Life Sciences and Agriculture

Polish Journal of Veterinary Sciences

Content

Polish Journal of Veterinary Sciences | 2023 | vol. 26 | No 3

Download PDF Download RIS Download Bibtex

Abstract

Neonatal calf diarrhea (NCD) is one of the most important concerns in cattle production. Escherichia coli is the most important bacterial agent of NCD. Although vaccination and antibiotic treatment are common in NCD, the high antigenic diversity of E. coli and the increase in antibiotic resistance cause difficulties in the control. The study aimed to investigate the rate of E. coli in calf diarrhea, isolate an agent of the NCD E. coli strain, determine antimicrobial resistance, and find out about some surface antigens. Fecal samples (n=115) were analyzed to isolate pathogenic E. coli strains with nine mixed infections; sixty-one strains isolate from fifty diarrhoeic calves. Among the isolates from diseased animals, 22 K99+STa+F41, 3 K99+STa, 3 strains F41, 2 strains Stx1, one strain K99, one strain eae, and one strain Stx2+eae were detected. 27 strains of F17- associated fimbriae have been identified. 17 strains F17a, 6 strains F111, 3 strains F17c, one strain carrying the F17a and F17c gene regions, whereas subfamily typing of one strain could not be performed. Serotypes were determined by molecular and serological methods: 32/61 (52.5%) isolates were O101 and 2/61 (3.3%) isolates were O9 serotypes. But 27 strain serotypes could not be detected. The antibiotic resistance profiles of the isolates were determined by the disc diffusion method. The resistance rates to antibiotics were trimethoprim- sulphamethoxazole 91.7%, ampicillin 86.7%, enrofloxacin 86.7%, gentamicin 45%, tobramycin 41.7%, cefotaxime 3.3%, and ceftazidime 1.7%. Due to increasing antibiotic resistance, prophylaxis is gaining importance. In further research, E. coli surface antigenic structures should be examined in detail, and it should form the basis for vaccine and hyperimmunization studies to be developed.
Go to article

Bibliography

  1. Algammal AM, El-Kholy AW, Riad EM, Mohamed HE, Elhaig MM, Yousef SaA, Hozzein WN, Ghobashy MOI (2020) Genes Encoding the Virulence and the Antimicrobial Resistance in Enterotoxigenic and Shiga-Toxigenic E. coli Isolated from Diarrheic Calves. Toxins 12: 383.
  2. Andersson DI, Hughes D (2011) Persistence of antibiotic resistance in bacterial populations. FEMS Microbiol Rev 35: 901-911.
  3. Bendali F, Bichet H, Schelcher F, Sanaa M (1999) Pattern of diarrhoea in newborn beef calves in south-west France. Vet Res 30: 61-74.
  4. Bertin Y, Martin C, Oswald E, Girardeau J-P (1996) Rapid and specific detection of F17-related pilin and adhesin genes in diarrheic and septicemic Escherichia coli strains by multiplex PCR. J Clinic Microbiol 34: 2921-2928.
  5. Bielaszewska M, Schmidt H, Liesegang A, Prager R, Rabsch W, TschäPe H, Cízek A, Janda J, Bláhová K, Karch H (2000) Cattle can be a reservoir of sorbitol-fermenting Shiga toxin-producing Escherichia coli O157: H− strains and a source of human diseases. J Clin Microbiol 38: 3470-3473.
  6. Boucher HW, Talbot GH, Bradley JS, Edwards JE, Gilbert D, Rice LB, Scheld M, Spellberg B, Bartlett J (2009) Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis 48: 1-12.
  7. Caffarena RD, Casaux ML, Schild CO, Fraga M, Castells M, Colina R, Maya L, Corbellini LG, Riet-Correa F, Giannitti F (2021) Causes of neonatal calf diarrhea and mortality in pasture-based dairy herds in Uruguay: a farm-matched case-control study. Braz J Microbiol 52: 977-988.
  8. Cengiz S, Adiguzel MC (2020) Determination of virulence factors and antimicrobial resistance of E. coli isolated from calf diarrhea, part of eastern Turkey. Ankara Univ Vet Fak Derg 67: 365-371.
  9. Cho YI, Yoon KJ (2014) An overview of calf diarrhea - infectious etiology, diagnosis, and intervention. J Vet Sci 15: 1-17.
  10. Coura FM, Freitas MD, Ribeiro J, De Leme RA, De Souza C, Alfieri AA, Facury Filho EJ, De Carvalho AU, Silva MX, Lage AP, Heinemann MB (2015) Longitudinal study of Salmonella spp., diarrheagenic Escherichia coli, Rotavirus, and Coronavirus isolated from healthy and diarrheic calves in a Brazilian dairy herd. Trop Anim Health Prod 47: 3-11.
  11. Debroy C, Maddox C (2001) Identification of virulence attributes of gastrointestinal Escherichia coli isolates of veterinary significanc. Anim Health Res Rev 1: 12.
  12. Donovan GA, Dohoo R, Montgomery DM, Bennett FL (1998) Calf and disease factors affecting growth in female Holstein calves in Florida, USA. Prev Vet Med 33: 10.
  13. Dubreuil JD, Isaacson RE, Schifferli DM (2016) Animal Enterotoxigenic Escherichia coli. EcoSal Plus 7: 47.
  14. Fidock DA, Mcnicholas PA, Lehrbach PR (1989) Nucleotide sequence of the F41 fimbriae subunit gene in Escherichia coli B41. Nucleic Acids Res 17: 2849.
  15. Foster DM, Smith GW (2009) Pathophysiology of diarrhea in calves. Vet Clin North Am Food Anim Pract 25: 13-36, xi.
  16. Franck SM, Bosworth BT, Moon HW (1998) Multiplex PCR for enterotoxigenic, attaching and effacing, and Shiga toxin-producing Escherichia coli strains from calves. J Clini Microbiol 36: 1795-1797.
  17. Guler L, Gunduz K, Ok U (2008) Virulence factors and antimicrobial susceptibility of Escherichia coli isolated from calves in Turkey. Zoonoses Public Health 55: 249-257.
  18. Gulliksen SM, Lie KI, Loken T, Osteras O (2009) Calf mortality in Norwegian dairy herds. J Dairy Sci 92: 2782-2795.
  19. Hang BPT, Wredle E, Börjesson S, Sjaunja KS, Dicksved J, Duse A (2019) High level of multidrug-resistant Escherichia coli in young dairy calves in southern Vietnam. Trop Anim Health Prod 51: 1405-1411.
  20. Hur T-Y, Jung Y-H, Choe C-Y, Cho Y-I, Kang S-J, Lee H-J, Ki K-S, Baek K-S, Suh G-H (2013) The dairy calf mortality: the causes of calf death during ten years at a large dairy farm in Korea. Korean J Vet Res 53: 103-108.
  21. Iguchi A, Iyoda S, Seto K, Morita-Ishihara T, Scheutz F, Ohnishi M (2015) Escherichia coli O-genotyping PCR: a comprehensive and practical platform for molecular O serogrouping. J Clini Microbiol 53: 2427-2432.
  22. Jerse AE, Yu J, Tall BD, Kaper JB (1990) A genetic locus of enteropathogenic Escherichia coli necessary for the production of attaching and effacing lesions on tissue culture cells. Proc Nat Acad Sci 87: 7839-7843.
  23. Mohammed S, Marouf S, Erfana AM, El JK, Hessain AM, Dawoud TM, Kabli SA, Moussa IM (2019) Risk factors associated with E. coli causing neonatal calf diarrhea. Saudi J Bio Sci 26: 1084-1088.
  24. Paton AW, Beutin L, Paton JC (1995) Heterogeneity of the amino-acid sequences of Escherichia coli Shiga-like toxin type-I operons. Gene 153: 71-74.
  25. Paton AW, Paton JC, Manning PA (1993) Polymerase chain reaction amplification, cloning and sequencing of variant Escherichia coli Shiga-like toxin type II operons. Microb Pathog 15: 77-82.
  26. Pervez A, Anjum FR, Bukhari AA, Anam S, S. R, Arshad MI (2018) Isolation and virulence genes characterization of diarrheagenic Escherichia coli from calves. Pak Vet J 38: 5.
  27. Prieto A, Lopez-Novo C, Diaz P, Diaz-Cao JM, Lopez-Lorenzo G, Anton C, Remesar S, Garcia-Dios D, Lopez C, Panadero R, Diez-Banos P, Morrondo P, Fernandez G (2022) Antimicrobial susceptibility of enterotoxigenic Escherichia coli from diarrhoeic neonatal calves in Spain. Animals (Basel) 12: 12.
  28. Roosendaal B, Gaastra W, De Graaf FK (1984) The nucleotide sequence of the gene encoding the K99 subunit of enterotoxigenic Escherichia coli. FEMS Microbiol Lett 22: 253-258.
  29. Sekizaki T, Akashi H, Terakado N (1985) Nucleotide sequences of the genes for Escherichia coli heat-stable enterotoxin I of bovine, avian, and porcine origins. Am J Vet Res 46: 909-912.
  30. Shahrani M, Dehkordi FS, Momtaz H (2014) Characterization of Escherichia coli virulence genes, pathotypes and antibiotic resistance properties in diarrheic calves in Iran. Biol Res 47: 13.
  31. Smith DR (2012) Field disease diagnostic investigation of neonatal calf diarrhea. Vet Clin North Am Food Anim Pract 28: 465-481.
  32. Thiry D, Saulmont M, Takaki S, De Rauw K, Duprez J-N, Iguchi A, Piérard D, Mainil JG (2017) Enteropathogenic Escherichia coli O80: H2 in young calves with diarrhea, Belgium. Emerg Infect Dis 23: 3.
  33. To S (1984) F41 antigen among porcine enterotoxigenic Escherichia coli strains lacking K88, K99, and 987P pili. Infect Immun 43: 549-554.
  34. Umpiérrez A, Acquistapace S, Fernández S, Oliver M, Acuña P, Reolón E, Zunino P (2016) Prevalence of Escherichia coli adhesion-related genes in neonatal calf diarrhea in Uruguay. J Infect Dev Ctries 10: 472-477.
  35. Usda (2008) Dairy 2007, Part II: Changes in the U.S. Dairy Cattle Industry, 1991–2007. Fort Collins, CO: 57-60.
  36. Van Boeckel TP, Glennon EE, Chen D, Gilbert M, Robinson TP, Grenfell BT, Levin SA, Bonhoeffer S, Laxminarayan R (2017) Reducing antimicrobial use in food animals. Science 357: 1350-1352.
  37. Wani S, Bhat M, Samanta I, Nishikawa Y, Buchh A (2003) Isolation and characterization of Shiga toxin‐producing Escherichia coli (STEC) and enteropathogenic Escherichia coli (EPEC) from calves and lambs with diarrhoea in India. Lett Appl Microbiol 37: 121-126.
  38. Yu J, Kaper JB (1992) Cloning and characterization of the eae gene of enterohaemorrhagic Escherichia coli O157: H7. Mol Microbiol 6: 411-417.
  39. Yuyama Y, Yoshimatsu K, Ono E, Saito M, Naiki M (1993) Postnatal change of pig intestinal ganglioside bound by Escherichia coli with K 99 fimbriae. J Biochem 113: 488-492.
Go to article

Authors and Affiliations

M.R. Coşkun
1
M. Şahin
2

  1. Department of Microbiology, Faculty of Veterinary Medicine, Kafkas University, 36100, Kars, Turkey
  2. Department of Microbiology, Faculty of Veterinary Medicine, Kyrgyz-Turkish Manas University, 720038, Bishkek, Kyrgyzstan
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study is to determine the protective efficacy of anise in cerebral ischemia and reperfusion injury in rats. In this study, 28 Wistar Albino rats, weighing 250-300 grams (g), were used. Four groups were formed with 7 rats in each group. Group 1 (n=7): Control group, Group 2 (n=7): Anise group, 5 mL/kg/day of anise aqueous extract prepared according to Gamberini’s protocol was given orally by gavage for 30 days. Group 3 (n=7): Cerebral ischemia reperfusion (CIR) group, at the beginning of the experiment, 30 minutes of cerebral ischemia and 1 hour of reperfusion were induced and the animals were sacrificed by exanguination. Group 4 (n=7): Anise+ CIR group, After administering 30 days of anise’s aqueous extract, CIR was induced and the study was terminated. TOS values of the Anise+ CIR group was significantly lower than that of the CIR group (p<0.05). Il-6 and TNF-α values of the CIR group were significantly higher than the Anise+ CIR group (p<0,05). Our study revealed that anise ameliorates oxidative damage and inflammation due to cerebral ischemia/reperfusion, by reducing the levels of inflammatory cytokines (TNF-α, Il-6).
Go to article

Bibliography

  1. Cao Y, Mao X, Sun C, Zheng P, Gao J, Wang X, Min D, Sun H, Xie N, Cai J (2011) Baicalin attenuates global cerebral ischemia/ reperfusion injury in gerbils via antioxidative and anti-apoptotic pathways. Brain Res Bull 85: 396-402.
  2. Crack PJ, Taylor JM (2005) Reactive oxygen species and the modulation of stroke. Free Radic Biol Medi 38: 1433-1444
  3. Earnest MP, Yarnell PR, Merrill SL, Knapp GL (1980) Long-term survival and neurologic status after resuscitation from out-of-hospital cardiac arrest. Neurology 30: 1298-1302.
  4. Erel O (2004) A novel automated method to measure total antioxidant response against potent free radical reactions. Clin Biochem 37: 112-119.
  5. Erel O (2005) A new automated colorimetric method for measuring total oxidant status. Clin Biochem 38: 1103-1111.
  6. Gamberini MT, Rodrigues DS, Rodrigues D, Pontes VB (2015) Effects of the aqueous extract of Pimpinella anisum L. seeds on exploratory activ-ity and emotional behavior in rats using the open field and elevated plus maze tests. J. Ethnopharmacol 168: 45-49.
  7. Jeon YT, Hwang JW, Lim YJ, Park SK, Park HP (2013) Postischemic sevoflurane offers no additional neuroprotective benefit to preischemic dexmedetomidine. J Neurosurg Anesthesiol 25: 184-190.
  8. Johansson BB (2003) Environmental influence on recovery after brain lesions – experimental and clinical data. J Rehabil Med 41: 11-16.
  9. Kalogeris T, Baines C, Krenz M, Korthuis R (2012) Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol 298: 229-317.
  10. Kalogeris T, Baines CP, Krenz M, Korthuis RJ (2016) Ischemia/ Reperfusion. Compr Physiol 7: 113-170.
  11. Kucukkurt I, Avcı G, Eryavuz A, Bayram I, Cetingul IS, Akkaya AB, Uyarlar C (2009) Uyarlar effects of supplementation of aniseed (Pimpinella anisum L.) at various amounts to diets on lipid peroxidation, antioxidant activity and some biochemical parameters in laying quails (Coturnix coturnix japonica); Kocatepe Vet J 2: 1-5
  12. Lopalco A, Lopedota AA, Laquintana V, Denora N, Stella VJ (2020) Boric acid, a Lewis acid with unique and unusual properties: formulation implications. J Pharm Sci 109: 2375-2386.
  13. Lopez-Neblina F, Toledo AH, Toledo-Pereyna LH (2005) Molecular biology of apoptosis in ischemia and reperfusion. J Invest Surg. 18: 335-350.
  14. Özcan MM, Chalchat JC (2006) Chemical composition and antifungal effect of anise (Pimpinella anisum L.) fruit oil at ripening stage. Ann Micro-biol 56: 353-358.
  15. Rabuffetti M, Scioratti C, Tarozzo G, Clementi E, Manfredi AA, Beltramo M. (2000) Inhibition of Caspase-1-Like Activity by Ac-Tyr-Val-Ala-Asp-Chloromethyl Ketone Induces Long-Lasting Neuroprotection in Cerebral Ischemia through Apoptosis Reduction and Decrease of Proinflammatory Cytokines. J Neurosci 20: 4398-4404
  16. Selakovic V, Korenic A, Radenovic L (2011) Spatial and temporal patterns of oxidative stress in the brain of gerbils submitted to different duration of global cerebral ischemia. Int J Dev Neurosci 29: 645-654.
  17. Shojaii A, Fard MA (2012) Review of pharmacological properties and chemical constituents of Pimpinella anisum. ISRN Pharm. 2012; 2012: 510795.
  18. Sugawara T, Fujimura M, Noshita N, Kim GW, Saito A, Hayashi T, Narasimhan P, Maier CM, Chan PH (2004) Neuronal death/survival signaling pathways in cerebral ischemia. NeuroRx 1: 17-25
  19. Sun J, Li YZ, Ding YH, Wang J, Geng J, Yang H, Ren J, Tang JY, Gao J (2014) Neuroprotective effects of gallic acid against hypoxia/reoxygenation-induced mitochondrial dysfunctions in vitro and cerebral ischemia/reperfusion injury in vivo. Brain Res 1589: 126-139.
  20. Wu L, Tan JL, Wang ZH, Chen YX, Gao L, Liu JL, Shi YH, Endoh M, Yang HT (2015) ROS generated during early reperfusion contribute to intermittent hypobaric hypoxiaafforded cardioprotection against postischemia-induced Ca+2 overload and contractile dysfunction via the JAK2/ /STAT3 pathway. J Mol Cell Cardiol 81: 150-161.
Go to article

Authors and Affiliations

E. Yildizhan
1
H. Ozesmer
2
M.M. Inan
2
F. Tatli
2
M. Rencber
3
A. Akbas
2
M. Kamar
2
E. Gündüz
4
N. Baksi
5
B.V. Ulger
2
M. Akkuş
1
A. Kaydu
6

  1. Department of Histology and Embryology, Faculty of Medicine, Dicle University, 21280, Diyarbakır, Turkey
  2. Department of General Surgery, Faculty of Medicine, Dicle University, 21280, Diyarbakır, Turkey
  3. Department of General Surgery, Viransehir State Hospital, 63700, Viransehir, Şanlıurfa, Turkey
  4. Department of Emergency Medicine, Faculty of Medicine, Dicle University, 21280, Diyarbakır, Turkey
  5. Department of Laboratuary Animals, Faculty of Veterinary, Dicle University, 21280, Diyarbakır, Turkey
  6. Department of Anesthesia, Faculty of Medicine, Dicle University, 21280, Diyarbakır, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Neospora caninum ( N. caninum) is the etiologic agent of neosporosis, a potential cause of severe reproductive disorders in cattle, small ruminants, equines, wild animals and canids across the world. The current study is performed to estimate molecular prevalence of N. caninum in small ruminants and equines that had abortion in Kurdistan region of Iraq. A total of 64 tissue samples (brain, placenta, heart, lung and liver) were taken from aborted foetuses, with a total of 122 dam blood samples taken from 63 sheep, 39 goats, 12 mares and 8 jennies in local breed fields. Besides, a risk factor analysis for N. caninum positive animals was performed. The observed prevalence of N. caninum DNA in the blood of sheep, goats, horses and donkeys were 20.6%, 17.9%, 21.4% and 25.0%, respectively, and 19.3%, 17.6%, 18.1 and 20.0% in the aborted foetuses of the animals, respectively. Moreover, occurrence of N. caninum was 20.3% in the blood of aborted dams, while it was 18.7% in their aborted foetuses. Confirmatory analysis was also done through constructing a phylogenetic tree to compare the partial sequences of the Nc-5 gene in our isolates (OP771519, OP771520, OP771521 and OP771522) with the GenBank sequences. This showed 98-100% sequence identity with other N. caninum strains in the GenBank database. Older small ruminants and equines had a higher risk of being positive for N. caninum and exposure to dogs were considered as significant risk factors for N. caninum infection in the studied animals (p<0.05). Thus, the results of this study suggest that N. caninum is one of the microbial abortive agents in small ruminants and equines in Kurdistan region of Iraq. It is hoped that the results of this study will help to control animal abortion in livestock and reduce the economic losses.
Go to article

Bibliography

  1. Ahmed NE, Al-Akabway LM, Ramadan MY, El-Gawad SM, Moustafa MM (2017) Serological and PCR-sequencing assays for diagnosis of Toxoplasma gondii and Neospora caninum infecting camels in Egypt. Benha Vet Med J 33: 200-210.
  2. AL-Badrani BA, AL-Farwachi MI, AL-Hankawai OK (2012) Detection of Toxoplasma gondii and Neospora caninum antibodies in cattle in Mosul city, Iraq. Al-Qadisiyah J Vet Med Sci 11: 46-50.
  3. Al-Farwachi MI, Al-Badrani BA, Al-Khafaji WS (2018) Serodiagnosis of ovine neosporosis in Mosul city , Iraq. Eurasian J Vet Sci 28: 190-193.
  4. Al-Farwachi MI, Al-Hankawe OK, Al-Khafaji WS (2012) Serodiagnosis of Bovine Neosporosis in Mosul City, Iraq. Assiut Vet Med J 58: 1-4.
  5. Al-Shaeli SJ, Ethaeb AM, Gharban HA (2020) Molecular and histopathological identification of ovine neosporosis (Neospora caninum) in aborted ewes in Iraq. Vet World 13: 597-603.
  6. Almería S, Serrano-Perez B, Darwich L, Domingo M, Mur-Novales R, Regidor-Cerrillo J, Cabezón O, Pérez-Maillo M, Lopez-Helguera I, Fernández-Aguilar X, Puig-Ribas M, Ortega-Mora LM, García-Ispierto I, Dubey JP, López-Gatius F (2016) Foetal death in naive heifers inoculated with Neospora caninum isolate Nc-Spain7 at 110 days of pregnancy. Exp Parasitol 168: 62-69.
  7. Amdouni Y, Abedennebi I, Amairia S, Abdelkader A, Chandoul W, Gharbi M (2022) First molecular detection of Neospora caninum from naturally infected slaughtered camels in Tunisia. Vet Med Sci 8: 2241-2247.
  8. Amdouni Y, Rjeibi MR, Awadi S, Rekik M, Gharbi M (2018) First detection and molecular identification of Neospora caninum from naturally infected cattle and sheep in North Africa. Transbound Emerg Dis 65: 976-982.
  9. Anderson JA, Alves DA, Cerqueira-Cézar CK, da Silva AF, Murata FH, Norris JK, Howe DK, Dubey JP (2019) Histologically, immunohistochemically, ultrastructurally, and molecularly confirmed neosporosis abortion in an aborted equine fetus. Vet Parasitol 270: 20-24.
  10. Antonello AM, Pivoto FL, Camillo G, Braunig P, Sangioni LA, Pompermayer E, Vogel FS (2012) The importance of vertical transmission of Neospora sp. in naturally infected horses. Vet Parasitol 187: 367-370.
  11. Bartley PM, Guido S, Mason C, Stevenson H, Chianini F, Carty H, Innes EA, Katzer F (2019) Detection of Neospora caninum DNA in cases of bovine and ovine abortion in the South-West of Scotland. Parasitol 146: 979-982.
  12. Bártová E, Sedlák K, Kobédová K, Budíková M, Joel Atuman Y, Kamani J (2017) Seroprevalence and risk factors of Neospora spp. and Toxoplasma gondii infections among horses and donkeys in Nigeria, West Africa. Acta Parasitol 62: 606-609.
  13. Basso W, Holenweger F, Schares G, Müller N, Campero LM, Ardüser F, Moore-Jones G, Frey CF, Zanolari P (2022) Toxoplasma gondii and Neospora caninum infections in sheep and goats in Switzerland: Seroprevalence and occurrence in aborted foetuses. Food Waterborne Parasitol 28: e00176.
  14. Basso W, Venturini L, Venturini MC, Hill DE, Kwok OCH, Shen SK, Dubey JP (2001) First isolation of Neospora caninum from the feces of a naturally infected dog. J Parasitol 87: 612-618.
  15. Beck HP, Blake D, Dardé ML, Felger I, Pedraza-Díaz S, Regidor-Cerrillo J, Gómez-Bautista M, Ortega-Mora LM, Putignani L, Shiels B, Tait A, Weir W (2009) Molecular approaches to diversity of populations of apicomplexan parasites. Int J Parasitol 39: 175-189.
  16. Cong W, Nie LB, Qin SY, Wang WL, Qian AD, Meng QF (2018) Prevalence of Neospora spp. in donkeys in China. Parasite 25, 16.
  17. Costa RC, Mesquita LP, Nunes MV, Oliveira IM, Oliveira LF, Souza AR, Maiorka PC, Varaschin MS (2018) Neospora caninum bioassay in gerbils using placental tissues from naturally infected goats. Vet Parasitol 249: 70-73.
  18. Dahourou LD, Gbati OB, Savadogo M, Yougbare B, Dicko A, Combari AH, Kamga-Waladjo AR (2019) Prevalence of Toxoplasma gondii and Neospora caninum infections in households sheep “Elevage en case” in Dakar, Senegal. Vet World 12: 1028-1032.
  19. Diakou A, Papadopoulos E, Panousis N, Karatzias C, Giadinis N (2013) Toxoplasma gondii and Neospora caninum seroprevalence in dairy sheep and goats mixed stock farming. Vet Parasitol 198: 387-390.
  20. Della Rosa P, Fiorentino MA, Morrell EL, Scioli MV, Paolicchi FA, Moore DP, Cantón GJ, Hecker YP (2021) Neospora caninum and Toxoplasma gondii as causes of reproductive losses in commercial sheep flocks from Argentina. Curr Res Parasitol Vector Borne Dis 1: 100057.
  21. Duarte PC, Conrad PA, Barr BC, Wilson WD, Ferraro GL, Packham AE, Carpenter TE, Gardner IA (2004) Risk of transplacental transmission of Sarcocystis neurona and Neospora hughesi in California horses. J Parasitol 90: 1345-1351.
  22. Dubey JP (2003) Review of Neospora caninum and neosporosis in animals. Korean J Parasitol 41: 1-16.
  23. Dubey JP, Buxton D, Wouda W (2006) Pathogenesis of Bovine Neosporosis. J Comp Pathol 134: 267-289.
  24. Dubey JP, Hemphill A, Calero-Bernal R, Schares G (2017) Neosporosis in animals, 1st ed., CRC Press, Taylor and Francis group.
  25. Dubey JP, Jenkins MC, Ferreira LR, Choudhary S, Verma SK, Kwok OC, Fetterer R, Butler E, Carstensen M (2014) Isolation of viable Neospora caninum from brains of wild gray wolves (Canis lupus). Vet Parasitol 201: 150-153.
  26. Dubey JP, Schares G (2011) Neosporosis in animals-the last five years. Vet Parasitol 180: 90-108.
  27. Dubey JP, Schares G, Ortega-Mora LM (2007) Epidemiology and control of neosporosis and Neospora caninum. Clin Microbiol Rev 20: 323-367.
  28. Faraj AA, Ghattof HH (2018) Diagnosis of Neospora caninum using ELIZA and study of histopathological changes in dairy goat in Wasit province: Iraq. J Entom Zool Stud 6: 1256-1259.
  29. Gazzonis AL, Garcia GA, Zanzani SA, Ortega Mora LM, Invernizzi A, Manfredi MT (2016) Neospora caninum infection in sheep and goats from north-eastern Italy and associated risk factors. Small Rumin Res 140: 7-12.
  30. Gennari SM, Pena HF, Lindsay DS, Lopes MG, Soares HS, Cabral AD, Vitaliano SN, Amaku M (2016) Prevalence of antibodies against Neospora spp. and Sarcocystis neurona in donkeys from northeastern Brazil. Rev Bras Parasitol Vet 25: 109-111.
  31. Gharekhani J, Yakhchali M, Berahmat R (2020) Neospora caninum infection in Iran (2004-2020): A review. J Parasit Dis 44: 671-686.
  32. Gharekhani J, Yakhchali M, Heidari R (2022) Molecular detection and phylogenetic analysis of Neospora caninum in various hosts from Iran. Comp Immunol Microbiol Infect Dis 80: 101737.
  33. Ghattof HH, Faraj AA (2015) Seroprevalence of Neospora caninum in goats in Wasit province Iraq. Int J Curr Microbiol Appl Sci 4: 182-191.
  34. Gondim LF (2006) Neospora caninum in wildlife. Trends Parasitol 22: 247-252.
  35. Gondim LF, McAllister MM (2022) Experimental Neospora caninum Infection in Pregnant Cattle: Different Outcomes Between Inoculation With Tachyzoites and Oocysts. Front Vet Sci 9: 911015.
  36. González-Warleta M, Castro-Hermida JA, Regidor-Cerrillo J, Benavides J, Álvarez-García G, Fuertes M, Ortega-Mora LM, Mezo M (2014) Neospora caninum infection as a cause of reproductive failure in a sheep flock. Vet Res 45: 88.
  37. Irehan B, Sonmez A, Atalay MM, Ekinci AI, Celik F, Durmus N, Ciftci AT, Simsek S (2022) Investigation of Toxoplasma gondii, Neospora caninum and Tritrichomonas foetus in abortions of cattle, sheep and goats in Turkey: Analysis by real-time PCR, conventional PCR and histopathological methods. Comp Immunol Microbiol Infect Dis 89: 101867.
  38. Japa O, Morand S, Karnchanabanthoeng A, Chaisiri K, Ribas A (2018) Detection of Neospora caninum (Toxoplasmatidae) in wild small mammals from Thailand. Folia Parasitologica 65.
  39. King JS, Jenkins DJ, Ellis JT, Fleming P, Windsor PA, Šlapeta J (2011) Implications of wild dog ecology on the sylvatic and domestic life cycle of Neospora caninum in Australia. Vet J 188: 24-33.
  40. Langoni H, Greca HJ, Guimarães FF, Ullmann LS, Gaio FC, Uehara RS, Rosa EP, Amorim RM, Da Silva RC (2011) Serological profile of Toxoplasma gondii and Neospora caninum infection in commercial sheep from São Paulo State, Brazil. Vet Parasitol 177: 50-54.
  41. Locatelli-Dittrich R, Dittrich JR, Richartz RR, Gasino JM, Antunes J, Pinckney RD, Deconto I, Hoffmann DC, Thomaz-Soccol V (2006) Investigation of Neospora sp. and Toxoplasma gondii antibodies in mares and in precolostral foals from Parana State, Southern Brazil. Vet Parasitol 135: 215-221.
  42. Machačová T, Bártová E, Di Loria A, Sedlák K, Guccione J, Fulgione D, Veneziano V (2013) Seroprevalence and risk factors of Neospora spp. in donkeys from Southern Italy. Vet Parasitol 198: 201-204.
  43. Manca R, Ciccarese G, Scaltrito D, Chirizzi D (2022) Detection of Anti-Neospora caninum Antibodies on Dairy Cattle Farms in Southern Italy. Vet Sci 9: 87
  44. Marsh AE, Barr BC, Packham AE, Conrad PA (1998) Description of a new neospora species (Protozoa: apicomplexa: sarcocystidae). J Parasitol 5: 84-91.
  45. Mazuz ML, Mimoun L, Schvartz G, Tirosh-Levy S, Savitzki I, Edery N, Blum SE, Baneth G, Pusterla N, Steinman A (2020) Detection of Neospora caninum infection in aborted equine fetuses in Israel. Pathogenes 9: 1-11.
  46. Mendoza-Morales LF, Lagorio V, Corigliano MG, Sánchez-López E, Ramos-Duarte VA, Clemente M, Sander VA (2022) Neosporosis in sheep: A systematic review and meta-analysis of global seroprevalence and related risk factors. Acta Trop 233: 106569.
  47. Moore DP, de Yaniz MG, Odeón AC, Cano D, Leunda MR, Späth EA, Campero CM (2007) Serological evidence of Neospora caninum infections in goats from La Rioja Province, Argentina. Small Rumin Res 73: 256-258.
  48. Moreira TR, Sarturi C, Stelmachtchuk FN, Andersson E, Norlander E, de Oliveira FL, Machado Portela J, Marcili A, Emanuelson U, Gennari SM, Minervino AH (2019) Prevalence of antibodies against Toxoplasma gondii and Neospora spp. in equids of Western Pará Brazil. Acta Trop 189: 39-45.
  49. Moreno B, Collantes-Fernández E, Villa A, Navarro A, Regidor-Cerrillo J, Ortega-Mora LM (2012) Occurrence of Neospora caninum and Toxoplasma gondii infections in ovine and caprine abortions. Vet Parasitol 187: 312-318.
  50. Müller N, Zimmermann V, Hentrich B, Gottstein B (1996) Diagnosis of Neospora caninum and Toxoplasma gondii infection by PCR and DNA hybridization immunoassay. J Clin Microbiol 34: 2850-2852.
  51. Nayeri T, Sarvi S, Moosazadeh M, Daryani A (2022) The Global Prevalence of Neospora caninum Infection in Sheep and Goats That Had an Abortion and Aborted Fetuses: A Systematic Review and Meta-Analysis. Front Vet Sci 9: 870904
  52. Nooruldeen MY, Jaafar SE, Salih AI (2021) Seroprevalence of Neospora caninum infections in cattle in Kirkuk province. Iraqi J Vet Sci 35: 331-334.
  53. Novoa MB, Soler JP, Cirone KM, Hecker YP, Valentini BS, Primo ME, Moore DP (2023) Use and comparison of serologic assays to detect anti-Neospora caninum antibodies in farmed red deer (Cervus elaphus). Vet Parasitol 313: 109839.
  54. Rahmani SS, Malekifard F, Tavassoli M (2022) Neospora caninum, a cause of abortion in donkeys (Equus asinus) in Iran. Parasitol Res 121: 367-372.
  55. Reichel MP, Ayanegui-Alcérreca MA, Gondim LF, Ellis JT (2013) What is the global economic impact of Neospora caninum in cattle - the billion dollar question. Int J Parasitol 43: 133-142.
  56. Rodrigues AA, Brito DR, Kono IS, Reis SS, de Souza Lima Nino B, Nascimento TV, Barros LD, Garcia JL, de Cunha IA (2022) Seroprevalence of Neospora caninum and risk factors associated with infection in water buffaloes (Bubalus bubalis) from Maranhão State, Brazil. Vet Parasitol Reg Stud Reports 27: 100661.
  57. Sedlák K, Bartova E, Machacova T (2014) Seroprevalence of Neospora caninum in cats from the Czech Republic. Acta Parasitol 59: 359-361.
  58. Špilovská S, Reiterová K (2008) Seroprevalence of Neospora caninum in aborting sheep and goats in the Eastern Slovakia. Folia Vet 52: 33-35.
  59. Tayyub M, Ali S, Javid A, Imran M (2022) Molecular detection of Toxoplasma gondii and Neospora caninum in rock pigeons (Columba livia) in Punjab, Pakistan. Parasitol Res 121: 1499-1505
  60. Tirosh-Levy S, Savitsky I, Blinder E, Mazuz ML (2022) The involvement of protozoan parasites in sheep abortions – a ten-year review of diagnostic results. Vet Parasitol 303: 109664.
  61. Tirosh-Levy S, Steinman A, Minderigiu A, Arieli O, Savitski I, Fleiderovitz L, Edery N, Schvartz G, Mazuz ML (2020) High Exposure to Toxoplasma gondii and Neospora Spp. in Donkeys in Israel: Serological Survey and Case Reports. Animals 10: 1921
  62. Waap H, de Oliveira UV, Nunes T, Gomes J, Gomes T, Bärwald A, Dias Munhoz A, Schares G (2020) Serological survey of Neospora spp. and Besnoitia spp. in horses in Portugal. Vet Parasitol Reg Stud Reports 20: 100391.
  63. Wouda W, van den Ingh TS, van Knapen F, Sluyter FJ, Koeman JP, Dubey JP (1992) Neospora abortion in cattle in The Netherlands. Tijdschr Diergeneeskd 117: 599-602.
  64. Yang J, Ai J, Qi T, Ni X, Xu Z, Guo L, Sun Y, Li Y, Kang M, Li J (2022) Toxoplasma gondii and Neospora caninum Infections in Stray Cats and Dogs in the Qinghai-Tibetan Plateau Area, China. Animals 12: 1390
  65. Zhao SS, Tao DL, Chen JM, Chen X, Geng XL, Wang JW, Yang X, Song JK, Liu Q, Zhao GH (2022) Neospora caninum infection activated autophagy of caprine endometrial epithelial cells via mTOR signaling. Vet Parasitol 304: 109685.
Go to article

