhibited the adherence of cells at MIC 12.5 mg/mL and essential oil at MIC 0.25 mg/mL. Degradation of the formed biofilm was detected at MIC 14.1 mg/mL for plant extract and at MIC 0.4 mg/mL for essential oil. Extracellular phospholipase production was most effectively inhibited by the essential oil. In particular, the number of isolates with intensive extracellular phospholipase production decreased significantly. Of the 12 isolates intensively producing extracellular phospholipase, only 1 isolate (4.5%) retained intense production. Essential oil caused up to a 100 % reduction in germ tubes formation and plant extract reduced their formation depending on the concentration as follows: 2.6% (0.8 mg/mL), 21.2 % (6.25 mg/mL), and 64.5 % (12.5 mg/mL) compared to the control.
Go to article

Bibliography

1. Brondani LP, da Silva Neto TA, Freitag RA, Lund RG (2018) Evaluation of anti-enzyme properties of Origanum vulgare essential oil against oral Candida albicans. J Mycol Med 28: 94-100.
2. CLSI, Clinical and Laboratory Standards Institute (2017) M27-A3: Reference method for broth dilution antifungal susceptibility testing of yeasts; approved standard. 4th ed. USA: Wayne, PA.
3. de Souza Ramos L, Barbedo LS, Braga-Silva LA, dos Santos AL, Pinto MR, da Graça Sgarbi DB (2015) Protease and phospholipase activities of Candida spp. isolated from cutaneous candidiasis. Rev Iberoam Micol 32: 122-125.
4. Doke SK, Raut JS, Dhawale S, Karuppayil SM (2014) Sensitization of Candida albicans biofilms to fluconazole by terpenoids of plant origin. J Gen Appl Microbiol 60: 163-168.
5. Edelmann A, Krüger M, Schmid J (2005) Genetic relationship between human and animal isolates of Candida albicans. J Clin Micro-biol 43: 6164-6166.
6. Ellepola AN, Samaranayake LP, Khan ZU (2016) Extracellular phospholipase production of oral Candida albicans isolates from smok-ers, diabetics, asthmatics, denture wearers and healthy individuals following brief exposure to polyene, echinocandin and azole antimy-cotics. Braz J Microbiol 47: 911-916.
7. Fule SR, Das D, Fule RP (2015) Detection of phospholipase activity of Candida albicans and non albicans isolated from women of reproductive age with vulvovaginal candidiasis in rural area. Indian J Med Microbiol 33: 92-95.
8. Jin Y, Yip HK, Samaranayake YH, Yau JY, Samaranayake LP (2003) Biofilm-forming ability of Candida albicans is unlikely to con-tribute to high levels of oral yeast carriage in cases of human immunodeficiency virus infection. J Clin Microbiol 41: 2961-2967.
9. Kumamoto CA, Gresnigt MS, Hube B (2020) The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine. Curr Opin Microbiol 56: 7-15.
10. Lee H, Woo ER, Lee DG (2018) Apigenin induces cell shrinkage in Candida albicans by membrane perturbation. FEMS Yeast Res 18: 10.1093.
11. Martins N, Ferreira IC, Barros L, Silva S, Henriques M (2014) Candidiasis: predisposing factors, prevention, diagnosis and alternative treatment. Mycopathologia 177: 223-240.
12. Mattei AS, Alves SH, Severo CB, da Silva Guazzelli L, de Mattos Oliveira F, Severo LC (2013) Determination of germ tube, phos-pholipase, and proteinase production by bloodstream isolates of Candida albicans. Rev Soc Bras Med Trop 46: 340-342.
13. Mohandas V, Ballal M (2011) Distribution of Candida species in different clinical samples and their virulence: biofilm formation, pro-teinase and phospholipase production: a study on hospitalized patients in southern India. J Glob Infect Dis 3: 4-8.
14. Nagy M, Mučaji P, Grančai D (2017) Pharmacognosy. Biologically active plant metabolites and their sources, 2nd ed., Bratislava, Herba.
15. Pozzatti P, Loreto ES, Nunes Mario DA, Rossato L, Santurio JM, Alves SH (2010) Activities of essential oils in the inhibition of Can-dida albicans and Candida dubliniensis germ tube formation. J Mycol Med 20: 185-189.
16. Pristov KE, Ghannoum MA (2019) Resistance of Candida to azoles and echinocandins worldwide. Clin Microbiol Infect 25: 792-798.
17. Raut JS, Karuppayil SM (2014) A status review on the medicinal properties of essential oils. Ind Crop Prod 62: 250-264.
18. Rossoni RD, Barbosa JO, Vilela SF, dos Santos JD, Jorge AO, Junqueira JC (2013) Correlation of phospholipase and proteinase pro-duction of Candida with in vivo pathogenicity in Galleria mellonella. Braz J Oral Sci 12: 199-204.
19. Ruchi T, Sujata B, Anuradha D (2015) Comparison of phenotypic methods for the detection of biofilm production in uro-pathogens in a tertiary care hospital in India. Int J Curr Microbiol App Sci 4: 840-849.
20. Seyedmousavi S, Bosco SM, de Hoog S, Ebel F, Elad D, Gomes RR, Jacobsen ID, Jensen HE, Martel A, Mignon B, Pasmans F, Piecková E, Rodrigues AM, Singh K, Vicente VA, Wibbelt G, Wiederhold NP, Guillot J (2018) Fungal infections in animals: a patch-work of different situations. Med Mycol 56 (Suppl 1): 165-187.
21. Silva S, Rodrigues CF, Araújo D, Rodrigues ME, Henriques M (2017) Candida species biofilms’ antifungal resistance. J Fungi (Ba-sel) 3: 8.
22. Vitális E, Nagy F, Tóth Z, Forgács L, Bozó A, Kardos G, Majoros L, Kovács R (2020) Candida biofilm production is associated with higher mortality in patients with candidaemia. Mycoses 63: 352-360.
Go to article

Authors and Affiliations

P. Váczi
1
M. Proškovcová
1
E. Čonková
1
D. Marcinčáková
1
M. Bačkorová
2
M. Harčárová
3

  1. Department of Pharmacology and Toxicology
  2. Department of Pharmaceutical Technology, Pharmacognosy and Botany
  3. Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy, Komenského 73, Košice, 041 81, Slovakia
Download PDF Download RIS Download Bibtex

Abstract

Antiseptic agents used in the postoperative period affect the functions of many tissues in the body, including the testicles. In this study, the effect of dressings administered with different antiseptic agents on testicular functions in rats that underwent abdominal incisions was investigated. A total of 48 Sprague-Dawley rats were used in the study. Each of the rats in the study group underwent a 4 cm-long skin and muscle operation. The incision was then stitched immediately. Antiseptics, hemp seed oil, hemp leaf oil, and cannabidiol oil were then administered to the rats for 10 days to provide antisepsis. The rats were sacrificed 24 hours after the last administration, and testicular tissues were removed. Testicular tissues were used for histopathological examination and biochemical analysis, while epididymal tissue was used for sperm analysis. According to the results, the MDA level in the antiseptic-administered group was higher than in the other experimental groups (p<0.05). Levels of SOD, CAT activities, and GSH content were found to be lower in the antiseptic group than in the hemp seed oil, hemp leaf oil, and cannabidiol oil groups (p<0.05). In testicular histology, the SEED group had the highest Johnsen score, and the antiseptic group had the lowest score (p<0.05). While JAK, P-JAK2, STAT3, PSTAT3, and NF-κB were generally higher in the antiseptic group compared to the other groups, they were lower in the SEED group. Additionally, sperm total motility rate and epididymal sperm density were highest in the SEED group (p<0.05). As a result, it was determined that cannabidiol seed oil had a good effect on testicular histology and sperm quality in male rats during the wound healing process.
Go to article

Bibliography

1. Aebi H (1984) Catalase in vitro. Methods Enzymol 105: 121-126.
2. Akarsu SA, Acısu TC, Güngör İH, Cihangiroǧlu AÇ, Koca RH, Türk G, Sönmez M, Gür S, Fırat F, Duruel HE (2023) The effect of luteolin on spermatological parameters, apoptosis, oxidative stress rate in freezing rabbit semen. Pol J Vet Sci 26: 91-98.
3. Aksu EH, Kandemir FM, Küçükler S (2021) Ameliorative effect of hesperidin on streptozotocin‐diabetes mellitus‐induced testicular DNA damage and sperm quality degradation in Sprague-Dawley rats. J Food Biochem 45: e13938.
4. Aksu EH, Kandemir FM, Küçükler S (2021) The effects of hesperidin on colistin‐induced reproductive damage, autophagy, and apopto-sis by reducing oxidative stress. Andrologia 53: e13900.
5. Aksu EH, Kandemir FM, Yıldırım S, Küçükler S, Dörtbudak MB, Çağlayan C, Benzer F (2019) Palliative effect of curcumin on doxo-rubicin‐induced testicular damage in male rats. J Biochem Mol Toxicol 33: e22384.
6. Aksu E, Özkaraca M, Kandemir FM, Ömür AD, Eldutar E, Küçükler S, Çomaklı S (2016) Mitigation of paracetamol‐ -induced repro-ductive damage by chrysin in male rats via reducing oxidative stress. Andrologia 48: 1145-1154.
7. Bodnar RJ (2014) Anti-angiogenic drugs: involvement in cutaneous side effects and wound-healing complication. Adv Wound Caref 3: 635-646.
8. Carette C, Levy R, Eustache F, Baron G, Coupaye M, Msika S, Barrat C, Cohen R, Catheline JM, Brugnon F, Slim K, Barsamian C, Chevallier JM, Bretault M, Bouillot JL, Antignac JP, Rives-Lange C, Ravaud P, Czernichow S (2019) Changes in total sperm count after gastric bypass and sleeve gastrectomy: the BARIASPERM prospective study. Surg Obes Relat Dis 15: 1271-1279.
9. Carvalho RK, Rocha TL, Fernandes FH, Gonçalves BB, Souza MR, Araújo AA, Barbosa CC, Silva DM, Campos HM, Tomazett MV, Ghedini PC, Guimarães FS, Andersen ML, A Santos FC, Mazaro-Costa R (2022) Decreasing sperm quality in mice subjected to chronic cannabidiol exposure: New insights of cannabidiol-mediated male reproductive toxicity. Chem Biol Interact 351: 109743.
10. Çoban S, Ali A, Türkoğlu A R, Güzelsoy M, Üstündağ Y, Öztürk M, Demirci H (2018) Are there any ef fect of povidone iodine used in genitourinary surgery on thyroid hormones? New J Urol 13: 34-38.
11. Donkin I, Barrès R (2018) Sperm epigenetics and influence of environmental factors. Mol Metab 14: 1-11.
12. Farinon B, Molinari R, Costantini L, Merendino N (2020) The seed of industrial hemp (Cannabis sativa L.): Nutritional quality and po-tential functionality for human health and nutrition. Nutrients 12: 1935.
13. Fouda MA, Ghovanloo MR, Ruben PC (2020) Cannabidiol protects against high glucose‐induced oxidative stress and cytotoxicity in cardiac voltage‐gated sodium channels. Br J Pharmacol 177: 2932-2946.
14. Graczyk M, Lewandowska AA, Dzierżanowski T (2021) The therapeutic potential of cannabis in counteracting oxidative stress and in-flammation. Molecules 26: 4551.
15. Gur C, Akarsu, SA, Akaras N, Tuncer SC, Kandemir FM (2023) Carvacrol reduces abnormal and dead sperm counts by attenuating sodium arsenite‐induced oxidative stress, inflammation, apoptosis, and autophagy in the testicular tissues of rats. Environ Toxicol 38: 1265-1276.
16. Hassan E, Kahilo K, Kamal T, El-Neweshy M, Hassan M (2019) Protective effect of diallyl sulfide against lead-mediated oxidative damage, apoptosis and down-regulation of CYP19 gene expression in rat testes. Life Sci 226: 193-201.
17. Heymann WR (2000) Potassium iodide and the Wolff-Chaikoff effect: relevance for the dermatologist. J Am Acad Dermatol 42: 490-492.
18. Islamov R, Kustova T, Nersesyan A, Ilin A (2020) Subchronic toxicity of the new iodine complex in dogs and rats. Front Vet Sci 7: 184.
19. Johnsen SG (1970) Testicular biopsy score count – a method for registration of spermatogenesis in human testes: normal values and results in 335 hypogonadal males. Horm Res Paediatr 1: 2-25.
20. Kalkan Y, Kapakin KAT, Kara A, Atabay T, Karadeniz A, Simsek N, Karakuş E, Can I, Yıldırım S, Özkanlar S, Sengul E (2012) Protective effect of Panax ginseng against serum biochemical changes and apoptosis in kidney of rats treated with gentamicin sulphate. J Mol His 43: 603-613.
21. Karlidag R, Unal S, Sezer OH, Bay Karabulut A, Battaloğlu B, But A, Ozcan C (2006) The role of oxidative stress in postoperative de-lirium. Gen Hosp Psychiatry 28: 418-423.
22. Kiralan M, Gül V, Kara SM (2010) Fatty acid composition of hempseed oils from different locations in Turkey. Span J Agric Res 8: 385-390.
23. Koksal M, Oğuz E, Baba F, Ali Eren M, Ciftci H, Demir ME, Kurcer Z, Aral F, Ocak AR, Aksoy N, Ulas T (2012) Effects of melatonin on testis histology, oxidative stress and spermatogenesis after experimental testis ischemia-reperfusion in rats. Eur Rev Med Pharmacol Sci 16: 582-588.
24. Maccarrone M, Bab I, Bíró T, Cabral GA, Dey SK, Di Marzo V, Konje JC, Kunos G, Mechoulam R, Pacher P, Sharkey KA, Zimmer A (2015) Endocannabinoid signaling at the periphery: 50 years after THC. Trends Pharmacol Sci 36: 277-296.
25. Matkovics B (1988) Determination of enzyme activity in lipid peroxidation and glutathione pathways. Laborator Diagnos 15: 248-250.
26. Matthäus B, Brühl L (2008) Virgin hemp seed oil: An interesting niche product. Eur J Lipid Sci Technol 110: 655-661.
27. Menghini L, Ferrante C, Carradori S, D’Antonio M, Orlando G, Cairone F, Cesa S, Filippi A, Fraschetti C, Zengin G, AK G, Tacchini M, Iqbal K (2021) Chemical and bioinformatics analyses of the anti-leishmanial and anti-oxidant activities of hemp essential oil. Biomol-ecules 11: 272.
28. Noble D, Jablonka E, Joyner MJ, Müller GB, Omholt SW (2014) Evolution evolves: physiology returns to centre stage. J Physiol 592: 2237.
29. Peeters E, Spiessens C, Oyen R, De Wever L, Vanderschueren D, Miserez M (2014) Sperm motility after laparoscopic inguinal hernia repair with lightweight meshes: 3-year follow-up of a randomised clinical trial. Hernia 18: 361-367.
30. Placer ZA, Cushman LL, Johnson BC (1966) Estimation of product of lipid peroxidation (malonyl dialdehyde) in biochemical sys-tems. Anal Biochem 16: 359-364.
31. Sedlak J, Lindsay RH (1968) Estimation of total, protein-bound, and nonprotein sulfhydryl groups in tissue with Ellman’s reagent. Anal Biochem 25: 192-205.
32. Shafiey SI, Ahmed KA, Abo-Saif AA, Abo-Youssef AM, Mohamed WR (2023) Galantamine mitigates testicular injury and disturbed spermatogenesis in adjuvant arthritic rats via modulating apoptosis, inflammatory signals, and IL-6/JAK/STAT3/SOCS3 signaling. In-flammopharmacology: doi: 10.1007/s10787-023-01268-z
33. Sun Y, Oberley LW, Li Y (1988) A simple method for clinical assay of superoxide dismutase. Clinic Chem 34: 497-500.
34. Thomas GW, Rael LT, Bar-Or R, Shimonkevitz R, Mains CW, Slone DS, Craun ML Bar-Or D (2009) Mechanisms of delayed wound healing by commonly used antiseptics. J Trauma 66: 82-91.
35. Zheng Z, Qi J, Hu L, Quyang D, Wang H, Sun Q, Lin L, You Q, Tang B (2022) A cannabidiol-containing alginate based hydrogel as novel multifunctional wound dressing for promoting wound healing. Biomater Adv 134: 112560.
Go to article

Authors and Affiliations

G.D. Akarsu
1 2
E. Erbaş
3
S.A. Akarsu
4

  1. Division of Molecular Medicine, Laboratory for Advanced Genomics, Institut Ruđer Bošković, Zagreb, Croatia
  2. Department of Pharmacy Services, Vocational School of Health Servıces, Yozgat Bozok Unıversıty, Atatürk Road 7.Km, Bilal Şahin West Campus 66100, Yozgat, Turkey
  3. Department of Histology and Embryology, Faculty of Veterinary Medicine, Ataturk University, Yakutiye 25030, Erzurum, Turkey
  4. Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Yakutiye 25030, Ataturk University, Erzurum, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Cystic endometrial hyperplasia-pyometra complex (CEH-P) is a common disease in sexually mature bitches. Disease progression leads to oxidative stress, resulting in the depletion of uterine antioxidants and lipid peroxidation of associated cells, which further aggravates the condition. The concentration of antioxidant enzymes, the level of lipid peroxidation within the uterine tissue, and its reflection in the serum and urine need to be elucidated. The aim of this study was to analyze the concentration of antioxidants such as superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), glutathione peroxidase (GPx), and the lipid peroxidation marker malonaldehyde (MDA) in three types of samples, i.e., serum, urine, and uterine tissue. For this purpose, 58 pyometra-affected and 44 healthy bitches were included in the present study. All animals underwent ovariohysterectomy (OVH). Our data indicated highly significant difference (p<0.01) in the antioxidant concentrations of uterine, serum and urine samples. Furthermore, there was a highly significant (p<0.01) difference in the serum levels of ferric reducing antioxidant power (FRAP) and free radical scavenging activity (FRSA) indicated poor capacity to overcome oxidative stress in the CEH-Pyometra condition. We showed that CEH-P induces oxidative stress, which further depletes the antioxidant enzyme reserves in the uterus. Thus, the weak antioxidant defence predisposes to uterine damage and disease progression. The simultaneous depletion of antioxidants and an increase in lipid peroxidation in the serum and urine may also act as early indicators of uterine pathology.
Go to article