Authors and Affiliations

R.R. Mohammed
1
M. Tavassoli
1
K.R. Sidiq
2
B. Esmaeilnejad
1

  1. Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Nazloo Campus, PO Box 1177, Urmia, West Azerbaijan, Iran
  2. Department of Medical Laboratory Science, College of Medical and Applied Sciences, Charmo University, 46023 Chamchamal, Sulaimani, Kurdistan Region, Iraq
Download PDF Download RIS Download Bibtex

Abstract

Cryptosporidium spp., and Giardia duodenalis are intestinal protozoan parasites known to infect humans and various animals and cause diarrhea. This study aimed at determining the prevalence and genotype of Cryptosporidium spp. and Giardia duodenalis in sheep in different locations of Siirt province. The fecal material for this study was collected from 500 sheep in different locations of Siirt province, Turkey. Fecal samples obtained from sheep were examined for Cryptosporidium spp. by Kinyoun Acid Fast staining and the Nested PCR method. Microscopic and Nested PCR methods revealed a prevalence of 2.4% (12/500) and 3.6% (18/500), respectively. Sequence analysis revealed the presence of C. ryanae, C. andersoni, and zoonotic C. parvum. In terms of Giardia duodenalis, 8.4% (42/500) and 10.2% (51/500) prevalence was determined using Nativ-Lugol and Nested PCR methods, respectively. Using sequence analysis, zoonotic assemblages A and B as well as assemblages E and D were detected. As a result of this study, both the prevalence of Cryptosporidium spp. and Giardia duodenalis and the presence of species that appear to be host-specific, as well as those known to be zoonotic, were revealed. A large-scale study is needed to understand the impact of these agents on sheep farming and their consequences on human health.
Go to article

Bibliography

  1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ (1990) Basic local alignment search tool. J Mol Biol 215: 403-410.
  2. Çelik BA, Çelik ÖY, Ayan A, Akyıldız G, Kılınç ÖO, Ayan ÖO, Ercan K (2023) Molecular Prevalence of Giardia duodenalis and Subtype Distribution (Assemblage E and B) in Calves in Siirt, Turkey. Egypt J Vet Sci 54: 457-463.
  3. Ballweber LR, Xiao L, Bowman DD, Kahn G, Cama VA (2010) Giardiasis in dogs and cats: update on epidemiology and public health significance. Trends Parasitol 26: 180-189.
  4. Barker IK, Carbonell PL (1974) Cryptosporidium agni sp. n. from lambs, and Cryptosporidium bovis sp. n. from a calf, with observations on the oocyst. Z Parasitenkd 44: 289-298.
  5. Caccio SM, De Giacomo M, Pozio E (2002) Sequence analysis of the beta-giardin gene and development of a polymerase chain reaction-restriction fragment length polymorphism assay to genotype Giardia duodenalis cysts from human faecal samples. Int J Parasitol 32: 1023-1030.
  6. Castro-Hermida JA, González-Warleta M, Mezo M (2007) Natural infection by Cryptosporidium parvum and Giardia duodenalis in sheep and goats in Galicia (NW Spain). Small Rumin Res 72: 96-100.
  7. Dessì G, Tamponi C, Varcasia A, Sanna G, Pipia AP, Carta S, Salis F, Díaz P, Scala A (2020) Cryptosporidium infections in sheep farms from Italy. Parasitol Res 119: 4211-4218.
  8. Fayer R, Santín M (2009) Cryptosporidium xiaoi n. sp.(Apicomplexa: Cryptosporidiidae) in sheep (Ovis aries). Vet Parasitol 164: 192-200.
  9. Gharekhani J, Heidari H, Youssefi M (2014) Prevalence of Cryptosporidium infection in sheep in Iran. Turkiye Parazitol Derg 38: 22-25.
  10. Giangaspero A, Paoletti B, Iorio R, Traversa D (2005) Prevalence and molecular characterization of Giardia duodenalis from sheep in central Italy. Parasitol Res 96: 32-37.
  11. Goma FY, Geurden T, Siwila J, Phiri IGK, Gabriël S, Claerebout E, Vercruysse J (2007) The prevalence and molecular characterisation of Cryptosporidium spp. in small ruminants in Zambia. Small Rumin Res 72: 77-80.
  12. Hall TA (1999) BioEdit: a user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucleic Acids Symposium Series 41:95-98.
  13. Jafari H, Jalali MH, Shapouri MS, Hajikolaii MR (2014) Determination of Giardia duodenalis genotypes in sheep and goat from Iran. J Parasit Dis 38: 81-84.
  14. Janoff EN, Reller LB (1987) Cryptosporidium species, a protean protozoan. J Clin Microbiol 25: 967-975.
  15. Jian Y, Zhang X, Li X, Karanis G, Ma L, Karanis P (2018) Prevalence and molecular characterization of Giardia duodenalis in cattle and sheep from the Qinghai-Tibetan Plateau Area (QTPA), northwestern China. Vet Parasitol 250: 40-44.
  16. Kiani-Salmi N, Fattahi-Bafghi A, Astani A, Sazmand A, Zahedi A, Firoozi Z, Ebrahimi B, Dehghani-Tafti A, Ryan U, Akrami-Mohajeri F (2019) Molecular typing of Giardia duodenalis in cattle, sheep and goats in an arid area of central Iran. Infect Genet Evol 75: 104021.
  17. Kılınç ÖO, Ayan A, Çelik BA, Çelik ÖY, Yüksek N, Akyıldız G, Oğuz FE (2023) The Investigation of Giardiasis (Foodborne and Waterborne Diseases) in Buffaloes in Van Region, Türkiye: First Molecular Report of Giardia duodenalis Assemblage B from Buffaloes. Pathogens 12: 106.
  18. Kızıltepe Ş, Ayvazoğlu C (2022) Investigation of Diarrhea Factors in Neonatal Lambs in Iğdır Region. ISPEC J Agrİ Sci 6: 189-194.
  19. Koçhan A, Şimşek A, İpek-Sayın DN, İçen H (2020) Severe Bloody Diarrhea in a Calf Infected with Giardia duodenalis. Dicle Üniv Vet Fak Derg 13: 179-182.
  20. Koinari M, Lymbery AJ, Ryan UM (2014) Cryptosporidium species in sheep and goats from Papua New Guinea. Exp Parasitol 141: 134-137.
  21. Lalle M, Pozio E, Capelli G, Bruschi F, Crotti D, Cacciò SM (2005) Genetic heterogeneity at the beta giardin locus among human and animal isolates of Giardia duodenalis and identification of potentially zoonotic subgenotypes. Int J Parasitol 35: 207-213.
  22. Majeed QA, El-Azazy OM, Abdou NE, Al-Aal ZA, El-Kabbany AI, Tahrani LM, AlAzemi MS, Wang Y, Feng Y, Xiao L (2018) Epidemiological observations on cryptosporidiosis and molecular characterization of Cryptosporidium spp. in sheep and goats in Kuwait. Parasitol Res 117: 1631-1636.
  23. Majewska AC, Werner A, Sulima P, Luty T (2000) Prevalence of Cryptosporidium in sheep and goats bred on five farms in west-central region of Poland. Vet Parasitol 89: 269-275.
  24. Minh BQ, Nguyen MA, Von Haeseler A (2013) Ultrafast approximation for phylogenetic bootstrap. Mol Biol Evol 30: 1188-1195.
  25. Mondebo JA, Abah AE, Awi-Waadu GD (2022) Cryptosporidium infection in cattle, goat and ram in Yenagoa abattoir Bayelsa State, Nigeria. Anim Res Int 19: 4499-4506.
  26. Olson ME, Thorlakson CL, Deselliers L, Morck DW, McAllister TA (1997) Giardia and Cryptosporidium in Canadian farm animals. Vet Parasitol 68: 375-381.
  27. Ozmen O, Yukari BA, Haligur M, Sahinduran S (2006) Observations and immunohistochemical detection of Coronavirus, Cryptosporidium parvum and Giardia intestinalis in neonatal diarrhoea in lambs and kids. Schweiz Arch Tierheilkd 148: 357-364.
  28. Paz e Silva FM, Lopes RS, Bresciani KD, Amarante AF, Araujo JP (2014) High occurrence of Cryptosporidium ubiquitum and Giardia duodenalis genotype E in sheep from Brazil. Acta parasitol 59: 193-196.
  29. Rekha KMH, Puttalakshmamma GC, D’Souza PE (2016) Comparison of different diagnostic techniques for the detection of cryptosporidiosis in bovines. Vet World 9: 211–215.
  30. Robertson LJ, Gjerde BK, Hansen EF (2010) The zoonotic potential of Giardia and Cryptosporidium in Norwegian sheep: a longitudinal investigation of 6 flocks of lambs. Vet Parasitol 171: 140-145.
  31. Romero-Salas D, Alvarado-Esquivel C, Cruz-Romero A, Aguilar-Domínguez M, Ibarra-Priego N, Merino-Charrez JO, Pérez de León AA, Hernández-Tinoco J (2016) Prevalence of Cryptosporidium in small ruminants from Veracruz, Mexico. BMC Vet Res 12: 14.
  32. Ryan U, Cacciò SM (2013) Zoonotic potential of Giardia. Int J Parasitol 43: 943-956.
  33. Sahraoui L, Thomas M, Chevillot A, Mammeri M, Polack B, Vallée I, Follet J, Ain-Baaziz H, Adjou KT (2019) Molecular characterization of zoonotic Cryptosporidium spp. and Giardia duodenalis pathogens in Algerian sheep. Vet Parasitol Reg Stud Reports 16: 100280.
  34. Santín M, Trout JM, Fayer R (2007) Prevalence and molecular characterization of Cryptosporidium and Giardia species and genotypes in sheep in Maryland. Vet Parasitol 146: 17-24.
  35. Soltane R, Guyot K, Dei-Cas E, Ayadi A (2007) Prevalence of Cryptosporidium spp.(Eucoccidiorida: Cryptosporiidae) in seven species of farm animals in Tunisia. Parasite 14: 335-338.
  36. Trifinopoulos J, Nguyen L-T, von Haeseler A, Minh BQ (2016) W-IQ-TREE: a fast online phylogenetic tool for maximum likelihood analysis. Nucleic Acids Res 44: W232-W235.
  37. Ulutas B, Voyvoda H (2004) Cryptosporidiosis in Diarrhoeic Lambs on a Sheep Farm. Türkiye Parazitol Derg 28:15-17.
  38. Wang H, Qi M, Zhang K, Li J, Huang J, Ning C, Zhang L (2016) Prevalence and genotyping of Giardia duodenalis isolated from sheep in Henan Province, central China. Infect Genet Evol 39: 330-335.
  39. Wilson JM, Hankenson FC (2010) Evaluation of an inhouse rapid ELISA test for detection of Giardia in domestic sheep (Ovis aries). J Am Assoc Lab Anim Sci 49: 809-813.
  40. Xiao L (2010) Molecular epidemiology of cryptosporidiosis: an update. Exp Parasitol 124: 80-89.
  41. Xiao L, Singh A, Limor J, Graczyk TK, Gradus S, Lal A (2001) Molecular characterization of Cryptosporidium oocysts in samples of raw surface water and wastewater. Appl Environ Microbiol 67: 1097-1101.
  42. Yang F, Ma L, Gou JM, Yao HZ, Ren M, Yang BK, Lin Q (2022) Seasonal distribution of Cryptosporidium spp., Giardia duodenalis and Enterocytozoon bieneusi in Tibetan sheep in Qinghai, China. Parasites Vectors 15: 394.
  43. Yang R, Jacobson C, Gardner G, Carmichael I, Campbell AJ, Ng-Hublin J, Ryan U (2014) Longitudinal prevalence, oocyst shedding and molecular characterisation of Cryptosporidium species in sheep across four states in Australia. Vet Parasitol 200: 50-58.
Go to article

Authors and Affiliations

B. Aslan Çelik
1
Ö.Y. Çelik
2
A. Ayan
3
Ö. Orunç Kılınç
4
G. Akyıldız
5
K. İrak
6
M.A. Selçuk
1
K. Ercan
2
V. Baldaz
2
Ö. Oktay Ayan
7

  1. Department of Parasitology, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  2. Department of Internal Medicine, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  3. Department of Genetics, Faculty of Veterinary Medicine, Van Yüzüncü Yıl University, Van, 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. Department of Biochemistry, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  7. Department of Parasitology, Van Yüzüncü Yıl University, Faculty of Medicine, Van, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Montmorillonite (MMT), a natural absorbent agent, has widely been accepted for its antidiarrhea function in human and farm animals; however, its specific physicochemical property limits its biological function in practical use. In the current study, raw MMT was loaded by andrographolide, namely andrographolide loaded montmorillonite (AGP-MMT). The microstructure of AGP-MMT was observed by scanning electron microscope (SEM) and X-ray diffraction (XRD). The effect of AGP-MMT on the growth performance, intestinal barrier and inflammation was investigated in an enterotoxigenic Escherichia coli (ETEC) challenged mice model. The results show that the microstructure of MMT was obviously changed after andrographolide modification: AGP-MMT exhibited a large number of spheroid particles, and floccule aggregates, but lower interplanar spacing compared with MMT. ETEC infection induced body weight losses and intestinal barrier function injury, as indicated by a lower villus height and ratio of villus height/crypt depth, whereas the serum levels of diamine oxidase (DAO), D-xylose and ETEC shedding were higher in the ETEC group compared with the CON group. Mice pretreated with AGP-MMT showed alleviated body weight losses and the intestinal barrier function injury induced by ETEC challenge. The villus height and the ratio of villus height/crypt depth, were higher in mice pretreated with AGP-MMT than those pretreated with equal levels of MMT. Pretreatment with AGP-MMT also alleviated the increased concentration of serum tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and the corresponding genes in the jejunum induced by ETEC infection in mice. The protein and mRNA levels of IL-1β were lower in mice pretreated with AGP-MMT than those with equal levels of MMT. The results indicate that AGP-MMT was more effective in alleviating intestinal barrier injury and inflammation in mice with ETEC challenge than MMT.
Go to article

Bibliography

  1. Ala’a A, Canatan H, and Ezeamuzie CI (2009) In vitro and in vivo anti-inflammatory effects of andrographolide. Int Immunopharmacol 9: 313-318.
  2. Almeida JAS, Liu Y, Song M, Lee JJ, Gaskins HR, Maddox CW, Osuna O, Pettigrew JE (2013) Escherichia coli challenge and one type of smectite alter intestinal barrier of pigs. J Anim Sci Biotechnol 4: 52.
  3. Brubaker J, Zhang X, Bourgeois AL, Harro C, Sack DA, Chakraborty S (2021) Intestinal and systemic inflammation induced by symptomatic and asymptomatic enterotoxigenic E. coli infection and impact on intestinal colonization and ETEC specific immune responses in an experimental human challenge model. Gut Microbes 13: 1891852.
  4. Burgos RA, Alarcón P, Quiroga J, Manosalva C, Hancke J (2021) Andrographolide, an anti-inflammatory multitarget drug: all roads lead to cellular metabolism. Molecules 26: 5.
  5. Bywater RJ (2005) Identification and surveillance of antimicrobial resistance dissemination in animal production. Poultry Sci 84: 644-648.
  6. Chang FY, Lu CL, Chen CY, Luo JC (2007) Efficacy of dioctahedral smectite in treating patients of diarrhea‐predominant irritable bowel syndrome. J Gastroen Hepatol 22: 2266-2272.
  7. Chen J, Wan CM, Gong ST, Fang F, Sun M, Qian Y, Huang Y, Wang BX, Xu CD, Ye LY, Dong M, Jin Y, Huang ZH, Wu QB, Zhu CM, Fang YH, Zhu QR, Dong YS (2018) Chinese clinical practice guidelines for acute infectious diarrhea in children. World J Pediatr 14: 429-436.
  8. Cheng G, Hao H, Xie S, Wang X, Dai M, Huang L, Yuan Z (2014) Antibiotic alternatives: the substitution of antibiotics in animal husbandry? Front microbiol 5: 217.
  9. Delbem, MF, Valera TS, Valenzuela-Diaz FR, Demarquette N (2010) Modification of a brazilian smectite clay with different quaternary ammonium salts. Quim Nova 33: 309-315.
  10. Gaastra W, Svennerholm AM (1996) Colonization factors of human enterotoxigenic Escherichia coli (ETEC). Trends Microbiol 4: 444-452.
  11. Ghosh P, Mondal S, Bera T (2016) Preparation and characterization of andrographolide nanoparticles for visceral leishmaniasis chemotherapy: In vitro and in vivo evaluations. Int J Pharm Pharmac Sci 8: 102-107.
  12. Guarino A, Ashkenazi S, Gendrel D, Vecchio AL, Shamir R, Szajewska H (2014) European Society for Pediatric Gastroenterology, Hepatology, and Nutrition/European Society for Pediatric Infectious Diseases Evidence-Based Guidelines for the Management of Acute Gastroenteritis in Children in Europe: Update 2014. J Pediatr Gastr Nutr 59: 132-152.
  13. Guarino A, Bisceglia M, Castellucci G, Iacono G, Casali LG, Bruzzese E, Musetta A, Greco L (2001) Smectite in the treatment of acute diarrhea: a nationwide randomized controlled study of the italian society of pediatric gastroenterology and hepatology (SIGEP) in collaboration with primary care pediatricians. J Pediatr Gastroenterol Nutr 32: 71-75.
  14. Guarino A, Vecchio AL, Pirozzi MR (2009) Clinical role of diosmectite in the management of diarrhea. Expert Opin Drug Metab Toxicol 5: 433-440.
  15. Guo X, Zhang LY, Wu SC, Xia F, Fu YX, Wu YL, Leng CQ, Yi PF, Shen HQ, Wei XB, Fu BD (2014) Andrographolide interferes quorum sensing to reduce cell damage caused by avian pathogenic Escherichia coli. Vet Microbiol 174: 496-503.
  16. Han C, Song J, Hu J, Fu H, Feng Y, Mu R, Xing Z, Wang Z, Wang L, Zhang J, Wang C, Dong L (2021) Smectite promotes probiotic biofilm formation in the gut for cancer immunotherapy. Cell Rep 34: 108706.
  17. Hu C, Song J, You Z, Luan Z, Li W (2012) Zinc oxide-montmorillonite hybrid influences diarrhea, intestinal mucosal integrity, and digestive enzyme activity in weaned pigs. Biol Trace Elem Res 149: 190-196.
  18. Jiang N, Wei Y, Cen Y, Shan L, Zhang Z, Yu P, Wang Y, Xu L (2020) Andrographolide derivative AL-1 reduces intestinal permeability in dextran sulfate sodium (DSS)-induced mice colitis model. Life Sci 241: 117164.
  19. Jiao L, Lin F, Cao S, Wang C, Wu H, Shu M, Hu C (2017) Preparation, characterization, antimicrobial and cytotoxicity studies of copper/zinc- loaded montmorillonite. J Anim Sci Biotechno 8: 27.
  20. Kim N, Lertnimitphun P, Jiang Y, Tan H, Zhou H, Lu Y, Xu H (2019) Andrographolide inhibits inflammatory responses in LPS-stimulated macrophages and murine acute colitis through activating AMPK. Biochem Pharmacol 170: 113646.
  21. Liu H, Wang C, Gu X, Zhao J, Nie C, Zhang W, Ma X (2020) Dietary Montmorillonite Improves the Intestinal Mucosal Barrier and Optimizes the Intestinal Microbial Community of Weaned Piglets. Front Microbiol 11: 593056.
  22. Ma T, Peng W, Liu Z, Gao T, Liu W, Zhou D, Yang K, Guo R, Duan Z, Liang W, Bei W, Yuan F, Tian Y (2021) Tea polyphenols inhibit the growth and virulence of ETEC K88. Microb Pathogenesis 152: 104640.
  23. Massaro M, Colletti CG, Lazzara G, Riela S (2018) The Use of Some Clay Minerals as Natural Resources for Drug Carrier Applications. J Funct Biomater 9: 58.
  24. Ren M, Cai S, Zhou T, Zhang S, Li S, Jin E, Che C, Zeng X, Zhang T, Qiao S (2019) Isoleucine attenuates infection induced by E. coli challenge through the modulation of intestinal endogenous antimicrobial peptide expression and the inhibition of the increase in plasma endotoxin and IL-6 in weaned pigs. Food Funct 10: 3535-3542.
  25. Rodas C, Mamani R, Blanco J, Blanco JE, Wiklund G, Svennerholm AM, Sjöling A, Iniguez V (2011) Enterotoxins, colonization factors, serotypes and antimicrobial resistance of enterotoxigenic Escherichia coli (ETEC) strains isolated from hospitalized children with diarrhea in Bolivia. Braz J Infect Dis 15: 132-137.
  26. Rodea GE, Montiel-Infante FX, Cruz-Córdova A, Saldaña-Ahuactzi Z, Ochoa SA, Espinosa-Mazariego K, Hernández-Castro R, Xicohtencatl-Cortes J (2017) Tracking bioluminescent ETEC during in vitro BALB/c mouse colonization. Front Cell Infect Microbiol 7: 187.
  27. Sargeant HR, McDowall KJ, Miller HM, Shaw MA (2010) Dietary zinc oxide affects the expression of genes associated with inflammation: Transcriptome analysis in piglets challenged with ETEC K88. Vet Immunol Immunop 137: 120-129.
  28. Su HM, Mo JL, Ni JD, Ke HH, Bao T, Xie JH, Xu Y, Xie LH, Chen W (2020) Andrographolide exerts antihyperglycemic effect through strengthening intestinal barrier function and increasing microbial composition of akkermansia muciniphila. Oxid Med and Cell Longev 2020: 6538930.
  29. Vaseeharan B, Thaya R (2014) Medicinal plant derivatives as immunostimulants: an alternative to chemotherapeutics and antibiotics in aquaculture. Aquacult Int 22: 1079-1091.
  30. Wang P, Chen Q, Gan L, Du X, Li Q, Qiao H, Zhao Y, Huang J, Wang J (2022) Marginal zinc deficiency aggravated intestinal barrier dysfunction and inflammation through ETEC virulence factors in a mouse model of diarrhea. Vet Sci 9: 507.
  31. Wang P, Yuan P, Lin S, Zhong H, Zhang X, Zhuo Y, Li J, Che L, Feng B, Lin Y, Xu S, Wu D, Burrin DG, Fang ZF (2022) Maternal and fetal bile acid homeostasis regulated by sulfated progesterone metabolites through FXR signaling pathway in a pregnant sow model. Int J Mol Sci 23: 6496.
  32. Wang Q, Zhan X, Wang B, Wang F, Zhou Y, Xu S, Li X, Tang L, Jin Q, Li W, Gong L, Fu A (2022) Modified montmorillonite improved growth performance of broilers by modulating intestinal microbiota and enhancing intestinal barriers, anti-inflammatory response, and antioxidative capacity. Antioxidants (Basel) 11: 1799.
  33. Xia Y, Chen S, Zhao Y, Chen S, Huang R, Zhu G, Yin Y, Ren W, Deng J (2019) GABA attenuates ETEC-induced intestinal epithelial cell apoptosis involving GABA(A)R signaling and the AMPK-autophagy pathway. Food Funct 10: 7509-7522.
  34. Yan F, Liu L, Cao H, Moore DJ, Washington MK, Wang B, Peek R, Acra SA, Polk DB (2017) Neonatal colonization of mice with LGG promotes intestinal development and decreases susceptibility to colitis in adulthood. Mucosal Immunol 10: 117-127.
  35. Zhu Q, Zheng P, Chen X, Zhou F, He Q, Yang Y (2018) Andrographolide presents therapeutic effect on ulcerative colitis through the inhibition of IL-23/IL-17 axis. Am J Transl Res 10: 465-473.
Go to article