Bibliography

1. Agarwal A, Gupta S, Sharma RK (2005) Role of oxidative stress in female reproduction. Reprod Biol Endocrinol 3: 28.
2. Al-Gubory KH, Fowler PA, Garrel C (2010) The roles of cellular reactive oxygen species, oxidative stress and antioxidants in pregnancy outcomes. Int J Biochem Cell Biol 42: 1634-1650.
3. Biswas SK (2016) Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxid Med Cell Longev 2016: 5698931.
4. Clemo FA (1998) Urinary enzyme evaluation of nephrotoxicity in the dog. Toxicol Pathol 26: 29-32.
5. Dringen R (2000) Metabolism and functions of glutathione in brain. Prog Neurobiol 62: 649-671.
6. Ďuračková Z (2010) Some current insights into oxidative stress. Physiol Res 59: 459-469.
7. Egenvall A, Hagman R, Bonnett BN, Hedhammar A, Olson P, Lagerstedt AS (2001) Breed risk of pyometra in insured dogs in Sweden. J Vet Intern Med 15: 530-538.
8. El-Bahr SM, El-Deeb WM (2016) Acute-phase proteins, oxidative stress biomarkers, proinflammatory cytokines, and cardiac troponin in Arabian mares affected with pyometra. Theriogenology 86: 1132-1136
9. Emanuelli MP, Martins DB, Wolkmer P, Antoniazzi AQ, Emanuelli T, de Vargas AC, dos Anjos Lopes ST (2012) Complete blood count, total plasma protein, neutrophil oxidative metabolism, and lipid peroxidation in female dogs with pyometra associated with Esche-richia coli. Comp Clin Pathol 21: 309-313.
10. Ercan N, Fidancı UR (2012) Urine 8-Hydroxy-2’-Deoxyguanosine (8-OHdG) Levels of Dogs in Pyoderma. Ankara Univ Vet Fak Derg 59: 163-168.
11. Ghezzi P (2011) Role of glutathione in immunity and inflammation in the lung. Int J Gen Med 4: 105-113.
12. Gogoi J, Leela V, Suganya G, Shafiuzama M, Vairamuthu S, Rajamanickam K, Pandiyan AS (2018) Effect of ovariohysterectomy on oxidative stress markers in pyometra affected bitches. Int J Chem Stud 6: 994-998.
13. Halliwell B (2006) Reactive species and antioxidants. Redox biology is a fundamental theme of aerobic life. Plant Physiol 141: 312-322.
14. Halliwell B, Whiteman M (2004) Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol 142: 231-255.
15. Hussain T, Tan B, Yin Y, Blachier F, Tossou MC, Rahu N (2016) Oxidative stress and inflammation: what polyphenols can do for us? Oxid Med Cell Longev 7432797.
16. Jitpean S, Hagman R, Ström Holst B, Höglund OV, Pettersson A, Egenvall A (2012) Breed variations in the incidence of pyometra and mammary tumours in Swedish dogs. Reprod Domest Anim 47(Suppl): 347-350.
17. Jitpean S, Ström-Holst B, Emanuelson U, Höglund OV, Pettersson A, Alneryd-Bull C, Hagman R (2014) Outcome of pyometra in fe-male dogs and predictors of peritonitis and prolonged postoperative hospitalization in surgically treated cases. BMC Vet Res 10: 6.
18. Kumar A, Saxena A (2018) Canine pyometra: Current perspectives on causes and management – a review. Ind J Vet Sci Biotechnol 14: 52-56.
19. Nielsen F, Mikkelsen BB, Nielsen JB, Andersen HR, Grandjean P (1997) Plasma malondialdehyde as biomarker for oxidative stress: reference interval and effects of life-style factors. Clin Chem 43: 1209-1214.
20. Prasad VD, Kumar PR, Sreenu M (2017) Pyometra in bitches: a review of literature. Res Rev J Vet Sci Technol 6: 12-20.
21. Rivers BJ, Walter PA, O’Brien TD, King VL, Polzin DJ (1996) Evaluation of urine gamma-glutamyl transpeptidase-to-creatinine ratio as a diagnostic tool in an experimental model of aminoglycoside-induced acute renal failure in the dog. J Am Anim Hosp Assoc 32: 323–336.
22. Santos C, dos Anjos Pires M, Santos D, Payan-Carreira R (2016) Distribution of superoxide dismutase 1 and glutathione peroxidase 1 in the cyclic canine endometrium. Theriogenology 86: 738-748.
23. Sasidharan JK, Patra MK, Khan JA, Singh AK, Karikalan M, De UK, Saxena AC, Dubal ZB, Singh SK, Kumar H, Krishnaswamy N (2023) Differential expression of inflammatory cytokines, prostaglandin synthases and secretory leukocyte protease inhibitor in the en-dometrium and circulation in different graded CEH-pyometra in bitch. Theriogenology 197: 139-149.
24. Smith FO (2006) Canine pyometra. Theriogenology 66: 610-612.
25. Szczubial M, Dabrowski R, Bochniarz M, Brodzki P (2019) Uterine non-enzymatic antioxidant defence mechanisms (glutathione, vita-min C, copper and zinc) in diagnosis of canine pyometra. Pol J Vet Sci 22: 549-555.
26. Szczubiał MA, Dąbrowski RO (2009) Activity of antioxidant enzymes in uterine tissues of bitches with pyometra. Bull Vet Inst Puławy 53: 673-676.
27. Todorova I, Simeonova G, Kyuchukova D, Dinev D, Gadjeva V (2005) Reference values of oxidative stress parameters (MDA, SOD, CAT) in dogs and cats. Comp Clin Pathol 13: 190-194.
28. Toydemir Karabulut TS (2018) SOD, CAT, TBARS, and TNF-α concentrations in uterine tissues of bitches with pyometra and dioestrus bitches. Acta Vet Eurasia 44: 59-62.
29. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J (2007) Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 39: 44-84.
30. Woźna-Wysocka M, Rybska M, Błaszak B, Jaśkowski BM, Kulus M, Jaśkowski JM (2021) Morphological changes in bitches endome-trium affected by cystic endometrial hyperplasia-pyometra complex – the value of histopathological examination. BMC Vet Res 17: 174.

Go to article

Authors and Affiliations

A. Kumar
1 4
J.K. Prasad
2
S. Verma
3
A. Gattani
5
G.D. Singh
6
V.K. Singh
6

  1. Department of Veterinary Gynaecology and Obstetrics, Bihar Veterinary College, Bihar Animal Sciences University, Patna, Bihar 800014, India
  2. Dean, Bihar Veterinary College, Bihar Animal Sciences University, Patna, Bihar 800014, India
  3. Department of Veterinary Medicine, College of Veterinary Science and Animal Husbandry, Deen-dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-anusandhan Sansthan (DUVASU), Mathura, U.P. 281001 India
  4. Department of Veterinary Biochemistry, Bihar Veterinary College, Bihar Animal Sciences University, Patna, Bihar 800014, India
  5. Department of Veterinary Physiology and Biochemistry, College of Veterinary Science and Animal Husbandry, Nanaji Deshmukh Veterinary Science University (NDVSU), Jabalpur, M.P., 483220 India
  6. Veterinary Clinical Complex, Bihar Veterinary College, Bihar Animal Sciences University, Patna, Bihar 800014, India
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to determine the effects of an ohmic heating (OH) process with different electric field intensities on Listeria monocytogenes inactivation in protein-enriched cow milk. Protein powder was added at rates of 2.5%, 5% and 7.5% in 1.5% fat content milk, and L. monocytogenes (ATCC 13932) strain was then inoculated into the samples. The OH process was carried out in a laboratory-type pilot unit created using stainless steel electrodes, a K-type thermocouple, a datalogger and power supply providing AC current at 0-250 V, 10 A. The inoculated milk samples were heated to 63°C by applying an electric field intensity of 10V/cm and 20V/cm. L. monocytogenes counts, pH, color measurement and hydroxymethylfurfurol levels were then determined. OH applied with an electric field intensity of 10 V/cm caused an average decrease of 5 logs in L. monocytogenes level in the samples containing 2.5% protein and decreased below the detection limit (<1 log) at the 9th minute (p<0.05). Similarly, application of an electric field intensity of 20 V/cm in milk containing 2.5% and 5% protein caused the L.monocytogenes level to decrease below the detection limit (<1 log) at 2 minutes 30 seconds (p<0.05). No change was observed in the L* (brightness) values of the samples but it was determined that there was a slight increase in pH, a* (redness) and b* (yellowness) values compared to the control group. It was observed that the inactivation of L. monocytogenes by OH depends on the duration of the OH process, protein concentration in the milk and the applied voltage gradient.
Go to article

Bibliography

1. Ahmad T, Butt MZ, Aadil RM, Inam‐ur‐Raheem M, Abdullah Bekhit AE, Guimarães JT, Balthazar CF, Rocha RS, Esmerino EA, Freitas MQ, Silva MC, Sameen A, Cruz AG (2019) Impact of nonthermal processing on different milk enzymes. Int J Dairy Tech 72: 481-495.
2. Altuntas S, Korukluoğlu M (2018) Listeria monocytogenes in food facilities and new approaches for struggle. J Food 43:101-113.
3. Assiry AM, Gaily MH, Alsamee M, Sarifudin A (2010) Electrical conductivity of seawater during ohmic heating. Desalination 260: 9-17.
4. Awuah GB, Ramaswamy HS, Economides A (2007) Thermal processing and quality: Principles and overview. Chem Eng Process 46: 584-602.
5. Balpetek D, Gürbüz U (2015) Application of ohmic heating system in meat thawing. Procedia Soc 195: 2822-2828.
6. Belfort M, Cherkerzian S, Bell K, Soldateli B, Cordova Ramos E, Palmer C, Steele T, Pepin H, Ellard D, Drouin K, Inder, T (2020) Macronutrient intake from human milk, infant growth, and body composition at term equivalent age: a longitudinal study of hospitalized very preterm infants. Nutrients 12: 2249.
7. Cevik M, Icier F (2020) Characterization of viscoelastic properties of minced beef meat thawed by ohmic and conventional meth-ods. Food Sci Technol Int 26: 277-290.
8. Cho WI, Kim EJ, Hwang HJ, Cha YH, Cheon HS, Choi JB, Chung MS (2017) Continuous ohmic heating system for the pasteurization of fermented red pepper paste. Innov Food Sci Emerg Technol 42: 190-196.
9. Coimbra LO, Vidal VA, Silva R, Rocha RS, Guimarães JT, Balthazar CF, Cruz AG (2020) Are ohmic heating-treated whey dairy beverages an innovation? Insights of the Q methodology. LWT 134: 110052.
10. Costa NR, Cappato LP, Ferreira MV, Pires RP, Moraes J, Esmerino EA, Silva R, Neto RP, Tavares MI, Freitas MQ, Júnior RN, Rodrigues FN, Bisaggio RC, Cavalcanti RN, Raices RSL, Silva MC, Cruz AG (2018) Ohmic Heating: A potential technology for sweet whey processing. Food Res Int 106: 771-779.
11. Inmanee P, Kamonpatana P, Pirak T (2019) Ohmic heating effects on Listeria monocytogenes inactivation, and chemical, physical, and sensory characteristic alterations for vacuum packaged sausage during postpasteurization. LWT 108: 183-189.
12. Jaeger H, Janositz A, Knorr D (2010) The Maillard reaction and its control during food processing. The potential of emerging technologies. Pathol Biol 58: 207-213.
13. Kallipolitis BH, Gahan CG, Piveteau P (2020) Factors contributing to Listeria monocytogenes transmission and impact on food safety. Curr Opin Food Sci 36: 9-17.
14. Kim SS, Kang DH (2015) Effect of milk fat content on the performance of ohmic heating for inactivation of Escherichia coli O157: H7, Salmonella enterica serovar Typhimurium and Listeria monocytogenes. J Appl Microbiol 119: 475-486.
15. Knirsch MC, Dos Santos CA, de Oliveira Soares AA, Penna TC (2010) Ohmic heating–a review. Trends Food Sci Technol 21: 436-441.
16. Lee JY, Kim SS, Kang DH (2015) Effect of pH for inactivation of Escherichia coli O157: H7, Salmonella Typhimurium and Listeria monocytogenes in orange juice by ohmic heating. LWT 62: 83-88.
17. Morales FJ, Jiménez-Pérez S (1999) HMF formation during heat treatment of milk type products as related to milkfat content. J Food Sci 64: 855–859.
18. Morales FJ, Jiménez-Pérez S (2001) Hydroxymethylfurfural determination in infant milk-based formulas by micellar electrokinetic capillary chromatography. Food Chem 72: 525-531.
19. Özkale S, Kahraman HA (2023) Determination of the effect of milk fat on the inactivation of Listeria monocytogenes by ohmic heat-ing. Ankara Univ Vet Fak Derg 70: 277-283.
20. Park IK, Kang DH (2013) Effect of electropermeabilization by ohmic heating for inactivation of Escherichia coli O157:H7, Salmonella enterica serovar Typhimurium, and Listeria monocytogenes in buffered peptone water and apple juice. Appl Environ Microbiol 79: 7122-7129.
21. Parmar P, Singh AK, Meena GS, Borad S, Raju PN (2018) Application of ohmic heating for concentration of milk. J Food Sci Tech-nol 55: 4956-4963.
22. Pereira MO, Guimarães JT, Ramos GL, do Prado-Silva L, Nascimento JS, Sant’Ana AS, Cruz AG (2020) Inactivation kinetics of Lis-teria monocytogenes in whey dairy beverage processed with ohmic heating. LWT 127: 109420.
23. Pires RP, Cappato LP, Guimarães JT, Rocha RS, Silva R, Balthazar CF, Freitas MQ, Silva PHF, Neto RPC, Tavares MIB, Granato D, Raices RSL, Silva MC, Cruz AG (2020) Ohmic heating for infant formula processing: Evaluating the effect of different voltage gra-dient. J Food Eng 280: 109989.
24. Sagong HG, Lee SY, Chang PS, Heu S, Ryu S, Choi YJ, Kang DH (2011) Combined effect of ultrasound and organic acids to reduce Escherichia coli O157: H7, Salmonella Typhimurium, and Listeria monocytogenes on organic fresh lettuce. Int J Food Microbiol 145: 287-292.
25. Sakr M, Liu S (2014) A comprehensive review on applications of ohmic heating (OH). Renewable Sustainable Energy Rev 39: 262-269.
26. Salas AA, Jerome M, Finck A, Razzaghy J, Chandler-Laney P, Carlo WA (2022) Body composition of extremely preterm infants fed protein-enriched, fortified milk: a randomized trial. Pediatr Res 91: 1231-1237.
27. Shi C, Jia Z, Chen Y, Yang M, Liu X, Sun Y, Zheng, Z, Zhang X, Song K, Cui L, Baloch AB, Xia X (2015) Inactivation of Cronobacter sakazakii in reconstituted infant formula by combination of thymoquinone and mild heat. J Appl Microbiol 119: 1700-1706.
28. Somavat R, Mohamed HM, Chung YK, Yousef AE, Sastry SK (2012) Accelerated inactivation of Geobacillus stearothermophilus spores by ohmic heating. J Food Eng 108: 69-76.
29. Somavat R, Mohamed HM, Sastry SK (2013). Inactivation kinetics of Bacillus coagulans spores under ohmic and conventional heating. LWT 54: 194–198.
30. Tian X, Shao L, Yu Q, Liu Y, Li X, Dai R (2019) Evaluation of structural changes and intracellular substance leakage of Escherichia coli O157: H7 induced by ohmic heating. J Appl Microbiol 127: 1430-1441.
31. Urgu M, Saatli TE, Türk A, Koca N (2017) Determination of hydroxymethylfurfural content of heat-treated milk (pasteurized, UHT and lactose-hydrolised UHT milk). Academic Food 15: 249-255.
32. Wang C, Llave Y, Sakai N, Fukuoka M (2021) Analysis of thermal processing of liquid eggs using a high frequency ohmic heating: Experimental and computer simulation approaches. Innov Food Sci Emerg Technol 73: 102792


Go to article

Authors and Affiliations

R.Y. Ayyıldız
1
H.A. Kahraman
2

  1. Department of Food Hygiene and Technology, Institute of Health Sciences, University of Burdur Mehmet Akif Ersoy, 15030, Burdur, Turkey
  2. Department of Food Hygiene and Technology, Faculty of Veterinary Medicine, University of Burdur Mehmet Akif Ersoy, 15030, Burdur, Turkey
Download PDF Download RIS Download Bibtex