Authors and Affiliations

P. Wang
1
L. Li
1
L. Gan
1
Q. Chen
1
H. Qiao
1
W. Gao
1
Y. Zhang
1
J. Wang
1

  1. College of Biology Engineering, Henan University of Technology, Zhengzhou, China
Download PDF Download RIS Download Bibtex

Abstract

This study aimed to evaluate the impact of betaine (Bet) and protected calcium butyrate (PCB) supplementation individually and together on the performance, carcass traits, blood biochemistry, and meat quality of growing Japanese quails ( Coturnix coturnix Japonica) from 1 to 42 days old. 144 one-day-old unsexed Japanese quails were randomly assigned to four dietary treatments with six replicates each. All birds were fed a maize-soybean meal diet for 42 days. The control group received no feed additives, while the treatment groups received 1.2 g/kg Bet, 1.0 g/kg PCB, or a combination of both in their diets. The results indicated that Bet and PCB supplementation individually and together did not differ performance, relative weights of heart, gizzard, proventriculus, bursa of Fabricius and pancreas, water holding capacity (WHC), cooking loss (CL), blood biochemical values except for glucose and triglyceride. Bet supplementation significantly increased relative liver weights, while PCB supplementation decreased glucose levels in serum. Moreover, carcass yield was increased and triglyceride value in blood serum, malondialdehyde (MDA), and the pH levels of breast meats both on the 1st and 30st day of post-mortem were decreased in all treatment groups. Therefore, based on these results, the combination of betaine and butyrate improves both carcass yield and meat quality in growing Japanese quails. More research is needed to determine the impact of betaine and butyrate on the structure of amino acids in meat, antioxidant enzyme activity, and the immune system in poultry.
Go to article

Bibliography


  1. Abd El‐Wahab A, Mahmoud RE, Ahmed MF, Salama MF (2019) Effect of dietary supplementation of calcium butyrate on growth performance, carcass traits, intestinal health and pro‐inflammatory cytokines in Japanese quails. Anim Physiol Anim Nutr 103: 1768-1775.
  2. Abudabos AM, Suliman GM, Al-Owaimer AN, Sulaiman AR, Alharthi AS (2021) Effects of nano emulsified vegetable oil and betaine on growth traits and meat characteristics of broiler chickens reared under cyclic heat stress. Animals 11:1911.
  3. Al-Abdullatif AA, Al-Sagan AA, Hussein EO, Saadeldin IM, Suliman GM, Azzam MM, Al-Mufarrej SI, Alhotan RA (2021) Betaine could help ameliorate transport associated water deprivation stress in broilers by reducing the expression of stress-related transcripts and modulating water channel activity. Ital J Anim Sci 20:14-25.
  4. Al-Sagan AA, Al-Yemni AH, Abudabos AM, Al-Abdullatif AA, Hussein EO (2021) Effect of different dietary betaine fortifications on performance, carcass traits, meat quality, blood biochemistry, and hematology of broilers exposed to various temperature patterns. Animals 11: 1555.
  5. Carpenter KJ, Clegg KM (1956) The metabolizable energy of poultry feeding stuffs in relation to their chemical composition. J Sci Food Agric 7: 45-51.
  6. Chamba F, Puyalto M, Ortiz A, Torrealba H, Mallo JJ, Riboty R (2014) Effect of partially protected sodium butyrate on performance, digestive organs, intestinal villi and E. coli development in broilers chickens. Int J Poult Sci 13: 390-396.
  7. Chen R, Yang M, Song YD, Wang RX, Wen C, Liu Q, Zhou YM, Zhuang S (2022) Effect of anhydrous betaine and hydrochloride betaine on growth performance, meat quality, postmortem glycolysis, and antioxidant capacity of broilers. Poult Sci 101: 101687.
  8. Czerwiński J, Højberg O, Smulikowska S, Engberg RM, Mieczkowska A (2012) Effects of sodium butyrate and salinomycin upon intestinal microbiota, mucosal morphology and performance of broiler chickens. Arch Anim Nutr 66: 102-116.
  9. Dawood MA, Eweedah NM, Elbialy ZI, Abdelhamid AI (2020) Dietary sodium butyrate ameliorated the blood stress biomarkers, heat shock proteins, and immune response of Nile tilapia (Oreochromis niloticus) exposed to heat stress. J Therm Biol 88: 102500.
  10. Deepa K, Purushothaman MR, Vasanthakumar P, Sivakumar K (2017) Serum biochemical parameters and meat quality influenced due to supplementation of sodium butyrate in broiler chicken. Int J Livest Res 7: 108-116.
  11. Egbuniwe IC, Uchendu CN, Obidike IR (2021) Ameliorative effects of betaine and ascorbic acid on endocrine and erythrocytic parameters of sexually-maturing female Japanese quails during the dry season. J Therm Biol 96: 102812.
  12. El-Bahr SM, Shousha S, Khattab W, Shehab A, El-Garhy O, El-Garhy H, Mohamed S, Ahmed-Farid O, Hamad A, Sabike I (2021) Impact of dietary betaine and metabolizable energy levels on profiles of proteins and lipids, bioenergetics, peroxidation and quality of meat in Japanese quail. Animals 11: 117.
  13. Elnesr SS, Ropy A, Abdel-Razik AH (2019) Effect of dietary sodium butyrate supplementation on growth, blood biochemistry, haematology and histomorphometry of intestine and immune organs of Japanese quail. Animal 13: 1234-1244.
  14. Esteve-Garcia E, Mack S (2000) The effect of dl-methionine and betaine on growth performance and carcass characteristics in broilers. Anim Feed Sci Technol 87: 85-93.
  15. Gümüş E, Küçükersan S, Bayraktaroğlu AG, Sel T (2021) Effect of dietary supplementation of some natural antioxidants and coated calcium butyrate on carcass traits, some serum biochemical parameters, lipid peroxidation in meat and intestinal histomorphology in broilers. Ankara Univ Vet Fak Derg 68: 237-244.
  16. Jiang Y, Zhang W, Gao F, Zhou G (2015) Micro-encapsulated sodium butyrate attenuates oxidative stress induced by corticosterone exposure and modulates apoptosis in intestinal mucosa of broiler chickens. Anim Prod Sci 55: 587.
  17. Leeson S, Namkung H, Antongiovanni M, Lee EH (2005) Effect of butyric acid on the performance and carcass yield of broiler chickens. Poult Sci 84: 1418-1422.
  18. Liu J, Song R, Su S, Qi N, Li Q, Xie Z, Yu S (2022) Betaine promotes fat accumulation and reduces injury in Landes Goose hepatocytes by regulating multiple lipid metabolism pathways. Animals 12: 1530.
  19. Mátis G, Petrilla J, Kulcsár A, van den Bighelaar H, Boomsma B, Neogrády Z, Fébel H (2019) Effects of dietary butyrate supplementation and crude protein level on carcass traits and meat composition of broiler chickens. Arch Anim Breed 62: 527-536.
  20. National Research Council (1994) Nutrient Requirements of Poultry, 9th ed., The National Academies Press, Washington DC.
  21. Pascual A, Trocino A, Birolo M, Cardazzo B, Bordignon F, Balarin C, Carraro L, Xiccato G (2020) Dietary supplementation with sodium butyrate: growth, gut response at different ages, and meat quality of female and male broiler chickens. Ital J Anim Sci 19: 1134-1145.
  22. Pillai PB, Fanatico AC, Beers KW, Blair ME, Emmert JL (2006) Homocysteine remethylation in young broilers fed varying levels of methionine, choline, and betaine. Poult Sci 85: 90-95.
  23. Rama Rao SV, Raju MV, Panda AK, Saharia P, Sunder GS (2011) Effect of supplementing betaine on performance, carcass traits and immune responses in broiler chicken fed diets containing different concentrations of methionine. Asian-Australas J Anim Sci 24: 662-669.
  24. Rice EM, Aragona KM, Moreland SC, Erickson PS (2019) Supplementation of sodium butyrate to postweaned heifer diets: Effects on growth performance, nutrient digestibility, and health. J Dairy Sci 102: 3121-3130.
  25. Shihab MA, Olgun O, Abdulqader AF (2020) The effect of supplementation of sodium butyrate to diets with different levels of metabolic energy contents on performance, carcass and some blood parameters in growing quails. Journal of Bahri Dagdas Animal Research 9: 79-87.
  26. Su SY, Dodson MV, Li XB, Li QF, Wang HW, Xie Z (2009) The effects of dietary betaine supplementation on fatty liver performance, serum parameters, histological changes, methylation status and the mRNA expression level of Spot14α in Landes goose fatty liver. Comp Biochem Physiol A Mol Integr Physiol 154: 308-314.
  27. Uzunoğlu K, Yalçın S (2019) Effects of dietary supplementation of betaine and sepiolite on performance and intestinal health in broilers. Ankara Univ Vet Fak Derg 66: 221-229.
  28. Yang M, Chen R, Song YD, Zhou YM, Liu Q, Zhuang S (2022a) Effects of dietary betaine supplementation on growth performance, meat quality, muscle fatty acid composition and antioxidant ability in slow-growing broiler chickens. Br Poult Sci 63: 351-359.
  29. Yang Q, Chen B, Robinson K, Belem T, Lyu W, Deng Z, Ramanathan R, Zhang G (2022b) Butyrate in combination with forskolin alleviates necrotic enteritis, increases feed efficiency, and improves carcass composition of broilers. J Animal Sci Biotechnol 13: 3.
  30. Yin F Yu H, Lepp D, Shi X, Yang X, Hu J, Leeson S, Yang C, Nie S, Hou Y, Gong J (2016) Transcriptome analysis reveals regulation of gene expression for lipid catabolism in young broilers by butyrate glycerides. Plos One 11: e0160751.
  31. Zeb A, Ullah F (2016) A simple spectrophotometric method for the determination of thiobarbituric acid reactive substances in fried fast foods. J Anal Chem 2016: 9412767.
  32. Zhang W, Jiang Y, Zhu QM, Gao F, Dai S, Chen JC, Zhou G (2011a) Sodium butyrate maintains growth performance by regulating the immune response in broiler chickens. Br Poult Sci 52: 292-301.
  33. Zhang W, Gao F, Zhu QM, Zhou G, Jiang Y, Dai S (2011b) Dietary sodium butyrate alleviates the oxidative stress induced by corticosterone exposure and improves meat quality in broiler chickens. Poult Sci 90: 2592-2599.
Go to article

Authors and Affiliations

E. Gümüş
1
B. Sevim
2
O. Olgun
3
S. Küçükersan
4

  1. Department of Veterinary, Eskil Vocational School, Aksaray University, Şehit Recep Bozdağ Cad., 68800 Eskil, Aksaray, Turkey
  2. Department of Food Processing, Technical Sciences Vocational School, Aksaray University, Hacılar Harmanı Mah, 12. Bulvar No:2, Merkez, 68100 Aksaray, Turkey
  3. Department of Animal Science, Faculty of Agriculture, Selçuk University, Alaeaddin Keykubat Yerleşkesi, 42130, Selcuklu, Konya, Turkey
  4. Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Ankara University, Zübeyde Hanım Mahallesi Şehit Ömer Halisdemir Bulvarı No: 9/C, 06070, Altındağ, Ankara, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to evaluate the plasma levels of chemokines CCL2 and CXCL12 in female dogs with malignant mammary gland tumours without and with metastases. The concentrations of CCL2 and CXCL12 were determined in 25 female dogs with malignant mammary gland tumours (15 without metastases and 10 with metastases) and 10 healthy control animals using a specific canine ELISA assay. The mean plasma concentrations of CCL2 and CXCL12 were significantly higher (p<0.05) in the metastatic group compared to the control group. Moreover, the concentrations of these chemokines were markedly higher in the dogs with metastases than in those without metastases; however, a statistically significant difference was not found. The concentrations of both tested chemokines were numerically increased in the dogs with grade 2 and grade 3 carcinomas compared to the dogs with grade 1 carcinomas but the differences did not reach statistical significance. In conclusion, the results of our study demonstrate that plasma concentrations of chemokines CCL2 and CXCL12 are significantly increased in the dogs with metastatic malignant mammary gland tumours compared to the healthy dogs and show an upward trend compared to those without metastases. However, clarifying whether the increase of these chemokines is a cause or an effect of metastasis in female dogs with malignant mammary gland tumours as well as their potential role in metastatic process requires further research.
Go to article

Bibliography


  1. Abdelmegeed SM, Mohammed S (2018) Canine mammary tumors as a model for human disease. Oncol Lett 15: 8195-8205.
  2. Aljurany AN, Samarrai OR (2022) Evaluation of chemokine CXCL12 level with oxidative stress in breast cancer. Int J Health Sci 6: 6382-6392.
  3. Allende C, Higgins B, Johns J (2020) Comparison of serum cytokine concentrations between healthy dogs and canine osteosarcoma patients at the time of diagnosis. Vet Immunol Immunopathol 227: 110084.
  4. Ariyarathna H, Thomson N, Aberdein D, Munday JS (2020) Chemokine gene expression influences metastasis and survival time of female dogs with mammary carcinoma. Vet Immunol Immunopathol 227: 110075.
  5. Dehqanzada ZA, Storrer CE, Hueman MT, Foley RJ, Harris KA, Jama YH, Shriver CD, Ponniah S, Peoples GE (2007) Assessing serum cytokine profiles in breast cancer patients receiving a HER2/neu vaccine using Luminex technology. Oncol Rep 17: 687-694.
  6. Deshmane SL, Kremlev S, Amini S, Sawaya BE (2009) Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res 29: 313-326.
  7. Elston CW, Ellis IO (1998) Assessment of histological grade. In: Systemic Pathology. Churchill Livingstone, London, pp 356-384.
  8. Feng L, Qi Q, Wang P, Chen H, Chen Z, Meng Z, Liu L (2020) Serum level of CCL2 predicts outcome of patients with pancreatic cancer. Acta Gastroenterol Belg 83: 295-299.
  9. Ferrari SM, Elia G, Piaggi S, Baldini E, Ulisse S, Miccoli M, Materazzi G, Antonelli A, Fallahi P (2018) CCL2 is modulated by cytokines and PPAR-γ in anaplastic thyroid cancer. Anticancer Agents Med Chem 18: 458-466.
  10. Gao D, Fish EN (2018) Chemokines in breast cancer: Regulating metabolism. Cytokine 109: 57-64.
  11. Goldschmidt M, Pena L, Rasotto R, Zappulli V (2011) Classification and grading of canine mammary tumors. Vet Pathol 48: 117-131.
  12. Hayasaka H, Yoshida J, Kuroda Y, Nishiguchi A, Matsusaki M, Kishimoto K, Nishimura H, Okada M, Shimomura Y, Kobayashi D, Shimazu Y, Taya Y, Akashi M, Miyasaka M (2022) CXCL12 promotes CCR7 ligand-mediated breast cancer cell invasion and migration toward lymphatic vessels. Cancer Sci 113: 1338-1351.
  13. Hefler L, Tempfer C, Heinze G, Mayerhofer K, Breitenecker G, Leodolter S, Reinthaller A, Kainz C (1999) Monocyte chemoattractant protein-1 serum levels in ovarian cancer patients. Br J Cancer 81: 855-859.
  14. Im KS, Graef AJ, Breen M, Lindblad-Toh K, Modiano JF, Kim JH (2017) Interactions between CXCR4 and CXCL12 promote cell migration and invasion of canine hemangiosarcoma. Vet Comp Oncol 15: 315-327.
  15. Ishioka K, Suzuki Y, Tajima K, Ohtaki S, Miyabe M, Takasaki M, Mori A, Momota Y, Azakami D, Sako T (2013) Monocyte chemoattractant protein-1 in dogs affected with neoplasia or inflammation. J Vet Med Sci 75: 173-177.
  16. Janssens R, Struyf S, Proost P (2018) The unique structural and functional features of CXCL12. Cell Mol Immunol 15: 299-311.
  17. Kohli K, Pillarisetty VG, Kim TS (2022) Key chemokines direct migration of immune cells in solid tumors. Cancer Gene Ther 29: 10-21.
  18. Li H, Wu M, Zhao X (2022) Role of chemokine systems in cancer and inflammatory diseases. MedComm 3: e147.
  19. Lim SY, Yuzhalin AE, Gordon-Weeks AN, Muschel RJ (2016) Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget 7: 28697-28710.
  20. Lu X, Kang Y (2009) Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone. J Biol Chem 284: 29087-29096.
  21. Lu X, Qian CN, Mu YG, Li NW, Li S, Zhang HB, Li SW, Wang FL, Guo X, Xiang YQ (2011) Serum CCL2 and serum TNF-α – Two new biomarkers predict bone invasion, post-treatment distant metastasis and poor overall survival in nasopharyngeal carcinoma. Eur J Cancer 47: 339-346.
  22. Lubowicka E, Przylipiak A, Zajkowska M, Piskór BM, Malinowski P, Fiedorowicz W, Ławicki S (2018) Plasma chemokine CCL2 and its receptor CCR2 concentrations as diagnostic biomarkers for breast cancer patients. Biomed Res Int 2018: 2124390.
  23. Maekawa N, Konnai S, Asano Y, Sajiki Y, Deguchi T, Okagawa T, Watari K, Takeuchi H, Takagi S, Hosoya K, Kim S, Ohta H, Kato Y, Suzuki Y, Murata S, Ohashi K (2022) Exploration of serum biomarkers in dogs with malignant melanoma receiving anti-PD-L1 therapy and potential of COX-2 inhibition for combination therapy. Sci Rep 12: 9265.
  24. Marques CS, Santos AR, Gameiro A, Correia J, Ferreira F (2018) CXCR4 and its ligand CXCL12 display opposite expression profiles in feline mammary metastatic disease, with the exception of HER2-overexpressing tumors. BMC Cancer 18: 741.
  25. Marques CS, Soares M, Santos A, Correia J, Ferreira F (2017) Serum SDF-1 levels are a reliable diagnostic marker of feline mammary carcinoma, discriminating HER2-overexpressing tumors from other subtypes. Oncotarget 8: 105775-105789.
  26. Nielsen LN, Kjelgaard-Hansen M, Kristensen AT (2013) Monocyte chemotactic protein-1 and other inflammatory parameters in Bernese Mountain dogs with disseminated histiocytic sarcoma. Vet J 198: 424-428.
  27. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA (2005) Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell 121: 335-348.
  28. Pan YW, Zhou ZG, Wang M, Dong JQ, Du KP, Li S, Liu YL, Lv PJ, Gao JB (2016) Combination of IL6, IL-10, and MCP-1 with traditional serum tumor markers in lung cancer diagnosis and prognosis. Genet Mol Res 15: gmr15048949.
  29. Pena L, De Andres PJ, Clemente M, Cuesta P, Perez-Alenza MD (2013) Prognostic value of histological grading in noninflammatory canine mammary carcinomas in a prospective study with two-year follow-up: relationship with clinical and histological characteristics. Vet Pathol 50: 94-105.
  30. Perry JA, Thamm DH, Eickhoff J, Avery AC, Dow SW (2011) Increased monocyte chemotactic protein-1 concentration and monocyte count independently associate with a poor prognosis in dogs with lymphoma. Vet Comp Oncol 9: 55-64.
  31. Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, Kaiser EA, Snyder LA, Pollard JW (2011) CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475: 222-225.
  32. Raman D, Baugher PJ, Thu YM, Richmond A (2007) Role of chemokines in tumor growth. Cancer Lett 256: 137-165.
  33. Rybicka A, Eyileten C, Taciak B, Mucha J, Majchrzak K, Hellmen E, Krol M (2016) Tumour-associated macrophages influence canine mammary cancer stem-like cells enhancing their pro-angiogenic properties. J Physiol Pharmacol 67: 491-500.
  34. Salas Y, Marquez A, Diaz D, Romero L (2015) Epidemiological study of mammary tumors in female dogs diagnosed during the period 2002-2012: a growing animal health problem. PLoS One 10: e0127381.
  35. Sanchez-Martin L, Estecha A, Samaniego R, Sanchez-Ramon S, Vega MA, Sanchez-Mateos P (2011) The chemokine CXCL12 regulates monocyte-macrophage differentiation and RUNX3 expression. Blood 117: 88-97.
  36. Shi Y, Riese DJ 2nd, Shen J (2020) The role of the CXCL12/CXCR4/CXCR7 chemokine axis in cancer. Front Pharmacol 11: 574667.
  37. Shimizu N, Hamaide A, Dourcy M, Noёl S, Clercx C, Teske E (2019) Evaluation of urinary and serum level of chemokine (C-C motif) ligand 2 as a potential biomarker in canine urothelial tumours. Vet Comp Oncol 17: 11-20.
  38. Sleeckx N, de Rooster H, Veldhuis Kroeze EJ, Van Ginneken C, Van Brantegem L (2011) Canine mammary tumours, an overview. Reprod Domest Anim 46: 1112-1131.
  39. Soria G, Ben-Baruch A (2008) The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett 267: 271-285.
  40. Tang CH, Tsai CC (2012) CCL2 increases MMP-9 expression and cell motility in human chondrosarcoma cells via the Ras/Raf/MEK/ERK/NF-κB signaling pathway. Biochem Pharmacol 83: 335-344.
  41. Wu J, Liu X, Wang Y (2013) Predictive value of preoperative serum CCL2, CCL18, and VEGF for the patients with gastric cancer. BMC Clin Pathol 13: 15.
  42. Yao M, Yu E, Staggs V, Fan F, Cheng N (2016) Elevated expression of chemokine C-C ligand 2 in stroma is associated with recurrent basal-like breast cancers. Mod Pathol 29: 810-823.
  43. Zielińska KA, Katanaev VL (2020) The signaling duo CXCL12 and CXCR4: chemokine fuel for breast cancer tumorigenesis. Cancers (Basel) 12: 3071.
Go to article

Authors and Affiliations

M. Szczubiał
1
W. Łopuszyński
2
R. Dąbrowski
1
M. Jamioł
3
M. Bochniarz
1
P. Brodzki
1

  1. Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, 20-612 Lublin, Poland
  2. Department of Pathomorphology and Forensic Veterinary Medicine, Faculty of Veterinary Medicine, University of Life Sciences, Gleboka 30, 20-612 Lublin, Poland
  3. Department of Animal Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, 20-033 Lublin, Poland
Download PDF Download RIS Download Bibtex

Abstract

Immunoaffinity chromatography (IAC) is a fundamental isolation and purification tool which is incorporated in a substantial range of therapeutic and diagnostic applications. This study has reappraised the usefulness of immunoaffinity chromatography for the purification of polyclonal antibodies. Protein A based IAC is a convenient and reliable method for purification of IgG, from hyperimmunesera (HIS) raised in experimental animals such as rabbits, guinea pigs and mice to be utilized in pharmaceutics and diagnostics. The 146S fraction of Foot and Mouth Disease virus (FMDV) TCID50=10 5.6 was cultured on a baby hamster kidney cell line 21 (BHK-21), concentrated using salt precipitation method using PEG 6000, purified by size exclusion chromatography (SEC) using Sepharose-30 at 254nm absorbance. Purification of 146S FMDV was analyzed using 12% SDS-PAGE which provided two bands of light and heavy chains. The alum-based vaccine, consisting of ≥10μg of 146S FMDV, was applied in 10 male rabbits and 10 male guinea pigs and two animals of each group were taken as a negative control. The titer of serum was calculated using virus neutralization test. A Protein-A kit (Thermo scientific- 44667, 0528.2) was used to purify HIS raised against 146S FMDV and validated using 12% SDS PAGE in reducing condition. The data demonstrate that protein-A affinity chromatography is an efficient tool for the purification of antibodies from hyper-immune sera raised against 146S FMDV and can be used for the production of diagnostic kits e.g. Enzyme linked immuno-sorbent assay (ELISA) and radioimmunoassay.
Go to article

Bibliography


  1. Abi-Ghanem DA, Berghman LR (2015) Immunoaffinity chromatography: a review. Aff Chromatograph 95-103.
  2. Arora S, Saxena V, Ayyar BV (2017) Affinity chromatography: A versatile technique for antibody purification. Methods 116: 84-94.
  3. Ayyar BV, Arora S, Murphy C, O’Kennedy R (2012) Affinity chromatography as a tool for antibody purification. Methods 56: 116-129.
  4. Bergmann-Leitner ES, Mease RM, Duncan EH, Khan F, Waitumbi J, Angov E (2008) Evaluation of immunoglobulin purification methods and their impact on quality and yield of antigen-specific antibodies. Mala J 7: 1-10.
  5. Basagoudanavar SH, Hosamani M, Muthuchelvan D, Singh R, Santhamani R, Sreenivasa B, Saravanan P, Pandey A, Singh R, Venkataramanan R (2018) Baculovirus expression and purification of peste-des-petits-ruminants virus nucleocapsid protein and its application in diagnostic assay. Biologicals 55: 38-42.
  6. Chames P, Van Regenmortel M, Weiss E, Baty D (2009) Therapeutic antibodies: successes, limitations and hopes for the future. Br J Pharmacol 157: 220-233.
  7. Coelho LC, Santos AF, Napoleão TH, Correia MT, Paiva PM (2012) Protein purification by affinity chromatography. Intech.
  8. de Sousa P, Tavares P, Teixeira E, Dias N, Lima MdA, Luna F, Gondim D, de Azevedo D, Junior IS (2019) Experimental designs for optimizing the purification of immunoglobulin G by mixed-mode chromatography. J Chromatogr B. 1125, 121719.
  9. Eivazi S, Majidi J, Abdolalizadeh J, Yousefi M, Ahmadi M, Dadashi S, Moradi Z, Zolali E (2015) Production and purification of a polyclonal antibody against purified mouse IgG2b in rabbits towards designing mouse monoclonal isotyping kits. Adv Pharm Bull 5: 109.
  10. Hilbrig F, Freitag R (2003) Protein purification by affinity precipitation. J Chromatogr B 790: 79-90.
  11. Hosamani M, Gopinath S, Sreenivasa B, Behera S, Basagoudanavar SH, Boora A, Bora DP, Deka P, Bhanuprakash V, Singh RK (2022) A new blocking ELISA for detection of foot-and-mouth disease non-structural protein (NSP) antibodies in a broad host range. Appl Microbiol and Biotechnol 106: 6745-6757.
  12. Hossienizadeh SMJ, Bagheri M, Alizadeh M, Rahimi M, Azimi SM, Kamalzade M, Es-Haghi A, Ghassempour A (2021) Two Dimensional Anion Exchange-Size Exclusion Chromatography Combined with Mathematical Modeling for Downstream Processing of Foot and Mouth Disease Vaccine. J Chromatogr A 1643: 462070.
  13. Huang S, Cheng SY, Zhang SY, Yan YL, Cai SL, Li XL, Zheng SR, Fan J, Zhang WG. (2020) Protein A-mesoporous silica composites for chromatographic purification of immunoglobulin G. New J Chem 44: 7884-7890.
  14. Huse K, Böhme HJ, Scholz GH (2002) Purification of antibodies by affinity chromatography. J Bioch Bioph Meth 51: 217-231.
  15. Ma Z, Ramakrishna S. (2008) Electrospun regenerated cellulose nanofiber affinity membrane functionalized with protein A/G for IgG purification. J Memb Sci 319: 23-28.
  16. Rathore AS, Narnaware S (2022) Purification of therapeutic antibodies by protein a affinity chromatography. Methods Mol Biol 2313, pp 169-177.
  17. Rižner TL (2014) Teaching the structure of immunoglobulins by molecular visualization and SDS‐PAGE analysis. Biochem Mol Biol Educ 42: 152-159.
  18. Roque AC, Silva CS, Taipa MÂ (2007) Affinity-based methodologies and ligands for antibody purification: advances and perspectives. J Chromatogr A 1160: 44-55.
  19. Sadeghi S, Aghebati Maleki L, Nozari S, Majidi J (2018) A methodological approach for production and purification of polyclonal antibody against dog IgG. Vet Res Forum.
  20. Subramanian A (2002) Immunoaffinity chromatography. Mol Biotechnol 20: 41-47.
  21. Verdoliva A, Pannone F, Rossi M, Catello S, Manfredi V (2002) Affinity purification of polyclonal antibodies using a new all-D synthetic peptide ligand: comparison with protein A and protein G. J Immunol Meth 271:77-88.
  22. Wang Y, Zhang P, Liu S, Zhang Y, Zhao T, Huang W, He C, Yu Y, Wang L, Wan M (2011) Purification of IgG from sera of rabbit and guinea pig by flow-through mode ion-exchange chromatography using DEAE sepharose fast flow column. Chromatographia 74: 209-214.
  23. Wu M, Wang X, Zhang Z, Wang R (2011) Isolation and purification of bioactive proteins from bovine colostrum; Progress in Molecular and Environmental Bioengineering-From Analysis and Modeling to Technology Applications; IntechOpen; 347-366.
  24. Yang L, Harding JD, Ivanov AV, Ramasubramanyan N, Dong DD (2015) Effect of cleaning agents and additives on Protein A ligand degradation and chromatography performance. J Chromatogr A 1385: 63-68.
Go to article