Abstract

This study aimed to develop an equine-derived hyperimmune serum against SARS-CoV-2 and evaluate its efficacy as a potential immunotherapy tool for the treatment of known and potential variants of COVID-19 in preclinical trials.
The novelty of this study is the whole virus and ALUM gel adjuvant formula. The horses were immunized using a whole inactivated SARS-CoV-2 antigen, and the final purified hyperimmune serum showed high plaque reduction neutralization (PRNT 50) neutralizing titers. The efficacy of the hyperimmune serum was evaluated histopathologically and biochemically in the lungs, hearts, and serum of K18 hACE2 transgenic mice (n=45), which is an accepted model organism for SARS-CoV-2 studies and was challenged with live SARS-CoV-2.
Serum treatment improved the general condition, resulting in lower levels of proinflammatory cytokines in the blood plasma, as well as reduced viral RNA titers in the lungs and hearts. Additionally, it reduced oxidative stress significantly and lessened the severity of interstitial pneumonia in the lungs when compared to infected positive controls.
The study concluded that equine-derived anti-SARS-CoV-2 antibodies could be used for COVID-19 prevention and treatment, especially in the early stages of the disease and in combination with antiviral drugs and vaccines. This treatment will benefit special patient populations such as immunocompromised individuals, as specific antibodies against SARS-CoV-2 can neutralize the virus before it enters host cells. The rapid and cost-effective production of the serum allows for its availability during the acute phase of the disease, making it a critical intervention in preventing the spread of the disease and saving lives in new variants where a vaccine is not yet developed.
Go to article

Bibliography

1. Botosso VF, Jorge SAC, Astray RM, Guimaraes AMS, Mathor MB, Carneiro PS, Durigon EL, Covas D, Oliveira DBL, Oliveira RN, Maria DA, Eto SF, Gallina NMF, Pidde G, Squaiella-Baptistão CS, Silva DT, Villas-Boas IM, Fernandes DC, Auada AVV, Banari AC, Filho AFS, Bianconi C, Utescher CLA, Oliveira DCA, Mariano DOC, Barbosa FF, Rondon G, Kapronezai J, Silva J G, Goldfeder MB, Comone P, Junior REC, Pereira TTS, Wen FH, Tambourgi DV, Chudzinski-Tavassi AM (2022) Anti-SARS-CoV-2 equine F (Ab′)2 immunoglobulin as a possible therapy for COVID-19. Sci Rep 12: 3890.
2. De Vito A, Colpani A, Saderi L, Puci M, Zauli B, Fiore V, Fois M, Meloni MC, Bitti A, Di Castri C, Maida I, Babudieri S, Sotgiu G, Madeddu G. (2022) Impact of early SARS-CoV-2 antiviral therapy on disease progression. Viruses 15: 71.
3. Dong W, Mead H, Tian L, Park JG, Garcia JI, Jaramillo S, Barr T, Kollath DS, Coyne VK, Stone NE, Jones A, Zhang J, Li A, Wang LS, Milanes-Yearsley M, Torrelles JB, Martinez-Sobrido L, Keim PS, Barker BM, Caligiuri MA, Yu J (2022) The K18-Human ACE2 transgenic mouse model recapitulates non-severe and severe COVID-19 in response to an infectious dose of the SARS-CoV-2 virus. J Virol: e0096421.
4. Golden JW, Cline CR, Zeng X, Garrison AR, Carey BD, Mucker EM, White LE, Shamblin JD, Brocato RL, Liu J, Babka AM, Rauch HB, Smith JM, Hollidge BS, Fitzpatrick C, Badger CV, Hooper JW (2020) Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight 5: e142032.
5. Jha A, Barker D, Lew J, Manoharan V, Kessel JV, Haupt R, Toth D, Frieman M, Falzarano D, Kodihalli S (2022) Efficacy of COVID-HIGIV in animal models of SARS-CoV-2 infection. Sci Rep 12: 16956.
6. León G, Herrera M, Vargas M, Arguedas M, Sánchez A, Segura A, Gómez A, Solano G, Aguilar E C, Risner K, Narayanan A, Bailey C, Villalta M, Hernández A, Sánchez A, Cordero D, Solano D, Durán G, Segura E, Cerdas M, Umaña D, Moscoso E, Estrada R, Gu-tiérrez J, Méndez M, Castillo AC, Sánchez L, Sánchez R, Gutiérrez JM, Díaz C, Alape A (2021) Development and characterization of two equine formulations towards SARS-CoV-2 proteins for the potential treatment of COVID-19. Sci Rep 11: 9825.
7. Li E, Han Q, Bi J, Wei S, Wang S, Zhang Y, Liu J, Feng N, Wang T, Wu J, Yang S, Zhao Y, Liu B, Yan F, Xia X (2023) Therapeutic equine hyperimmune antibodies with high and broad-spectrum neutralizing activity protect rodents against SARS-CoV-2 infection. Front Immunol 14: 1066730.
8. Luis Eduardo Cunha, Stolet AA, Strauch MA, Pereira VAR, Dumard CH, Gomes AMO, Souza PNC, Fonseca JG, Pontes FE, Meirelles LGR, Albuquerque JWM, Sacramento CQ, Rodrigues NF, Lima TM, Alvim RGF, Marsili FF, Caldeira MM, Higa LM, Monteiro FL, Zingali RB, Oliveira GAP, Souza TML, Tanuri A, Oliveira AC, Guedes H L M, Castilho L R , Silva J L (2020) Potent neutralizing equine antibodies raised against recombinant SARS-CoV-2 spike protein for COVID-19 passive immunization therapy. Cold Spring Harbor Laboratory, bioRxiv 17: 254375
9. Maccio U, Zinkernagel AS, Shambat SM, Zeng X, Cathomas G, Ruschitzka F, Schuepbach RA, Moch H, Varga Z. SARS-CoV-2 (2021) Leads to a small vessel endotheliitis in the heart. EBioMedicine 63: 103182.
10. Moreira-Soto A, Arguedas M, Brenes H, Buján W, Corrales-Aguilar E, Díaz C, Echeverri A, Flores-Díaz M, Gómez A, Hernández A, Herrera M, León G, Macaya R, Kühne A, Molina-Mora JA, Mora J, Sanabria A, Sánchez A, Sánchez L, Segura Á, Segura E, Solano D, Soto C, Stynoski JL, Vargas M, Villalta M, Reusken CBEM, Drosten C, Gutiérrez JM, Alape-Girón A, Drexler JF (2021) High efficacy of therapeutic equine hyperimmune antibodies against SARS-CoV-2 variants of concern. Frontiers in Medicine. 8: 735853.
11. Onen EA, Sonmez K, Yildirim F, Demirci EK, Gurel A (2022) Development, analysis, and preclinical evaluation of inactivated vaccine candidate for prevention of Covid-19 disease. All Life 15: 771-793.
12. Pan X, Zhou P, Fan T, Wu Y, Zhang J, Shi X, Shang W, Fang L, Jiang X, Shi J, Sun Y, Zhao S, Gong R, Chen Z, Xiao G (2020) Im-munoglobulin fragment F(ab’)2 against RBD potently neutralizes SARS-CoV-2 in vitro. Antiviral Res 182: 104868.
13. Schoell A, Heyde B, Weir D, Po-Chang C, Yiding H, Tung D (2009) Euthanasia method for mice in rapid time-course pulmonary. Pharmacokinetic Studies 48: 506.
14. Suvarna KS, Layton C, Bancroft JD (2018) Bancroft’s theory and practice of histological techniques E-Book. Elsevier health sciences, Philadelphia, pp 286-291.
15. Winkler ES, Bailey AL, Kafai NM, Nair S, McCune BT, Yu J, Fox JM, Chen RE, Earnest JT, Keeler SP, Ritter JH, Kang L, Dort S, Robichaud A, Head R, Holtzman MJ, Diamond MS (2020) SARS-CoV-2 infection of human ACE2-transgenic mice causes severe lung inflammation and impaired function. Nat Immunol 21: 1327-1335.
16. Xu J, Xu X, Jiang L, Dua K, Hansbro PM, Liu G (2020) SARS-CoV-2 induces transcriptional signatures in human lung epithelial cells that promote lung fibrosis. Respir Res 21: 182.
17. Yu P, Deng W, Bao L, Qu Y, Xu Y, Zhao W, Han Y, Qin C (2022) Comparative pathology of the nasal epithelium in K18-hACE2 Tg mice, hACE2 Tg mice, and hamsters infected with SARS-CoV-2. Vet Pathol 59: 602-612.
18. Zhang Q, Wang Y, Qi C, Shen L, Li J (2020) Clinical trial analysis of 2019-nCoV therapy registered in China. J Med Virol 92: 540-545.

Go to article

Authors and Affiliations

E.A. Onen
1
E.K. Demirci
2

  1. Kocak Pharmaceutical Company, Biotechnology and Vaccine R&D, Tekirdag, Turkey
  2. Histology and Embryology Department, Istanbul Faculty of Medicine,Istanbul University, Istanbul, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The trapezius muscle (TRAP) belongs to the scapulothoracic group of muscles, which play a crucial role in the integrity and strength of the upper limb, trunk, head, and neck movements and, thus, in maintaining balance. Combined retrograde tracing (using fluorescent tracer Fast Blue, FB) and double-labelling immunohistochemistry were applied to investigate the chemical coding of motoneurons projecting to the porcine TRAP. FB-positive (FB+) motoneurons supplying the cervical (c-TRAP) and thoracic part (th-TRAP) of the right (injected with the tracer) TRAP were located within the IX-th Rexed lamina in the ipsilateral ventral horn of the grey matter of the spinal medulla. Immunohistochemistry revealed that nearly all the neurons were cholinergic in nature [choline acetyltransferase (CHAT)- or vesicular acetylcholine transporter (VACHT)-positive]. Many retrogradelly labelled neurons displayed also immunoreactivity to calcitonin gene-related peptide (CGRP; approximately 68% of FB+ neurons). The smaller number of nerve cells (5%, 3%, 2% or 1%, respectively) stained for nitric oxide synthase (n-NOS), vasoactive intestinal polypeptide (VIP), neuropeptide Y (NPY) and substance P (SP). The retrogradely labelled neurons were closely apposed by nerve fibres expressing immunoreactivity to CHAT, VACHT, CGRP, SP, DβH, VIP, n-NOS, NPY, GAL, Leu-Enk and Met-Enk. Taking into account the clinical relevance of TRAP, the present results may be useful in designing further research aimed at the management of various dysfunctions of the muscle.
Go to article

Bibliography

1. Akamatsu FE, Ayres BR, Saleh SO, Hojaij F, Andrade M, Hsing WT, Jacomo AL (2015) Trigger points: an anatomical substratum. Biomed Res Int 2015: 623287.
2. Arita H, Sakamoto M, Hirokawa Y, Okado N (1993) Serotonin innervation patterns differ among the various medullary motoneuronal groups involved in upper airway control. Exp Brain Res 95: 100-110.
3. Arlotta M, Lovasco G, McLean L (2011) Selective recruitment of the lower fibers of the trapezius muscle. J Electromyogr Kinesiol 21: 403-410.
4. Arluison M, Conrath-Verrier M, Tauc M, Mailly P, De la Manche IS, Cesselin F, Bourgoin S, Hamon M (1983a) Different localizations of Met-enkephalin-like immunoreactivity in rat forebrain and spinal cord using hydrogen peroxide and Triton X-100. Light microscopic study. Brain Res Bull 11: 555-571.
5. Arluison M, Conrath-Verrier M, Tauc M, Mailly P, De la Manche IS, Dietl M, Cesselin F, Bourgoin S, Hamon M (1983b) Met-enkephalin-like immunoreactivity in rat forebrain and spinal cord using hydrogen peroxide and Triton X-100. Ultrastructural study. Brain Res Bull 11: 573-586.
6. Atoji Y, Kusindarta DL, Hamazaki N, Kaneko A (2005) Innervation of the rat trachea by bilateral cholinergic projections from the nucle-us ambiguus and direct motor fibers from the cervical spinal cord: a retrograde and anterograde tracer study. Brain Res 1031: 90-100.
7. Barber RP, Phelps PE, Houser CR, Crawford GD, Salvaterra PM, Vaughn JE (1984) The morphology and distribution of neurons con-taining choline acetyltransferase in the adult rat spinal cord: an immunocytochemical study. J Comp Neurol 229: 329-346.
8. Boehm I, Alhindi A, Leite AS, Logie C, Gibbs A, Murray O, Farrukh R, Pirie R, Proudfoot C, Clutton R, Wishart TM, Jones RA, Gillingwater TH (2020) Comparative anatomy of the mammalian neuromuscular junction. J Anat 237: 827-836.
9. Chan PK, Hems TE (2006) Clinical signs of accessory nerve palsy. J Trauma, 60: 1142-1144.
10. Chu J (1995) Dry needling (intramuscular stimulation) in myofascial pain related to lumbosacral radiculopathy. Eur J Phys Rehabil Med 5: 106-121.
11. Chiocchetti R, Grandis A, Bombardi C, Clavenzani P, Spadari A, Gentile A, Bortolami R (2005) Localization, morphology, and im-munohistochemistry of spinal cord and dorsal root ganglion neurons that innervate the gastrocnemius and superficial digital flexor mus-cles in cattle. Am J Vet Res 66: 710-720.
12. Csillik B, Tajti L, Kovacs T, Kukla E, Rakic P, Knyihar-Csillik E (1993) Distribution of calcitonin gene-related peptide in vertebrate neu-romuscular junctions: relationship to the acetylcholine receptor. J Histochem Cytochem 41: 1547-1555.
13. De Meulemeester K, Calders P, De Pauw R, Grymonpon I, Govaerts A, Cagnie B (2017) Morphological and physiological differences in the upper trapezius muscle in patients with work-related trapezius myalgia compared to healthy controls: A systematic review. Muscu-loskelet Sci Pract 29: 43-51.
14. Dudek A, Sienkiewicz W (2009) Immunohistochemical characterisation of the motoneurones supplaying trapezius muscle (musculus trapezius) in the rat. Abstract Book of the 29-th Congress of Polish Anatomical Society, Bydgoszcz, September 3-5, p 23.
15. Dudek A, Sienkiewicz W, Kaleczyc J (2015) Contribution of the dorsal branch of the accessory nerve to the innervation of the trapezius muscle in the pig - a retrograde tracing study. Acta Zoologica 96: 519-529.
16. Dudek A, Sienkiewicz W, Marczak M, Kaleczyc J (2011) Immunohistochemical properties of motoneurons supplying the trapezius mus-cle in the rat. Pol J Vet Sci 14: 199-205.
17. Eberhorn AC, Ardeleanu P, Buttner-Ennever JA, Horn AK (2005) Histochemical differences between motoneurons supplying multiply and singly innervated extraocular muscle fibers. J Comp Neurol 491: 352-366.
18. Eberhorn AC, Buttner-Ennever JA, Horn AK (2006) Identification of motoneurons supplying multiply- or singly-innervated extraocular muscle fibers in the rat. Neuroscience 137: 891-903.
19. Fernandez HL, Chen M, Nadelhaft I, Durr JA (2003) Calcitonin gene-related peptides: their binding sites and receptor accessory proteins in adult mammalian skeletal muscles. Neuroscience 119: 335-345.
20. Gardan D, Gondret F, Louveau I (2006) Lipid metabolism and secretory function of porcine intramuscular adipocytes compared with subcutaneous and perirenal adipocytes. Am J Physiol Endocrinol Metab 291: 372-380.
21. Grozdanovic Z (2001) NO message from muscle. Microsc Res Tech 55: 148-153.
22. Grozdanovic Z, Baumgarten HG (1999) Nitric oxide synthase in skeletal muscle fibers: a signaling component of the dystro-phin-glycoprotein complex. Histol Histopathol 14: 243-256.
23. Handel SE, Stickland NC (1986) “Giant” muscle fibres in skeletal muscle of normal pigs. J Comp Pathol 96: 447-457.
24. Hietanen M, Pelto-Huikko M, Rechardt L (1990) Immunocytochemical study of the relations of acetylcholinesterase, enkephalin-, sub-stance P-, choline acetyltransferase- and calcitonin gene-related peptide-immunoreactive structures in the ventral horn of rat spinal cord. Histochemistry 93: 473-477.
25. Hisa Y, Tadaki N, Koike S, Bamba H, Uno T (1998) Calcitonin gene-related peptide-like immunoreactive motoneurons innervating the canine intrinsic laryngeal muscles. Ann Otol Rhinol Laryngol 107: 1029-1032.
26. Hisa Y, Tadaki N, Uno T, Okamura H, Taguchi J, Ibata Y (1994) Calcitonin gene-related peptide-like immunoreactive motoneurons in-nervating the canine inferior pharyngeal constrictor muscle. Acta Otolaryngol 114: 560-564.
27. Holtman JR Jr, Norman WP, Skirboll L, Dretchen KL, Cuello C, Visser TJ, Hokfelt T, Gillis RA (1984) Evidence for 5-hydroxytryptamine, substance P, and thyrotropin-releasing hormone in neurons innervating the phrenic motor nucleus. J Neurosci 4: 1064-1071.
28. Homonko DA, Theriault E (2000) Downhill running preferentially increases CGRP in fast glycolytic muscle fibers. J Appl Physiol (1985 ) 89: 1928-1936.
29. Hou N, Du X, Wu S (2022) Advances in pig models of human diseases. Animal Model Exp Med 5: 141-152.
30. Houser CR, Crawford GD, Barber RP, Salvaterra PM, Vaughn JE (1983) Organization and morphological characteristics of cholinergic neurons: an immunocytochemical study with a monoclonal antibody to choline acetyltransferase. Brain Res 266: 97-119.
31. Iaizzo PA, Lehmann-Horn F (1989) The in vitro determination of susceptibility to malignant hyperthermia. Muscle Nerve 12: 184-190.
32. Ichikawa T, Shimizu T (1998) Organization of choline acetyltransferase-containing structures in the cranial nerve motor nuclei and spinal cord of the monkey. Brain Res 779: 96-103.
33. Jones SW, Parr T, Sensky PL, Scothern GP, Bardsley RG, Buttery PJ (1999) Fibre type-specific expression of p94, a skeletal mus-cle-specific calpain. J Muscle Res Cell Motil 20: 417-424.
34. Lefaucheur L (1990) Changes in muscle fiber populations and muscle enzyme activities in the primiparous lactating sow. Reprod Nutr Dev 30: 523-531.
35. Lu IC, Wang HM, Kuo YW, Shieh CF, Chiang FY, Wu CW, Tsai CJ. (2010) Electromyographic study of differential sensitivity to suc-cinylcholine of the diaphragm, laryngeal and somatic muscles: a swine model. Kaohsiung J Med Sci 26: 640-646.
36. Lunney JK, Van Goor A, Walker KE, Hailstock T, Franklin J, Dai C (2021) Importance of the pig as a human biomedical model. Sci Transl Med 13 (621): eabd5758.
37. Maley B, Elde R (1981) Localization of substance P-like immunoreactivity in cell bodies of the feline dorsal vagal nucleus. Neurosci Lett 27: 187-191.
38. Merighi A, Kar S, Gibson SJ, Ghidella S, Gobetto A, Peirone SM, Polak JM (1990) The immunocytochemical distribution of seven peptides in the spinal cord and dorsal root ganglia of horse and pig. Anat Embryol (Berl) 181: 271-280.
39. Neumann DA (2017) Kinesiology of the Musculoskeletal System: Foundations for Rehabilitation. 3rd ed., Elsevier, St Louis.
40. Piehl F, Arvidsson U, Hokfelt T, Cullheim S (1993) Calcitonin gene-related peptide-like immunoreactivity in motoneuron pools inner-vating different hind limb muscles in the rat. Exp Brain Res 96: 291-303.
41. Popper P, Micevych PE (1989) Localization of calcitonin gene-related peptide and its receptors in a striated muscle. Brain Res 496: 180-186.
42. Rock E, Kozak-Reiss G. (1987) Effect of halothane on the Ca2+-transport system of surface membranes isolated from normal and ma-lignant hyperthermia pig skeletal muscle. Arch Biochem Biophys 256: 703-707.
43. Rock E, Sidi Mammar M, Thomas MA, Viret J, Vignon X (1990) Halothane-induced functional and structural modifications in sarco-plasmic reticulum membranes from pig skeletal muscle. Biochimie 72: 245-250.
44. Sakanaka M (1992) Development of neuronal elements with substance P-like immunoreactivity in the central nervous system. Ontogeny of transmitters and peptides in the CNS. Handbook of chemical neuroanatomy. Elsevier, Amsterdam-London-New york-Tokyo, pp 197-255.
45. Schiaffino S, Reggiani C. (2011) Fiber types in mammalian skeletal muscles. Physiol Rev 91: 1447-1531.
46. Senba E, Shiosaka S, Hara Y, Inagaki S, Sakanaka M, Takatsuki K, Kawai Y, Tohyama M (1982) Ontogeny of the peptidergic system in the rat spinal cord: immunohistochemical analysis. J Comp Neurol 208: 54-66.
47. Sienkiewicz W, Dudek A, Kaleczyc J, Chroszcz A (2010) Immunohistochemical characterization of neurones in the hypoglossal nucleus of the pig. Anat Histol Embryol 39: 152-159.
48. Swindle MM, Makin A, Herron AJ, Clubb FJ Jr, Frazier KS (2012) Swine as models in biomedical research and toxicology testing. Vet Pathol 49: 344-356.
49. Vazquez C, Anesetti G, Martinez PL (1999) Transient expression of nitric oxide synthase in the hypoglossal nucleus of the rat during early postnatal development. Neurosci Lett 275: 5-8.
50. Villar MJ, Huchet M, Hokfelt T, Changeux JP, Fahrenkrug J, Brown JC (1988) Existence and coexistence of calcitonin gene-related peptide, vasoactive intestinal polypeptide- and somatostatin-like immunoreactivities in spinal cord motoneurons of developing embryos and post-hatch chicks. Neurosci Lett 86: 114-118.
51. Villar MJ, Roa M, Huchet M, Hokfelt T, Changeux JP, Fahrenkrug J, Brown JC, Epstein M, Hersh L (1989) Immunoreactive calcitonin gene-related peptide, vasoactive intestinal polypeptide, and somatostatin in developing chicken spinal cord motoneurons. Eur J Neurosci 1: 269-287.
52. Vizzard MA, Erdman SL, Roppolo JR, Forstermann U, de Groat WC (1994) Differential localization of neuronal nitric oxide synthase immunoreactivity and NADPH-diaphorase activity in the cat spinal cord. Cell Tissue Res 278: 299-309.
53. Wessendorf MW, Elde RP (1985) Characterization of an immunofluorescence technique for the demonstration of coexisting neurotrans-mitters within nerve fibers and terminals. J Histochem Cytochem 33: 984-994.
54. Wiater JM, Bigliani LU (1999) Spinal accessory nerve injury. Clin Orthop Relat Res 368: 5-16.
Go to article

Authors and Affiliations

A. Dudek
1
W. Sienkiewicz
1
E. Lepiarczyk
2
J. Kaleczyc
1

  1. Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland
  2. Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum,University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082, Olsztyn, Poland
Download PDF Download RIS Download Bibtex

Abstract

Anaplasmosis and ehrlichiosis are important tick-borne rickettsial diseases of medical and veterinary importance that cause economic losses in livestock. In this study, the prevalence of Anaplasma ovis, Ehrlichia canis and Ehrlichia chaffeensis was investigated in ticks collected from sheep in various farms in Van province, which is located in the Eastern Anatolian Region of Turkey. The ticks used in this study were collected by random sampling in 26 family farm business in 13 districts of Van province. A total of 688 ticks were collected from 88 sheep and 88 tick pools were created. All ticks identified morphologically as Rhipicephalus bursa. Phylogenetic analysis of Chaperonin and 16S rRNA gene sequences confirmed A. ovis, E. canis and E. chaffeensis in this study. Of the 88 tick pools tested, 28.41% (25/88) were positive for at least one pathogen. Anaplasma DNA was detected in five of the 88 pools (5.68%), E. canis DNA was detected in 19 of the 88 pools (21.59%), and E. chaffeensis DNA was detected in one of the 88 pools (1.14%) of R. bursa ticks. To our knowledge, this is the first report describing the presence of A. ovis, E. canis, and E. chaffeensis in R. bursa ticks collected from sheep in Turkey. Further studies are needed to investigate other co-infections in sheep in Turkey.
Go to article

Bibliography

1. Aktas M, Altay K, Dumanli N, Kalkan A (2009) Molecular detection and identification of Ehrlichia and Anaplasma species in ixodid ticks. Parasitol Res 104: 1243-1248.
2. Alessandra T, Santo C (2012) Tick-borne diseases in sheep and goats: Clinical and diagnostic aspects. Small Rumin Res 106: S6-S11.
3. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ (1990) Basic local alignment search tool. J Mol Biol 215: 403-410.
4. Anderson BE, Sims KG, Olson JG, Childs JE, Piesman JF, Happ CM, Maupin GO, Johnson BJ (1993) Amblyomma americanum: a potential vector of human ehrlichiosis. Am J Trop Med Hyg 49: 239-244.
5. Aubry P, Geale DW (2011) A review of bovine anaplasmosis. Transbound Emerg Dis. 58: 1-30.
6. Belkahia H, Ben Said M, El Hamdi S, Yahiaoui M, Gharbi M, Daaloul-Jedidi M, Mhadhbi M, Jedidi M, Darghouth MA, Klabi I, Zribi L, Messadi L (2014) First molecular identification and genetic characterization of Anaplasma ovis in sheep from Tunisia. Small Rumin Res 121: 404-410.
7. Cao WC, Gao YM, Zhang PH, Zhang XT, Dai QH, Dumler JS, Fang LQ, Yang H (2000) Identification of Ehrlichia chaffeensis by nested PCR in ticks from Southern China. J Clin Microbiol 38: 2778-2780.
8. Cicculli V, Masse S, Capai L, De Lamballerie X, Charrel R, Falchi A (2019) First detection of Ehrlichia minasensis in Hyalomma mar-ginatum ticks collected from cattle in Corsica, France. Vet Med Sci 5: 243-248.
9. Cohen SB, Yabsley MJ, Freye JD, Dunlap BG, Rowland ME, Huang J, Dunn JR, Jones TF, Moncayo AC (2010) Prevalence of Ehr-lichia chaffeensis and Ehrlichia ewingii in ticks from Tennessee. Vector Borne Zoonotic Dis 10: 435-440.
10. Dahmani M, Davoust B, Rousseau F, Raoult D, Fenollar F, Mediannikov O (2017) Natural Anaplasmataceae infection in Rhipicephalus bursa ticks collected from sheep in the French Basque Country. Ticks Tick Borne Dis 8: 18-24.
11. Dugan VG, Little SE, Stallknecht DE, Beall AD (2000) Natural infection of domestic goats with Ehrlichia chaffeensis. J Clin Microbiol 38: 448-449.
12. Enkhtaivan B, Narantsatsral S, Davaasuren B, Otgonsuren D, Amgalanbaatar T, Uuganbayar E, Zoljargal M, Myagmarsuren P, Suganuma K, Molefe NI, Sivakumar T, Inoue N, Battur B, Battsetseg B, Yokoyama N (2019) Molecular detection of Anaplasma ovis in small ruminants and ixodid ticks from Mongolia. Parasitol Int 69: 47-53.
13. Estrada-Peña A, Bouattour A, Camicas JL, Walker AR (2004) Ticks of domestic animals in the Mediterranean region. A guide to identi-fication of species. University of Zaragoza, Spain 123-126.
14. Haigh JC, Gerwing V, Erdenebaatar J, Hill JE (2008) A novel clinical syndrome and detection of Anaplasma ovis in Mongolian reindeer (Rangifer tarandus). J Wildl Dis 44: 569-577.
15. Hall TA (1999) BioEdit: a user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucleic Ac-ids Symposium Series 41: 95-98.
16. Jalali SM, Khaki Z, Kazemi B, Bandehpour M, Rahbari S, Razi Jalali MR, Yasini SP (2013) Molecular detection and identification of Anaplasma species in sheep from Ahvaz, Iran. Iran J Vet Res 14: 50-56.
17. Johnson EM, Ewing SA, Barker RW, Fox JC, Crow DW, Kocan KM (1998) Experimental transmission of Ehrlichia canis (Rickettsiales: Ehrlichieae) by Dermacentor variabilis (Acari: Ixodidae). Vet Parasitol 74: 277-288.
18. Kocan KM, Fuente JL, Blouin EF (2008) Advances toward understanding the molecular biology of the Anaplasma-tick interface. Front Biosci 13: 7032-7045.
19. Kramer VL, Randolph MP, Hui LT, Irwin WE, Gutierrez AG, Vugia DJ (1999) Detection of the agents of human ehrlichioses in ixodid ticks from California. Am J Trop Med Hyg 60: 62-65.
20. Lee SO, Na DK, Kim CM, Li YH, Cho YH, Park JH, Lee JH, Eo SK, Klein TA, Chae JS (2005) Identification and prevalence of Ehr-lichia chaffeensis infection in Haemaphysalis longicornis ticks from Korea by PCR, sequencing and phylogenetic analysis based on 16S rRNA gene. J Vet Sci 6: 151-155.
21. Li Y, Galon EM, Guo Q, Rizk MA, Moumouni PF, Liu M, Li J, Ji S, Chahan B, Xuan X (2020) Molecular detection and identification of Babesia spp., Theileria spp., and Anaplasma spp. in sheep from border regions, Northwestern China. Front Vet Sci 7: 630.
22. Liu Z, Ma M, Wang Z, Wang J, Peng Y, Li Y, Guan G, Luo J, Yin H (2011) Molecular survey and genetic identification of Anaplasma species in goats from central and southern China. Appl Environ Microbiol 78: 464-470.
23. Ma M, Liu Z, Sun M, Yang J, Guan G, Li Y, Luo J, Yin H (2011) Development and evaluation of a loop-mediated isothermal amplifica-tion method for rapid detection of Anaplasma ovis. J Clin Microbiol 49: 2143-2146.
24. Macieira DB, Messick JB, Cerqueira AM, Freire IM, Linhares GF, Almeida NK, Almosny NR (2005) Prevalence of Ehrlichia canis in-fection in thrombocytopenic dogs from Rio de Janeiro, Brazil. Vet Clin Pathol 34: 44-48.
25. Makino H, Sousa VR, Fujimori M, Rodrigues JY, Dias AF, Dutra V, Nakazato L, de Almeida AB (2015) Ehrlichia canis detection in dogs from Várzea Grande: a comparative analysis of blood and bone marrow samples. Cienc Rural 46: 310-314.
26. Masala G, Chisu V, Foxi C, Socolovschi C, Raoult D, Parola P (2012) First detection of Ehrlichia canis in Rhipicephalus bursa ticks in Sardinia, Italy. Ticks Tick Borne Dis 3: 396-397.
27. Minh BQ, Nguyen MA, Von Haeseler A (2013) Ultrafast approximation for phylogenetic bootstrap. Mol Biol Evol 30: 1188-1195.
28. Murphy GL, Ewing SA, Whitworth LC, Fox JC, Kocan AA (1998) A molecular and serologic survey of Ehrlichia canis, E. chaffeensis, and E. ewingii in dogs and ticks from Oklahoma. Vet Parasitol 79: 325-339.
29. Noaman V (2012) Identification of hard ticks collected from sheep naturally infected with Anaplasma ovis in Isfahan province, central Iran. Comp Clin Pathol 21: 367-369.
30. Ojeda-Chi MM, Rodriguez-Vivas RI, Esteve-Gasent MD, Perez de León AA, Modarelli JJ, Villegas-Perez SL (2019) Ehrlichia canis in dogs of Mexico: Prevalence, incidence, co-infection and factors associated. Comp Immunol Microbiol Infect Dis 67: 101351.
31. Piratae S, Pimpjong K, Vaisusuk K, Chatan W (2015) Molecular detection of Ehrlichia canis, Hepatozoon canis and Babesia canis vogeli in stray dogs in Mahasarakham province, Thailand. Ann Parasitol 61: 183-187.
32. Renneker S, Abdo J, Salih DE, Karagenç T, Bilgiç H, Torina A, Oliva AG, Campos J, Kullmann B, Ahmed J, Seitzer U (2013) Can Anaplasma ovis in small ruminants be neglected any longer? Transbound Emerg Dis 60 (Suppl 2): 105-112.
33. Roland WE, Everett ED, Cyr TL, Hasan SZ, Dommaraju CB, McDonald GA (1998) Ehrlichia chaffeensis in Missouri ticks. Am J Trop Med Hyg 59: 641-643.
34. Santino I, Iori A, Sessa R, Sulli C, Favia G, Del Piano M (1998) Borrelia burgdorferi s.l. and Ehrlichia chaffeensis in the National Park of Abruzzo. FEMS Microbiol Lett 164: 1-6.
35. Selim A, Abdelhady A, Alahadeb J (2020) Prevalence and first molecular characterization of Ehrlichia canis in Egyptian dogs. Pak Vet J 41: 117-121.
36. Solano‐Gallego L, Trotta M, Razia L, Furlanello T, Caldin M (2006) Molecular survey of Ehrlichia canis and Anaplasma phagocytophi-lum from blood of dogs in Italy. Ann N Y Acad Sci 1078: 515-518.
37. Torina A, Alongi A, Naranjo V, Estrada-Pena A, Vicente J, Scimeca S, Marino AM, Salina F, Caracappa S, de la Fuente J (2008) Prev-alence and genotypes of Anaplasma species and habitat suitability for ticks in a Mediterranean ecosystem. Appl Environ Microbiol 74: 7578-7584.
38. Trifinopoulos J, Nguyen LT, von Haeseler A, Minh BQ (2016) W-IQ-TREE: a fast online phylogenetic tool for maximum likelihood analysis. Nucleic Acids Res 44: W232-W235.
39. TUİK (2023) Number of animals in Turkey. Retrieved from Turkish Statistical Institute https://biruni.tuik.gov.tr/
bolgeselistatistik/sorguSayfa.do?target=tablo. Access Date: 10.07.2023.
40. Tumwebaze MA, Lee SH, Moumouni PF, Mohammed-Geba K, Sheir SK, Galal-Khallaf A, Latif HM, Morsi DS, Bishr NM, Galon EM, Byamukama B, Liu M, Li J, Li Y, Ji S, Ringo AE, Rizk MA, Suzuki H, Ibrahim HM, Xuan X (2020) First detection of Anaplasma ovis in sheep and Anaplasma platys-like variants from cattle in Menoufia governorate, Egypt. Parasitol Int 78: 102150.
41. Walker JB, Keirans JE, Horak IG (2000) The genus Rhipicephalus (Acari, Ixodidae): a guide to the brown ticks of the world: Cam-bridge: Cambridge University Press, England.
42. Wang Y, Zhang Q, Han S, Li Y, Wang B, Yuan G, Zhang P, Yang Z, Zhang H, Sun Y, Chen J, Han X, He H (2021) Ehrlichia chaffeensis and four Anaplasma species with veterinary and public health significance identified in Tibetan sheep (Ovis aries) and yaks (Bos grunniens) in Qinghai, China. Front Vet Sci 8: 727166.
43. Yılmaz AB, Deger MS (2011) Determination and Seasonal Distribution of Tick Species on Cattle and Sheep in the Van and Erciş Re-gion. YYU Vet Fak Derg 22: 133-137.
44. Yousefi A, Rahbari S, Shayan P, Sadeghi-dehkordi Z, Bahonar A (2017) Molecular detection of Anaplasma marginale and Anaplasma ovis in sheep and goat in west highland pasture of Iran. Asian Pac J Trop Biomed 7: 455-459.
45. Zhang H, Chang Z, Mehmood K, Wang Y, Rehman MU, Nabi F, Sabir AJ, Liu X, Wu X, Tian X, Zhou D (2017) First report of Ehr-lichia infection in goats, China. Microb Pathog 110: 275-278.
46. Zhang L, Liu H, Xu B, Zhang Z, Jin Y, Li W, Lu Q, Li L, Chang L, Zhang X, Fan D, Cao M, Bao M, Zang Y, Guan Z, Cheng X, Tian L, Wang S, Yu H, Yu Q, Wang Y, Zhang Y, Tang X, Yin J, Lao S, Wu B, Li J, Li W, Xu Q, Shi Y, Huang F (2014) Rural residents in China are at increased risk of exposure to tick-borne pathogens Anaplasma phagocytophilum and Ehrlichia chaffeensis. Biomed Res Int 2014:313867.