Authors and Affiliations

A. Munir
1
A.A. Anjum
1
I. Altaf
2
A.R. Awan
3

  1. Institute of Microbiology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Outfall road, Lahore, Pakistan
  2. Quality Operations Laboratory, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Outfall road, Lahore, Pakistan
  3. Department of Biochemistry and Biotechnology, Faculty of Veterinary Sciences, University of Veterinary and Animal Sciences, Outfall road, Lahore, Pakistan
Download PDF Download RIS Download Bibtex

Abstract

Parasitosis in horses may be uncontrolled and expose breeders and owners to serious financial losses or, possibly, to the loss of animals. Therefore, the prevention and monitoring of the development of parasitic diseases should play an important role in the breeding process. The aim of this study was to confirm the influence of factors such as age, breed, herd size, deworming program, and type of anthelmintics, on the prevalence and intensity of parasites (helminths) in domestic horses in Lower Silesia. The study was carried out between August and November of 2020. The samples of horse feces were collected from 50 different stables in the area of Lower Silesia, Poland. A total of 286 individuals from various breeds were examined. Detailed analysis revealed that the mean age of infected horses was significantly lower than in uninfected horses. The mean time since the last deworming procedure was almost twice as low in uninfected horses than in infected ones. Additionally, the deworming agent affects the prevalence of infection. The analysis was also performed for the same factors in reference to quantitative data. The mean EPG was four-fold higher in juvenile horses than in adults and three-fold higher when the horses were dewormed with the use of fenbendazole instead of ivermectin or ivermectin with praziquantel combined.
Go to article

Bibliography


  1. Francisco I, Arias M, Cortinas FJ, Francisco R, Mochales E, Dacal V, Suárez JL, Uriarte J, Morrondo P, Sánchez-Andrade R, Díez-Banos P, Paz-Silva A (2009) Intrinsic factors influencing the infection by helminth parasites in horses under an oceanic climate area (NW Spain). J Parasitol Res 2009: 616173.
  2. Gawor J (1995) The prevalence and abundance of internal parasites in working horses autopsied in Poland. Vet Parasitol 58: 99-108.
  3. Gawor J, Kornaś S, Kharchenko V, Nowosad B, Skalska M (2006) Intestinal parasites and health problems in horses in different breeding systems (in Polish). Med Weter 62: 331-334.
  4. Kaplan RM (2002) Anthelmintic resistance in nematodes of horses. Vet Res 33: 491-507.
  5. Kaplan RM, Nielsen MK (2010) An evidence-based approach to equine parasite control: it ain’t the 60s anymore. Equine Vet Educ 22: 306-316.
  6. Kaplan RM, Vidyashankar AN (2012) An inconvenient truth: global worming and anthelmintic resistance. Vet Parasitol 186: 70-78.
  7. Kornaś S, Nowosad B, Skalska M (2004) Intestinal parasite infections in horses from different types of environments (in Polish). Med Weter 60: 853-856.
  8. Kornaś S, Cabaret J, Skalska M, Nowosad B (2010). Horse infection with intestinal helminths in relation to age, sex, access to grass and farm system. Vet Parasitol 174: 285-291.
  9. Kuzmina T, Kornaś S, Basiaga M, Kharchenko V, Vyniarska A (2011) Biodiversity of strongylids (Nematoda: Strongylidae) communities in domestic horses from Poland and Ukraine. Helminthologia 48: 77-84.
  10. Kuzmina TA. Dzeverin I, Kharchenko VA (2016) Strongylids in domestic horses: Influence of horse age, breed and deworming programs on the strongyle parasite community. Vet Parasitol 227: 56-63.
  11. Kuzmina TA, Kharchenko VO (2008) Anthelmintic resistance in cyathostomins of brood horses in Ukraine and influence of anthelmintic treatments on strongylid community structure. Vet Parasitol 154: 277-288.
  12. Lichtenfels JR, Kharchenko VA, Dvojnos GM. (2008) Illustrated identification keys to strongylid parasites (Strongylidae: Nematoda) of horses, zebras and asses (Equidae). Vet Parasitol 156: 4-161.
  13. Love S, Murphy D, Mellor D (1999) Pathogenicity of cyathostome infection. Vet Parasitol 85: 113-121.
  14. Lyons ET, Tolliver SC, Drudge JH (1999) Historical perspective of cyathostomes: prevalence, treatment and control programs. Vet Parasitol 85: 97-112.
  15. Martin F, Svansson V, Eydal M, Oddsdóttir C, Ernback M, Persson I, Tydén E (2021) First report of resistance to ivermectin in Parascaris univalens in Iceland. J Parasitol 107: 16-22.
  16. Osterman Lind E, Hoglund J, Ljungstrom BL, Nilson O, Uggla A (1999) A field survey on the distribution of strongyle infections of horses in Sweden and factors affecting faecal egg counts. Equine Vet J 31: 68-72.
  17. Petney TN, Andrews RM (1998) Multiparasite communities in animals and humans: frequency: structure and pathogenic significance. Int J Parasitol 28: 377-393.
  18. Roepstorff A, Nansen P (1998) Epidemiology, diagnosis and control of helminth parasites of swine. FAO Animal health manual. Food and Agriculture Organization of the United Nations, Rome, p 161.
  19. Saeed K, Qadir Z, Ashraf K, Ahmad N (2010) Role of intrinsic and extrinsic epidemiological factors on strongylosis in horses. J Anim Plant Sci 20: 277-280.
  20. Sallé G, Kornaś S, Basiaga M (2018) Equine strongyle communities are constrained by horse sex and species dipersal-fecundity trade-off. Parasit Vectors 11: 279.
  21. Selzer PM, Epe C (2021) Antiparasitics in animal health: Quo Vadis? Trends Parasitol 37(1): 77-89.
  22. Slivinska K, Kharchenko V, Wroblewski Z, Gawor J, Kuzmina T (2016) Parasitological survey of Polish primitive horses (Equus caballus gmelini Ant.): influence of age, sex and management strategies on the parasite community. Helminthologia 53: 233-242.
  23. Studzińska MB, Demkowska-Kutrzepa M, Bogucki J, Roczeń-Karczmarz M, Tomczuk K (2017) Influence of horse management systems in south-western Poland on the prevalence and intensity of gastrointestinal parasites (in Polish). Med Weter 73: 721-725.
  24. Vadlejch J, Petrtýl M, Zaichenko I, Čadková Z, Jankovská I, Langrová I, Moravec M (2011) Which McMaster egg counting technique is the most reliable. Parasitol Res 109: 1387-1394.
  25. Zak A, Siwinska N, Slowikowska M, Borowicz H, Kubiak K, Hildebrand J, Popiolek M, Niedzwiedz A (2017) Searching for ivermectin resistance in a Strongylidae population of horses stabled in Poland. BMC Vet Res 13: 210.
  26. Zeileis A, Kleiber C, Jackman S (2008) Regression Models for Count Data in R. J Stat Soft 27: 1-25.
Go to article

Authors and Affiliations

W. Hildebrand
1
P. Zielińska
2
J. Hildebrand
3
G. Zaleśny
4

  1. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wroclaw, Poland
  2. Department of Surgery, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Plac Grunwaldzki 51, 50-366 Wroclaw, Poland
  3. Department of Parasitology, Faculty of Biological Sciences, University of Wrocław, Przybyszewskiego 63, Wrocław, 51-148, Poland
  4. Department of Invertebrate Systematics and Ecology, Institute of Environmental Biology, Wroclaw University of Environmental and Life Sciences, Kożuchowska 5B, 51-631 Wroclaw, Poland
Download PDF Download RIS Download Bibtex

Abstract

Babesiosis is a parasitic disease caused by intraerythrocytic parasites of the genus Babesia, which infect both wild and domestic animals. Merozoite surface antigens (MSAs) have been identified as efficient immunogens in Babesia-infected animals. MSAs play a key role in the invasion process and have been proposed as potential targets for vaccine development. Epitope-based vaccines offer several advantages over whole protein vaccines as the immunogenic proteins are small and can induce both Th1 and Th2 immune responses, which are desirable for protection. However, the MSA, particularly gp45, is polymorphic in Babesia bigemina, posing a challenge to vaccine development. The purpose of this study was to develop a recombinant gpME (gp45-multi-epitope) for a vaccine against Babesia bigemina. B-cell, T-cell, and HLA epitope predictions were used to synthesize the gpME sequence from the consensus sequence of gp45. The gpME sequence was synthesized and cloned in the pET28α vector through the commercial biotechnology company to get pET28-gpME. The plasmid cloned with the gpME sequence comprising 1068 bp was expressed in a bacterial expression system. A band of 39 kDa of rec-gpME was obtained via SDS-PAGE and Western blotting. Rec-gpME @200ng was injected in calves 3 times at 2 weeks interval. The humoral response was evaluated through the indirect ELISA method. The ELISA with rec-gp45 protein showed a significant value of optical density. The recombinant protein containing multiple epitopes from the MSA gp45 may represent a promising candidate for a vaccine against Babesia bigemina.
Go to article

Bibliography

  1. Adu-Bobie J, Capecchi B, Serruto D, Rappuoli R, Pizza M (2003) Two years into reverse vaccinology. Vaccine 21: 605-610.
  2. Bock RE, De Vos AJ (2001) Immunity following use of Australian tick fever vaccine: a review of the evidence. Aust Vet J 79: 832-839.
  3. Callow LL, Dalgliesh RJ, De Vos AJ (1997) Development of effective living vaccines against Bovine Babesiosis – the longest field trial? Int J Parasitol 27: 747-767.
  4. Carcy B, Précigout E, Schetters T, Gorenflot A (2006) Genetic basis for GPI-anchor merozoite surface antigen polymorphism of Babesia and resulting antigenic diversity. Vet Parasitol 138: 33-49.
  5. Caro-Gomez E, Gazi M, Goez Y, Valbuena G (2014) Discovery of novel cross-protective Rickettsia prowazekii T-cell antigens using a combined reverse vaccinology and in vivo screening approach. Vaccine 32: 4968-4976.
  6. Çiloğlu A, Abdullah IN, Yildirim A, Önder Z, Düzlü Ö (2018) Molecular characterization and expression of the apical membrane antigen-1 from in vivo and in vitro isolates of Babesia bigemina Kayseri/Turkey strain. Ankara Univ Vet Fak Derg 65: 239-246.
  7. de Castro JJ (1997) Sustainable tick and tickborne disease control in livestock improvement in developing countries. Vet Parasitol 71: 77-97.
  8. Debierre-Grockiego F, Schwarz RT (2010) Immunological reactions in response to apicomplexan glycosylphosphatidylinositols. Glycobiology 20: 801-811.
  9. Debierre-Grockiego F, Smith TK, Delbecq S, Ducournau C, Lantier L, Schmidt J, Brès V, Dimier-Poisson I, Schwarz RT, Cornillot E (2019) Babesia divergens glycosylphosphatidylinositols modulate blood coagulation and induce Th2-biased cytokine profiles in antigen presenting cells. Biochimie 167: 135-144.
  10. Delbecq S, Hadj-Kaddour K, Randazzo S, Kleuskens J, Schetters T, Gorenflot A, Précigout E (2006) Hydrophobic moeties in recombinant proteins are crucial to generate efficient saponin-based vaccine against Apicomplexan Babesia divergens. Vaccine 24: 613-621.
  11. Djokic V, Akoolo L, Parveen N (2018) Babesia microti infection changes host spleen architecture and is cleared by a Th1 immune response. Front Microbiol 9:85
  12. Durrani A, Kamal N (2008) Identification of ticks and detection of blood protozoa in Friesian cattle by polymerase chain reaction test and estimation of blood parameters in district Kasur, Pakistan. Trop Anim Health Prod 40: 441-447.
  13. Echaide IE, De Echaide ST, Guglielmone AD (1993) Live and soluble antigens for cattle protection to Babesia bigemina. Vet Parasitol 51: 35-40.
  14. Esmaeilnejad B, Tavassoli M, Asri-Rezaei S, Dalir-Naghadeh B, Mardani K, Golabi M, Arjmand J, Kazemnia A, Jalilzadeh G (2015) Determination of prevalence and risk factors of infection with Babesia ovis in small ruminants from West Azerbaijan Province, Iran by polymerase chain reaction. J Arthropod Borne Dis 9: 246-252.
  15. Ezediuno LO, Onile OS, Oladipo EK, Majolagbe ON, Jimah EM, Senbadejo TY (2021) Designing multi-epitope subunit vaccine for ocular trachoma infection using Chlamydia trachomatis polymorphic membrane proteins G. Inform Med Unlocked 26: 100764.
  16. Fisher TG, McElwain TF, Palmer GH (2001) Molecular basis for variable expression of merozoite surface antigen gp45 among American isolates of Babesia bigemina. Infect Immun 69: 3782-3790.
  17. Gaafar B, Ali SA, Abd-Elrahman KA, Almofti YA (2019) Immunoinformatics approach for multiepitope vaccine prediction from H, M, F, and N proteins of Peste des Petits ruminants virus. J Immunol Res 2019. 6124030
  18. Gomara MJ, Haro I (2007) Synthetic peptides for the immunodiagnosis of human diseases. Curr Med Chem 14: 531-546.
  19. Hajissa K, Zakaria R, Suppian R, Mohamed Z (2019) Epitope-based vaccine as a universal vaccination strategy against Toxoplasma gondii infection: A mini-review. J Adv Vet Anim Res 6: 174-182.
  20. Jabbar A, Abbas T, Sandhu Z, Saddiqi HA, Qamar MF, Gasser RB (2015) Tick-borne diseases of bovines in Pakistan: major scope for future research and improved control. Parasit Vectors 8:.283
  21. Jorgensen WK, De Vos AJ, Dalgliesh RJ (1989) Comparison of immunogenicity and virulence between Babesia bigemina parasites from continuous culture and from a splenectomised calf. Aust Vet J 66: 371-372.
  22. Kalita J, Padhi AK, Tripathi T (2020a) Designing a vaccine for fascioliasis using immunogenic 24 kDa mu-class glutathione s-transferase. Infect Genet Evol 83: 104352.
  23. Kalita P, Padhi AK, Zhang KY, Tripathi T (2020b) Design of a peptide-based subunit vaccine against novel coronavirus SARS-CoV-2. Microb Pathog 145: 104236.
  24. Kar PP, Srivastava A (2018) Immuno-informatics analysis to identify novel vaccine candidates and design of a multi-epitope based vaccine candidate against Theileria parasites. Front Immunol 9: 2213.
  25. Karim S, Budachetri K, Mukherjee N, Williams J, Kausar A, Hassan MJ, Adamson S, Dowd SE, Apanskevich D, Arijo A, Sindhu ZU (2017) A study of ticks and tick-borne livestock pathogens in Pakistan. PLOS Negl Trop Dis 11: e0005681.
  26. Kearse M, Moir R, Wilson A, Stones-Havas S, Cheung M, Sturrock S, Buxton S, Cooper A, Markowitz S, Duran C, Thierer T ( 2012) Geneious Basic: an integrated and extendable desktop software platform for the organization and analysis of sequence data. Bioinformatics 28: 1647-1649.
  27. Khatoon N, Ojha R, Mishra A, Prajapati VK (2018) Examination of antigenic proteins of Trypanosoma cruzi to fabricate an epitope-based subunit vaccine by exploiting epitope mapping mechanism. Vaccine 36: 6290-6300.
  28. Klafke G, Webster A, Agnol BD, Pradel E, Silva J, de La Canal LH, Becker M, Osório MF, Mansson M, Barreto R, Scheffer R (2017) Multiple resistance to acaricides in field populations of Rhipicephalus microplus from Rio Grande do Sul state, Southern Brazil. Ticks Tick Borne Dis 8: 73-80.
  29. Kumar P, Lata S, Shankar UN, Akif M (2021) Immunoinformatics based designing of a multi- epitope chimeric vaccine from multi-domain outer surface antigens of Leptospira. Front Immunol 12: 735373.
  30. Lew-Tabor AE, Valle MR (2016) A review of reverse vaccinology approaches for the development of vaccines against ticks and tick borne diseases. Ticks Tick Borne Dis 7: 573-585.
  31. List C, Qi W, Maag E, Gottstein B, Müller N, Felger I (2010) Serodiagnosis of Echinococcus spp. infection: explorative selection of diagnostic antigens by peptide microarray. PLOS Negl Trop Dis 4: e771.
  32. Maiorano AM, Giglioti R, Oliveira MC, Oliveira HN, Cyrillo JN, Mercadante ME, Silva JA (2018) Resistance to the tick Rhipicephalus microplus and Babesia bovis infection levels in beef heifers raised in an endemic area of Sao Paulo state, Brazil Anim Prod Sci 59: 938-944.
  33. Mangold AJ, Vanzini VR, Echaide IE, De Echaide ST, Volpogni MM, Guglielmone AA (1996) Viability after thawing and dilution of simultaneously cryopreserved vaccinal Babesia bovis and Babesia bigemina strains cultured in vitro. Vet Parasitol 61: 345- 348.
  34. McElwain TF, Perryman LE, Musoke AJ, McGuire TC (1991) Molecular characterization and immunogenicity of neutralization-sensitive Babesia bigemina merozoite surface proteins. Mol Biochem Parasitol 47: 213-222.
  35. Mehla K, Ramana J (2016) Identification of epitope-based peptide vaccine candidates against enterotoxigenic Escherichia coli: a comparative genomics and Immunoinformatics approach. Mol Biosyst 12: 890-901.
  36. Menezes-Souza D, Mendes TA, Nagem RA, Santos TT, Silva AL, Santoro MM, de Carvalho SF, Coelho EA, Bartholomeu DC, Fujiwara RT (2014) Mapping B-cell epitopes for the peroxidoxin of Leishmania (Viannia) braziliensis and its potential for the clinical diagnosis of tegumentary and visceral leishmaniasis. PloS One 9: e99216.
  37. Mercado-Uriostegui MA, Castro-Sánchez LA, Batiha GE, Valdez-Espinoza UM, Falcón-Neri A, Ramos-Aragon JA, Hernández-Ortiz R, Kawazu SI, Igarashi I, Mosqueda J (2022) The GP-45 Protein, a Highly Variable Antigen from Babesia bigemina, Contains Conserved B-Cell Epitopes in Geographically Distant Isolates. Pathogens 11: 591.
  38. Mishra VS, McElwain TF, Dame JB, Stephens EB (1992) Isolation, sequence and differential expression of the p58 gene family of Babesia bigemina. Mol Biochem Parasitol 53: 149-158.
  39. Mosqueda J, Olvera-Ramirez A, Aguilar-Tipacamu G, Canto GJ (2012) Current advances in detection and treatment of babesiosis. Curr Med Chem 19: 1504-1518.
  40. Moubri K, Kleuskens J, Van de Crommert J, Scholtes N, Van Kasteren T, Delbecq S, Grenflot A, Schetters T (2018) Discovery of a recombinant Babesia canis supernatant antigen that protects dogs against virulent challenge infection. Vet Parasitol 249: 21-29.
  41. Mucci J, Carmona SJ, Volcovich R, Altcheh J, Bracamonte E, Marco JD, Nielsen M, Buscaglia CA, Agüero F (2017) Next-generation ELISA diagnostic assay for Chagas Disease based on the combination of short peptidic epitopes. PLoS Negl Trop Dis 11: e0005972.
  42. Nabi H, Rashid I, Ahmad N, Durrani A, Akbar H, Islam S, Bajwa AA, Shehzad W, Ashraf K, Imran N (2017) Induction of specific humoral immune response in mice immunized with ROP18 nanospheres from Toxoplasma gondii. Parasitol Res 116: 359-370.
  43. Naeem H, Sana M, Islam S, Khan M, Riaz F, Zafar Z, Akbar H, Shehzad W, Rashid MI (2018) Induction of Th1 type-oriented humoral response through intranasal immunization of mice with SAG1- Toxoplasma gondii polymeric nanospheres. Artif Cells Nanomed Biotechnol 46: 1025-1034.
  44. Patarroyo MF, Cifuentes G, Bermudez A, Patarroyo MA (2008) Strategies for developing multi‐epitope, subunit‐based, chemically synthesized anti‐malarial vaccines. J Cell Mol Med 12: 1915-1935.
  45. Rahman SU, Akbar H, Shabbir MZ, Ullah U, Rashid MI (2021) Development of Human Toxo IgG ELISA Kit, and False-Positivity of Latex Agglutination Test for the Diagnosis of Toxoplasmosis. Pathogens 10: 1111.
  46. Rashid I, Hedhli D, Moiré N, Pierre J, Debierre-Grockiego F, Dimier-Poisson I, Mévélec MN (2011) Immunological responses induced by a DNA vaccine expressing RON4 and by immunogenic recombinant protein RON4 failed to protect mice against chronic toxoplasmosis. Vaccine 29: 8838-8846.
  47. Rauf U, Shabir S, Khan M, Rashid MI, Akbar H, Durrani AZ (2020) Identification of 23 kD immunogen from native antigens of Babesia bigemina in splenectomized calf. Int J Agric Biol 24: 1788-1794.
  48. Rauf U, Suleman M, Abid A, Jamil H, Menghwar H, Durrani AZ, Rashid MI, Akbar H (2020) Humoral and cell-mediated immune response validation in calves after a live attenuated vaccine of Babesia bigemina. Pathogens 9: 936.
  49. Rehman A, Conraths FJ, Sauter‐Louis C, Krücken J, Nijhof AM (2019) Epidemiology of tick‐ borne pathogens in the semi‐arid and the arid agro‐ecological zones of Punjab province, Pakistan. Transbound Emerg Dis 66: 526-536.
  50. Rodríguez-Camarillo SD, Quiroz-Castañeda RE, Aguilar-Díaz H, Vara-Pastrana JE, Pescador- Pérez D, Amaro-Estrada I, Martínez-Ocampo F (2020) Immunoinformatic analysis to identify proteins to be used as potential targets to control bovine anaplasmosis. Int J Microbiol 2020: 88.
  51. Islam MS, Aryasomayajula A, Selvaganapathy PR (2017) A review on macroscale and microscale cell lysis methods. Micromachines 8: 83
  52. Shkap V, Leibovitz B, Krigel Y, Hammerschlag J, Marcovics A, Fish L, Molad T, Savitsky I, Mazuz M (2005) Vaccination of older Bos taurus bulls against bovine babesiosis. Vet Parasitol 129: 235-242.
  53. Siddique RM, Sajid MS, Iqbal Z, Saqib M (2020) Association of different risk factors with the prevalence of babesiosis in cattle and buffalos. Pak J Agri Sci 57: 517-524.
  54. Silva MG, Knowles DP, Suarez CE (2016) Identification of interchangeable cross-species function of elongation factor-1 alpha promoters in Babesia bigemina and Babesia bovis. Parasit Vectors 9: 576.
  55. Singh H, Mishra AK, Rao JR, Tewari AK (2009) Comparison of indirect fluorescent antibody test (IFAT) and slide enzyme linked immunosorbent assay (SELISA) for diagnosis of Babesia bigemina infection in bovines. Trop Anim Health Prod 41: 153- 159.
  56. Teimouri A, Modarressi MH, Shojaee S, Mohebali M, Rezaian M, Keshavarz H (2019) Development, optimization, and validation of an in-house Dot-ELISA rapid test based on SAG1 and GRA7 proteins for serological detection of Toxoplasma gondii infections. Infect Drug Resist 12: 2657-2669.
  57. Tuvshintulga B, Sivakumar T, Yokoyama N, Igarashi I (2019) Development of unstable resistance to diminazene aceturate in Babesia bovis. Int J Parasitol: Drugs Drug Resist 9: 87-92.
  58. Uilenberg G (2006) Babesia—a historical overview. Vet Parasitol 138: 3-10.
  59. Wagner GG, Holman P, Waghela S (2002) Babesiosis and heart water: threats without boundaries. Vet Clin North Am Food Anim Pract 18: 417-430.
  60. Wieser SN, Schnittger L, Florin-Christensen M, Delbecq S, Schetters T (2019) Vaccination against babesiosis using recombinant GPI-anchored proteins. Int J Parasitol 49: 175-181.
  61. Wright IG, Goodger BV, Leatch G, Aylward JH, Rode-Bramanis K, Waltisbuhl DJ (1987) Protection of Babesia bigemina-immune animals against subsequent challenge with virulent Babesia bovis. Infect Immun 55: 364-368.
  62. Yadav S, Prakash J, Shukla H, Das KC, Tripathi T, Dubey VK (2020) Design of a multi- epitope subunit vaccine for immune-protection against Leishmania parasite. Pathog Glob Health 114: 471-481.
  63. Yoshida M, Reyes SG, Tsuda S, Horinouchi T, Furusawa C, Cronin L (2017) Time- programmable drug dosing allows the manipulation, suppression and reversal of antibiotic drug resistance in vitro. Nat Commun 8: 15589.
Go to article

Authors and Affiliations

Z. Ul Rehman
1
M. Suleman
2
K. Ashraf
1
S Ali
1
S. Rahman
1
M.I. Rashid
1

  1. Department of Parasitology, University of Veterinary and Animal Sciences, Syed Abdul Qadir Jillani (Out Fall) Road, Lahore, 54000, Pakistan
  2. University Diagnostic Laboratory, Institute of Microbiology, University of Veterinary and Animal Sciences, Syed Abdul Qadir Jillani (Out Fall) Road, Lahore, 54000, Pakistan
Download PDF Download RIS Download Bibtex

Abstract

This study analyzed the internal and external quality traits of eggs derived from hens of different breeds, including Silkie, Sultan, Cochin Bantam, Brahma and White Leghorn. The highest mean weight was noted for eggs originating from the White Leghorns breed, and the lowest was for eggs from the Cochin Bantams. Simultaneously, both a positive correlation between the egg weight and the percentage of albumen (r = 0.876) and a negative correlation between egg weight and the percentage of yolk (r = - 0.842) were found. The eggshell composition varied significantly in mean phosphorus amount, whereas the calcium content did not differ significantly. Despite this, eggshell strength varied significantly between breeds. Regarding cholesterol and fatty acid levels, the highest amount of cholesterol was noted in the Cochin Bantam breed, and the lowest was in the White Leghorn, although Leghorn was the breed characterized by the highest saturated fatty acid levels, and Cochin Bantam was the lowest. Regarding the polyunsaturated fatty acids (which have been proven to positively influence the cardiovascular system), the highest levels were obtained by Leghorn eggs, and the lowest were obtained by Silkie eggs. In conclusion, the study indicates that ornamental chicken breeds are a source of high-quality products, which could be attractive to consumers, additionally supporting traditional farming and animal genetic resources.
Go to article

Bibliography

  1. Ajayi FO (2010) Nigerian indigenous chicken: A valuable genetic resource for meat and egg production. Asian J Poult Sci 4: 164-172.
  2. Allonby JIH, Wilson PB (2018) Large fowls and bantams. In: British poultry standards: complete specifications and judging points of all standardized breeds and varieties of poultry as compiled by the specialist affiliated breed clubs and recognized by the Poultry Club of Great Britain. Wiley-Blackwell, UK, pp 31-344.
  3. Altuntaş E, Şekeroğlu A (2008) Effect of egg shape index on mechanical properties of chicken eggs. J Food Eng 85: 606 - 612.
  4. Arpášová H, Halaj M, Halaj P (2010) Eggshell quality and calcium utilization in feed of hens in repeated laying cycles. Czech J Anim Sci 55: 66-74.
  5. Arthur JA, O’Sullivan N (2005) Breeding chickens to meet egg quality needs. Int Hatch Pract 19:7-9.
  6. Aygun A, Yetisir R (2010) The Relationship among egg quality characteristics of different hybrid layers to forced molting programs with and without feed withdrawal. J Anim Vet Adv 9 : 710-715.
  7. Bain MM (2005) Recent advances in the assessment of eggshell quality and their future application. Worlds Poult Sci J 61: 268-277.
  8. Basmacioglu H, Ergul M (2005) Characteristic of egg in laying hens. The effect of genotype and rearing system. Turk J Vet Anim Sci 29: 157-164.
  9. Bernacki Z, Kaszynski B (2013) Assessment of egg quality and hatch results of different origin hens. Acta Sci Pol Zootech 12: 3-14.
  10. Campbell AM, Johnson AM, Persia ME, Jacobs L (2022) Effects of Housing System on Anxiety, Chronic Stress, Fear, and Immune Function in Bovan Brown Laying Hens. Animals (Basel) 12: 1803.
  11. Crowe-White KM, Cardel MI, Burkhalter HH, Huo T, Fernández JR (2018) Higher n-6: n-3 Fatty Acid Intake Is Associated with Decreased Cardiometabolic Risk Factors in a Racially Diverse Sample of Children. Current Dev Nutr 2: nzy014.
  12. Drabik K, Karwowska M, Wengerska K, Próchniak T, Adamczuk A, Batkowska J (2021) The Variability of Quality Traits of Table Eggs and Eggshell Mineral Composition Depending on Hens’ Breed and Eggshell Color. Animals (Basel) 11: 1204.
  13. Drażbo A, Mikulski D, Zduńczyk Z, Szmatowicz B, Rutkowski A, Jankowski J (2014) Fatty acid composition, physicochemical and sensory properties of eggs from laying hens fed diets containing blue lupine seeds. Europ Poult. Sci 78: 245-252.
  14. Duman M, Sekeroglu A, Yildirim A, Eleroglu H, Camci O (2016) Relation between egg shape index and egg quality characteristics. Europ Poult Sci 80: 1-9.
  15. Enjoji M, Nakamuta M (2010) Is the control of dietary cholesterol intake sufficiently effective to ameliorate nonalcoholic fatty liver disease? World J Gastroenterol 16: 800-803.
  16. Escobedo del Bosque CI, Spiller A, Risius A (2021) Who Wants Chicken? Uncovering Consumer Preferences for Produce of Alternative Chicken Product Methods. Sustainability 13: 2440.
  17. European Commission. Agriculture and rural development. Available online: https://agriculture.ec.europa.eu/farming/animal-products/eggs_en (accessed on 16th October 2022).
  18. Folch J, Lees M, Sloane Stanley GH (1957) A simple method for the isolation and purification of total lipids from animal tissues. J Biol Chem 226: 497-509.
  19. Fraeye I, Bruneel C, Lemahieu C, Buyse J, Muylaert K, Foubert I (2012) Dietary enrichment of eggs with omega-3 fatty acids: A review. Food Res Int 48: 961-969.
  20. Fraser D (2008) Animal welfare and the intensification of animal production. In: Thompson PB (ed) Ethics of Intensification: Agricultural Development and Cultural Change; FAO, Italy, pp 167-189.
  21. Funk EM (1948) The relation of the Yolk Index Determined in Natural Position to the Yolk Index as Determined after Separating the Yolk from the Albumen. Poult Sci 27: 367.
  22. Goto T, Tsudzuki M (2017) Genetic mapping of quantitative trait loci for egg production and egg quality traits in chickens: A review. J Poult Sci 54: 1-12.
  23. Grashorn M (2016) Feed additives for influencing chicken meat and egg yolk color. In: Carle R, Schweiggert R (eds) Handbook on Natural Pigments in Food and Beverages. Woodhead Publishing: United Kingdom, pp 283-302.
  24. Hamady GA (2013) Effects of different ratios of dietary omega-6 to omega-3 fatty acids on laying performance and egg quality of Lohmann brown hens. Egypt Poult Sci J 33: 957-969.
  25. Hamidu JA, Brown CA, Adjepong M (2022) Improving the cognitive development of children in rural areas as development tool. In: de Salvo P, Vaquero Pineiro M (eds) Rural Development - Education, Sustainability, Multifunctionality. IntechOpen, United Kingdom, pp 67-82.
  26. Hammershøj M, Johansen NF (2016) Review: The effect of grass and herbs in organic egg production on egg fatty acid composition, egg yolk colour and sensory properties. Livest Sci 194: 37-43.
  27. Hanusova E, Hrnčár C, Hanus A, Oravcová M (2015) Effect of breed on some parameters of egg quality in laying hens. Acta Fytot Zoo 18: 20-24.
  28. Haugh H (1937) The Haugh unit for measuring egg quality. US Egg Poult Mag 43: 552-555, 572-573.
  29. Heng Y, Peterson HH, Li X (2013) Consumer attitudes toward farm-animal welfare: the case of laying hens. J Agric Res Econ 38: 418-434.
  30. Hisasaga C, Griffin SE, Tarrant KJ (2020) Survey of egg quality in commercially available table eggs. Poult Sci 99: 7202-7206.
  31. Hrnčár C, Gašparovič M, Gálik B, Bujko, J (2015) Egg Traits, Fertility and Hatchability of Brahma, Cochin and Orpington Chicken Breeds. Anim Sci Biotechnol 48: 137-141.
  32. Hrnčár C, Hässlerová M, Bujko J (2013) The effect of oviposition time on egg quality parameters in Brown Leghorn, Oravka and Brahma hens. Anim Sci Biotechnol 46: 53-57.
  33. Ianni A, Bartolini D, Bennato F, Martino G (2021) Egg quality from Nera Atriana, a local poultry breed of the Abruzzo Region (Italy), and ISA Brown hens reared under free range conditions. Animals (Basel) 11: 257.
  34. International Atomic Energy Agency. Genetic characterization of indigenous chicken breeds in search for unique properties of immune-related genes. Available online: https://www.iaea.org/resources/news-article/genetic-characterizationof-indigenous-chicken-breeds-in-search-for-unique-propertiesof-immune-related-genes (accessed on 2nd December 2022)
  35. International Dairy Federation (1992) Provisional Standard (159): Milk and Milk Fat Product-Determination of Cholesterol Content; IDF: Brussels, Belgium.
  36. Jones D (2012) Haugh unit: Gold standard of egg quality. Natl Egg Quality Sch Proc 7: 47-51.
  37. Jones DR, Musgrove MT (2005) Effects of extended storage on egg quality factors. Poult Sci 84: 1774-1777.
  38. Kaszynski B, Bernacki Z (2014) Assessment of egg quality and hatch results of two show hen breeds raised for fancy. J Cent Eur Agric 15: 1-11.
  39. Kaur N, Chugh V, Gupta AK (2014) Essential fatty acids as functional components of foods- A review. J Food Sci Technol 51: 2289-2303.
  40. Ketta M, Tumova E (2017) Eggshell characteristics and cuticle deposition in three laying hen genotypes housed in enriched cages and on litter. Czech J Anim Sci 63: 11-16.
  41. Kosewski G, Kowalówka M, Dobrzyńska M, Jagielski P, Przysławski J (2021) Profile of fatty acids in the yolks of chicken eggs, including DHA, depending on hen breeding method. Bromatol Chem Toksykol 54: 66-72.
  42. Krauss RM, Eckel RH, Howard B, Appel LJ, Daniels SR, Deckelbaum RJ, Erdman JW Jr, Kris-Etherton P, Goldberg IJ, Kotchen TA, Lichtenstein AH, Mitch WE, Mullis R, Robinson K, Wylie-Rosett J, St Jeor S, Suttie J, Tribble DL, Bazzarre TL (2001) Revision 2000: a statement for healthcare professionals from the Nutrition Committee of the American Heart Association. J Nutr 131: 132-146.
  43. Krawczyk J (2017) Effect of genotype and age of hens on egg quality of Leghorn hens (lines G-99 and H-22) and Sussex (line S-66). Apar Badaw Dydakt 22: 94-100. (in Polish).
  44. Lemos Teixeira D, Larraín R, Hötzel MJ (2018) Are views towards egg farming associated with Brazilian and Chilean egg consumers’ purchasing habits? PLoS One 13: e0203867.
  45. Liswaniso S, Qin N, Shan X, Im C, Sun X, Xu R (2020) Quality Characteristics, Phenotypic correlations and Principal Component Analysis of Indigenous Free Range Chicken Eggs in Lusaka; Zambia Int J Environ Agri Res 6: 29-35.
  46. Lordelo M, Cid J, Cordovil CM, Alves SP, Bessa RJ, Carolino I (2020) A comparison between the quality of eggs from indigenous chicken breeds and that from commercial layers. Poult Sci 99: 1768-1776.
  47. Lusk JL (2019) Consumer preferences for cage-free eggs and impacts of retailer pledges. Agribusiness 35: 129-148.
  48. Malomane DK, Simianer H, Weigend A, Reimer C, Schmitt AO, Weigend S (2019) The SYNBREED chicken diversity panel: a global resource to assess chicken diversity at high genomic resolution. BMC Genom 20: 345.
  49. Mariamenatu AH, Abdu EM (2021) Overconsumption of omega-6 polyunsaturated fatty acids (PUFAs) versus deficiency of omega-3 PUFAs in modern-day diets: the disturbing factor for their “balanced antagonistic metabolic functions” in the human body. J Lipids: 8848161.
  50. Milinsk MC, Murakami AE, Gomes STM, Matsushita M, de Souza NE (2003) Fatty acid profile of egg yolk lipids from hens fed diets rich in n-3 fatty acids. Food Chem 83: 287-292.
  51. Moula N, Antoine-Moussiaux N, Farnir F, Leroy P (2009) Comparison of egg composition and conservation ability in two Belgian local breeds and one commercial strain. Int J Poult Sci 8: 768-774.
  52. Neijat M, Ojekudo O, House JD (2016) Effect of flaxseed oil and microalgae DHA on the production performance, fatty acids and total lipids of egg yolk and plasma in laying hens. Prostaglandins Leukot. Essent. Fatty Acids 115: 77-88.
  53. Neunzehn J, Szuwart T, Wiesmann HP (2015) Eggshells as natural calcium carbonate source in combination with hyaluronan as beneficial additives for bone graft materials, an in vitro study. Head Face Med 11: 11-12.
  54. Nogueira GC, Bragagnolo N (2002) Assessment of methodology for the enzymatic assay of cholesterol in egg noodles. Food Chem 79: 267-270.
  55. Nolte T, Jansen S, Weigend S, Moerlein D, Halle I, Simianer H, Sharifi AR (2021) Genotypic and Dietary Effects on Egg Quality of Local Chicken Breeds and Their Crosses Fed with Faba Beans. Animals (Basel) 11: 1947.
  56. Nys Y, Sauveur B (2004) Valeur nutritionnelle des oeufs. Prod Anim 17: 385-393.
  57. Park JA, Sohn SH (2018) The Influence of Hen Aging on Eggshell Ultrastructure and Shell Mineral Components. Korean J Food Sci Anim Resour 38: 1080-1091.
  58. Patterson E, Wall R, Fitzgerald GF, Ross RP, Stanton C (2012) Health implications of high dietary omega-6 polyunsaturated fatty acids. J Nutr Metab 2012: 539426.
  59. Polat ES, Citil OB, Garip M (2013) Fatty acid composition of yolk of nine poultry species kept in their natural environment. Anim Sci Pap Rep 31: 363-368.
  60. Rafea MT (2019) Prediction of the Haugh unit through albumen height and egg weight. Mesop J Agric 47: 37-43
  61. Réhault-Godbert S, Guyot N, Nys Y (2019) The Golden Egg: Nutritional Value, Bioactivities, and Emerging Benefits for Human Health. Nutrients 11: 684.
  62. Rizzi C, Chiericato GM (2010) Chemical composition of meat and egg yolk of hybrid and Italian breed hens reared using an organic production system. Poult Sci 89: 1239-1251.
  63. Rodríguez-Navarro A, Kalin O, Nys Y, Garcia-Ruiz JM (2002) Influence of the microstructure on the shell strength of eggs laid by hen of different ages. Br Poult Sci 43: 395-403.
  64. Rodriguez-Navarro AB, Yebra A, Nys Y, Jimenez-Lopez C, Garcia-Ruiz JM (2007) Analysis of avian eggshell microstructure using X-ray area detectors. Eur J Mineral 19: 391-398.
  65. Sanlier N, Üstün D (2021) Egg consumption and health effects: A narrative review. J Food Sci 86: 4250-4261.
  66. Shaker AS, Kirkuki SM, Aziz SR, Jalal BJ (2017) Influence of genotype and hen age on the egg shape index. Int J Biochem Biophy Mol Biol 2: 68-70.
  67. Shi SR, Wang KH, Dou TC, Yang HM (2009) Egg weight affects some quality traits of chicken eggs. J Food Agr Environ 7: 432-434.
  68. Song KT, Choi SH, Oh HR (2000) A comparison of egg quality of pheasant, chukar, quail and guinea fowl. Asian-Aus. J Anim Sci 13: 986-990.
  69. Stadelman WJ (1995) Quality identification of shell eggs. In: Stadelman WJ, Cotterill OJ (eds) Egg science and technology. Food Products Press: USA, pp 39-66.
  70. Świątkiewicz S, Arczewska-Włosek A, Krawczyk J, Szczurek W, Puchała M, Józefiak D (2018) Effect of selected feed additives on egg performance and eggshell quality in laying hens fed a diet with standard or decreased calcium content. Ann Anim Sci 18: 167-183.
  71. U.S. Department of Agriculture. Agricultural Research Service, FoodData Central. Available online: https://fdc.nal.usda.gov/fdc-app.html#/food-details/748967/nutrients (accessed on 16th October 2022).
  72. Wilson PB (2017) Recent advances in avian egg science: A review. Poult Sci 96: 3747-3754.
  73. Yan YY, Sun CJ, Lian L, Zheng JX, Xu GY, Yang N (2014) Effect of uniformity of eggshell thickness on eggshell quality in chickens. J Poult Sci 51: 338-342.
  74. Zemková Ľ, Simeonovová J, Lichovníková M, Somerlíková K (2007) The effects of housing systems and age of hens on the weight and cholesterol concentration of the egg. Czech J Anim Sci 52: 110-115.
Go to article

Authors and Affiliations

J. Tyc
1
B. Wysok
1
A. Drażbo
2
J. Naczmański
2
Ł. Szymański
2

  1. Department of Veterinary Public Health, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 14, Olsztyn, 10-718, Poland
  2. Department of Poultry Science, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Oczapowskiego 14, Olsztyn, 10-718, Poland
Download PDF Download RIS Download Bibtex

Abstract

The main purpose of the study was to determine the safety of oclacitinib (OCL), a Janus kinase inhibitor, with respect of its effect on CD4 + and CD8 + T cells as well as B cells in the lymphoid tissue. The mice were treated orally with OCL at a dose of 2.7 mg/kg for 14 days and peripheral blood, head and neck lymph nodes (HNLNs), mediastinal lymph nodes (MLNs) and spleen were collected. The study found that OCL induced depletion of CD4 + T cells in the HNLNs and MLNs, while it did not affect the absolute count of CD8 + T cells in these tissues. Also OCL caused a loss of B cells in the HNLNs, although not in the MLNs. Moreover, OCL depleted B cells in the peripheral blood, but did not affect the absolute count of CD4 + and CD8 + T cells. Thus, it can be concluded that OCL may induce a depletive effect on CD4 + and CD8 + T cells as well as B cells in the lymphoid tissue. This effect should be seen as an unfavorable one, especially in patients with infections. Therefore, a clinical implication is that in such patients, the benefit/risk ratio should be thoroughly considered by clinicians. Moreover, OCL reduced the absolute count of eosinophils, basophils, neutrophils and monocytes. However, it is uncertain whether this effect should be considered to be of clinical importance because the levels of these cells were within the physiological range. It is possible that the depletive effect of OCL toward T and B cells, as well as eosinophils and basophils may contribute to the beneficial effects of the drug in the treatment of skin allergic diseases.
Go to article

Bibliography

  1. Apoquel® Summary of Product Characteristics available here: www.ema.europa.eu/en/documents/product-information/apoquel-epar-product-information_en.pdf.
  2. Banovic F, Tarigo J, Gordon H, Barber JP, Gogal RM Jr (2019) Immunomodulatory in vitro effects of oclacitinib on canine T-cell proliferation and cytokine production. Vet Dermatol 30: 17-e6.
  3. Barry M, Bleackley RC (2002) Cytotoxic T lymphocytes: all roads lead to death. Nat Rev Immunol 2: 401-409.
  4. Benucci M, Bernardini P, Coccia C, De Luca R, Levani J, Economou A, Damiani A, Russo E, Amedei A, Guiducci S, Bartoloni E, Manfredi M, Grossi V, Infantino M, Perricone C (2023) JAK inhibitors and autoimmune rheumatic diseases. Autoimmun Rev 22: 103276.
  5. Cetkovic-Cvrlje M, Olson M, Ghate K (2012) Targeting Janus tyrosine kinase 3 (JAK3) with an inhibitor induces secretion of TGF-β by CD4+ T cells. Cell Mol Immunol 9: 350-360.
  6. De Caro Martins G, da Costa-Val AP, Coura FM, Diamantino GM, Nogueira MM, de Oliveira Melo-Junior OA, Giunchetti RC, da Silveira-Lemos D, Melo MM (2022) Immunomodulatory effect of long-term oclacitinib maleate therapy in dogs with atopic dermatitis. Vet Dermatol 33: 142-e40.
  7. Denti D, Caldin M, Ventura L, De Lucia M (2022) Prolonged twice-daily administration of oclacitinib for the control of canine atopic dermatitis: a retrospective study of 53 client-owned atopic dogs. Vet Dermatol 33: 149-e42.
  8. Gonzales AJ, Bowman JW, Fici GJ, Zhang M, Mann DW, Mitton-Fry M (2014) Oclacitinib (APOQUEL(®)) is a novel Janus kinase inhibitor with activity against cytokines involved in allergy. J Vet Pharmacol Ther 37: 317-324.
  9. Gottlieb SL, Martin DH, Xu F, Byrne GI, Brunham RC (2010) Summary: The natural history and immunobiology of Chlamydia trachomatis genital infection and implications for Chlamydia control. J Infect Dis 201 Suppl 2: S190-204.
  10. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, van Lier RA (1997) Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med 186: 1407-1418.
  11. Hashimoto T, Yokozeki H, Karasuyama H, Satoh T (2023) IL-31–generating network in atopic dermatitis comprising macrophages, basophils, thymic stromal lymphopoietin, and periostin. J Allergy Clin Immunol 151: 737-746.
  12. James JM, Kagey-Sobotka A, Sampson HA (1993) Patients with severe atopic dermatitis have activated circulating basophils. J Allergy Clin Immunol 91: 1155-1162.
  13. Jasiecka-Mikołajczyk A, Jaroszewski JJ, Maślanka T (2018) Oclacitinib depletes canine CD4+ and CD8+ T cells in vitro. Res Vet Sci 121: 124-129.
  14. Jasiecka-Mikołajczyk A, Jaroszewski JJ, Maślanka T (2021) Oclacitinib, a Janus Kinase Inhibitor, Reduces the Frequency of IL-4- and IL-10-, but Not IFN-γ-, Producing Murine CD4+ and CD8+ T Cells and Counteracts the Induction of Type 1 Regulatory T Cells. Molecules 26: 5655.
  15. Lee S, Shah T, Yin C, Hochberg J, Ayello J, Morris E, van de Ven C, Cairo MS (2018) Ruxolitinib significantly enhances in vitro apoptosis in Hodgkin lymphoma and primary mediastinal B-cell lymphoma and survival in a lymphoma xenograft murine model. Oncotarget 9: 9776-9788.
  16. Majewska A, Dembele K, Dziendzikowska K, Prostek A, Gajewska M (2022) Cytokine and Lymphocyte Profiles in Dogs with Atopic Dermatitis after Allergen-Specific Immunotherapy. Vaccines (Basel) 10: 1037.
  17. Majewska A, Gajewska M, Dembele K, Maciejewski H, Prostek A, Jank M (2016) Lymphocytic, cytokine and transcriptomic profiles in peripheral blood of dogs with atopic dermatitis. BMC Vet Res 12: 174.
  18. Maślanka T, Otrocka-Domagała I, Zuśka-Prot M, Mikiewicz M, Przybysz J, Jasiecka A, Jaroszewski JJ (2016) IκB kinase β inhibitor, IMD-0354, prevents allergic asthma in a mouse model through inhibition of CD4(+) effector T cell responses in the lung-draining mediastinal lymph nodes. Eur J Pharmacol 775: 78-85.
  19. Nuttall TJ, Knight PA, McAleese SM, Lamb JR, Hill PB (2002) Expression of Th1, Th2 and immunosuppressive cytokine gene transcripts in canine atopic dermatitis. Clin Exp Allergy 32: 789-795.
  20. Olivry T, Dean GA, Tompkins MB, Dow JL, Moore PF (1999) Toward a canine model of atopic dermatitis: amplification of cytokine-gene transcripts in the skin of atopic dogs. Exp Dermatol 8: 204-211.
  21. Olivry T, Naydan DK, Moore PF (1997) Characterization of the cutaneous inflammatory infiltrate in canine atopic dermatitis. Am J Dermatopathol 19: 477-486.
  22. Ramirez GA, Yacoub MR, Ripa M, Mannina D, Cariddi A, Saporiti N, Ciceri F, Castagna A, Colombo G, Dagna L (2018) Eosinophils from Physiology to Disease: A Comprehensive Review. Biomed Res Int 2018: 9095275.
  23. Reagan-Shaw S, Nihal M, Ahmad N (2007) Dose translation from animal to human studies revisited. FASEB J 22: 659-661.
  24. Schlotter YM, Rutten VP, Riemers FM, Knol EF, Willemse T (2011) Lesional skin in atopic dogs shows a mixed Type-1 and Type-2 immune responsiveness. Vet Immunol Immunopathol 143: 20-26.
  25. Sinke JD, Thepen T, Bihari IC, Rutten VP, Willemse T (1997) Immunophenotyping of skin-infiltrating T-cell subsets in dogs with atopic dermatitis. Vet Immunol Immunopathol 57: 13-23.
  26. Stosović R, Bogić M (1998) The role of eosinophilic leukocytes in allergic inflammation. Srp Arh Celok Lek 126: 130-137.
  27. Wada T, Ishiwata K, Koseki H, Ishikura T, Ugajin T, Ohnuma N, Obata K, Ishikawa R, Yoshikawa S, Mukai K, Kawano Y, Minegishi Y, Yokozeki H, Watanabe N, Karasuyama H (2010) Selective ablation of basophils in mice reveals their nonredundant role in acquired immunity against ticks. J Clin Invest 120: 2867-2875.
  28. Wang S, Li H, Lian Z, Deng S (2023) The Role of m6A Modifications in B-Cell Development and B-Cell-Related Diseases. Int J Mol Sci 24: 4721.
  29. Weller PF, Spencer LA (2017) Functions of tissue-resident eosinophils. Nat Rev Immunol 17: 746-760.
Go to article

Authors and Affiliations

A. Jasiecka-Mikołajczyk
1
T. Maślanka
1

  1. Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-718 Olsztyn, Poland
Download PDF Download RIS Download Bibtex

Abstract

Applications of cold atmospheric plasma/nitric oxide (CAP/NO) gas have recently garnered popularity when treating impaired wound healing in patients with diabetes. In this study, we aimed to investigate the effects of NO gas application for 60 and 120 s on wound healing in diabetic rats. A dorsal excision 3 cm in diameter was performed in 15 diabetic rats; these rats were categorized into the following 3 groups: DC (untreated diabetic control); DNO/60 (exposure to 200 ppm NO gas for 60 s/day); and DNO/120 (exposure to 200 ppm NO gas for 120 s/day). Wound contraction on days 0, 3, 7, 11, and 14 and wound contraction rate between days 0 and 14 were evaluated. On day 14, tissue samples were collected for histopathologic assessment of inflammation, epithelial regeneration, angiogenesis congestion, and collagen fiber organization. Normality of distribution was assessed using the Shapiro-Wilk test, and intergroup comparisons were performed using the Mann-Whitney U test (NPar Test) and the Kruskal-Wallis test (non-parametric ANOVA). Wound contraction during treatment days 7-14 was significantly greater in the NO-treatment groups than in the DC group (p<0.05). The NO60 s and NO120 s groups showed a significantly higher wound contraction rate than the DC group (p=0.033, p=0.049, respectively). Significant differences were noted between the control and NO groups in terms of inflammation (p<0.05) and between the control group and DNO/60 and DNO/120 groups in terms of collagen organization (p<0.05, p<0.01, respectively). Evaluation of epithelialization revealed significant intergroup differences between the control and NO treatment groups (p<0.01). In this study, the application of NO once a day for 60 seconds and 120 seconds in diabetic wounds contributed equally to wound healing.
Go to article

Bibliography

  1. Ahmed R, Augustine R, Chaudhry M, Akhtar UA, Zahid AA, Tariq M, Falahati M, Ahmad IS, Hasan A (2022) Nitric oxide-releasing biomateri-als for promoting wound healing in impaired diabetic wounds: State of the art and recent trends. Biomed Pharmacother 149: 112707.
  2. Anuk T, Öztürk S, Özaydın İ, Kahramanca Ş, Yayla S, Aksoy, Demirkan I (2016) Comparison of Three Fixation Methods for the prevention of wound contractions in diabetic and non-diabetic mice with full-thickness skin excision. Kafkas Univ Vet Fak Derg 22: 647-651.
  3. Bae SH, Bae YC, Nam SB, Choi SJ (2012) A skin fixation method for decreasing the influence of wound contraction on wound healing in a rat model. Arch Plast Surg 39: 457-462.
  4. Bryan NS (2015) Nitric oxide enhancement strategies. Future Sci OA 1: FSO48.
  5. Burgess JL, Wyant WA, Abujamra B, Kirsner RS, Jozic I (2021) Diabetic wound-healing science. Medicina (Kaunas) 57: 1072.
  6. Caskey RC, Liechty KW (2013) Novel animal models for tracking the fate and contributions of bone marrow derived cells in diabetic healing. Methods: 99-115.
  7. Förstermann U, Sessa WC (2012) Nitric oxide synthases: Regulation and function. Eur Heart J 33: 829-837
  8. Fridman G, Friedman G, Gutsol A, Shekhter AB, Vasilets VN, Fridman A (2008) Applied Plasma Medicine. Plasma Process Polym 5: 503-533.
  9. Ghaffari A, Jalili R, Ghaffari M, Miller C, Ghahary A (2007) Efficacy of gaseous nitric oxide in the treatment of skin and soft tissue infec-tions. Wound Repair Regen 15: 368-377.
  10. Ghaffari A, Miller CC, McMullin B, Ghahary A (2006) Potential application of gaseous nitric oxide as a topical antimicrobial agent. Nitric Ox-ide 14: 21-29.
  11. Ghaffari A, Neil DH, Ardakani A, Road J, Ghahary A, Miller CC (2005) A direct nitric oxide gas delivery system for bacterial and mammalian cell cultures. Nitric Oxide 12: 129-140.
  12. Gronbach M, Mitrach F, Lidzba V, Müller B, Möller S, Rother S, Schulz-Siegmund M (2020) Scavenging of Dickkopf-1 by macromer-based biomaterials covalently decorated with sulfated hyaluronan displays pro-osteogenic effects. Acta Biomater 114: 76-89.
  13. Güngör GÇ, Gültekin Ç, Kükner A, Etikan İ, Temizel M, Özgencil FE (2022) Effect of topical insulin and ozonized cream for the treatment of full-thickness dermal burn injuries: A clinical and histopathological study in diabetic rats. Pak Vet J 42: 229-235.
  14. Krausz A, Friedman AJ (2015) Nitric oxide as a surgical adjuvant. Future Sci OA 1: FSO56.
  15. Luo JD, Chen AF (2005) Nitric oxide: A newly discovered function on wound healing. Acta Pharmacol Sin 26: 259-264.
  16. Malone-Povolny MJ, Maloney SE, Schoenfisch MH (2019) Nitric oxide therapy for diabetic wound healing. Adv Healthc Mater 8: e1801210.
  17. Grada A, Mervis J, Falanga V (2018) Research techniques made simple: Animal models of wound healing. J Invest Dermatol 138: 2095-2105..
  18. Mieczkowski M, Mrozikiewicz-Rakowska B, Kowara M, Kleibert M, Czupryniak L (2022) The problem of wound healing in diabetes-from mo-lecular pathways to the design of an animal model. Int J Mol Sci 23: 7930.
  19. Miersch S, Espey MG, Chaube R, Akarca A, Tweten R, Ananvoranich S, Mutus B (2008) Plasma membrane cholesterol content affects nitric oxide diffusion dynamics and signaling. J Biol Chem 283: 18513-18521.
  20. Miller CC, Miller MK, Ghaffari A, Kunimoto B (2004) Treatment of chronic nonhealing leg ulceration with gaseous nitric oxide: A case study. J Cutan Med Surg 8: 233-238.
  21. Pekshev AV, Shekhter AB, Vagapov AB, Sharapov NA, Vanin AF (2018) Study of plasma-chemical NO-containing gas flow for treatment of wounds and inflammatory processes. Nitric Oxide 73: 74-80.
  22. Powers JG, Higham C, Broussard K, Phillips TJ (2016) Wound healing and treating wounds: Chronic wound care and management. J Am Acad Dermatol 74: 607-625.
  23. Moeen Rezakhanlou A, Miller C, McMullin B, Ghaffari A, Garcia R, Ghahary A (2011) Gaseous nitric oxide exhibits minimal effect on skin fibroblast extracellular matrix gene expression and immune cell viability. Cell Biol Int 35: 407-415.
  24. Shekhter AB, Pekshev AV, Vagapov AB, Telpukhov VI, Panyushkin PV, Rudenko TG, Fayzullin AL, Sharapov NA, Vanin AF (2019) Physi-cochemical parameters of NO-containing gas flow affect wound healing therapy. An experimental study. Eur J Pharm Sci 128: 193-201.
  25. Shekhter AB, Serezhenkov VA, Rudenko TG, Pekshev AV, Vanin AF (2005) Beneficial effect of gaseous nitric oxide on the healing of skin wounds. Nitric Oxide 12: 210-219.
  26. Wu M, Lu Z, Wu K, Nam C, Zhang L, Guo J (2021) Recent advances in the development of nitric oxide-releasing biomaterials and their application potentials in chronic wound healing. J Mater Chem B 9: 7063-7075.
  27. Yang Y, Qi PK, Yang ZL, Huang N (2015) Nitric oxide based strategies for applications of biomedical devices. Biosurf Biotribol 1: 177-201.
  28. Young LH, Ikeda Y, Lefer AM (2001) Caveolin-1 peptide exerts cardioprotective effects in myocardial ischemia-reperfusion via nitric oxide mechanism. Am J Physiol Heart Circ Physiol 280: H2489-H2495.
Go to article

Authors and Affiliations

G. Tatlıcıoğlu
1
A. Çürükoğlu
1
G. Akan
2
G. Yeşilovalı
1
G. Öğütçü
3
R. Aktaş
1
A. Kükner
3
M. Temizel
4
Z.K. Sarıtaş
5
F.E. Özgencil
1

  1. Surgery Department, Near East University, Faculty of Veterinary Medicine, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10 – Turkey
  2. DESAM Institute, Near East University, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10 – Turkey
  3. Histology Department, Near East University, Faculty of Medicine, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10 – Turkey
  4. Experimental Animal Research Center, Near East University Faculty of Veterinary Medicine, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10- Turkey
  5. Surgery Department, Afyon Kocatepe University, Faculty of Veterinary Medicine, ANS Campus, Erenler, Afyonkarahisar/Turkey
Download PDF Download RIS Download Bibtex