Go to article

Authors and Affiliations

A. Ayan
1
B. Aslan Çelik
2
Ö.Y. Çelik
3
Ö. Orunç Kılınç
4
G. Akyıldız
5
A.B. Yılmaz
6
D.N. Sayın İpek
7
Ö. Oktay Ayan
8
A.R. Babaoğlu
9

  1. Department of Genetics, Faculty of Veterinary Medicine, Van Yuzuncu Yil University, Van, Turkey
  2. Department of Parasitology, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  3. Department of Internal Medicine, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  4. Özalp Vocational School, Van Yüzüncü Yıl University, Van, Turkey
  5. Department of Basic Health Sciences, Faculty of Health Sciences, Marmara University, İstanbul, Turkey
  6. Faculty of Health, Van Yuzuncu Yil University, Van, Turkey
  7. Department of Parasitology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Turkey
  8. Department of Parasitology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
  9. Department of Virology, Faculty of Veterinary Medicine, Van Yuzuncu Yil University, Van, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Arsenic is an important metalloid that can cause poisoning in humans and domestic animals. Exposure to arsenic causes cell damage, increasing the production of reactive oxygen species. Chitosan is a biopolymer obtained by deacetylation of chitin with antioxidant and metal ion chelating properties. In this study, the protective effect of chitosan on arsenic-induced nephrotoxicity and oxidative damage was investigated. 32 male Wistar-albino rats were divided into 4 groups of 8 rats each as control group (C), chitosan group (CS group), arsenic group (AS group), and arsenic+chitosan group (AS+CS group). The C group was given distilled water by oral gavage, the AS group was given 100 ppm/day Na-arsenite ad libitum with drinking water, the CS group was given 200 mg/kg/day chitosan dissolved in saline by oral gavage, the AS+CS group was given 100 ppm/day Na-arsenite ad libitum with drinking water and 200 mg/kg/day chitosan dissolved in saline by oral gavage for 30 days. At the end of the 30-day experimental period, 90 mg/kg ketamine was administered intraperitoneally to all rats, and blood samples and kidney tissues were collected. Urea, uric acid, creatinine, P, Mg, K, Ca, Na, Cystatin C (CYS-C), Neutrophil Gelatinase Associated Lipocalin (NGAL) and Kidney Injury Molecule 1 (KIM-1) levels were measured in serum samples. Malondialdehyde (MDA), Glutathione (GSH), Catalase (CAT) and Superoxide dismutase (SOD) levels in the supernatant obtained from kidney tissue were analyzed by ELISA method. Compared with AS group, uric acid and creatinine levels of the AS+CS group were significantly decreased (p<0.001), urea, KIM-1, CYS-C, NGAL, and MDA levels were numerically decreased and CAT, GSH, and SOD levels were numerically increased (p>0.05). In conclusion, based on both biochemical and histopathological-immunohistochemical- immunofluorescence findings, it can be concluded that chitosan attenuates kidney injury and protects the kidney.
Go to article

Bibliography

1. Aboulthana WM, Ibrahim NE (2018) A renoprotective role of chitosan against lithium-induced renal toxicity in rats. Bull Natl Res Cent 42: 1-11.
2. Akao Y, Yamada H, Nakagawa Y (2000) Arsenic-induced apoptosis in malignant cells in vitro. Leuk Lymphoma 37: 53-63.
3. Aleksunes LM, Augustine LM, Scheffer GL, Cherrington NJ, Manautou JE (2008) Renal xenobiotic transporters are differentially ex-pressed in mice following cisplatin treatment. Toxicology 250: 82-88.
4. Aras S, Gerin F, Aydin B, Ustunsoy S, Sener U, Turan BC, Armutcu F (2015) Effects of sodium arsenite on the some laboratory signs and therapeutic role of thymoquinone in the rats. Eur Rev Med Pharmacol Sci 19: 658-663.
5. Bagga A, Bajpai A, Menon S (2005) Approach to renal tubular disorders. Indian J Pediatr 72: 771-776.
6. Bakan M (2019) Arsenic Metabolism and Arsenolipid Biosynthesis. MedFar 2: 83-88.
7. Büget Mİ, Özkilitçi E, Küçükgergin C, Seçkin Ş, Küçükay S, Yenigün Y, Orhun G, Akıncı İÖ, Özcan PE (2014) Early Diagnosis in Acute Kidney Failure: Neutrophil Gelatinase Associated Lipocain (NGAL), Kidney Injury Molecule-1 (KIM-1), Interleukine-18 (IL-18), Cystatin C. J Turk Soc Intens Care 12: 94-100.
8. Cai X, Yu Y, Huang Y, Zhang L, Jia PM, Zhao Q, Chen Z, Tong JH, Dai W, Chen GQ (2003) Arsenic trioxide-induced mitotic arrest and apoptosis in acute promyelocytic leukemia cells. Leukemia 17: 1333-1337.
9. Chou CK, Li YC, Chen SM, Shih YM, Lee JA (2015) Chitosan prevents gentamicin-induced nephrotoxicity via a carbonyl stress-dependent pathway. Biomed Res Int 2015: 675714.
10. Danışman B, Çiçek B, Yıldırım S, Yüce N, Bolat I (2023) Gastroprotective effects of bromelain on indomethacin-induced gastric ulcer in rats. GSC Biological and Pharmaceutical Sciences 23: 277-286.
11. Demir A, Seventekin N (2009) Chitin, Chitosan And General Application Areas. TTED 3: 92-103.
12. Devarajan P (2010) Review: neutrophil gelatinase-associated lipocalin: a troponin-like biomarker for human acute kidney injury. Neph-rology (Carlton) 15: 419-428.
13. Dhondup T, Qian Q (2017) Electrolyte and acid-base disorders in chronic kidney disease and end-stage kidney failure. Blood Purif 43: 179-188.
14. Dieterle F, Perentes E, Cordier A, Roth DR, Verdes P, Grenet O, Pantano S, Moulin P, Wahl D, Mahl A, End P, Staedtler F, Legay F, Carl K, Laurie D, Chibout SD, Vonderscher J, Maurer G (2010) Urinary clusterin, cystatin C, beta2-microglobulin and total protein as markers to detect drug-induced kidney injury. Nat Biotechnol 28: 463-U114.
15. Duker AA, Carranza EJ, Hale M (2005) Arsenic geochemistry and health. Environ Int 31: 631-641.
16. Elieh-Ali-Komi D, Hamblin MR (2016) Chitin and chitosan: production and application of versatile biomedical nanomaterials. Int J Adv Res (Indore) 4: 411–427.
17. Ewere EG, Okolie NP, Eze GI, Jegede DA (2019) Irvingia gabonensis leaves mitigate arsenic-induced renal toxicity in Wistar rats. Asian J Biomed Pharmaceut Sci 9: 17-25.
18. Flora SJ (2011) Arsenic-induced oxidative stress and its reversibility. Free Radic Biol Med 51: 257-281.
19. Florea AM, Yamoah EN, Dopp E (2005) Intracellular calcium disturbances induced by arsenic and its methylated derivatives in relation to genomic damage and apoptosis induction. Environ Health Perspect 113: 659-664.
20. Galanis A, Karapetsas A, Sandaltzopoulos R (2009) Metal-induced carcinogenesis, oxidative stress and hypoxia signalling. Mutat Res 674: 31-35.
21. Ghys L, Paepe D, Smets P, Lefebvre H, Delanghe J, Daminet S (2014) Cystatin C: a new renal marker and its potential use in small ani-mal medicine. J Vet Intern Med 28: 1152-1164.
22. Hughes MF (2002) Arsenic toxicity and potential mechanisms of action. Toxicol Lett 133: 1-16.
23. Ibaokurgil F, Aydin H, Yildirim S, Sengul E (2023) Melatonin alleviates oxidative stress, inflammation, apoptosis, and DNA damage in acrylamide–induced nephrotoxicity in rats. Asian Pac J Trop Biomed 13: 121-130.
24. Ishaq A, Gulzar H, Hassan A, Kamran M, Riaz M, Parveen A, Chattha MS, Walayat N, Fatima S, Afzal S, Fahad S (2021) Ameliorative mechanisms of turmeric-extracted curcumin on arsenic (As)-induced biochemical alterations, oxidative damage, and impaired organ func-tions in rats. Environ Sci Pollut Res Int 28: 66313-66326.
25. Jeon TI, Hwang SG, Park NG, Jung YR, Shin SI, Choi SD, Park DK (2003) Antioxidative effect of chitosan on chronic carbon tetra-chloride induced hepatic injury in rats. Toxicology 187: 67-73.
26. Jia G, Sone H, Nishimura N, Satoh M, Tohyama C (2004) Metallothionein (I/II) suppresses genotoxicity caused by dimethylarsinic acid. Int J Oncol 25: 325-333.
27. Jomova K, Valko M (2011) Advances in metal-induced oxidative stress and human disease. Toxicology 283: 65-87.
28. Kamran M, Malik Z, Parveen A, Huang L, Riaz M, Bashir S, Mustafa A, Abbasi GH, Xue B, Ali U (2020) Ameliorative effects of bio-char on rapeseed (Brassica napus L.) growth and heavy metal immobilization in soil irrigated with untreated wastewater. J Plant Growth Regul 39: 266-281.
29. Kaya Z, Eraslan G (2013) The effects of evening primrose oil on arsenic-induced oxidative stress in rats. Toxicol Environ Chem 95: 1416-1423.
30. Kitchin KT, Conolly R (2010) Arsenic-induced carcinogenesis oxidative stress as a possible mode of action and future research needs for more biologically based risk assessment. Chem Res Toxicol 23: 327-335.
31. Liu J, Waalkes MP (2008) Liver is a target of arsenic carcinogenesis. Toxicol Sci 105: 24-32.
32. Liu YC, Huang H (1997) Involvement of calcium-dependent protein kinase C in arsenite-induced genotoxicity in chinese hamster ovary cells. J Cell Biochem 64: 423-433.
33. Mehrzadi S, Fatemi I, Malayeri AR, Khodadadi A, Mohammadi F, Mansouri E, Rashno M, Goudarzi M (2018) Ellagic acid mitigates sodium arsenite-induced renal and hepatic toxicity in male Wistar rats. Pharmacol Rep 70: 712-719.
34. Mussap M, Plebani M (2004) Biochemistry and clinical role of human cystatin C. Crit Rev Clin Lab Sci 41: 467-550.
35. Muzaffar S, Khan J, Srivastava R, Gorbatyuk MS, Athar M (2023) Mechanistic understanding of the toxic effects of arsenic and warfare arsenicals on human health and environment. Cell Biol Toxicol 39: 85-110.
36. Namgung U, Xia Z (2001) Arsenic induces apoptosis in rat cerebellar neurons via activation of JNK3 and p38 MAP kinases. Toxicol Appl Pharmacol 174: 130-138.
37. Narayana KR, Reddy MS, Chaluvadi MR, Krishna DR (2001) Bioflavonoids classification, pharmacological, biochemical effects and therapeutic potential. Indian J Pharmacol 33: 2-16.
38. Noman AS, Dilruba S, Mohanto NC, Rahman L, Khatun Z, Riad W, Al Mamun A, Alam S, Aktar S, Chowdhury S, Saud ZA, Rahman Z, Hossain K, Haque A (2015) Arsenic-induced histological alterations in various organs of mice. J Cytol Histol 6: 323.
39. Nomier YA, Alshahrani S, Elsabahy M, Asaad GF, Hassan A, El-Dakroury WA (2022) Ameliorative effect of chitosan nanoparticles against carbon tetrachloride-induced nephrotoxicity in Wistar rats. Pharm Biol 60: 2134-2144.
40. Nozohour Y, Jalilzadeh-Amin G (2019) Histopathological changes and antioxidant enzymes status in oxidative stress induction using Sodium arsenite in rats. J Appl Biotechnol Rep 6: 40-44.
41. Özdek U, Toz H, Kömüroğlu AU, Mis L, Huyut Z, Değer Y (2019) Protective Effect of Chitosan Against Lead-Induced Oxidative Stress in Rat Kidney. Van Vet J 30: 187-191.
42. Patel HV, Kalia K (2010) Sub-chronic arsenic exposure aggravates nephrotoxicity in experimental diabetic rats. Indian J Exp Biol 48: 762-768.
43. Peters BA, Hall MN, Liu X, Neugut YD, Pilsner JR, Levy D, Ilievski V, Slavkovich V, Islam T, Factor-Litvak P, Graziano JH, Gamble MV (2014) Creatinine, arsenic metabolism, and renal function in an arsenic-exposed population in Bangladesh. PLoS One 9: e113760.
44. Poręba R, Gać P, Poręba M, Antonowicz-Juchniewicz J, Andrzejak R (2011) Relation between occupational exposure to lead, cadmium, arsenic and concentration of cystatin C. Toxicology 283: 88-95.
45. Robles-Osorio ML, Sabath-Silva E, Sabath E (2015) Arsenic-mediated nephrotoxicity. Ren Fail 37: 542-547.
46. Roy M, Roy S (2011) Ameliorative potential of Psidium guajava in induced arsenic toxicity in Wistar rats. Vet World 4: 82-83.
47. Selby LA, Case AA, Osweiler GD, Hayes HM Jr (1977) Epidemiology and toxicology of arsenic poisoning in domestic ani-mals. Environ Health Perspect 19: 183-189.
48. Sharma S, Kaur T, Sharma AK, Singh B, Pathak D, Yadav HN, Singh AP (2022) Betaine attenuates sodium arsenite-induced renal dys-function in rats. Drug Chem Toxicol 45: 2488-2495.
49. Shen ZY, Shen WY, Chen MH, Shen J, Cai WJ, Yi Z (2002) Nitric oxide and calcium ions in apoptotic esophageal carcinoma cells in-duced by arsenite. World J Gastroenterol 8: 40-43.
50. Shen ZY, Shen J, Cai WJ, Hong CQ, Zheng MH (2000) The alteration of mitochondria is an early event of arsenic trioxide induced apoptosis in esophageal carcinoma cells. Int J Mol Med 5: 155-158.
51. Singh MK, Yadav SS, Yadav RS, Singh US, Shukla Y, Pant KK, Khattri S (2014) Efficacy of crude extract of Emblica officinalis (amla) in arsenic-induced oxidative damage and apoptosis in splenocytes of mice. Toxicol Int 21: 8-17.
52. Sinha M, Manna P, Sil PC (2008) Arjunolic acid attenuates arsenic-induced nephrotoxicity. Pathophysiology 15: 147-156.
53. Smith DL, Harris AD, Johnson JA, Silbergeld EK, Morris JG Jr (2002) Animal antibiotic use has an early but important impact on the emergence of antibiotic resistance in human commensal bacteria. Proc Natl Acad Sci USA 99: 6434-6439.
54. Tilako BH, Ogbodo SO, Okonkwo IN, Nubila IN, Shuneba IL, Ogbonna E, Odoma S, Gali RM, Bassey BE, Shu EN (2020) Distribu-tion and interactions of priority heavy metals with some antioxidant micronutrients in inhabitants of a lead-zinc mining community of eb-onyi state, Nigeria. Adv Toxicol Toxic Effects 4: 011-017.
55. Toz H, Değer Y (2018) The Effect of Chitosan on the Erythrocyte Antioxidant Potential of Lead Toxicity-Induced Rats. Biol Trace Elem Res 184:114-118
56. Wang Z, Yan Y, Yu X, Li W, Li B, Qin C (2016) Protective effects of chitosan and its water-soluble derivatives against lead-induced oxidative stress in mice. Int J Biol Macromol 83: 442-449.
57. Wan Muhamad Salahudin WS, Norlelawati AT, Nor ZA, Aung S, Asmah HH, Zunariah B (2021) Histopathological changes in chronic low dose organic arsenic exposure in rats kidney. IIUM Med J Malays 20: 91-98.
58. Wu KY, Wu M, Fu ML, Li H, Yang Y, Zhang H, Cheng C, Wang ZZ, Wang XY, Lu XB, Liu DG, Li H, Gao R (2006) A novel chi-tosan CpG nanoparticle regulates cellular and humoral immunity of mice. Biomed Environ Sci 19: 87-95.
59. Yuan WP, Liu B, Liu CH, Wang XJ, Zhang MS, Meng XM, Xia XK (2009) Antioxidant activity of chito-oligosaccharides on pancreatic islet cells in streptozotocin-induced diabetes in rats. World J Gastroenterol 15: 1339.
60. Zalups RK (1997) Reductions in renal mass and the nephropathy induced by mercury. Toxicol Appl Pharmacol 143: 366-379.
61. Zeng L, Qin C, Wang W, Chi W, Li W (2008) Absorption and distribution of chitosan in mice after oral administra-tion. Carbohydr Polym 71: 435-440.
62. Zhang Z, Lu B, Sheng X, Jin N (2011) Cystatin C in prediction of acute kidney injury: a systemic review and meta-analysis. Am J Kid-ney Dis 58: 356-365.
63. Zheng L, Kuo CC, Fadrowski J, Agnew J, Weaver VM, Navas-Acien A (2014) Arsenic and chronic kidney disease: a systematic re-view. Curr Environ Health Rep 1: 192-207.

Go to article

Authors and Affiliations

K. İrak
1
Ö.Y. Çelik
2
M. Bolacalı
3
T. Tufan
4
S. Özcan
4
S. Yıldırım
5
İ. Bolat
5

  1. Department of Biochemistry, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  2. Department of Internal Medicine, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  3. Kırsehir Ahi Evran University, Faculty of Medicine, Department of Biostatistics and Medical Informatics, Kirsehir, Turkey
  4. Department of Animal Nutrition and Nutritional Disease, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  5. Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Our main aim was to investigate the predictive value of prepartum behaviors such as total daily rumination (TDR), total daily activity (TDA) and dry matter intake (DMI) as early indicators to detect cows at risk for hyperketonemia (HYK), hypoglycemia (HYG) or high non-esterified fatty acid (NEFA) status in the first (wk1) and second week (wk2) postpartum. In a case control study, 64 Holstein cows were enrolled 3 weeks before the expected time of calving and monitored until 15 days in milk (DIM). Postpartum blood samples were taken at D3 and D6 for wk1 and at D12 and D15 for wk2 to measure beta-hydroxybutyrate, NEFA and glucose concentration. Ear-mounted accelerometers were used to measure TDR and TDA. DMI and milk yield were obtained from farm records. Relationships between the average daily rate of change in prepartum TDR (ΔTDR), TDA (ΔTDA), and DMI (ΔDMI) with postpartum HYK, HYG and NEFA status in wk1 and wk2 post-partum were evaluated using linear regression models. Models were adjusted for potential confounding variables, and covariates retained in the final models were determined by backward selection. No evidence was found to support the premise that prepartum ΔTDR, ΔTDA or ΔDMI predicted postpartum HYK, HYG or NEFA status in wk1 or in wk2. Overall, prepartum ΔTDR, ΔTDA and ΔDMI were not effective predictors of HYK, HYG or NEFA status in the first 2 weeks postpartum.
Go to article

Bibliography

1. Adewuyi AA, Gruys E, van Eerdenburg FJ (2005) Non esterified fatty acids (NEFA) in dairy cattle. A review. Vet Q 27: 117-126.
2. Bauman DE, Currie WB (1980) Partitioning of Nutrients During Pregnancy and Lactation: A Review of Mechanisms Involving Homeo-stasis and Homeorhesis. J Dairy Sci 63: 1514-1529.
3. Bell AW (1995) Regulation of organic nutrient metabolism during transition from late pregnancy to early lactation. J Anim Sci 73: 2804-2819.
4. Berckmans D (2015) Smart farming for Europe: value creation through precision livestock farming. In: Ilan Halachmi (ed) Precision Livestock Farming Applications, Wageningen Academic, brill, pp 139-147.
5. Bikker JP, van Laar H, Rump P, Doorenbos J, van Meurs K, Griffioen GM, Dijkstra J (2014) Technical note: Evaluation of an ear-attached movement sensor to record cow feeding behavior and activity. J Dairy Sci 97: 2974-2979.
6. Caixeta LS, Ospina PA, Capel MB, Nydam DV (2015) The association of subclinical hypocalcemia, negative energy balance and disease with bodyweight change during the first 30 days post-partum in dairy cows milked with automatic milking systems. Vet J 204: 150-156.
7. Constable PD, Hinchcliff KW, Done SH, Gruenberg W (2016) Veterinary Medicine: A Textbook of the Diseases of Cattle, Horses, Sheep, Pigs and Goats. 11th ed.,. Elsevier Health Sciences, pp 1662-1726.
8. Contreras GA, O’Boyle NJ, Herdt TH, Sordillo LM (2010) Lipomobilization in periparturient dairy cows influences the composition of plasma nonesterified fatty acids and leukocyte phospholipid fatty acids. J Dairy Sci 93: 2508-2516.
9. Duffield TF, Lissemore KD, McBride BW, Leslie KE (2009) Impact of hyperketonemia in early lactation dairy cows on health and pro-duction. J Dairy Sci 92: 571-580.
10. Edwards JL, Tozer PR (2004) Using Activity and Milk Yield as Predictors of Fresh Cow Disorders. J Dairy Sci 87: 524-531.
11. Emam MH, Shepley E, Mahmoud MM, Ruch M, Elmaghawry S, Abdelrazik W, Abdelaal AM, Crooker BA, Caixeta LS (2023) The association between prepartum rumination time, activity and dry matter intake and subclinical hypocalcemia and hypomagnesemia in the first 3 days postpartum in Holstein dairy cows. Animals 13:1621
12. Goff JP (2006) Macromineral physiology and application to the feeding of the dairy cow for prevention of milk fever and other peripar-turient mineral disorders. Anim Feed Sci Technol 126: 237-257.
13. Goff JP (2008) The monitoring, prevention, and treatment of milk fever and subclinical hypocalcemia in dairy cows. Vet J 176: 50-57.
14. Goldhawk C, Chapinal N, Veira DM, Weary DM, von Keyserlingk MA. (2009) Prepartum feeding behavior is an early indicator of sub-clinical ketosis. J Dairy Sci 92: 4971-4977.
15. González LA, Tolkamp BJ, Coffey MP, Ferret A, Kyriazakis I (2008) Changes in feeding behavior as possible indicators for the auto-matic monitoring of health disorders in dairy cows. J Dairy Sci 91: 1017-1028.
16. Grummer RR (1995) Impact of changes in organic nutrient metabolism on feeding the transition dairy cow. J Anim Sci 73: 2820-2833.
17. Hammon DS, Evjen IM, Dhiman TR, Goff JP, Walters JL (2006) Neutrophil function and energy status in Holstein cows with uterine health disorders. Vet Immunol Immunopathol 113: 21-29.
18. Hansen LB, Young CW, Miller KP, Touchberry RW (1979) Health Care Requirements of Dairy Cattle. I. Response to Milk Yield Selec-tion. J Dairy Sci 62: 1922-1931.
19. Hayirli A, Grummer RR, Nordheim EV, Crump PM (2002) Animal and dietary factors affecting feed intake during the prefresh transi-tion period in holsteins. J Dairy Sci 85: 3430-3443.
20. Hebbali A (2020) Olsrr: Tools for Building OLS Regression Models, R Package Version 0.5.3.; R Foundation for Statistical Compu-ting:Vienna, Austria.
21. Herdt TH (2000) Ruminant adaptation to negative energy balance. Influences on the etiology of ketosis and fatty liver.Vet Clin North Am Food Anim Pract 16: 215-230.
22. Huzzey JM, Veira DM, Weary DM, Von Keyserlingk MA (2007) Prepartum behavior and dry matter intake identify dairy cows at risk for metritis. J Dairy Sci 90: 3220-3233.
23. Kabir M, Hasan MM, Tanni NS, Parvin MS, Asaduzzaman M, Ehsan MA, Islam MT (2022) Metabolic profiling in periparturient dairy cows and its relation with metabolic diseases. BMC Res Notes 15: 231.
24. Kaufman EI, LeBlanc SJ, McBride BW, Duffield TF, DeVries TJ (2016) Association of rumination time with subclinical ketosis in tran-sition dairy cows. J Dairy Sci 99: 5604-5618.
25. LeBlanc SJ, Leslie KE, Duffield TF (2005) Metabolic predictors of displaced abomasum in dairy cattle. J Dairy Sci 88: 159-170.
26. Liboreiro DN, Machado KS, Silva PR, Maturana MM, Nishimura TK, Brandão AP, Endres MI, Chebel RC (2015) Characterization of peripartum rumination and activity of cows diagnosed with metabolic and uterine diseases. J Dairy Sci 98: 6812-6827.
27. McArt JA, Nydam DV, Oetzel GR (2012) Epidemiology of subclinical ketosis in early lactation dairy cattle. J Dairy Sci 95: 5056-5066.
28. Ospina PA, McArt JA, Overton TR, Stokol T, Nydam DV (2013) Using nonesterified fatty acids and beta hydroxybutyrate concentra-tions during the transition period for herd-level monitoring of increased risk of disease and decreased reproductive and milking perfor-mance. Vet Clin North Am Food Anim Pract 29: 387-412.
29. Ospina PA, Nydam DV, Stokol T, Overton TR (2010) Association between the proportion of sampled transition cows with increased nonesterified fatty acids and beta hydroxybutyrate and disease incidence, pregnancy rate, and milk production at the herd level. J Dairy Sci 93: 3595-3601.
30. Overton TR (2001) Transition cow programs. The good, the bad, and how to keep them from getting ugly. Adv Dairy Tech 13: 17-26.
31. Paudyal S (2021) Using rumination time to manage health and reproduction in dairy cattle: a review. Vet Q 41: 292-300.
32. Ruoff J, Borchardt S, Heuwieser W (2017) Short communication: Associations between blood glucose concentration, onset of hyper-ketonemia, and milk production in early lactation dairy cows. J Dairy Sci 100: 5462-5467.
33. Schirmann K, Weary DM, Heuwieser W, Chapinal N, Cerri RL, von Keyserlingk MA (2016) Short communication: Rumination and feeding behaviors differ between healthy and sick dairy cows during the transition period. J Dairy Sci 99: 9917-9924.
34. Soriani N, Trevisi E, Calamari L (2012) Relationships between rumination time, metabolic conditions, and health status in dairy cows during the transition period. J Anim Sci 90: 4544-4554.
35. Stevenson JS, Banuelos S, Mendonça LG. (2020) Transition dairy cow health is associated with first postpartum ovulation risk, meta-bolic status, milk production, rumination, and physical activity. J Dairy Sci 103: 9573-9586.
36. Mepham TB (1993) The development of ideas on the role of glucose in regulating milk secretion. Australian J Agric Res 44: 509-522.
37. van Hoeij RJ, Kok A, Bruckmaier RM, Haskell MJ, Kemp B, van Knegsel AT (2019) Relationship between metabolic status and behav-ior in dairy cows in week 4 of lactation. Animal 13: 640-648.
38. von Keyserlingk MA, Rushen J, de Passillé AM, Weary DM (2009) Invited review: The welfare of dairy cattle – Key concepts and the role of science. J Dairy Sci 92: 4101-4111.
39. Wathes CM, Kristensen HH, Aerts JM, Berckmans D (2008) Is precision livestock farming an engineer’s daydream or nightmare, an animal’s friend or foe, and a farmer’s panacea or pitfall? Comp Electr 64: 2-10.
40. Weary DM, Huzzey JM, Von Keyserlingk MA (2009) Boardinvited review: Using behavior to predict and identify ill health in animals. J Anim Sci 87: 770-777.
41. Weber WJ, Wallaces CR, Hansen LB, Chester-Jones H, Crooker BA (2007) Effects of genetic selection for milk yield on somatotropin, insulin-like growth factor-I, and placental lactogen in Holstein cows. J Dairy Sci 90: 3314-3325.
42. Young CW (1977) Review of Regional Project NC-2. J Dairy Sci 60: 493-498.
Go to article

Authors and Affiliations

M.H. Emam
1 2
E. Shepley
1
M.M. Mahmoud
1 3
M. Ruch
1
S. Elmaghawry
2
W. Abdelrazik
2
A.M. Abdelaal
2
B.A. Crooker
4
L.S. Caixeta
1

  1. Department of Veterinary Population Medicine, University of Minnesota, Saint Paul, MN 55108, USA
  2. Department of Animal Medicine, Zagazig University, Zagazig 44511, Egypt
  3. Department of Animal Medicine, Beni-Suef University, Beni-Suef 62521, Egypt
  4. Department of Animal Science, University of Minnesota, Saint Paul, MN 55108, USA
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 experiment aimed to determine the effect of adaptive duration to saline water on behaviors, weight gain and blood biochemical parameters in growing goats. The experiment was arranged in a completely randomized design, which included four treatments with five animals per group. The goats were administered either fresh water (control) or seawater with a salinity of 1.5%, with varying durations of adaptation to seawater. The adaptive durations included an abrupt change (A0) from fresh water to seawater with a salinity of 1.5% or stepwise adaptation either 4 (A4) or 7 (A7) days of increasing saline concentrations. The results showed that dry matter intake in the non-adapted goats (A0 group) was lower than that of the control group or the adapted goats throughout the experiment (p<0.05). In contrast, water intake from drinking saline water was greater than that in the control group (p<0.05). Body weigh did not differ among the treatments; however, non-adapted goats exhibited a lower weight gain than the adapted goats (p<0.05). The goats in the A0 and A4 groups exhibited increased plasma levels of urea, AST, and ALT compared with the control and A7 groups. However, blood electrolyte levels remained unchanged and were within the normal range for goats. Therefore, it is concluded that the stepwise adaptation to seawater with a salinity of 1.5% for 21 days has no influence on productivity and health status of goats.
Go to article

Bibliography

1. Al-Ramamneh D, Riek A, Gerken M (2012) Effect of water restriction on drinking behaviour and water intake in German black-head mutton sheep and Boer goats. Animal 6: 173-178.
2. Assad F, El-Sherif MMA (2002) Effect of drinking saline water and feed shortage on adaptive responses of sheep and camels. Small Rumin Res 45: 279-290.
3. Association of Official Analytical Chemists (AOAC) (1990) Official Method of Analysis, 15th ed., Virginia, USA. Association of Offi-cial Agricultural Chemists, Inc.
4. Bahman A, Rooket J, Topps J (1993) The performance of dairy cows offered drinking water of low or high salinity in a hot arid climate. Animal Science 57(1): 23-28.
5. Do Nguyen DK, Semsirmboon S, Chaiyabutr N, Thammacharoen S (2022) Effects of Low Dietary Cation and Anion Difference on Blood Gas, Renal Electrolyte, and Acid Excretions in Goats in Tropical Conditions. Animals (Basel) 12: 3444.
6. Cardoso EDA, Furtado DA, Ribeiro NL, Saraiva EP, do Nascimento JWB, de Medeiros AN, Pereira PHB (2021) Intake salinity water by creole goats in a controlled environment: ingestive behavior and physiological variables. Trop Anim Health Prod 53: 1-7.
7. Costa RG, Ribeiro NL, Nobre PT, Carvalho FFR, Medeiros AN, Martins FE (2019) Ingestive behavior and efficacy of male sheep housed in different stocking densities. Rev Bras de Zootec 46: e20180219
8. Custodio SAS, Tomaz MPP, Silva DAL, Goulart RO, Dias KM, Carvalho ER (2017) Feeding behavior of beef cattle fed different for-ages and housed in individual or collective pens. Anim Behav Biometeorol 5: 20-28.
9. Dunson WA (1974) Some aspects of salt and water balance of feral goats from arid islands. Am J Physiol 226: 662-669.
10. Eltayeb EE (2006) Effect of salinity of drinking water and dehydration on thermo regulation, blood and urine composition in nubian goats. In: M. Vet. Sc. Thesis Faculty of Veterinary Medicine, University of Khartoum, Sudan.
11. Enke N, Brinkmann L, Runa RA, Südekum KH, Tholen E, Gerken M (2022) Drinking behaviour of llamas (Lama glama) in choice tests for fresh or saline water. Small Rumin Res 216: 1-9.
12. Fahmy AA, Youssef KM, El Shaer HM (2010) Intake and nutritive value of some salt-tolerant fodder grasses for sheep under saline conditions of South Sinai, Egypt. Small Rumin Res 91:110–115.
13. Furtado DA, Carvalho Júnior SB, Souza BB, Dantas NLB, Rodrigues LR (2021) Ingestive behavior of santa inês sheep under ther-moneutrality and thermal stress upon consumption of saline water. Engenharia Agrícola, 41(1), 19-24.
14. Ítavo LCV, Souza S R M B O, Rimoli J, Ítavo C C B F, Dias AM (2008) Diurnal intake behavior of bovines in continuous or rotational grazing. Arch de Zootec 57: 43-52.
15. Jackson PGG, Cockcroft PD (2002) Clinical examination of farm animals. Blackwell Science, Oxford, UK, pp 303-305.
16. Kattnig RM, Pordomingo AJ, Schneberger AG, Duff GC, Wallance JD (1992) Influence of saline water on intake, digesta kinetics, and serum profiles of steers. J Range Manage 45: 514-518.
17. Kii WY, Dryden GM (2005) Effect of drinking saline water on food and water intake, food digestibility, and nitrogen and mineral bal-ances of rusa deer stags (Cervus timorensis russa). Anim Sci 81: 99-105.
18. López A, Arroquy J, Juárez Sequeira A, García M, Nazareno M, Coria H, Distel R (2014) Effect of protein supplementation on tropical grass hay utilization by beef steers drinking saline water. J Anim Sci 92: 2152- 2160.
19. McGregor BA (2004) Water quality and provision for goats. Australian Government. Rural Industries Research and Development Cor-poration. p 19.
20. Mdletshe ZM, Chimonyo M, Marufu MC, Nsahlai IV (2017) Effects of saline water consumption on physiological responses in Nguni goats. Small Rumin Res 153: 209-211.
21. Metwally NH (2001) Studies on some physiological and behavioural aspects in camels. M.Sc. Thesis. Monoufia University, Egypt.
22. Nguyen NA (2017) Historic drought and salinity intrusion in the Mekong Delta in 2016: Lessons learned and response solutions. Viet J Sci Technol Eng 59: 93-96.
23. Nguyen T, Truong VK, Nguyen VH, Nguyen TN, Thammacharoen S (2022a) Effects of high salinity in drinking water on behaviors, growth and renal electrolyte excretion in crossbred Boer goats under tropical conditions. Vet World 15: 834-840.
24. Nguyen T, Nguyen TN, Nguyen THN, Thammacharoen S (2022b) The effects of high saline water on physiological responses, nutrient digestibility and milk yield in lactating crossbred goats. Livest Res Rural Develop 34: 01-08.
25. Nguyen T, Truong VK, Nguyen TN, Thammacharoen S (2023) Salt tolerance threshold and physiological responses in Bach Thao goats drank diluted seawater under tropical conditions. Vet World 16: 1714-1720.
26. Runa RA, Brinkmann L, Gerken M, Riek A (2019) Adaptive capacity of Boer goats to saline drinking water. Animal 13: 2268-2276.
27. Runa RA, Gerken M, Riek A, Brinkmann L (2020) Boer goats physiology adaptation to saline drinking water. Res Vet Sci 129: 120-128.
28. Semsirmboon S, Do Nguyen DK, Chaiyabutr N, Poonyachoti S, Lutz TA, Thammacharoen S (2023) High dietary cation and anion dif-ference and high-dose ascorbic acid modify acid-base and antioxidant balance in dairy goats fed under tropical conditions. Animals (Ba-sel) 13:970
29. Tsukahara Y, Puchala R, Sahlu T, Goetch AL (2016) Effects of level of brackish water on feed intake, digestion, heat energy, and blood constituents of growing Boer and Spanish goat wethers. J Anim Sci 94: 3864-3874.
30. Van Soest PJ, Robertson JB, Lewis BA (1991) Methods for dietary fiber neutral detergent fiber and non-starch polysaccharides in rela-tion to animal nutrition. J Dairy Sci 74: 3583-3597
31. Wilson AD (1975) Influence of water salinity on sheep performance while grazing on natural grassland and saltbush pastures. Australian J Experim Agricul 15: 760-765.
32. Yousfi I, Salem HB, Aouadi D, Abidi S (2016) Effect of sodium chloride, sodium sulfate or sodium nitrite in drinking water on intake, digestion, growth rate, carcass traits and meat quality of Barbarine lamb. Small Rumin Res 143: 43-52.
33. Zoidis E, Hadjigeorgiou I (2018) Effects of drinking saline water on food and water intake, blood and urine electrolytes and biochemical and haematological parameters in goats: a preliminary study. Anim Prod Sci 58: 1822-1828.
Go to article