Abstract

Otitis externa is a canine disease of multifactorial etiology in which bacteria plays a significant role. Due to the predominant bacterial etiology otitis is usually treated with antibiotics. However, non-prudent use of antibiotics promotes the emergence of antibiotic-resistant bacteria thus compromising the therapy effectiveness. Currently, the increase of antimicrobial resistance (AMR) is one of the biggest threats to global health. For this reason, the aim of the study was to investigate prevalence of the microbiological causes of canine otitis externa and the antibiotic susceptibility of the isolated bacterial strains. The research and sampling were conducted at Veterinary Clinics for small pets in Serbia. Samples were sent to laboratory for bacteriological and mycological testing. Additionally, the sensitivity of the isolated bacteria to antibiotics was evaluated using disc diffusion method. Sixty dogs with otitis externa clinical symptoms were included in the study. Out of a total of 53 positive samples for pathogen presence, bacteria were present in 40. The most prevalent bacteria was Staphylococcus pseudintermedius, followed by Pseudomonas aeruginosa and Proteus spp., while Malassezia pachydermatis was the only isolated yeast pathogen occurring in 36 samples. Generally, the lowest resistance against all bacteria showed enrofloxacin. On the contrary, high resistance to penicillin and amoxicillin was a common finding for G+ and G- bacteria. These results indicate the need for laboratory testing in terms of isolation, identification and antibiotic susceptibility testing, not only in the case of otitis externa in dogs, but in all diseases when it is possible, in order to enhance antimicrobial stewardship and consequently to contribute AMR reduction.
Go to article

Bibliography

  1. August JR (1988) Otitis externa: A disease of multifactorial etiology. Vet Clin North Am Small Anim Pract 18: 731-742.
  2. Ayrapetyan M, Williams T, Oliver JD (2018) Relationship between the Viable but Nonculturable State and Antibiotic Persister Cells. J Bacteriol 200: e00249-18.
  3. Bassetti M, Vena A, Croxatto A, Righi E, Guery B (2018) How to manage Pseudomonas aeruginosa infections. Drugs Context 7: 212527.
  4. Bornand V (1992) Bacteriology and mycology of otitis externa in dogs. Schweiz Arch Tierheilkd 134: 341-348.
  5. Bourély C, Cazeau G, Jarrige N, Leblond A, Madec JY, Haenni M, Gay E (2019) Antimicrobial resistance patterns of bacteria isolated from dogs with otitis. Epidemiol Infect 147: e121.
  6. Bugden DL (2012) Identification and antibiotic susceptibility of bacterial isolates from dogs with otitis externa in Australia. Aust Vet J 91: 43-46.
  7. De Martino L, Nocera FP, Mallardo K, Nizza S, Masturzo E, Fiorito F, Iovane G, Catalanotti P (2016) An update on microbiological causes of canine otitis externa in Campania Region, Italy. Asian Pac J Trop Biomed 6: 384-389.
  8. Dierikx CM, van der Goot JA, Smith HE, Kant A, Mevius DJ (2013) Presence of ESBL/AmpC- producing Escherichia coli in the broiler production pyramid: a descriptive study. PloS One 8: e79005.
  9. EMA/CVMP (2015) Reflection paper on the risk of antimicrobial resistance transfer from companion animals. European Medicines Agency, Amsterdam. Available from: https://www.ema.europa.eu/en/documents/scientific-guideline/ reflection-paper-risk-
  10. antimicrobial-resistance-transfer-companion-animals_en.pdf. (Accessed: 24.02.2023.)
  11. EUCAST. The European Committee on Antimicrobial Susceptibility Testing. Disk Diffusion Test Manual. Available from: https://www.eucast.org/ (Accessed: 17.02.2023.)
  12. Fernández G, Barboza G, Villalobos A, Parra O, Finol G, Ramírez RA (2006) Isolation and identification of microorganisms present in 53 dogs suffering otitis externa. Rev Cient 16: 23-30.
  13. Forster SL, Real T, Doucette KP, King SB (2018) A randomized placebo-controlled trial of the efficacy and safety of a terbinafine, florfenicol and betamethasone topical ear formulation in dogs for the treatment of bacterial and/or fungal otitis externa. BMC Vet Res 14: 262.
  14. Glavind AS, Kruse AB, Nielsen LR, Stege H (2022) Monitoring antimicrobial usage in companion animals: exploring the use of the Danish VetStat database. Acta Vet Scand 64: 27.
  15. Greene CE (1998) Infectious diseases of the dog and cat. 2nd ed., Philadelphia, WB Saunders, pp 551.
  16. Guardabassi L, Loeber ME, Jacobson A (2004) Transmission of multiple antimicrobial-resistant Staphylococcus intermedius between dogs affected by deep pyoderma and their owners. Vet Microbiol 98: 23-27.
  17. Gwenzi W, Chaukura N, Muisa-Zikali N, Teta C, Musvuugwa T, Rzymski P, Abia AL (2021) Insects, rodents, and pets as reservoirs, vectors, and sentinels of antimicrobial resistance. Antibiotics (Basel) 10: 68.
  18. Hariharan H, Coles M, Poole D, Lund L, Page R (2006) Update on antimicrobial susceptibilities of bacterial isolates from canine and feline otitis externa. Can Vet J 47: 253-255.
  19. Hong JS, Song W, Park HM, Oh JY, Chae JC, Shin S, Jeong SH (2019) Clonal spread of extended-spectrum cephalosporin-resistant Enterobacteriaceae between companion animals and humans in South Korea. Front Microbiol 10: 1371.
  20. Hosseini J, Zdovc I, Golob M, Blagus R, Kušar D, Vengušt M, Kotnik T (2012) Effect of treatment with Tris-EDTA/ chlorhexidine topical solution on canine Pseudomonas aeruginosa otitis externa with or without concomitant treatment with oral fluoroquinolones. Slov Vet Res 49: 133-140.
  21. Hubbuch A, Schmitt K, Lehner C, Hartnack S, Schuller S, Schüpbach-Regula G, Mevissen M, Peter R, Müntener C, Naegeli H, Willi B (2020) Antimicrobial prescriptions in cats in Switzerland before and after the introduction of an online antimicrobial stewardship tool. BMC Vet Res 16: 229.
  22. Jacobson LS (2002) Diagnosis and medical treatment of otitis externa in the dog and cat. J S Afr Vet Assoc 73: 162-170.
  23. Jasovský D, Littmann J, Zorzet A, Cars O (2016) Antimicrobial resistance – a threat to the world’s sustainable development. Ups J Med Sci 121: 159-164.
  24. Kaspar U, von Lützau A, Schlattmann A, Roesler U, Köck R, Becker K (2018) Zoonotic multidrug-resistant microorganisms among small companion animals in Germany. PLoS One, 13: e0208364.
  25. Kiss G, Radvanyi S, Szigeti G (1997) New combination for the therapy of canine otitis externa. I. Microbiology of otitis externa. J Small Anim Pract 38: 51-56.
  26. Kumar S, Hussain K, Sharma R, Chhibber S, Sharma N (2014) Prevalence of canine otitis externa in Jammu. J Anim Res 4: 121-129.
  27. Kumar A, Singh K, Sharma A (2002) Prevalence of Malassezia pachydermatis and other organisms in healthy and infected dogs ears. Isr J Vet Med 57: 145-148.
  28. Lancet T (2022) Antimicrobial resistance: time to repurpose the Global Fund. Lancet 399: 335.
  29. Langendonk RF, Neill DR, Fothergill JL (2021) The building blocks of antimicrobial resistance in Pseudomonas aeruginosa: implications for current resistance-breaking therapies. Front Cell Infect Microbiol 11: 665759.
  30. Laxminarayan R (2022) The overlooked pandemic of antimicrobial resistance. Lancet 399: 606- 607.
  31. Lilenbaum W, Veras M, Blum E, Souza GN (2000) Antimicrobial susceptibility of staphylococci isolated from otitis externa in dogs. Lett Appl Microbiol 31: 42-45.
  32. Lyskova P, Vydrzalova M, Mazurova J (2007) Identification and antimicrobial susceptibility of bacteria and yeasts isolated from healthy dogs and dogs with otitis externa. J Vet Med A Physiol Pathol Clin Med 54: 559-563.
  33. Marques C, Gama LT, Belas A, Bergström K, Beurlet S, Briend-Marchal A, Broens EM, Costa M, Criel D, Damborg P, van Dijk MA, van Dongen AM, Dorsch R, Espada CM, Gerber B, Kritsepi-Konstantinou M, Loncaric I, Mion D, Misic D, Movilla R, Overesch G, Perreten V, Roura X, Steenbergen J, Timofte D, Wolf G, Zanoni RG, Schmitt S, Guardabassi L, Pomba C (2016) European multicenter study on antimicrobial resistance in bacteria isolated from companion animal urinary tract infections. BMC Vet Res 12: 213.
  34. Martín Barrasa JL, Lupiola Gómez P, González Lama Z, Tejedor Junco MT (2000) Antibacterial susceptibility patterns of Pseudomonas strains isolated from chronic canine otitis externa. J Vet Med B Infect Dis Vet Public Health 47: 191-196.
  35. Mateus A, Brodbelt DC, Barber N, Stärk KD (2011) Antimicrobial usage in dogs and cats in first opinion veterinary practices in the UK. J Small Anim Pract 52: 515-521.
  36. Morley PS, Apley MD, Besser TE, Burney DP, Fedorka-Cray PJ, Papich MG, Traub-Dargatz JL, Weese JS (2005) Antimicrobial Drug Use in Veterinary Medicine. J Vet Intern Med 19: 617-629.
  37. Morris DO (2004) Medical therapy of otitis externa and otitis media. Vet Clin North Am Small Anim Pract 34: 541-555.
  38. Murphy KM (2001) A review of techniques for the investigation of otitis externa and otitis media. Clin Tech Small Anim Pract 16: 236-241.
  39. Nuttall T (2016) Successful management of otitis externa. In Pract 38: 17-21.
  40. O’Neill DG, Volk AV, Soares T, Church DB, Brodbelt DC, Pegram C (2021) Frequency and predisposing factors for canine otitis externa in the UK–a primary veterinary care epidemiological view. Canine Med Genet.
  41. O’Neill J (2014) Antimicrobial resistance: tackling a crisis for the health and wealth of nations. Rev Antimicrob Resist pp 1-20.
  42. Patel SJ, Wellington M, Shah RM, Ferreira MJ (2020) Antibiotic stewardship in food-producing animals: challenges, progress, and opportunities. Clin Ther 42: 1649-1658.
  43. Paterson S (2016) Discovering the causes of otitis externa. In Pract 38: 7-11.
  44. Penna B, Thomé S, Martins R, Martins G, Lilenbaum W (2011) In vitro antimicrobial resistance of Pseudomonas aeruginosa isolated from canine otitis externa in Rio de Janeiro, Brazil. Braz J Microbiol 42: 1434-1436.
  45. Petrov V, Mihaylov G, Tsachev I, Zhelev G, Marutsov P, Koev K (2013) Otitis externa in dogs: microbiology and antimicrobial susceptibility. Revue Méd Vét 164: 18-22.
  46. Pinto Ferreira J, Battaglia D, Dorado García A, Tempelman K, Bullon C, Motriuc N, Caudell M, Cahill S, Song J, LeJeune J (2022) Achieving antimicrobial stewardship on the global scale: challenges and opportunities. Microorganisms 10: 1599.
  47. Pomba C, Rantala M, Greko C, Baptiste KE, Catry B, Van Duijkeren E, Mateus A, Moreno MA, Pyörälä S, Ružauskas M, Sanders P, Teale C, Threlfall EJ, Kunsagi Z, Edo JT, Jukes H, Törneke K (2017) Public health risk of antimicrobial resistance transfer from companion animals. J Antimicrob Chemother 72: 957-968.
  48. Prescott JF, Baggot JD, Walker RD (2000) Antimicrobial therapy in veterinary medicine. 3rd ed., Ames, Iowa: Iowa State University, pp 547.
  49. Rosser EJ Jr (2004) Causes of otitis externa. Vet Clin North Am Small Anim Pract 34: 459-468.
  50. Rubin JE, Chirino-Trejo M (2011) Prevalence, sites of colonization, and antimicrobial resistance among Staphylococcus pseudintermedius isolated from healthy dogs in Saskatoon, Canada. J Vet Diagn Invest 23: 351-354.
  51. Sahoo KC, Tamhankar AJ, Johansson E, Lundborg CS (2010) Antibiotic use, resistance development and environmental factors: a qualitative study among healthcare professionals in Orissa, India. BMC Public Health 10: 629.
  52. Saridomichelakis MN, Farmaki R, Leontides LS, Koutinas AF (2007) Aetiology of canine otitis externa: a retrospective study of 100 cases. Vet Dermatol 18: 341-347.
  53. Scarborough R, Bailey K, Galgut B, Williamson A, Hardefeldt L, Gilkerson J, Browning G (2020) Use of local antibiogram data and antimicrobial importance ratings to select optimal empirical therapies for urinary tract infections in dogs and cats. Antibiotics (Basel) 9: 924.
  54. Scarborough R, Hardefeldt L, Browning G, Bailey K (2021) Pet Owners and Antibiotics: Knowledge, Opinions, Expectations, and Communication Preferences. Antibiotics (Basel) 10: 1326.
  55. Schmiedel J, Falgenhauer L, Domann E, Bauerfeind R, Prenger-Berninghoff E, Imirzalioglu C, Chakraborty T (2014) Multiresistant extended-spectrum β-lactamase-producing Enterobacteriaceae from humans, companion animals and horses in central Hesse, Germany. BMC Microbiol 14: 187.
  56. Scott DW, Miller WH, Griffin CE (2001) External ear diseases. In: Small animal dermatology. 6th ed., Philadelphia, PA: WB Saunders, pp 1203-1235.
  57. Sharma C, Rokana N, Chandra M, Singh BP, Gulhane RD, Gill JP, Ray P, Puniya AK, Panwar H (2018) Antimicrobial resistance: its surveillance, impact, and alternative management strategies in dairy animals. Front Vet Sci 4: 237.
  58. da Silva KC, Knobel T, Moreno AM (2013) Antimicrobial resistance in veterinary medicine: mechanisms and bacterial agents with the greatest impact on human health. Braz J Vet Res Anim Sci 50: 171-183.
  59. So JH, Kim J, Bae IK, Jeong SH, Kim SH, Lim SK, Park YH, Lee K (2012) Dissemination of multidrug-resistant Escherichia coli in Korean veterinary hospitals. Diagn Microbiol Infect Dis 73: 195-199.
  60. Staroniewicz Z, Król J, Cierpisz J (1995) Bacterial and mycologic flora in dogs with otitis externa. Med Weter 51: 667-670.
  61. Święcicka N, Bernacka H, Fac E, Zawiślak J (2015) Prevalence and commonest causes for otitis externa in dogs from two Polish veterinary clinics. Bulg J Vet Med 18: 65-73.
  62. Terziev G, Urumova V (2018) Retrospective study on the etiology and clinical signs of canine otitis. Comp Clin Pathol 27: 7-12.
  63. Ungemach FR, Müller-Bahrdt D, Abraham G (2006) Guidelines for prudent use of antimicrobials and their implications on antibiotic usage in veterinary medicine. Int J Med Microbiol 296 (Suppl 2): 33-38.
  64. Walther B, Tedin K, Lübke-Becker A (2017) Multidrug-resistant opportunistic pathogens challenging veterinary infection control. Vet Microbiol 200: 71-78.
  65. Wong C, Epstein SE, Westropp JL (2015) Antimicrobial susceptibility patterns in urinary tract infections in dogs (2010-2013). J Vet Intern Med 29: 1045-1052.
  66. World Health Organization (WHO) (2019) Antimicrobial stewardship programmes in health- care facilities in low-and middle-income countries: a WHO practical toolkit. Available from: https://www.who.int/publications/i/item/ 9789241515481 (Accessed: 28.02.2023.)
  67. World Health Organization (WHO) (2018) Critically important antimicrobials for human medicine, 6th revision – Advisory Group on Integrated Surveillance of Antimicrobial Resistance (AGISAR) Available from: https://www.who.int/publications/i/item/9789241515528 (Accessed: 28.02.2023.)
  68. Zamankhan Malayeri H, Jamshidi S, Zahraei Salehi T (2010) Identification and antimicrobial susceptibility patterns of bacteria causing otitis externa in dogs. Vet Res Commun 34: 435-444.
  69. Ziółkowska G, Nowakiewicz A (2004) Occurrence of the genus Malassezia yeasts in the external ear canal of dogs. Med Weter 60: 310-313.
  70. Zur G, Lifshitz B, Bdolah-Abram T (2011) The association between the signalment, common causes of canine otitis externa and pathogens. J Small Anim Pract 52: 254-258.
Go to article

Authors and Affiliations

N. Tesin
1
D. Stojanovic
1
I. Stancic
1
N. Kladar
2 3
Z. Ružić
1
J. Spasojevic
1
D. Tomanic
1
Z. Kovacevic
1

  1. Department of Veterinary Medicine, Faculty of Agriculture, University of Novi Sad, Trg Dositeja Obradovica 8, 21000 Novi Sad, Serbia
  2. Center for Medical and Pharmaceutical Investigations and Quality Control, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
  3. Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia
Download PDF Download RIS Download Bibtex

Abstract

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

Bibliography

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

Authors and Affiliations

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

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

Abstract

This study was conducted with the aim of determining the growth characteristics and survival rate of Karacabey Merino lambs, as well as the non-genetic factors affecting these traits. The study included data from a total of 17659 lambs and 12263 ewes raised in 30 herds between the years 2011 and 2016 as part of the National Sheep and Goat Improvement Project. The average birth weight of the lambs was determined as 3.73±0.05 kg, the average 45th day live weight was 18.43±0.58 kg, the average weaning (average 91.8 days) weight was 31.83±0.24 kg, and the average daily live weight gain until weaning was 289.1±3.91 g. The average survival rate of lambs at weaning was calculated to be 95.67% ± 1.15. The effects of the factors herd, birth year, birth type, birth season and sex were found significant for all traits (p<0.01). It was established that the mortality rate in lambs in large herds was higher during 6 years in which the project was carried out. Due to the high twinning rate in large herds, the number of lambs per worker is increased, and as a result, they cannot be adequately cared for. For this reason, large farms may be encouraged to increase workmanship services in addition to being provided with protective health practices for lambs throughout the birth period. On the other hand, it was determined that the twinning rate was low in small farms. On farms with fewer sheep populations, it may be advised to flush or administer exogenous hormone treatments to ewes in order to increase fertility and help them bear twins.
Go to article

Bibliography

  1. Abdoli R, Zamani P, Mirhoseini SZ, Ghavi Hossein-Zahed N, Nadri S (2016) A review on prolificacy in sheep. Reprod Domest Anim 51: 631-637.
  2. Akpa GN, Ambali AL, Suleiman IO (2013) Body conformation, testicular and semen characteristics as influenced by age, hair type and body condition of Red Sokoto goat. NY Sci J 6: 44-58.
  3. Albial AM, Singh J, Singh DP, Niwas R (2014) Environmental influences on growth traits of Nali sheep. Indian J Anim Res 48: 75-77.
  4. Balasubramanyam D, Jaishankar S, Sivaselvam SN (2010) Performance of Madras Red sheep under farmer’s flock. Indian J Small Rumin 16: 217-220.
  5. Baneh H, Hafezian SH (2009) Effects of environmental factors on growth traits in Ghezel sheep. Afr J Biotechnol 8: 2903-2907.
  6. Baş S, Özsoy MK, Vanlı Y (1986) The effects of feeding at different durations pre ram-mating on fertility in sheep, growth and survival rate. Turk Vet Anim Sci 10: 221-234.
  7. Bela B, Haile A (2009) Factors affecting growth performance of sheep under village management conditions in the south western part of Ethiopia. Livest Res Rural Dev 21: 189.
  8. Brien FD, Hebart ML, Jaensch KS, Smith DH, Grimson RJ (2009) Genetics of lamb survival: A study of Merino resource flocks in South Australia. Proc Assoc Advmt Anim Breed Genet 18: 492-495.
  9. Cloete SW, Cloete JJ (2015) Production performance of Merino and Dohne Merino ewes and lambs in pure or crossbreeding systems. Proc Assoc Advmt Anim Breed Genet 21: 217-220.
  10. Dangi PS, Poonia JS (2006) Factors affecting weaning and six month body weight in crossbred sheep. Indian J Anim Res 40: 161-163.
  11. Devendran P, Cauveri D, Murali N, Kumarasamy P (2014) Growth profile of Madras Red sheep in farmer’s flock. Indian J Small Rumin 20: 20-23.
  12. Dixit SP, Singh G, Dhillon JS (2011) Genetic and environmental factors affecting fleece traits in Bharat Merino sheep. Indian J Anim Sci 81: 80-83.
  13. Gowane GR, Chopra A, Prince LL, Mishra AK, Arora AL (2011) Genetic analysis for growth traits of prolific Garole × Malpura (GM) sheep. Trop Anim Health Prod 43: 299-303.
  14. Güngör İ, Akçapınar H (2013) The production traits of Bafra sheep reared in Ankara conditions. Lalahan Hay Araşt Enst Derg 53: 59-73.
  15. Hatcher S, Atkins KD, Safari E (2010) Lamb survival in Australian Merino Sheep: A genetic analysis. J Anim Sci 88: 3198-3205.
  16. Iman NY, Slyter AL (1996) Lifetime lamb and wool production of targhee or Finn-Dorset-Targhee ewes managed as farm place or range flock: II. Cumulative lamb and wool production. J Anim Sci 74: 1765-1769.
  17. Kaymakçı M (2016) Advanced sheep breeding, 1-9; Meta Press, Izmir, Türkiye, ISBN 978-605-85998.
  18. Lalit, Malik ZS, Dalal DS, Dahiya S, Magotra A, Patil CS (2016) Genetics of growth traits in sheep: A review. IJRRLS 3: 12-18.
  19. Lupi TM, Nogales S, León JM, Barba C, Delgado JV (2015) Analysis of the non-genetic factors affecting the growth of Segureño sheep. Ital J Anim Sci 14: 124-131.
  20. Mane PM, Pachpute ST, Nimase RG (2014) Growth and reproductive performance of Deccani sheep in an organized farm. Indian J Small Rumin 20: 23-27.
  21. Minitab 2019. Minitab® 19 Statistical Software. https://www.minitab.com/en-us/
  22. Mishra AK, Arora AL, Prince LL, Kumar S (2008) Performance evaluation of Garole×Malpura half-bred sheep evolved in semiarid region of Rajasthan. Indian J Anim Sci 78: 746-750.
  23. Momoh OM, Rotimi EA, Dim NI (2013) Breed effect and non-genetic factors affecting growth performance of sheep in semi-arid region of Nigeria. J Appl Biosci 67: 5302-5307.
  24. Morris CA, Hickey SM, Clarke JN (2000) Genetic and environmental factors affecting lamb survival at birth and through to weaning. New Zealand J Agric Res 43: 515-524.
  25. Mourad M, Gbanamou G, Balde IB (2000) Performance of West African dwarf goats under the extensive system of production in Faranah, Guinea. In: Proceedings of the 7th International conference on goats, 15-21 May, France, pp. 227-230.
  26. Mousa-Balabel TM (2010) The relationship between sheep management and lamb mortality. WASET 4: 372-377.
  27. Nirban LK, Joshi RK, Narula HK, Singh H, Bhakar S (2015) Genetic and non-genetic factors affecting body weights in Marwari sheep. Indian J Small Rumin 21: 106-108.
  28. Parihar K, Yadav SB, Narula HK, Pannu U, Bais B (2016) Factors affecting in growth performance and greasy fleece yield in Magra sheep under arid condition of Rajasthan. Int J Agric Sci 9: 4095-4096.
  29. Petrovic MP, Muslic DR, Petrovic VC, Maksimovic N (2011) Influence of environmental factors on birth weight variability of indigenous Serbian breeds of sheep. Afr J Biotechnol 10: 4673-4676.
  30. Prakash V, Prince LL, Gowane GR, Arora AL (2012) Factors affecting post-weaning average daily gain and kleiber ratios in Malpura sheep. Indian J Anim Sci 82: 1598-1600.
  31. Rather MA, Bashir I, Ahanger SA (2020) Effect of genetic and non-genetic factors on birth weight of Corriedale sheep in an organized farm in Kashmir. Anim Sci Quarterly 1: 13-16.
  32. Sezenler T, Köycü E, Özder M (2008) The effect of body condition score in Karacabey Merino at lambing on the lamb growth. J Tekirdag Agric Fac 5: 45-53.
  33. Sezenler T, Soysal D, Yıldırır M, Yüksel MA, Ceyhan A, Yaman Y, Erdoğan İ, Karadağ O, (2013) Influence of some environmental factors on litter size and lamb growth performance in Karacabey Merino sheep. J Tekirdag Agric Fac 10: 40-47.
  34. Susic V, Pavic V, Mioc B, Stokovic I, Kabalin A E (2005) Seasonal variations in lamb birth weight and mortality. Vet Arhiv 5: 375-381.
  35. Tesema Z, Deribe B, Lakew M, Getachew T, Tilahun M, Belayneh N, Kefale A, Shibesh M, Zegeye A, Yizengaw L, Alebachew GW, Tiruneh S, Kiros S, Asfaw M, Bishaw M (2022) Genetic and non-genetic parameter estimates for growth traits and Kleiber ratios in Dorper × indigenous sheep. Animal 16(6): 100533.
  36. Thiruvenkadan AK, Karunanithi K, Muralidharan J, Babu RN (2011) Genetic analysis of pre-weaning and post-weaning growth traits of Mecheri sheep under dry land farming conditions. Asian-Aust J Anim Sci 24: 1041-1047.
  37. Ullah F, Javed K, Salim M, Rehman IU, Khan M, Ali S, Rehman ZU, Bhunesh Babar A, Hussain MA, Khan S, Rahim N, Hosseini SM (2020) Identification of non-genetic factors affecting birth weight, weaning weight, pre-weaning weight, yearling weight and greasy fleece performance of Kajli Sheep at two ecologies in Pakistan. Ann Agric Crop Sci 5: 1065.
  38. Ünal N, Akçapınar H, Atasoy F, Aytaç M (2006) Some reproductive and growth traits of crossbred genotypes produced by crossing local sheep breeds of Kivircik × White Karaman and Chios × White Karaman in steppe conditions. Arch Tierz 49: 55-63.
  39. Ürüşan H, Emsen H (2010) Effect of lambing season, lamb genotype, maternal and birth related factors on growth and viability of lambs. J Tekirdag Agric Fac 7: 163-172.
  40. Vivekanand Joshi RK, Narula HK, Singh H, Chopra A (2014) Effect of genetic and non-genetic factor on growth of Magra sheep in arid region of rajasthan. Indian J Small Rumin 20: 19-22.
Go to article

Authors and Affiliations

H. Hanoglu Oral
1
S. Ozis Altincekic
2
S. Duru
2

  1. Muş Alparslan University, Faculty of Applied Sciences, Department of Animal Production and Technologies, Mus, 49250, Türkiye
  2. Bursa Uludag University, Faculty of Agriculture, Department of Animal Science, 16059, Bursa, Türkiye
Download PDF Download RIS Download Bibtex

Abstract

The objectives of this research were to highlight the main factors, which have relevant significance for etiology of myopathies and to assess the incidence of myopathies in a representative population of broilers raised in Lithuania. Eighteen flocks were evaluated to assess the incidence of musculus pectoralis major myopathies (PMM) (total 54,000 broilers) and dorsal cranial myopathy (DCM) (total 124,200 broilers). Thirteen flocks (total 19,500 broilers) were evaluated to find out deep pectoral myopathy (DPM) occurrence in Lithuania. Investigated parameters of each flock were: average broiler live body weight (BW) at slaughter, average slaughter age, treatment and seasons. A correlation analysis was used to measure the strength of the linear relationship between the investigated traits and incidence of these myopathies. Overall, the incidence of PMM in Lithuania was 18.19%. DCM and DPM were 5.16% and 0.27%, respectively. The percentage of PMM in flocks was strongly associated with average broiler live BW at slaughter (r=0.898, p<0.001) and age at slaughter (r=0.693, p<0.001). The percentage of PMM in flocks was negatively related with treatment of broilers (rs=-0.535, p<0.05). The percentage of DCM was positively associated with average broiler live BW at slaughter (r=0.537, p<0.05) and with seasons (rs=0.658, p<0.01). However, our study results revealed, that the analyzed parameters are not so important in DPM etiology. Furthermore, predisposing factors of PMM, DCM and DPM are different. These findings suggest that not only broiler’s heavy weight and age at slaughter could have influence for etiology of myopathies.
Go to article