Authors and Affiliations

T. Nguyen
1
N. Nguyen Trong
2
N. Chaiyabutr
3
S. Thammacharoen
3

  1. Department of Agricultural Technology, College of Rural Development, Can Tho University, 3/2 street, Can Tho city 94000, Vietnam
  2. Department of Animal Science, College of Agriculture, Can Tho University, 3/2 street, Can Tho city 94000, Vietnam
  3. Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, HenriDunang street, Bangkok 10330, Thailand
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to investigate the antimycobacterial activity of 39 free terpenes and their activity in combination with streptomycin. Antimicrobial activity was first evaluated by screening 39 free terpenes at concentrations from 1.56 to 400 μg/mL. None of these exhibited positive effects against any of the nontuberculous mycobacteria (NTM) strains tested. However, six of the 39 terpenes (isoeugenol, nerol, (+)-α-terpineol, (1R)-(−)-myrtenol, (+)-terpinen-4-ol, and eugenol) were shown to enhance the activity of streptomycin against the NTM strains isolated from diseased ornamental fish.
Go to article

Bibliography

1. Barbosa CR, Scherf JR, de Freitas TS, de Menezes IR, Pereira RL, dos Santos JF, de Jesus SS, Lopes TP, Silveira Z, Oliveira-Tintino CD, Junior JP, Coutinho HD, Tintino SR, da Cunha FA (2021) Effect of carvacrol and thymol on NorA efflux pump inhibition in multi-drug-resistant (MDR) Staphylococcus aureus strains. J Bioenerg Biomembr 53: 489-498.
2. Cantrell CL, Franzblau SG, Fischer NH (2001) Antimycobacterial plant terpenoids. Planta Med 67: 685–694.
3. Coêlho ML, Ferreira JH, de Siqueira-Júnior JP, Kaatz GW, Barreto HM, de Carvalho Melo Cavalcante AA (2016) Inhibition of the NorA multi-drug transporter by oxygenated monoterpenes. Microb Pathog 99: 173–177.
4. Decostere A, Hermans K, Haesebrouck F (2004) Piscine mycobacteriosis: A literature review covering the agent and the disease it causes in fish and humans. Vet Microbiol 99: 159-166.
5. Dias KJ, Miranda GM, Bessa JR, de Araújo AC, Freitas PR, Almeida RS, Paulo CL, Neto JB, Coutinho HD, Ribeiro-Filho J (2022) Terpenes as bacterial efflux pump inhibitors: A systematic review. Front Pharmacol 13: 953982.
6. De Rossi E, Aínsa JA, Riccardi G (2006) Role of mycobacterial efflux transporters in drug resistance: an unresolved question. FEMS Microbiol Rev 30: 36–52.
7. Gupta, AK, Chauhan DS, Srivastava K, Das R, Batra S, Mittal M, Goswami P, Singhal N, Sharma VD, Venkatesan K, Hasnain SE, Katoch VM (2006) Estimation of efflux mediated multi-drug resistance and its correlation with expression levels of two major efflux pumps in mycobacteria. J Commun Dis 38: 246–254.
8. Guz L, Grądzki Z, Krajewska M, Lipiec M, Zabost A, Augustynowicz-Kopeć E, Zwolska Z, Szulowski K (2013) Occurrence and anti-microbial susceptibility of Mycobacterium peregrinum in ornamental fish. Bull. Vet Inst Pulawy 57: 489-492.
9. Guz L, Puk K (2022) Antibiotic susceptibility of mycobacteria isolated from ornamental fish. J Vet Res 66: 69-76.
10. Jin J, Zhang JY, Guo N, Sheng H, Li L, Liang JC, Wang XL, Li Y, Liu MY, Wu XP, Yu L (2010) Farnesol, a potential efflux pump in-hibitor in Mycobacterium smegmatis. Molecules 15: 7750-7762.
11. Kumar G, Karthik L, Bhaskara Rao KV (2011) Haemolytic activity of Indian medicinal plants toward human erythrocytes: an in vitro study. Elixir Appl Botany 40: 5534-5537.
12. Kumar M, Singh SK, Singh PP, Singh VK, Rai AC, Srivastava AK, Shukla L, Kesawat MS, Jaiswal AK, Chung SM, Kumar A (2021) Potential anti-Mycobacterium tuberculosis activity of plant secondary metabolites: Insights with molecular docking interactions. Antioxi-dans 10: 1990.
13. Mahizan NA, Yang SK, Moo CL, Song AA, Chong CM, Chong CW, Abushelaibi A, Lim SH, Lai KS (2019) Terpene derivatives as a potential agent against antimicrobial resistance (AMR) pathogens. Molecules 24: 2631.
14. Marquez B (2005) Bacterial efflux systems and efflux pumps inhibitors. Biochimie 87: 1137-1147.
15. Martins A, Vasas A, Viveiros M, Molnár J, Hohmann J, Amaral L (2011) Antibacterial properties of compounds isolated from Car-pobrotus edulis. Int J Antimicrob Agents 37: 438-444.
16. Muniz DF, dos Santos Barbosa CR, de Menezes IR, de Sousa EO, Pereira RL, Júnior JT, Pereira PS, de Matos YM, da Costa RH, de Morais Oliveira-Tintino CD, Coutinho HD, Filho JM, de Sousa GR, Filho JR, Siqueira-Junior JP, Tintino SR (2021) In vitro and in sil-ico inhibitory effects of synthetic and natural eugenol derivatives against the NorA efflux pump in Staphylococcus aureus. Food Chem 337: 127776.
17. Puk K, Banach T, Wawrzyniak A, Adaszek Ł, Ziętek J, Winiarczyk S, Guz L (2018) Detection of Mycobacterium marinum, M. pere-grinum, M. fortuitum and M. abscessus in aquarium fish. J Fish Dis 41: 153-156.
18. Puk K, Guz L (2020) Occurrence of Mycobacterium spp. in ornamental fish. Ann Agric Environ Med 27: 535-539.
19. Puk K, Guz L (2022) Effect of alkaloid berberine on the susceptibility of nontuberculous mycobacteria to antibiotics. Pol J Vet Sci 25: 479-481.
20. Puk K, Wawrzykowski J, Guz L (2023) Evaluation of the anti-mycobacterial activity and composition of Carlina acaulis L. root extracts. Pol J Vet Sci 26: 57-63.
21. Sieniawska E, Sawicki R, Swatko-Ossor M, Napiorkowska A, Przekora A, Ginalska G, Augustynowicz-Kopeć E (2018) The effect of combining natural terpenes and antituberculous agents against reference and clinical Mycobacterium tuberculosis strains. Molecules 23: 176.
22. Sieniawska E, Swatko-Ossor M, Sawicki R, Skalicka-Woźniak K, Ginalska G (2017) Natural terpenes influence the activity of antibiot-ics against isolated Mycobacterium tuberculosis. Med Princ Pract 26: 108-112 .
23. Szmygin-Milanowska K, Grzywa-Celińska A, Zwolska Z, Krawczyk P, Guz L, Milanowski J (2016) ‘TB or not TB’ Problems of dif-ferentia diagnosis of cutaneous mycobacteriosis and tuberculosis – A case study and interdisciplinary discussion. Ann Agric Environ Med 23: 97-102.
24. Vasconcelos SS, Caleffi-Ferracioli KR, Hegeto LA, Baldin VP, Nakamura CV, Stefanello TF, Gauze GF, Yamazaki DA, Scodro RB, Siqueira VL, Cardoso RF (2018) Carvacrol activity and morphological changes in Mycobacterium tuberculosis. Future Microbiol 13: 877–888.
Go to article

Authors and Affiliations

L. Guz
1
K. Puk
1
D. Szwajgier
2
A. Pastuszka
1

  1. Department of Biology and Fish Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland
  2. Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8, 20-704 Lublin, Poland
Download PDF Download RIS Download Bibtex

Abstract

Avian gastric yeast ( Macrorhabdus ornithogaster) is a microorganism that infects aviary birds worldwide, both captive and wild. A total number of 352 birds, belonging to 18 avian species, were examined from 2019 to 2022 for M. ornithogaster, using fecal smears of live birds or cytological samples of the proventriculus taken at necropsy. These cytological samples were taken from birds that died from different causes. Some of the birds exhibited symptoms such as lethargy, regurgitation, weight loss and anorexia. Faecal samples were collected from all the birds and analysed for gastric yeast using a direct smear and Gram-staining method. The microorganism was diagnosed most frequently in budgerigars (55.5%), the African gray parrot (33.3%), and nymphs (34.3%). The prevalence of M. ornithogaster in canaries was 10%. The infection was detected in 31% of the examined birds, which shows that the occurrence of M. ornithogaster in exotic birds is common. No clinical signs were observed in the vast majority of birds that tested positive for gastric yeast.
Go to article

Bibliography

1. Amer MM, Mekky HM (2020) Avian gastric yeast (AGY) infection (macrorhabdosis or megabacteriosis). Bulg J Vet Med 23: 397-410.
2. Antinoff N (2004) Diagnosis and treatment options for mega-bacteria (Macrorhabdus ornithogaster). J Avian Med Surg 18: 189-195.
3. Baron HR, Stevenson BC, Phalen DN (2021) Comparison of In-Clinic Diagnostic Testing Methods for Macrorhabdus ornithogaster. J Avian Med Surg 35: 37-44.
4. Behnke EL, Fletcher OJ (2011) Macrorhabdus ornithogaster (megabacterium) infection in adult hobby chickens in North America. Avian Dis 55: 331-334.
5. Herck H, Duijser T, Zwart P, Dorrestein GM, Buitelaar M, Van Der Hage MH (1984) A bacterial proventriculitis in canaries (Serinus canaria). Avian Pathol 13: 561-572.
6. Lanzarot P, Blanco JL, Alvarez-Perez S, Abad C, Cutuli MT, Garcia ME (2013) Prolonged fecal shedding of ‘megabacteria’ (Macrorhabdus ornithogaster) by clinically healthy canaries (Serinus canaria). Med Mycol 51: 888-889.
7. Marlier D, Leroy C, Sturbois M, Delleur V, Poulipoulis A, Vindevogel H (2006) Increasing incidence of megabacteriosis in canaries (Serinus canarius domesticus). Vet J 172: 549-552.
8. Phalen DN (2014): Update on the diagnosis and management of Macrorhabdus ornithogaster (formerly Megabacteria) in avian patients. Vet Clin North Am Exot Anim Pract, 17: 203-210.
9. Piasecki T, Prochowska S, Celmer Z, Sochacka A, Bednarski M (2012) Occurrence of Macrorhabdus ornithogaster in exotic and wild birds in Poland. Med Weter 68: 245-249.
10. Powers LV, Mitchell MA, Garner MM (2019) Macrorhabdus ornithogaster Infection and Spontaneous Proventricular Adenocarcinoma in Budgerigars (Melopsittacus undulatus). Vet Pathol 56: 486-493.
11. Scanlan CM, Graham DL (1990) Characterization of a grampositive bacterium from the proventriculus of budgerigars (Melopsittacus undulatus). Avian Dis 34: 779-786.
12. Sullivan PJ, Ramsay EC, Greenacre CB, Cushing AC, Zhu X, Jones MP (2017) Comparison of two methods for determining prevalence of Macrorhabdus ornithogaster in the flock of captive Budgerigars (Melopsittacus undulatus). J Avian Med Surg 31: 128-131.
13. Tsai SS, Park JH, Hirai K, Itakura C (1992) Catarrhal proventriculitis associated with a filamentous organism in pet birds. Jpn J Vet Res 40: 143-148.
Go to article

Authors and Affiliations

B. Blagojević
1
I. Davidov
1
A. Galfi Vukomanović
1
D. Tekić
4
M. Došenović Marinković
2
V. Vidović
3

  1. Department of Veterinary Medicine, Faculty of Agriculture, University of Novi Sad, Trg Dositeja Obradovića 8, Novi Sad 21000, Serbia
  2. Academy of Applied Studies Šabac, Dobropoljska 5, 15000 Šabac, Serbia
  3. Department of Medical Oncology, Oncology Institute of Vojvodina, Put doktora Goldmana 4, Sremska Kamenica, Serbia
  4. Department of Agricultural Economics and Rural Sociology, Faculty of Agriculture, University of Novi Sad, Trg Dositeja Obradovića 8, Novi Sad 21000, Serbia
Download PDF Download RIS Download Bibtex

Abstract

Porcine epidemic diarrhea (PED) is a disease extremely harmful to pig health. Intramuscular and Houhai acupoint injections are the main immunization routes to prevent and control PED. This study aimed to evaluate the efficacy of these two routes in pregnant sows based on serum IgG, IgA, and neutralizing antibody levels. PED virus (PEDV) immunoprophylaxis with live-attenuated and inactivated vaccines was administered. The vaccinations for the intramuscular injections elevated IgG and neutralizing antibody levels more than Houhai acupoint injections at most timepoints after immunization. However, the anti-PEDV IgA antibodies induced by vaccination with the two immunization routes did not differ significantly. In conclusion, intramuscular injections are better than Houhai acupoint injections for PEDV vaccination of pregnant sows.
Go to article

Bibliography

1. Brown J, Poonsuk K, Cheng TY, Rademacher C, Kalkwarf E, Tian L, McKeen LA, Wang C, Gimenez-Lirola L, Baum D, Karriker LA (2023) Comparison of two diagnostic assays for the detection of serum neutralizing antibody to porcine epidemic diarrhea virus. Animals (Basel) 13:757.
2. Hsueh FC, Chang YC, Kao CF, Hsu CW, Chang HW (2020) Intramuscular immunization with chemokine-adjuvanted inactive porcine epidemic diarrhea virus induces substantial protection in pigs. Vaccines (Basel) 8:102.
3. Jin H, Wu Y, Bi S, Xu Y, Shi F, Li X, Ma X, Hu S (2020) Higher immune response induced by vaccination in Houhai acupoint relates to the lymphatic drainage of the injection site. Res Vet Sci 130: 230-236.
4. Jung K, Saif LJ, Wang Q (2020) Porcine epidemic diarrhea virus (PEDV): An update on etiology, transmission, pathogenesis, and pre-vention and control. Virus Res 286: 198045.
5. Krishna VD, Kim Y, Yang M, Vannucci F, Molitor T, Torremorell M, Cheeran MC (2020) Immune responses to porcine epidemic diar-rhea virus (PEDV) in swine and protection against subsequent infection. PLoS One 15: e0231723.
6. Langel SN, Paim FC, Alhamo MA, Buckley A, Van Geelen A, Lager KM, Vlasova AN, Saif LJ (2019) Stage of gestation at porcine epidemic diarrhea virus infection of pregnant swine impacts maternal immunity and lactogenic immune protection of neonatal suckling piglets. Front Immunol 10: 727.
7. Lee C (2015) Porcine epidemic diarrhea virus: an emerging and re-emerging epizootic swine virus. Virol J 12: 193.
8. Lv C, Xiao Y, Li X, Tian K (2016) Porcine epidemic diarrhea virus: current insights. Virus Adapt Treat 8: 1-12.
9. Shibata I, Tsuda T, Mori M, Ono M, Sueyoshi M, Uruno K (2000) Isolation of porcine epidemic diarrhea virus in porcine cell cultures and experimental infection of pigs of different ages. Vet Microbiol 72: 173-182.
10. Sun D, Wang X, Wei S, Chen J, Feng L (2016) Epidemiology and vaccine of porcine epidemic diarrhea virus in China: a mini-review. J Vet Med Sci 78: 355-363.
11. Xu W, Hu S (2021) Administration of infectious bursal disease vaccine in Houhai acupoint promotes robust immune responses in chickens. Res Vet Sci 142: 149-153.
Go to article

Authors and Affiliations

C. Hu
1
X. Xie
2
D. Zhao
3
H. Liu
1
ORCID: ORCID
X. Liu
4
T. Yang
5
W. Sun
6

  1. Pulike Biological Engineering Inc., Luoyang, Henan, 471000, China
  2. Yiyang Vocational and Technical College, Yiyang, Hunan, 413055, China
  3. College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, 410128, China
  4. Xiangtan Center for Animal Disease Prevention and Control, Xiangtan, Hunan, 411104, China
  5. College of Life Sciences and Resource Environment, Yichun University, Yichun, Jiangxi, 336000, China
  6. Sinopharm Animal Health Corporation Ltd., Wuhan, Hubei, 430075, China
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to investigate the effect of different extenders on the post-thaw (PT) quality of sperm originating from the sperm-rich fraction (SRF) and post-sperm-rich fraction (PSRF) of boar ejaculate. Motility and velocity parameters, analyzed using a computer-assisted semen analysis (CASA) system, and membrane integrity parameters were markedly higher in frozen-thawed (FT) spermatozoa of the SRF in both the Belstville Thawing Solution (BTS) and Androhep Plus (AHP) extenders, irrespective of the post-thaw (PT) storage time. Furthermore, reduced cryo-survival was more marked in FT spermatozoa of the PSRF in both extenders following storage for 60 min. It was found that the SRF-stored samples in the AHP extender for 60 min exhibited significantly higher percentages of spermatozoa with total motility, mitochondrial function and acrosome integrity than those stored in the BTS extender. The findings of this study confirm that components of the ejaculate fractions and extender have varying effects on the cryo-survival of boar spermatozoa.
Go to article

Bibliography

1. Aquila S, Giordano F, Guido C, Rago V, Carpino A (2011) Nitric oxide involvement in the acrosome reaction triggered by leptin in pig sperm. Reprod Biol Endocrinol 9: 133.
2. Fraser L, Strzeżek J (2007) Effect of different procedures of ejaculate collection, extenders and packages on DNA integrity of boar sper-matozoa following freezing-thawing. Anim Reprod Sci 99: 317-329.
3. Kaeoket K, Chanapai P, Junchiyaphoom P, Chanapiwat P (2011) The effect of using long term and short term extenders during cooling process on the quality of frozen boar semen. Thai J Vet Med 41: 283-288.
4. Rodríguez-Martínez H, Martínez EA, Calvete JJ, Peña Vega FJ, Roca J (2021) Seminal plasma: relevant for fertility? Int J Mol Sci 22: 4368.
5. Rodríguez-Martínez H, Saravia F, Wallgren M, Roca J, Peña FJ (2008) Influence of seminal plasma on the kinematics of boar sperma-tozoa during freezing. Theriogenology 70: 1242-1250.
6. Saravia F, Wallgren M, Johannisson A, Calvete JJ, Sanz L, Pena FJ, Roca J, Rodríguez-Martínez H (2009) Exposure to the seminal plasma of different portions of the boar ejaculate modulates the survival of spermatozoa cryopreserved in MiniFlatPacks. Theriogenology 71: 662-675.
7. Thomas CA, Garner DL, DeJarnette JM, Marshall CE (1998) Effect of cryopreservation of bovine sperm organelle function and viability as determined by flow cytometry. Biol Reprod 58: 786-793.
8. Wasilewska K, Fraser L (2017) Boar variability in sperm cryo-tolerance after cooling of semen in different longterm extenders at various temperatures. Anim Reprod Sci 185: 161-173.
9. Wasilewska-Sakowska K, Zasiadczyk Ł, Fraser L (2019) Effect of fractionated seminal plasma on sperm characteristics following cryo-preservation of boar semen. Ann Anim Sci 19: 695-712.
10. Weitze KF (2014) Benefits of AndrohepPlus and AndrostarPlus long-term extenders for boar semen. (Minitüb Gmbh) Technical Report 5: 1-6.
11. Yeste M (2016) Sperm cryopreservation update: cryodamage, markers, and factors affecting the sperm freezability in pigs. Theriogenol-ogy 85: 47-64.
Go to article

Authors and Affiliations

Ł. Zasiadczyk
1
K. Kurpanik
1
L. Fraser
1
W. Kordan
1

  1. Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-719 Olsztyn, Poland
Download PDF Download RIS Download Bibtex

Abstract

Raw meat-based diets for pet nutrition are becoming increasingly popular. The percentage of meat content, composition of nutrients, and amount of additives started to play an important role in the recipe of a given food. However, the use of healthier and unprocessed food must also be balanced with the animal’s specific needs based on its anatomy, physiology, and behavior. There are many potential advantages and disadvantages of a biologically appropriate raw food (BARF) diet, and all of them should be considered before switching to this approach. Raw meat is considered a diet closest to nature and least processed. However, raw diets threaten pet health because of the potential for nutrient imbalances. The choice of raw meat in pets’ everyday diet should be supported by the veterinarian’s medical decision and preferably also with nutritionist help. Growing animals require a specific Ca:P ratio in their diet, which may be improper in raw meat. For cats, taurine levels must be carefully checked. In addition, an imbalanced raw-meat diet can be the cause of poor semen quality in males. Females are prone to inhibition of the estrus cycle, especially due to hyperthyroidism. Exogenous thyroid hormone intake is a real concern when feeding a neck/head meat with thyroid glands. There is also a possibility of bacterial or parasitic presence in raw meat. The present paper aims to summarize the current state of knowledge about the benefits and threats of eating a raw meat diet for the health concerns of companion animals.
Go to article

Bibliography

1. Ahsan U, Kamran Z, Raza I, Ahmad S, Babar W, Riaz MH, Iqbal Z (2014) Role of selenium in male reproduction – a review. Anim Re-prod Sci 146: 55-62.
2. Alessandri G, Milani C, Mancabelli L, Mangifesta M, Lugli GA, Viappiani A, Duranti S, Turroni F, Ossiprandi MC, van Sinderen D, Ventura M (2019) Metagenomic dissection of the canine gut microbiota: insights into taxonomic, metabolic and nutritional features. En-viron Microbiol 21: 1331-1343.
3. Anderson RC, Armstrong KM, Young W, Maclean P, Thomas DG, Bermingham EN (2018) Effect of kibble and raw meat diets on pe-ripheral blood mononuclear cell gene expression profile in dogs. Vet J 234: 7-10.
4. Axelsson E, Ratnakumar A, Arendt ML, Maqbool K, Webster MT, Perloski M, Liberg O, Arnemo JM, Hedhammar Å, Lindblad-Toh K (2013) The genomic signature of dog domestication reveals adaptation to a starch-rich diet. Nature 495: 360-364.
5. Bilezikian JP, Bandeira L, Khan A, Cusano NE (2018) Hyperparathyroidism. Lancet 391: 168-178.
6. Brozić D, Mikulec Ž, Samardžija M, Đuričić D, Valpotić H (2020) Raw meat-based diet (BARF) in dogs and cats nutrition. Vet J Rep Srp 19: 314-321.
7. Buff PR, Carter RA, Bauer JE, Kersey JH (2014) Natural pet food: a review of natural diets and their impact on canine and feline physi-ology. J Anim Sci 92: 3781-3791.
8. Cornelissen S, De Roover K, Paepe D, Hesta M, Van Der Meulen E, Daminet S (2014) Dietary hyperthyroidism in a rottweiler. Vlaams Diergeneeskundig Tijdschrift 83: 306-311.
9. Crissey SD, Swanson JA, Lintzenich BA, Brewer BA, Slifka KA (1997) Use of a raw meat-based diet or a dry kibble diet for sand cats (Felis margarita). J Anim Sci 75: 2154-2160.
10. Damodaran S (1996) Amino acids, peptides and proteins. In: Fennema RO (ed) Food Chemistry, 3rd ed., CRC Press, New York, pp 321-416 .
11. Davies RH, Lawes JR, Wales AD (2019) Raw diets for dogs and cats: a review, with particular reference to microbiological hazards. J Small Anim Pract 60: 329-339.
12. Dillitzer N, Becker N, Kienzle E (2011) Intake of minerals, trace elements and vitamins in bone and raw food rations in adult dogs. Br J Nutr 106 (Suppl 1): S53-56.
13. Di Cerbo A, Morales-Medina JC, Palmieri B, Pezzuto F, Cocco R, Flores G, Iannitti T (2017) Functional foods in pet nutrition: focus on dogs and cats. Res Vet Sci 112: 161-166.
14. Domosławska A, Zdunczyk S, Franczyk M, Kankofer M, Janowski T (2018) Selenium and vitamin E supplementation enhances the an-tioxidant status of spermatozoa and improves semen quality in male dogs with lowered fertility. Andrologia 50: e13023.
15. Domosławska A, Zduńczyk S, Nizański W, Jurczak A, Janowski T (2015) Effect of selenium and vitamin E supplementation on semen quality in dogs with lowered fertility. Bull Vet Inst Pulawy 59: 85-90.
16. Dyachenko V, Pantchev N, Gawlowska S, Vrhovec MG, Bauer C (2008) Echinococcus multilocularis infections in domestic dogs and cats from Germany and other European countries. Vet Parasitol 157: 244-253.
17. Empert-Gallegos A, Hill S, Yam PS (2020) Insights into dog owner perspectives on risks, benefits, and nutritional value of raw diets compared to commercial cooked diets. PeerJ 8: e10383.
18. Fleischer S, Sharkey M, Mealey K, Ostrander EA, Martinez M (2008) Pharmacogenetic and metabolic differences between dog breeds: their impact on canine medicine and the use of the dog as a preclinical animal model. AAPS J 10: 110-119.
19. Frantz LA, Bradley DG, Larson G, Orlando L (2020) Animal domestication in the era of ancient genomics. Nat Rev Gene 21: 449-460.
20. Freeman LM, Chandler ML, Hamper BA, Weeth LP (2013) Current knowledge about the risks and benefits of raw meat-based diets for dogs and cats. J Am Vet Med Assoc 243: 1549-1558.
21. Hamper BA (2014) Raw meat-based diets: current evidence regarding benefits and risks. In: Purina Companion Animal Nutrition Sum-mit: Nutrition for Life, Austin, pp 99-107.
22. Hielm-Björkman A, Virtanen J (2013) Exploratory study: 632 shared experiences from dog owners changing their dogs’ food to a raw food (barf) diet. Faculty of Veterinary Medicine, Department of Equine and Small Animal Medicine, University of Helsinki, Finland 206: 2-3.
23. Howard J, Allen ME (2008) Nutritional factors affecting semen quality in felids. In: Fowler ME, Miller RE (eds) Zoo and Wild Animal Medicine: Current Therapy VI. Saunders, Elsevier Science, pp 272-283.
24. Huang HF, Dyrenfurth I, Hembree WC (1983) Endocrine changes associated with germ cell loss during vitamin A-induced recovery of spermatogenesis. Endocrinology 112: 1163-1171.
25. Johnston SD (1991) Clinical approach to infertility in bitches with primary anestrus. Vet Clin North Am Small Anim Pract 21: 421-425.
26. Kawaguchi K, Braga I, Takahashi A, Ochiai K, Itakura C (1993) Nutritional secondary hyperparathyroidism occurring in a strain of German shepherd puppies. Jpn J Vet Res 41: 89-96.
27. Kawakami E, Kobayashi M, Hori T, Kaneda T (2016) Therapeutic effects of vitamin E supplementation in 4 dogs with poor semen qual-ity and low superoxide dismutase activity in seminal plasma. J Vet Med Sci 77: 1711-1714.
28. Kerr KR, Beloshapka AN, Morris CL, Parsons CM, Burke SL, Utterback PL, Swanson KS (2013) Evaluation of four raw meat diets using domestic cats, captive exotic felids, and cecectomized roosters. J Anim Sci 91: 225-237.
29. Kienzle E (1993) Carbohydrate metabolism of the cat 1. Activity of amylase in the gastrointestinal tract of the cat. J Anim Physiol Animal Nutr 69: 91-101
30. Knize MG, Salmon CP, Felton JS (2003) Mutagenic activity and heterocyclic amine carcinogens in commercial pet foods. Mutat Res 539: 195-201.
31. Köhler B, Stengel C, Neiger-Casas R (2012). Dietary hyperthyroidism in dogs. J Small Anim Pract 53: 182-184.
32. Lawler DF, Bebiak DM (1986) Nutrition and management of reproduction in the cat. Vet Clin North Am Small Anim Pract 16: 495-519.
33. Lejeune JT, Hancock DD (2001) Public health concerns associated with feeding raw meat diets to dogs. J Am Vet Assoc 219: 1222-1225.
34. Loughrill E, Wray D, Christides T, Zand N (2017) Calcium to phosphorus ratio, essential elements and vitamin D content of infant foods in the UK: possible implications for bone health. Matern Child Nutr 13: e12368.
35. Morelli G, Bastianello S, Catellani P, Ricci R (2019) Raw meat-based diets for dogs: survey of owners’ motivations, attitudes and prac-tices. BMC Vet Res 15: 74.
36. Morris JG (2002) Idiosyncratic nutrient requirements of cats appear to be diet-induced evolutionary adaptations. Nutr Res Rev 15: 153-168.
37. Oba PM, Utterback PL, Parsons CM, Templeman JR, Swanson KS (2023) Standardized amino acid digestibility and nitrogen-corrected true metabolizable energy of frozen and freeze-dried raw dog foods using precision-fed cecectomized and conventional rooster assays. J Anim Sci 101: skad311.
38. Oswald H, Sharkey M, Pade D, Martinez MN (2015) Canine gastrointestinal physiology: breeds variations that can influence drug ab-sorption. Eur J Pharm Biopharm 97: 192-203.
39. Overgaauw PA (2020) Parasite risks from raw meat-based diets for companion animals. Comp Anim 25: 261-267.
40. Owens TJ, Fascetti AJ, Calvert CC, Larsen JA (2021) Rabbit carcasses for use in feline diets: amino acid concentrations in fresh and frozen carcasses with and without gastrointestinal tracts. Front Vet Sci 7: 592753.
41. Rampelli S, Turroni S, Debandi F, Alberdi A, Schnorr SL, Hofman CA, Taddia A, Helg R, Biagi E, Brigidi P, D’Amico F, Cattani M, Candela M (2021) The gut microbiome buffers dietary adaptation in bronze age domesticated dogs. iScience 24: 102816.
42. Schuller-Levis G, Mehta PD, Rudelli R Sturman J (1990) immunologic consequences of taurine deficiency in cats. J Leukoc Biol 47: 321-331.
43. Sontas HB, Schwendenwein I, Schäfer-Somi S (2014) Primary anestrus due to dietary hyperthyroidism in a miniature pinscher bitch. Can Vet J 55: 6-7.
44. Spitze AR, Wong DL, Rogers QR, Fascetti AJ (2003) Taurine concentrations in animal feed ingredients; cooking influences taurine con-tent. J Anim Physiol Anim Nutr 87: 7-8.
45. Strohmeyer RA, Morley PS, Hyatt DR, Dargatz DA, Scorza AV, Lappin MR (2006). Evaluation of bacterial and protozoal contamina-tion of commercially available raw meat diets for dogs. J Am Vet Med Assoc 228: 537-542.
46. Sturman JA, Moretz RC, French JH, Wisniewski HM (1985) Taurine deficiency in the developing cat: persistence of the cerebellar ex-ternal granule cell layer. J Neurosci Res 13: 405-416.
47. Sugimura T, Wakabayashi K, Nakagama H, Nagao M (2004) Heterocyclic amines: mutagens/carcinogens produced during cooking of meet and fish. Cancer Sci 95: 290-299.
48. van Bree FP, Bokken GC, Mineur R, Franssen F, Opsteegh M, van der Giessen JW, Lipman LJ, Overgaauw PA (2018) Zoonotic bacte-ria and parasites found in raw meat-based diets for cats and dogs. Vet Rec 182: 50.
49. Verbrugghe A, Bakovic M (2013) Peculiarities of one-carbon metabolism in the strict carnivorous cat and the role in feline hepatic lipid-osis. Nutrients 5: 2811-2835.
50. Verbrugghe A, Hesta M (2017) Cats and carbohydrates: the carnivore fantasy? Vet Sci 4: 4: 55.
51. Westermarck E (1987) Treatment of pancreatic degenerative atrophy with raw pancreas homogenate and various enzyme preparations. Zentralb Veterinarmed A 34: 728-73
Go to article

Authors and Affiliations

D. Główny
1
N. Sowińska
2 4
A. Cieślak
3
M. Gogulski
4 5
K. Konieczny
1
M. Szumacher-Strabel
3

  1. Poznan University of Life Sciences, Faculty of Veterinary Medicine and Animal Science, Department of Internal Diseases and Diagnostics, Poznan, Poland
  2. Poznan University of Life Sciences, Faculty of Veterinary Medicine and Animal Science, Department of Genetics and Animal Breeding, Wołynska 33, 60-637 Poznan, Poland
  3. Poznan University of Life Sciences, Faculty of Veterinary Medicine and Animal Science, Department of Animal Nutrition, Wołynska 33, 60-637 Poznan, Poland
  4. Poznan University of Life Sciences, Faculty of Veterinary Medicine and Animal Science, University Centre of Veterinary Medicine, Poznan, Poland
  5. Poznan University of Life Sciences, Faculty of Veterinary Medicine and Animal Science, Department of Preclinical Sciences and Infectious Diseases, Poznan, Poland

This page uses 'cookies'. Learn more