Bibliography

  1. Amaral PC, Zimermann C, Santos LR, Noro M, Prá MD, Pilotto F, Rodrigues LB, Dickel EL (2017) Evaluation of physiological parameters of broilers with dorsal cranial myopathy. Braz J Poult Sci 19: 69-74.
  2. Barbut S (2019) Recent myopathies in broiler’s breast meat fillets. Worlds Poult Sci J 75: 559-582.
  3. Bianchi M, Petracci M, Franchini A, Cavani C (2006) The occurrence of deep pectoral myopathy in roaster chickens. Poult Sci 85: 1843-1846.
  4. Bilgili SF, Hess JB (2002) Green muscle disease in broilers increasing. World Poult 18: 42-43.
  5. Bilgili SF, Hess JB (2008) Green muscle disease: Reducing the incidence in broiler flocks. Aviagen Brief. March.
  6. Che S, Wang C, Varga C, Barbut S, Susta L (2022) Prevalence of breast muscle myopathies (spaghetti meat, woody breast, white striping) and associated risk factors in broiler chickens from Ontario Canada. PLoS One 17: e0267019.
  7. Dalle Zotte A, Tasoniero G, Puolanne E, Remignon H, Cecchinato M, Catelli E, Cullere M (2017) Effect of ‘Wooden Breast’ appearance on poultry meat quality, histological traits, and lesions characterization. Czech J Anim Sci 62: 51-57.
  8. Dinev I, Kanakov D (2011) Deep pectoral myopathy: Prevalence in 7 weeks old broiler chickens in Bulgaria. Revue Méd Vét 162: 279-283.
  9. Gratta F, Birolo M, Piccirillo A, Petracci M, Maertens L, Xiccato G, Trocino A (2017) Effects of the feeding system on performance and myopathy occurrence in two broiler chicken genotypes. Ital J Anim Sci 16: 48.
  10. Huang X, Ahn DU (2018) The incidence of muscle abnormalities in broiler breast meat - A review. Korean J Food Sci Anim Resour 38: 835-850.
  11. Kijowski J, Kupińska E, Stangierski J, Tomaszewska-Gras J, Szablewski T (2014) Paradigm of deep pectoral myopathy in broiler chickens. Worlds Poult Sci J 70: 125-138.
  12. Kuttappan VA, Brewer VB, Apple JK, Waldroup PW, Owens CM (2012) Influence of growth rate on the occurrence of white striping in broiler breast fillets. Poult Sci 91: 2677-2685.
  13. Kuttappan VA, Hargis BM, Owens CM (2016) White striping and woody breast myopathies in the modern poultry industry: a review. Poult Sci 95: 2724-2733.
  14. Lien RJ, Joiner KS, Hess JB, Bilgili SF (2011) Finding answers to ‘green muscle disease’. Watt Poult 5: 15-18.
  15. Lorenzi M, Mudalal S, Cavani C, Petracci M (2014) Incidence of white striping under commercial conditions in medium and heavy broiler chickens in Italy. J Appl Poult Res 23: 754-758.
  16. Malila Y, U-Chupaj J, Srimarut Y, Chaiwiwattrakul P, Uengwetwanit T, Arayamethakorn S, Punyapornwithaya V, Sansamur C, Kirschke CP, Huang L, Tepaamorndech S, Petracci M, Rungrassamee W, Visessanguan W (2018) Monitoring of white striping and wooden breast cases and impacts on quality of breast meat collected from commercial broilers (Gallus gallus). Asian-Australas J Anim Sci 31: 1807-1817.
  17. NCC (2019) National Chicken Council. https://www.national chickencouncil.org/ (Accessed on September 2022)
  18. Owens CM (2014) Identifying quality defects in poultry processing. Watt Poult 2014: 42-50.
  19. Pedrão MR, de Souza RM, Louvandini H, Louvandini P, de Souza RB, de Morais Leite N, Coró FA (2021) White striping and wooden breast myopathies in the poultry industry: An overview of changes in the skin, bone tissue and intestinal microbiota and their economic Impact. Advances in Poultry Nutrition Research. IntechOpen; 2021. Available from: http://dx.doi.org/10.5772/intechopen.96513
  20. Petracci M, Mudalal S, Soglia F, Cavani C (2015) Meat quality in fast-growing broiler chickens. Worlds Poult Sci J 71: 363-374.
  21. Petracci M, Soglia F, Madruga M, Carvalho L, Ida E, Estévez M (2019) Wooden‐Breast, White Striping, and Spaghetti Meat: Causes, consequences and consumer perception of emerging broiler meat abnormalities. Compr Rev Food Sci Food Saf 18: 565-583.
  22. Prado F, Orso C, Ebbing MA, Kipper M, Andretta I, Ribeiro AM. (2021) An in-situ assessment of Dorsal Cranial Myopathy in broilers, approaching regarding meteorological influences in South Brazil, classification, and appearance of the lesions during industrial processing. J Appl Poult Res 30: 100182.
  23. Russo E, Drigo M, Longoni C, Pezzotti R, Fasoli P, Recordati C (2015) Evaluation of white striping prevalence and predisposing factors in broilers at slaughter. Poult Sci 94: 1843-1848.
  24. Sihvo H-K, Immonen K, Puolanne E (2014) Myodegeneration with fibrosis and regeneration in the pectoralis major muscle of broilers. Vet Pathol 51: 619–623.
  25. Sihvo H-K, Lindén J, Airas N, Immonen K, Valaja J, Puolanne E (2017) Wooden breast myodegeneration of pectoralis major muscle over the growth period in broilers. Vet Pathol 54: 119-128.
  26. Siller WG (1985) Deep pectoral myopathy: A penalty of succesful selection for muscle growth. Poult Sci 64: 1591-1595.
  27. Tijare VV, Yang FL, Kuttappan VA, Alvarado CZ, Coon CN, Owens CM (2016) Meat quality of broiler breast fillets with white striping and woody breast muscle myopathies. Poult Sci 95: 2167-2173.
  28. Zimermann FC, Fallavena LC, Salle CT, Moraes HL, Soncini RA, Barreta MH, Nascimento VP (2012) Downgrading of heavy broiler chicken carcasses due to myodegeneration of the anterior latissimus dorsi: pathologic and epidemiologic studies. Avian Dis 56: 418-421.
  29. Valentine BA, McGavin MD (2007) Skeletal muscle. In: McGavin MD, Zachary JF (eds) Pathologic basis of veterinary disease. 4th ed. Mosby Elsevier, Philadelphia, pp 996-1039.
  30. Williams S (2008) Muscular system. In Flecher OJ, Aziz TA (eds) Avian histopathology. 3rd ed., The American Association of Avian Pathologists, Jacksonville, FL, pp 80-95.
  31. Xing T, Zhao X, Zhang L, Li JL, Zhou GH, Xu XL, Gao F (2019) Characteristics and incidence of broiler chicken wooden breast meat under commercial conditions in China. Poult Sci 99: 620-628.
Go to article

Authors and Affiliations

E. Lebednikaite
1
L. Anskiene
2
Z. Balciauskiene
3
A. Pockevicius
1

  1. Department of Veterinary Pathobiology, Faculty of Veterinary Medicine, Lithuanian University of Health Sciences, Tilzes 18, Kaunas, Lithuania
  2. Department of Animal Breeding, Faculty of Animal Science, Lithuanian University of Health Sciences, Tilzes 18, Kaunas, Lithuania
  3. Vilnius Department of the State Food and Veterinary Service, Konstitucijos 23b, Vilnius, Lithuania
Download PDF Download RIS Download Bibtex

Abstract

The aim of the study was to compare the serum protein profile of Bernese Mountain Dogs (BMDs) reacting positive for Bb in snap testing with the serum protein profile of dogs of other breeds (healthy and with clinical borreliosis) using the MALDI time-of-flight (MALDI-TOF) technique. The observations included five groups of dogs. BMDs reacting positively to Bb in snap serological testing and showing symptoms of borreliosis (group 1), BMDs for which no borreliosis symptoms were determined but with seropositivity for Bb determined with snap serological tests (group 2), clinically healthy BMDs with no antibodies for Bb found in the serum (group 3), five dogs of different breeds, reacting positively in serological testing, in which borreliosis symptoms were observed (group 4), clinically healthy dogs of different breeds with negative reaction in tests towards Bb (group 5). A proteomic analysis demonstrated the presence of five identical protein fractions among all five groups. An additional two protein fractions of approximately 7.630 and 15.260 kDa were found in all the serum samples obtained from the dogs positive for borrelia in a snap test, both in those exhibiting symptoms of borreliosis, and seropositive BMDs not presenting symptoms of the disease. These two additional protein fractions may be used to differentiate between seropositive and seronegative B. burgdorferi dogs and may be considered a seropositivity marker, however, it cannot be used to differentiate between animals with the clinical form of the disease and those that are only seropositive.
Go to article

Bibliography

  1. Boyer PH, Boulanger N, Nebbak A, Collin E, Jaulhac B, Almeras L (2017) Assessment of MALDI-TOF MS biotyping for Borrelia burgdorferi sl detection in Ixodes ricinus. PLoS One 12: e0185430.
  2. Calderaro A, Gorrini C, Piccolo G, Montecchini S, Buttrini M, Rossi S, Piergianni M, Arcangeletti MC, De Conto F, Chezzi C, Medici MC (2014) Identification of Borrelia species after creation of an in-house MALDI-TOF MS database. PLoS One 9: e88895
  3. Dzięgiel B, Adaszek Ł, Banach T, Winiarczyk S. (2016) Specificity of mass spectrometry (MALDI-TOF) in the diagnosis of Babesia canis regarding to other canine vector-borne diseases. Ann Parasitol 62: 101-105
  4. Dzięgiel B, Kubrak T, Adaszek Ł, Dębiak P, Wyłupek D, Bogucka-Kocka A, Lechowski J, Winiarczyk S (2014) Prevalence of Babesia canis, Borrelia burgdorferi sensu lato, and Anaplasma phagocytophilum in hard ticks collected from meadows of Lubelskie Voivodship (eastern Poland). Bull Vet Inst Pulawy 58: 29-33
  5. Fotso Fotso A, Mediannikov O, Diatta G, Almeras L, Flaudrops C, Parola P, Drancourt M (2014) MALDI-TOF mass spectrometry detection of pathogens in vectors: the Borrelia crocidurae/Ornithodoros sonrai paradigm. PLoS Negl Trop Dis. 8: 2984
  6. Gerber B, Eichenberger S, Wittenbrink MM, Reusch CE (2007) Increased prevalence of Borrelia burgdorferi infections in Bernese Mountain Dogs: a possible breed predisposition. BMC Vet Res 3: 15
  7. Gerber B, Haug K, Eichenberger S, Reusch CE, Wittenbrink MM. (2009a) Follow-up of Bernese Mountain dogs and other dogs with serologically diagnosed Borrelia burgdorferi infection: what happens to seropositive animals? BMC Vet Res. 5: 18
  8. Gerber B, Eichenberger S, Haug K, Wittenbrink MM, Reusch CE (2009b) Association of urine protein excretion and infection with Borrelia burgdorferi sensu lato in Bernese Mountain dogs. Vet J 182: 487-488
  9. Neumann-Cip AC, Fingerle V, Margos G, Straubinger RK, Overzier E, Ulrich S, Wieser A (2020) A novel rapid sample preparation method for MALDI-TOF MS permits Borrelia burgdorferi sensu lato species and isolate differentiation. Front Microbiol 11: 690
  10. Obama T, Kato R, Masuda Y, Takahashi K, Aiuchi T, Itabe H (2007) Analysis of modified apolipoprotein B-100 structures formed in oxidized low-density lipoprotein using LC-MS/MS. Proteomics 7: 2132-2141
  11. Signor L, Erba EB (2013) Matrix-assisted laser desorption/ /ionization time of flight (MALDI-TOF) mass spectrometric analysis of intact proteins larger than 100 kDa. J Vis Exp 9: 50635
  12. Stanek G, Reiter M (2011) The expanding Lyme Borrelia complex-clinical significance of genomic species. Clin Microbiol Infect 17: 487-493
  13. Tsao JI (2009) Reviewing molecular adaptations of Lyme borreliosis spirochetes in the context of reproductive fitness in natural transmission cycles. Vet Res 40: 36
  14. Zygner W, Jaros S, Wedrychowicz H (2008) Prevalence of Babesia canis, Borrelia afzelii, and Anaplasma phagocytophilum infection in hard ticks removed from dogs in Warsaw (central Poland). Vet Parasitol 153: 139-142
  15. Zygner W, Górski P, Wedrychowicz H (2009) Detection of the DNA of Borrelia afzelii, Anaplasma phagocytophilum and Babesia canis in blood samples from dogs in Warsaw. Vet Rec 164: 465-467
Go to article

Authors and Affiliations

M. Pisarek
1
M. Kalinowski
1
M. Skrzypczak
2
Ł. Mazurek
1
K. Michalak
1
D. Pietras-Ożga
1
B. Dokuzeylü
3
S. Winiarczyk
1
Ł. Adaszek
1

  1. Department of Epizoology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine of the University of Life Sciences in Lublin, ul. Głęboka 30, 20-612 Lublin, Poland
  2. Second Department of Gynecology, Medical University of Lublin, 20-954 Lublin, Poland
  3. Department of Internal Medicine, Veterinary Faculty, Istanbul University-Cerrahpasa, 34320 Avcilar Campus, Avcilar, Istanbul, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The main cause of sperm chromatin damage is oxidative stress related to embryo development failure and adult infertility in mammals and also avian. Oxidative stress results in lipid peroxidation (LPO) causing cell damage. Lipid peroxidation is the oxidation of polyunsaturated fatty acids (PUFAs) in biological systems and causes changes in the physical structure and characteristics of the cell membrane. Due to the high amounts of PUFAs in the avian sperm membrane, its sperm seem susceptible to pe-roxidative damage and is a substantial factor in the fertilization capacity of sperm. The most commonly used methods for measuring LPO or its by-products, such as malondialdehyde (MDA) and 4-hydroksy-2-nonenal (4-HNE), in bird semen are based on the colorimetric method TBARS (thiobarbituric acid reactive substances) and on the use of a fluorescence probe (CC 11-BODIPY 581/591) as a marker to evaluate membrane lipid peroxidation. This review aims first to introduce LPO in avian semen and its effects on avian sperm and second to summarize the commonly applied methods of evaluating LPO and its damage in fresh and stored avian semen.
Go to article

Bibliography

  1. Agarwal A, Makker K, Sharma R (2008) Clinical Relevance of Oxidative Stress in Male Factor Infertility: An Update. Am J Reprod Immunol 59: 2-11.
  2. Agarwal A, Prabakaran S A, Said T M (2005) Prevention of Oxidative Stress Injury to Sperm. J Androl 26: 654-660.
  3. Agarwal A, Saleh RA, Bedaiwy MA (2003) Role of reactive oxygen species in the pathophysiology of human reproduction. Fert Steril 79: 829-843.
  4. Agarwal A, Tvrda E, Sharma R (2014) Relationship amongst teratozoospermia, seminal oxidative stress and male infertility. Reprod Biol Endocrinol 12: 45.
  5. Ahmadi A, Ng S-C (1999) Fertilizing ability of DNA-damaged spermatozoa. J Exp Zool 284: 696-704.
  6. Aitken RJ, Baker MA (2002) Reactive oxygen species generation by human spermatozoa: a continuing enigma. Int J Androl 25: 191-194.
  7. Aitken RJ, Buckingham DW, Carreras A, Irvine DS (1996) Superoxide dismutase in human sperm suspensions: Relationship with cellular composition, oxidative stress, and sperm function. Free Radic Biol Med 21: 495-504.
  8. Aitken RJ, Clarkson JS, Fishel S (1989) Generation of Reactive Oxygen Species, Lipid Peroxidation, and Human Sperm Function. Biol Reprod 41: 183-197.
  9. Aitken RJ, Harkiss D, Buckingham DW (1993) Analysis of lipid peroxidation mechanisms in human spermatozoa. Mol Reprod Dev 35: 302-315.
  10. Aitken RJ, Wingate JK, De Iuliis GN, McLaughlin EA (2007) Analysis of lipid peroxidation in human spermatozoa using BODIPY C11. Mol Hum Reprod 13: 203-211.
  11. Almeida J, Ball BA (2005) Effect of alpha-tocopherol and tocopherol succinate on lipid peroxidation in equine spermatozoa. Anim Reprod Sci 87: 321-337.
  12. Alvarez JG, Storey BT (1982) Spontaneous Lipid Peroxidation in Rabbit Epididymal Spermatozoa: Its Effect on Sperm Motility. Biol Reprod 27: 1102-1108.
  13. Amini MR, Kohram H, Zare-Shahaneh A, Zhandi M, Sharideh H, Nabi MM (2015) The effects of different levels of catalase and superoxide dismutase in modified Beltsville extender on rooster post-thawed sperm quality. Cryobiology 70: 226-232.
  14. Bailey JL, Lessard C, Jacques J, Brèque C, Dobrinski I, Zeng W, Galantino-Homer HL (2008) Cryopreservation of boar semen and its future importance to the industry. Theriogenology 70: 1251-1259.
  15. Ball BA, Vo AT, Baumber J (2001) Generation of reactive oxygen species by equine spermatozoa. Am J Vet Res 62: 508-515.
  16. Barrera G, Pizzimenti S, Dianzani MU (2008) Lipid peroxidation: control of cell proliferation, cell differentiation and cell death. Mol Aspects Med 29: 1-8.
  17. Baumber J, Ball B A, Gravance C G, Medina V, Davies‐ -Morel MC (2000) The Effect of Reactive Oxygen Species on Equine Sperm Motility, Viability, Acrosomal Integrity, Mitochondrial Membrane Potential, and Membrane Lipid Peroxidation. J Androl 21: 895-902.
  18. Blesbois E, Grasseau I, Blum J (1993) Effects of vitamin E on fowl semen storage at 4°C. Theriogenology 39: 771-779.
  19. Blesbois E, Grasseau I, Hermier D (1999) Changes in lipid content of fowl spermatozoa after liquid storage at 2 to 5°C. Theriogenology 52: 325-334.
  20. Blesbois E, Grasseau I, Seigneurin F (2005) Membrane fluidity and the ability of domestic bird spermatozoa to survive cryopreservation. Reproduction 129: 371-378.
  21. Blesbois E, Lessire M, Grasseau I, Hallouis JM, Hermier D (1997) Effect of Dietary Fat on the Fatty Acid Composition and Fertilizing Ability of Fowl Semen. Biol Reprod 56: 1216-1220.
  22. Bréque C, Surai P, Brillard J-P (2003) Roles of antioxidants on prolonged storage of avian spermatozoa in vivo and in vitro. Mol Reprod Dev 66: 314-323.
  23. Brouwers JF, Gadella BM (2003) In situ detection and localization of lipid peroxidation in individual bovine sperm cells. Free Radic Biol Med 35: 1382-1391.
  24. Brouwers JF, Silva PF, Gadella BM (2005) New assays for detection and localization of endogenous lipid peroxidation products in living boar sperm after BTS dilution or after freeze–thawing. Theriogenology 63: 458-469.
  25. Cecil H, Bakst M (1993) In Vitro Lipid Peroxidation of Turkey Spermatozoa. Poult Sci 72: 1370-1378.
  26. Cerolini S, Zaniboni L, Maldjian A, Gliozzi T (2006) Effect of docosahexaenoic acid and α-tocopherol enrichment in chicken sperm on semen quality, sperm lipid composition and susceptibility to peroxidation. Theriogenology 66: 877-886.
  27. Chatterjee S, Gagnon C (2001) Production of reactive oxygen species by spermatozoa undergoing cooling, freezing, and thawing. Mol Reprod Dev 59: 451-458.
  28. Davies KJ (1987) Protein damage and degradation by oxygen radicals. I. general aspects. J Biol Chem 262: 9895-9901.
  29. de Lamirande D, Gagnon C (1993) A positive role for the superoxide anion in triggering hyperactivation and capacitation of human spermatozoa. Int J Androl 16: 21-25.
  30. de Lamirande E, Jiang H, Zini A, Kodama H, Gagnon C (1997) Reactive oxygen species and sperm physiology. Rev Reprod 2: 48-54.
  31. Donoghue A, Wishart G (2000) Storage of poultry semen. Anim Reprod Sci 62: 213-232.
  32. Douard V, Hermier D, Magistrini M, Blesbois E (2003) Reproductive period affects lipid composition and quality of fresh and stored spermatozoa in Turkeys. Theriogenology 59: 753-764.
  33. Douard V, Hermier D, Magistrini M, Labbé C, Blesbois E (2004) Impact of changes in composition of storage medium on lipid content and quality of turkey spermatozoa. Theriogenology 61: 1-13.
  34. Drummen GP, Van Liebergen LC, Op den Kamp JA, Post JA (2002) C11-BODIPY581/591, an oxidation-sensitive fluorescent lipid peroxidation probe: (micro)spectroscopic characterization and validation of methodology. Free Radic Biol Med 33: 473-490.
  35. Dutta S, Majzoub A, Agarwal A (2019) Oxidative stress and sperm function: A systematic review on evaluation and management. Arab J Urol 17: 87-97.
  36. Erdelmeier I, Gérard-Monnier D, Yadan J-C, Chaudière J (1998) Reactions of N-Methyl-2-phenylindole with Malondialdehyde and 4-Hydroxyalkenals. Mechanistic Aspects of the Colorimetric Assay of Lipid Peroxidation. Chem Res Toxicol 11: 1184-1194.
  37. Eslami M, Zadeh Hashem E, Ghaniei A, Sayyah-Atashbeig H (2018) Evaluation of linoleic acid on lipid peroxidative/ /antioxidative parameters, motility and viability of rooster spermatozoa during cold storage. Cell Tissue Bank 19: 799-807.
  38. Esterbauer H, Schaur RJ, Zollner H (1991) Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 11: 81-128.
  39. Evenson DP, Wixon R (2006) Clinical aspects of sperm DNA fragmentation detection and male infertility. Theriogenology 65: 979-991.
  40. Fattah A, Sharafi M, Masoudi R, Shahverdi A, Esmaeili V (2017) L-carnitine is a survival factor for chilled storage of rooster semen for a long time. Cryobiology 74: 13-18.
  41. Fattah A, Sharafi M, Masoudi R, Shahverdi A, Esmaeili V, Najafi A (2017) L -Carnitine in rooster semen cryopreservation: Flow cytometric, biochemical and motion findings for frozen-thawed sperm. Cryobiology 74: 148-153.
  42. Fujihara N, Howarth B (1978) Lipid Peroxidation in Fowl Spermatozoa. Poult Sci 57: 1766-1768.
  43. Fujihara N, Koga O (1984) Prevention of the production of lipid peroxide in rooster spermatozoa. Anim Reprod Sci 7: 385-390.
  44. Gaschler MM, Stockwell BR (2017) Lipid peroxidation in cell death. Biochem Biophys Res Commun 482: 419-425.
  45. Ghaniei A, Eslami M, Zadeh Hashem E, Rezapour R, Talebi A (2019) Quercetin attenuates H2O2‐induced toxicity of rooster semen during liquid storage at 4°C. J Anim Physiol Anim Nutr103: 713-722.
  46. Guthrie H, Welch G, Long J (2008) Mitochondrial function and reactive oxygen species action in relation to boar motility. Theriogenology 70: 1209-1215.
  47. Gutteridge JM (1995) Lipid peroxidation and antioxidants as biomarkers of tissue damage. Clinical Chemistry 41: 1819-1828.
  48. Halliwell B, Chirico S (1993) Lipid peroxidation: its mechanism, measurement, and significance. Am J Clin Nutr 57 (5 Suppl): 715S-725S.
  49. Halliwell B, Gutteridge J (1984) Lipid peroxidation, oxygen radicals, cell damage, and antioxidant therapy. Lancet 1: 1396-1397.
  50. Hamilton TR, de Castro LS, Delgado J de C, De Assis PM, Siqueira AFP, Mendes CM, Goissis MD, Muiño-Blanco T, Cebrián-Pérez JÁ, Nichi M, Visintin JA, D’Ávila Assumpção ME (2016) Induced lipid peroxidation in ram sperm: semen profile, DNA fragmentation and antioxidant status. Reproduction 151: 379-390.
  51. Higuchi Y (2004) Glutathione depletion-induced chromosomal DNA fragmentation associated with apoptosis and necrosis. J Cell Mol Med 8: 455-464.
  52. Holt WV (2000) Basic aspects of frozen storage of semen. Anim Reprod Sci 62: 3-22.
  53. Izanloo H, Soleimanzadeh A, Bucak MN, Imani M, Zhandi M (2021) The effects of varying concentrations of glutathione and trehalose in improving microscopic and oxidative stress parameters in Turkey semen during liquid storage at 5°C. Cryobiology 101: 12-19.
  54. Kelso KA, Cerolini S, Noble RC, Sparks NHC, Speake BK (1996) Lipid and antioxidant changes in semen of broiler fowl from 25 to 60 weeks of age. Reproduction 106: 201-206.
  55. Long JA (2006) Avian Semen Cryopreservation: What Are the Biological Challenges? Poult Sci 85: 232-236.
  56. Long JA, Kramer M (2003) Effect of vitamin E on lipid peroxidation and fertility after artificial insemination with liquid-stored turkey semen. Poult Sci 82: 1802-1807.
  57. Łukaszewicz E (1988) Studies on the Diluents for cock’s Semen Storage in the Light of Laboratory Estimation and Fertility Rates. Zeszt Nauk AR we Wrocławiu 168: 43-59.
  58. Lushchak VI (2011) Environmentally induced oxidative stress in aquatic animals. Aquat Toxicol 101: 13-30.
  59. Lushchak VI (2014) Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact 224: 164-175.
  60. Masoudi R, Asadzadeh N, Sharafi M (2021) Effects of freezing extender supplementation with mitochondria-targeted antioxidant Mito-TEMPO on frozen-thawed rooster semen quality and reproductive performance. Anim Reprod Sci 225: 106671.
  61. Mavi GK, Dubey PP, Cheema RS (2020) Association of antioxidant defense system with semen attributes vis a vis fertility in exotic and indigenous chicken breeds. Theriogenology 144: 158-163.
  62. Mehaisen GM, Partyka A, Ligocka Z, Niżański W (2020) Cryoprotective effect of melatonin supplementation on post-thawed rooster sperm quality. Anim Reprod Sci 212: 106238.
  63. Moghbeli M, Kohram H, Zare-Shahaneh A, Zhandi M, Sharafi M, Nabi MM, Zahedi V, Sharideh H (2016) Are the optimum levels of the catalase and vitamin E in rooster semen extender after freezing-thawing influenced by sperm concentration? Cryobiology 72: 264-268.
  64. Neild DM, Brouwers JF, Colenbrander B, Agüero A, Gadella BM (2005) Lipid peroxide formation in relation to membrane stability of fresh and frozen thawed stallion spermatozoa. Mol Reprod Dev 72: 230-238.
  65. Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95: 351-358.
  66. Park NC, Park HJ, Lee KM, Shin DG (2003) Free Radical Scavenger Effect of Rebamipide in Sperm Processing and Cryopreservation. Asian J Androl 5: 195-201.
  67. Partyka A, Jerysz A, Pokorny P (2007) Lipid Peroxidation in Fresh and Stored Semen of Green-Legged Partridge. EJPAU 10: 08.
  68. Partyka A, Łukaszewicz E, Niżański W (2012a) Lipid peroxidation and antioxidant enzymes activity in avian semen. Anim Reprod Sci 134: 184-190.
  69. Partyka A, Łukaszewicz E, Niżański W (2012b) Effect of cryopreservation on sperm parameters, lipid peroxidation and antioxidant enzymes activity in fowl semen. Theriogenology 77: 1497-1504.
  70. Partyka A, Łukaszewicz E, Niżański W, Twardoń J (2011) Detection of lipid peroxidation in frozen-thawed avian spermatozoa using C11-BODIPY(581/591). Theriogenology 75: 1623-1629.
  71. Partyka A, Niżański W (2021) Supplementation of Avian Semen Extenders with Antioxidants to Improve Semen Quality – Is It an Effective Strategy? Antioxidants (Basel) 10:1927.
  72. Partyka A, Niżański W, Łukaszewicz E (2010) Evaluation of fresh and frozen-thawed fowl semen by flow cytometry. Theriogenology 74: 1019-1027.
  73. Peris SI, Bilodeau J-F, Dufour M, Bailey JL (2007) Impact of cryopreservation and reactive oxygen species on DNA integrity, lipid peroxidation, and functional parameters in ram sperm. Mol Reprod Dev 74: 878-892.
  74. Rad HM, Eslami M, Ghanie A (2016) Palmitoleate enhances quality of rooster semen during chilled storage. Anim Reprod Sci 165: 38-45.
  75. Rosato MP, Centoducati G, Santacroce MP, Iaffaldano N (2012) Effects of lycopene on in vitro quality and lipid peroxidation in refrigerated and cryopreserved turkey spermatozoa. Br Poult Sci 53: 545-552.
  76. Rui BR, Shibuya FY, Kawaoku AJ, Losano JD, Angrimani DS, Dalmazzo A, Nichi M, Pereira RJ (2017) Impact of induced levels of specific free radicals and malondialdehyde on chicken semen quality and fertility. Theriogenology 90: 11-19.
  77. Sakkas D, Alvarez JG (2010) Sperm DNA fragmentation: mechanisms of origin, impact on reproductive outcome, and analysis. Fertil Steril 93: 1027-1036.
  78. Salehi M, Mahdavi AH, Sharafi M, Shahverdi A (2020) Cryopreservation of rooster semen: Evidence for the epigenetic modifications of thawed sperm. Theriogenology 142: 15-25.
  79. Simões R, Feitosa WB, Siqueira AF, Nichi M, Paula-Lopes FF, Marques MG, Peres MA, Barnabe VH, Visintin JA, Assumpção ME (2013) Influence of bovine sperm DNA fragmentation and oxidative stress on early embryo in vitro development outcome. Reproduction 146: 433-441.
  80. Słowińska M, Liszewska E, Judycka S, Konopka M, Ciereszko A (2018) Mitochondrial membrane potential and reactive oxygen species in liquid stored and cryopreserved turkey (Meleagris gallopavo) spermatozoa. Poult Sci 97: 3709-3717.
  81. Surai PF, Blesbois E, Grasseau I, Chalah T, Brillard J-P, Wishart GJ, Cerolini S, Sparks NH (1998) Fatty acid composition, glutathione peroxidase and superoxide dismutase activity and total antioxidant activity of avian semen. Comp Biochem Physiol B: Biochem Mol Biol 120: 527-533.
  82. Surai PF, Fujihara N, Speake BK, Brillard J-P, Wishart GJ, Sparks NH (2001) Polyunsaturated Fatty Acids, Lipid Peroxidation and Antioxidant Protection in Avian Semen – Review – Asian-Aust J Anim Sci 14: 1024-1050.
  83. Tamburrino L, Marchiani S, Montoya M, Elia Marino F, Natali I, Cambi M, Forti G, Baldi E, Muratori M (2011) Mechanisms and clinical correlates of sperm DNA damage. Asian J Androl 14: 24-31.
  84. Tesarik J Mendoza C, Greco E (2002) Paternal effects acting during the first cell cycle of human preimplantation development after ICSI. Hum Reprod 17: 184-189.
  85. Thananurak P, Chuaychu-Noo N, Thélie A, Phasuk Y, Vongpralub T, Blesbois E (2019) Sucrose increases the quality and fertilizing ability of cryopreserved chicken sperms in contrast to raffinose. Poult Sci 98: 4161-4171.
  86. Thananurak P, Chuaychu-Noo N, Thélie A, Phasuk Y, Vongpralub T, Blesbois E (2020) Different concentrations of cysteamine, ergothioneine, and serine modulate quality and fertilizing ability of cryopreserved chicken sperm. Poult Sci 99: 1185-1198.
  87. Thuwanut P, Axnér E, Johanisson A, Chatdarong K (2009) Detection of Lipid Peroxidation Reaction in Frozen-Thawed Epididymal Cat Spermatozoa Using BODIPY581/591C11. Reprod Domest Anim 44: 373-376.
  88. Trevizan JT, Carreira JT, Carvalho IR, Kipper BH, Nagata WB, Perri SH, Franco Oliveira ME, Pierucci JC, de Koivisto MB (2018) Does lipid peroxidation and oxidative DNA damage differ in cryopreserved semen samples from young, adult and aged Nellore bulls? Anim Reprod Sci 195: 8-15.
  89. Virro MR, Larson-Cook KL, Evenson DP (2004) Sperm chromatin structure assay (SCSA®) parameters are related to fertilization, blastocyst development, and ongoing pregnancy in in vitro fertilization and intracytoplasmic sperm injection cycles. Fertil Steril 81: 1289-1295.
  90. Watson PF (1995) Recent developments and concepts in the cryopreservation of spermatozoa and the assessment of their post-thawing function. Reprod Fertil Dev 7: 871.
  91. Wishart GJ (1984) Effects of lipid peroxide formation in fowl semen on sperm motility, ATP content and fertilizing ability. J Reprod Fertil 71: 113-118.
  92. Wolff SP, Dean RT (1986) Fragmentation of proteins by free radicals and its effect on their susceptibility to enzymic hydrolysis. Biochem J 234: 399-403.
  93. Zaniboni L, Cerolini S (2009) Liquid storage of turkey semen: Changes in quality parameters, lipid composition and susceptibility to induced in vitro peroxidation in control, n-3 fatty acids and alpha-tocopherol rich spermatozoa. Anim Reprod Sci 112: 51-65.
  94. Zini A, Garrels K, Phang D (2000) Antioxidant activity in the semen of fertile and infertile men. Urology 55: 922-926.
Go to article

Authors and Affiliations

A. Partyka
1
A. Babapour
2
M. Mikita
1
S. Adeniran
3
W. Niżański
1

  1. Wroclaw University of Environmental and Life Sciences, Faculty of Veterinary Medicine, Department of Reproduction and Clinic of Farm Animals, pl. Grunwaldzki 49, 50-366 Wroclaw, Poland
  2. University of Tabriz, Faculty of Veterinary Medicine, Department of Basic Sciences, Tabriz, Iran
  3. Mountain Top University, College of Basic and Applied Sciences, Department of Biological Sciences, Ogun State, Nigeria
Download PDF Download RIS Download Bibtex

Abstract

Home dental care is a key element of periodontal therapy in veterinary patients. Among many strategies of passive home dental care there is a supplementation of animal diet with seaweed Ascophyllum nodosum which have been shown to reduce both calculus and plaque accumulation after oral administration in both dogs and cats. Ascophyllum nodosum contains numerous biologically active ingredients, including micro-elements, vitamins, and several other compounds, however the exact mechanism of its beneficial action remains unclear. The very first metabolomic data suggest that it could change the composition of dog saliva. Several products containing Ascophyllum nodosum had been assessed clinically according to standards and requirements provided by the Veterinary Oral Health Council. The conducted clinical trials in dogs and cats revealed that Ascophyllum nodosum exerts the strongest preventive action as powder, followed by dental bites and dry pet food. The data concerning its curative action are limited to one study in cats in which no beneficial action has been observed. Based on available clinical data it is recommended to administer Ascophyllum nodosum to dogs and cats after oral cavity prophylactic procedure to reduce the recurrence of plaque and calculus formation.
Go to article

Bibliography

  1. Anthony JM, Weber LP, Alkemade S (2011) Blood glucose and liver function in dogs administered a xylitol drinking water additive at zero, one and five times dosage rates. Vet Sci Develop 1: e2.
  2. Bjone S, Brown W, Harris A, Genity PM (2007) Influence of chewing on dental health in dogs. In: Proc. 16th European Congress of Veterinary Dentistry, pp 45-46.
  3. Borah BM, Halter TJ, Xie B, Henneman ZJ, Siudzinski TR, Harris S, Elliott M, Nancollas GH (2014) Kinetics of canine dental calculus crystallization: an in vitro study on the influence of inorganic components of canine saliva. J Colloid Interface Sci. 425: 20-26.
  4. Boyce EN, Ching RJ, Logan EI, Hunt JH, Maseman DC, Gaeddert KL, King CT, Reid EE, Hefferren JJ (1995) Occurrence of gram-negative black-pigmented anaerobes in subgingival plaque during the development of canine periodontal disease. Clin Infect Dis 20 (Suppl 2): S317-S319.
  5. Bringel M, Jorge PK, Francisco PA, Lowe C, Sabino-Silva R, Colombini-Ishikiriama BL, Machado MAAM, Siqueira WL (2020) Salivary proteomic profile of dogs with and without dental calculus. BMC Vet Res. 16(1): 298.
  6. Brown WY, McGenity P (2005) Effective periodontal disease control using dental hygiene chews. J Vet Dent 22: 16-19.
  7. Capik I (2007) Periodontal health vs. different preventative means in toy breeds – clinical study. In: Proc.16th European Congress of Veterinary Dentistry, pp 31-34.
  8. Chapek CW, Reed OK, Ratcliff PA (1995) Reduction of bleeding on probing with oral-care products. Compend Contin Educ Dent 16: 188-192.
  9. Clarke DE (2006) Drinking water additive decreases plaque and calculus accumulation in cats. J Vet Dent 23: 79-82.
  10. Corba NH, Jansen J, Pilot T (1986) Artificial periodontal defects and frequency of tooth brushing in beagle dogs (II). Clinical findings after a period of healing. J Clin Periodontol 13: 186-189.
  11. Debowes LJ (2010) Problems with the gingiva. In: Niemiec BA (ed) Small animal dental, oral and maxillofacial disease, A color handbook; Manson, London, UK, pp 159-181.
  12. Della Riccia DN, Bizzini F, Perilli MG, Plimeni A, Trinchieri V, Amicosante G, Cifone MG (2007) Anti-inflammatory effects of lactobacillus brevis (CD2) on periodontal disease. Oral Dis 13: 376-385.
  13. Dunayer EK (2004) Hypoglycemia following canine ingestion of xylitol-containing gum. Vet Hum Toxicol 46: 87-88.
  14. Dunayer EK (2006) New findings on the effects of xylitol ingestion in dogs. Vet Med 101: 791-797.
  15. DuPont GA (1998) Prevention of periodontal disease. Vet Clin North Am Small Anim Pract. 28(5): 1129-1145.
  16. Fiorellini JP, Ishikawa SO, Kim DM (2006) Clinical Features of Gingivitis. In: Carranza’s Clinical Periodontology. WB Saunders, St. Louis, USA, pp 362-372.
  17. Gawor J, Jank M (2019) The curative usage of Ascophyllum nodosum powder in cats with periodontal disease (data not published, available in Authors)
  18. Gawor J, Jank M, Jodkowska K, Klim E, Svensson UK (2018) Effects of Edible Treats Containing Ascophyllum nodosum on the Oral Health of Dogs: A Double-Blind, Randomized, Placebo-Controlled Single-Center Study. Front Vet Sci 27: 168.
  19. Gawor J, Reiter AM, Jodkowska K, Kurski G, Wojtacki MP, Kurek A (2006) Influence of diet on oral health in cats and dogs. J Nutr 136: 2021S-2023S.
  20. Gawor JP, Wilczak J, Svensson UK, Jank M (2021) Influence of dietary supplementation with a powder containing Ascophyllum Nodosum algae on dog saliva metabolome. Front Vet Sci 8: 81951.
  21. Goñi O, Quille P, O’Connell S (2018) Ascophyllum nodosum extract biostimulants and their role in enhancing tolerance to drought stress in tomato plants. Plant Physiol Biochem 126: 63-73.
  22. Hale FA (2003) Home care for the veterinary dental patient. J Vet Dent 20: 52-54.
  23. Hamp SE, Emilson CG (1973) Some effects of chlorhexidine on the plaque flora of the beagle dog. J Periodontol Res 12: 28-35.
  24. Harvey CE (2005) Management of periodontal disease: understanding the options. Vet Clin North Am Small Anim Pract. 5(4): 819-836.
  25. Harvey CE, Shofer FS, Laster L (1996) Correlation of diet, other chewing activities, and periodontal disease in North American Client-owned dogs. J Vet Dent 13: 101-105.
  26. Hasturk H, Goguet-Surmenian E, Blackwood A, Andry C, Kantarci A (2009) 1-Tetradecanol complex: Therapeutic actions in experimental periodontitis. J Periodontol 80: 1103-1113.
  27. Hasturk H, Jones VL, Andry C, Kantarci A (2007) 1-Tetradecanol complex reduces progression of Porphyromonas gingivalis-induced experimental periodontitis in rabbits. J Periodontol 78: 924-932.
  28. Hennet P, Servet E, Salesse H, Soulard Y (2006) Evaluation of the Logan and Boyce Plaque Index for the Study of Dental Plaque Accumulation in Dogs. Res Vet Sci 80: 175-180.
  29. Hennet P, Servet E, Soulard Y, Biourge V (2007) Effect of pellet food size and polyphosphates in preventing calculus accumulation in dogs. J Vet Dent 24: 236-239.
  30. Hennet P, Servet E, Venet C (2006) Effectiveness of an oral hygiene chew to reduce dental deposits in small breed dogs. J Vet Dent 23: 6-12.
  31. Jank M (2021) Nutrition, oral health and feeding dental patients. In: Gawor J, Niemiec BA (eds) Veterinary Dental Patient. A Multidisciplinary approach. John Wiley and Sons, Hoboken, USA, pp 75-86.
  32. Jensen L, Logan E, Finney O, Lowry S, Smith M, Hefferren J, Simone A, Richardson D (1995) Reduction in accumulation of plaque, stain, and calculus in dogs by dietary means. J Vet Dent 12: 161-163.
  33. Kwon T, Lamster IB, Levin L (2021) Current concepts in the management of periodontitis. Int Dent J. 71(6): 462-476.
  34. Lage A, Lausen N, Tracy R, Allred E (1990) Effect of chewing rawhide and cereal biscuit on removal of dental calculus in dogs. J. Am Vet Med Assoc 197: 213-219.
  35. Lappin DF, Kjeldsen M, Sander L, Kinane DF (2000) Inducible nitric oxide synthase expression in periodontitis. J Periodontal Res 35: 369-373.
  36. Larsen J (2010) Oral products and dental disease. Comp Contin Educ Vet 32(9): E1-3.
  37. Liu H, Segreto VA, Baker RA, Vastola KA, Ramsey LL, Gerlach RW (2002) Anticalculus efficacy and safety of a novel whitening dentifrice containing sodium hexametaphosphate: a controlled six-month clinical trial. J Clin Dent 13: 25-28.
  38. Loe H (1967) The gingival index, the plaque index and the retention index systems. J Periodontol 38(6)Suppl: 610-616.
  39. Logan EI, Berg ML, Coffman L. et al. (1999) Dietary control of feline gingivitis: results of a six-month study. In: Proc. 13th Veterinary Dental Forum, pp 54.
  40. Logan EI, Boyce EN (1994) Oral health assessment in dogs: parameters and methods. J Vet Dent 11: 58-63.
  41. Logan EI, Finney O, Hefferren JJ (2002) Effects of a dental food on plaque accumulation and gingival health in dogs. J Vet Dent 19: 15-18.
  42. Logan EI, Proctor V, Berg ML, Coffman L, Hefferren JJ (2001) Dietary effect on tooth surface debris and gingival health in cats. In: Proc. 15th Annual American Veterinary Dental Forum, San Antonio, USA, p 377.
  43. Logan EI, Wiggs RB, Zetner K, Hefferren JJ (2000) Dental disease. In: Small animal clinical nutrition. 4th ed., Hand MS, Thacher CD, Remillard RL, Roudebush P (eds) Mark Morris Institute, Topeka KS, USA, pp 475-492.
  44. Matejka M, Partyka L, Ulm C, Solar P, Sinzinger H (1998) Nitric oxide synthesis is increased in periodontal disease. J Periodontal Res 33: 517-518.
  45. Milella L (2015) The negative effects of volatile sulphur compounds. J Vet Dent 32: 99–102.
  46. Moreira R, Sineiro J, Chenlo F, Arufe S, Díaz-Varela D (2017) Aqueous extracts of Ascophyllum nodosum obtained by ultrasound-assisted extraction: effects of drying temperature of seaweed on the properties of extracts. J Appl Phycol 29: 3191-3200.
  47. Needleman I, Suvan J, Moles DR, Pimlott J (2005) A systematic review of professional mechanical plaque removal for prevention of periodontal diseases. J Clin Periodontol 32 (Supp 6): 229-282.
  48. Niemiec B, Gawor J, Nemec A, Clarke D, McLeod K, Tutt C, Gioso M, Steagall PV, Chandler M, Morgenegg G, Jouppi R. McLeod K (2020) World small animal veterinary association global dental guidelines. J Small Anim Pract. 61(7): 395-403.
  49. Niemiec BA (2003) Professional teeth cleaning. J Vet Dent 20(3): 175-180.
  50. Niemiec BA (2021) Prophylactic Program for Oral Health. In: Gawor J, Niemiec BA (eds) Veterinary Dental Patient. A Multidisciplinary approach. John Wiley and Sons, Hoboken, USA, pp 59-62.
  51. Patent (2000) Oral preparation containing seaweed for reduction of plaque and calculus. International Patent Classification PCT/SE01/02083. 27.10.2000
  52. Paquette DW, Williams RC (2000) Modulation of host inflammatory mediators as a treatment strategy for periodontal diseases. Periodontology 24: 239-252.
  53. Payne WA, Page RC, Ogilvie AL, Hall WB (1975) Histopathologic features of the initial and early stages of experimental gingivitis in man. J Periodontal Res 10: 51.
  54. Perry DA (2006) Plaque control for the periodontal patient. In: Carranza’s Clinical Periodontology. WB Saunders, St. Louis, USA, pp 728-748.
  55. Pihlstrom BL, Michalowicz BS, Johnson NW (2005) Periodontal diseases. Lancet 366(9499): 1809-1820.
  56. Rawlings JM, Gorrel C, Markwell PJ (1998) Effect on Canine oral health of adding chlorhexidine to a dental hygiene chew. J Vet Dent 15: 129-134.
  57. Ray JD Jr, Eubanks DL (2009) Dental homecare: teaching your clients to care for their pet’s teeth. J Vet Dent 26(1): 57-60.
  58. Roudebush P, Logan E, Hale FA (2005) Evidence-based veterinary dentistry: a systematic review of homecare for prevention of periodontal disease in dogs and cats. J Vet Dent 22: 6-15.
  59. Shukla PS, Mantin EG, Adil M, Bajpai S, Critchley AT, Prithiviraj B (2019) Ascophyllum nodosum-Based Biostimulants: Sustainable Applications in Agriculture for the Stimulation of Plant Growth, Stress Tolerance, and Disease Management. Front Plant Sci 10: 655.
  60. Stookey GK, Warrick JM, Miller LL, Katz BP (1996) Hexametphosphate-coated snack biscuits significantly reduce calculus formation in dogs. J Vet Dent 13: 270-279.
  61. Tibbitts L, Kashiwa H (1970) A histochemical study of early plaque mineralization. J Dent Res 119: 202.
  62. van Dijken JW, Koistinen S, Ramberg P (2015) A randomized controlled clinical study of the effect of daily intake of Ascophyllum nodosum alga on calculus, plaque, and gingivitis. Clin Oral Investig 19: 1507-1518.
  63. Vrieling HE, Theyse LF, van Winkelhoff AJ, Dijkshoorn NA, Logan EI, Picavet P (2005) Effectiveness of feeding large kibbles with mechanical cleaning properties in cats with gingivitis. Tijdschr Diergeneeskd 130: 136-40.
  64. Warric JM, Stookey GK, Inskeep GA, Inskeep TK (2001) Reducing calculus accumulation in dogs using an innovative rawhide treat system coated with Hexametaphosphate. In: Proc. 15th Annual American Veterinary Dental Forum, San Antonio, USA, 379-382.
  65. White DJ, Cox ER, Suszcynskymeister E, Baig AA (2002) In vitro studies of the anticalculus efficacy of a sodium hexametaphosphate whitening dentifrice. J Clin Dent 13: 33-37.
  66. Wiggs RB, Lobprise HB (1997) Periodontology in veterinary dentistry, principals and practice. Lippincott – Raven: Philadelphia, USA, pp 186-231.
  67. VOHC Report (2018) Submission to VOHC for claims on Helps Control Plaque and Helps Control Tartar in dogs receiving product ProDen PlaqueOff Powder. Submitted 2018.10.22. www.vohc.org.
  68. VOHC Report (2020a) Submission to VOHC regarding application for use of the VOHC seal for the product Pro Den Plaque Off Powder from Swedencare AB. Submitted 2020.09.10. www.vohc.org.
  69. VOHC Report (2020b) Submission to VOHC regarding application for use of the VOHC seal for the product Canagan Dental For Dogs from Symply Pet Foods Ltd. Submitted 2020.10.01. www.vohc.org.
  70. Xia Z, He Y, Yu J (2009) Experimental acute toxicity of xylitol in dogs. J Vet Pharmacol Therapeutics 32: 465-469.
Go to article

Authors and Affiliations

J. Gawor
1
M. Jank
2

  1. Klinika Arka, Chłopska 2a, 30-806 Krakow, Poland
  2. Institute of Veterinary Medicine, Department of Pre-Clinical Sciences, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
Download PDF Download RIS Download Bibtex

Abstract

Viral infections are common causes of diseases in animals and appropriate methods are increasingly being required to detect viral pathogens in animals. In this regard, similar to antigen- -antibody interactions, aptamers have high affinity and specificity for their respective target molecules, and can be selected using the Systematic Evolution of Ligands by EXponential enrichment (SELEX) technique. Recently, significant progress has been made in the development of aptamer selection and aptamer-based sensors for viral detection, and here we review some of the recent advances in aptamer-based detection of viral infections in animals. This review will serve as a comprehensive resource for aptamer-based strategies in viral diagnostics.
Go to article

Bibliography

  1. Bai H, Wang RH, Hargis B, Lu HG, Li YB (2012) A SPR aptasensor for detection of avian influenza virus H5N1. Sensors 12: 12506-12518.
  2. Banerjee J, Nilsen-Hamilton M (2013) Aptamers: multifunctional molecules for biomedical research. J Mol Med 91: 1333-1342.
  3. Bruno JG, Carrillo MP, Phillips T (2008) Development of DNA aptamers to a foot-and-mouth disease peptide for competitive FRET-based detection. J Biomol Tech 19: 109-115.
  4. Chakraborty B, Das S, Gupta A, Xiong YY, Vushnavi T-V, Kizer ME, Duan JW, Chandrasekaran AR, Wang X (2022). Aptamers for viral detection and inhibition. ACS Infect Dis 8: 667-692.
  5. Chauhan VM, Elsutohy MM, McClure CP, Irving WL, Roddis N, Aylott JW (2021) Gold-Oligonucleotide nanoconstructs engineered to detect conserved enteroviral nucleic acid sequences. Biosensors 11:238.
  6. Chen CH, Zou Z, Chen L, Ji XH, He ZK (2016) Functionalized magnetic microparticle-based colorimetric platform for influenza A virus detection. Nanotechnology 27: 435102.
  7. Chen ZQ, Wu QH, Chen J, Ni XH, Dai JF (2020) A DNA aptamer based method for detection of SARS-CoV-2 nucleocapsid protein. Virol Sin 35: 351-354.
  8. D’Cruz RJ, Currier AW, Sampson VB (2020) Laboratory testing methods for novel Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2). Front Cell Dev Biol 8: 468.
  9. Damase TR, Miura TA, Parent CE, Allen PB (2018) Application of the Open qPCR Instrument for the in Vitro Selection of DNA aptamers against epidermal growth factor receptor and Drosophila C virus. ACS Comb Sci 20: 45-54.
  10. Darmostuk M, Rimpelova S, Gbelcova H, Ruml T (2015) Current approaches in SELEX: An update to aptamer selection technology. Biotechnol Adv 33: 1141-1161.
  11. Diba FS, Kim S, Lee HJ (2015) Amperometric bioaffinity sensing platform for avian influenza virus proteins with aptamer modified gold nanoparticles on carbon chips. Biosens Bioelectron 72: 355-361.
  12. Ellenbecker M, Sears L, Li P, Lanchy JM, Lodmell JS (2012) Characterization of RNA aptamers directed against the nucleocapsid protein of Rift Valley fever virus. Antiviral Res 93: 330-339.
  13. Ellington AD, Szostak JW (1990) In vitro selection of RNA molecules that bind specific ligands. Nature 346: 818-822.
  14. Hmila I, Wongphatcharachai M, Laamiri N, Aouini R, Marnissi B, Arbi M, Streevatsan S, Ghram A (2017) A novel method for detection of H9N2 influenza viruses by an aptamer-real time-PCR. J Virol Methods 243: 83-91.
  15. Hong KL, Sooter LJ (2015) Single-Stranded DNA aptamers against pathogens and toxins: identification and biosensing applications. Biomed Res Int 2015: 419318.
  16. Hwang SD, Midorikawa N, Punnarak P, Kikuchi Y, Kondo H, Hirono I, Aoki T (2012) Inhibition of Hirame rhabdovirus growth by RNA aptamers. J Fish Dis 35: 927-934.
  17. Iliuk AB, Hu LH, Tao WA (2011) Aptamer in bioanalytical applications. Anal Chem 83: 4440-4452.
  18. Jafari M, Rezaei M, Kalantari H, Tabarzad M, Daraei B (2018) DNAzyme-aptamer or aptamer-DNAzyme paradigm: Biochemical approach for aflatoxin analysis. Biotechnol Appl Biochem 65:274-280.
  19. Kacherovsky N, Yang LF, Dang HV, Cheng EL, Cardle II, Walls AC, McCallum M, Sellers DL, DiMaio F, Salipante SJ, Corti D, Veesler D, Pun SH (2021) Discovery and characterization of spike N-Terminal domain-binding aptamers for rapid SARS-CoV-2 detection. Angew Chem Int Ed Engl 60: 21211-21215.
  20. Kaur A, Kaur P, Ahuja S (2021) Förster resonance energy transfer (FRET) and applications thereof. Anal Methods 12:5532-5550.
  21. Kim YS, Gu MB (2014) Advances in aptamer screening and small molecule aptasensors. Adv Biochem Eng Biotechnol 140: 29-67.
  22. Labib M, Zamay AS, Muharemagic D, Chechik A, Bell JC, Berezovski MV (2012) Electrochemical sensing of aptamer-facilitated virus immunoshielding. Anal Chem 84: 1677-1686.
  23. Le TT, Adamiak B, Benton DJ, Johnson CJ, Sharma S, Fenton R, McCauley JW, Iqbal M, Cass, AEG (2014) Aptamer-based biosensors for the rapid visual detection of flu viruses. Chem Commun (Camb) 50: 15533-15536.
  24. Lee JL, Stovall GM, Ellington AD (2006) Aptamer therapeutics advance. Curr Opin Chem Biol 10: 282-289.
  25. Li JX, Zhang ZJ, Gu J, Stacey HD, Ang JC, Capretta A, Filipe CDM, Mossman KL, Balion C, Salena BJ, Yamamura D, Soleymani L, Miller MS, Brennan JD, Li YF (2021) Diverse high-affinity DNA aptamers for wild-type and B.1.1.7 SARS-CoV-2 spike proteins from a pre-structured DNA library. Nucleic Acids Res 49: 7267-7279.
  26. Li P, Zhou L, Wei J, Yu Y, Yang M, Wei S, Qin Q (2016) Development and characterization of aptamer-based enzyme-linked apta-sorbent assay for the detection of Singapore grouper iridovirus infection. J Appl Microbiol 121: 634-643.
  27. Lichty BD, Power AT, Stojdl DF, Bell JC (2004) Vesicular stomatitis virus: re-inventing the bullet. Trends Mol Med 10: 210-216.
  28. Liu JX, Qin QW, Zhang XY, Li C, Yu YP, Huang XH, Mukama O, Zeng LW, Wang SW (2020) Development of a novel lateral flow biosensor combined with aptamer-based isolation: application for rapid detection of grouper nervous necrosis virus. Front Microbiol 11: 886.
  29. Lou BB, Liu YF, Shi ML, Chen J, Li K, Tan YF, Chen LW, Wu YW, Wang T, Liu XQ, Jiang T, Peng DM, Liu ZB (2022) Aptamer-based biosensors for virus protein detection. Trends Analyt Chem 157: 116738.
  30. Lu TF, Ma Q, Yan WZ, Wang YZ, Zhang YY, Zhao LL, Chen HY (2018) Selection of an aptamer against Muscovy duck parvovirus for highly sensitive rapid visual detection by label-free aptasensor. Talanta 176: 214-220.
  31. Lum J, Wang RH, Hargis B, Tung S, Bottje W, Lu HG, Li YB (2015) An impedance aptasensor with microfluidic chips for specific detection of H5N1 avian influenza virus. Sensors 15: 18565-18578.
  32. Negri P, Chen GJ, Kage A, Nitsche A, Naumann D, Xu BQ, Dluhy RA (2012) Direct optical detection of viral nucleoprotein binding to an anti-influenza aptamer. Anal Chem, 84: 5501-5508.
  33. Ouellet E, Foley JH, Conway EM, Haynes C (2015) Hi-Fi SELEX: A high-fidelity digital-PCR based therapeutic aptamer discovery platform. Biotechnol Bioeng 112: 1506-1522.
  34. Park JW, Lee SJ, Choi EJ, Kim J, Song JY, Gu MB (2014) An ultra-sensitive detection of a whole virus using dual aptamers developed by immobilization-free screening. Biosens Bioelectron 51: 324-329.
  35. Pfeiffer F, Mayer G (2016) Selection and biosensor application of aptamers for small molecules. Front Chem 4: 25.
  36. Prabhakar PK, Lakhanpal J (2020) Recent advances in the nucleic acid-based diagnostic tool for coronavirus. Mol Biol Rep 47: 9033-9041.
  37. Reinemann C, Stoltenburg R, Strehlitz B (2009) Investigations on the specificity of DNA aptamers binding to ethanolamine. Anal Chem 81: 3973-3978.
  38. Robertson DL, Joyce GF (1990) Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA. Nature 344: 467-468.
  39. Romero-Lopez C, Berzal-Herranz A (2017) Aptamers: Biomedical interest and applications. Pharmaceuticals 10:32.
  40. Sett A, Das S, Bora U (2014) Functional nucleic-acid-based sensors for environmental monitoring. Appl Biochem Biotechnol 174: 1073-1091.
  41. Storch GA (2000) Diagnostic virology. Clin Infect Dis 31: 739-751.
  42. Tuerk C, Gold L (1990) Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249: 505-510.
  43. Wang HY, Wu SQ, Jiang L, Xiao RH, Li T, Mei L, Lv JZ, Liu JJ, Lin XM, Han XQ (2018) Establishment and optimization of a liquid bead array for the simultaneous detection of ten insect-borne pathogens. Parasit Vectors 11: 442.
  44. Wang RH, Li YB (2013) Hydrogel based QCM aptasensor for detection of avian influenza virus. Biosens Bioelectron 42: 148-155.
  45. Wang RH, Xu LZ, Li YB (2015) Bio-nanogate controlled enzymatic reaction for virus sensing. Biosens Bioelectron 67: 400-407.
  46. Wu JJ, Zhu YY, Xue F, Mei ZL, Yao L, Wang X, Zheng L, Liu J, Liu GD, Peng CF, Chen W (2014) Recent trends in SELEX technique and its application to food safety monitoring. Mikrochim Acta 181: 479-491.
  47. Zhang ZJ, Pandey R, Li JX, Gu J, White D, Stacey HD, Ang JC, Steinberg CJ, Capretta A, Filipe CDM, Mossman K, Balion C, Miller MS, Salena BJ, Yamamura D, Soleymani L, Brennan JD, Li YF (2021) High-affinity dimeric aptamers enable the rapid electrochemical detection of wild-type and B.1.1.7 SARS-CoV-2 in Unprocessed Saliva. Angew Chem Int Ed Engl 60: 24266-24274.
Go to article

Authors and Affiliations

W. Zhang
1
L. Xiao
2
J. Luo
1
M. Wu
2
Y. Zhu
2
F. Cong
2

  1. Guangdong Eco-Engineering Polytechnic, 297# Guangshan 1st Road, Guangzhou 510520, Guangdong, China
  2. Guangdong Laboratory Animals Monitoring institute and Guangdong Provincial Key Laboratory of Laboratory Animals, 11# Fengxin Road, Guangzhou 510033, Guangdong, China

This page uses 'cookies'. Learn more