Life Sciences and Agriculture

Polish Journal of Veterinary Sciences

Content

Polish Journal of Veterinary Sciences | 2025 | vol. 28 | No 2

Keywords: 175-181
Download PDF Download RIS Download Bibtex

Abstract

In this study, the effects of in-ovo injection of Chrysin (CR) into Japanese quail eggs on testicular histology, oxidant status and epididymal sperm quality were investigated. 720 Japa­nese quail eggs were divided into 4 groups and 0.1 mL saline was given to the control group, 0.25 mg CR to the 0.25 group, 0.50 mg CR to the 0.50 group and 0.75 mg CR to the 0.75 group. On the 60th day after the laying of eggs, 8 Japanese quails were randomly selected from each group and sacrificed under mild sevoflurane anesthesia. The abdominal cavity was opened and both testicular tissues and epididymal parts were removed. The right testicular tissue was used for histopathological examinations and the left one was used for biochemical analyses. The epididy­mal part at the tip of the vas deferens at the end of the right testis was trimmed in 100 μL saline at 37°C. Histopathological examinations showed that histological scoring was higher in the CR 50 and CR 75 groups. A decrease in oxidant status was observed in all CR groups compared to the control group. Spermatozoa density was higher in CR groups compared to the control group. Total motility value was statistically significantly higher in CR 50 and CR 75 groups compared to control and CR 25 groups. There was no statistical difference between the groups in terms of dead and abnormal spermatozoa. In-ovo CR injection at doses of 0.50 and 0.75 mg/egg improved testicular histological score, decreased oxidative stress, and increased epididymal sperm quality.

Go to article

Bibliography

Akintunde AO, Ndubuisi-Ogbonna LC, Ladele MM, Olorunfemi OA, Ojo OA, Oyewumi SO, Shobo BA, Akinboye OE (2023) Spermiogramic parameters of Japanese quails (Coturnix coturnix japonica) to aqueous administration of egg lime molasses mixture. Ovozoa: Journal of Animal Reproduction. 12:59-70

Akintunde AO, Toye AA, Ademola AA, Chimezie, VO. Ajayi OA (2021) Genotype-diet effect on comparative semen parameters of chickens fed graded levels of Moringa oleifera seed meal. Mal. J. Anim. Sci. 24: 41-55.

AL-Jomaily TK, Taha AT, (2020) Effects in-ovo injection of L-carnitine on hatchability, productive and physiological traits of Japanese quail (Coturnix Japonica). Plant Archives 20: 4051-4055.

Angelopoulou E, Pyrgelis ES, Piperi C (2020) Neuroprotective potential of chrysin in Parkinson’s disease: Molecular mechanisms and clinical implications. Neurochem International 132: 104612.

Aygun A (2016) The effects of in-ovo injection of propolis on egg hatchability and starter live performance of Japanese quails. Brazil J Poult Sci 18: 83-89.

Cerutti C, Leroux S, Gourichon D, Labrune Y, David I, Zerjal T, Coustham V, Devailly G, Pitel F (2022) Short communication: Effects of in-ovo injection of endocrine disruptors and methyltransferase inhibitor on quail growth and egg-laying performances. Animal 16: 100464.

Chen W, Wang R, Xiong XL, Wan HF, Xu J, Peng J (2010) Influence of in ovo injection of disaccharides, glutamine and β-hydroxy-β-methylbutyrate on the development of small intestine in duck embryos and neonates. Br Poult Sci 51: 592-601.

Çeribaşı S, Türk G, Özçelik M, Doğan G, Çeribaşı AO, Mutlu İflazoğlu S, Erişir Z, Güvenç M, Güngören G, Acısu TC, Akarsu SA, Özer Kaya Ş, Sönmez M, Yüce A, Çiftçi M, Çambay Z, Bağcı E, Azman MA, Şimşek ÜG (2020) Negative effect of feeding with high energy diets on testes and metabolic blood parameters of male Japanese quails, and positive role of milk thistle seed. Theriogenology 144: 74-81.

Demir Ayazoğlu E, Mentese A, Demir S, Küçük H, Türkmen N, Aliyazıcıoğlu Y (2023) Evaluation of therapeutic effect of chrysin against 5-fluorouracil-induced ovarian damage in rats. Farabi Med J 2(1): 1-7.

El-Kazaz SE, Abo-Samaha MI Hafez MH, El-Shobokshy SA, Wirtu G (2020) Dietary supplementation of nano-selenium improves reproductive performance, sexual behavior and deposition of selenium in the testis and ovary of Japanese quail. J Adv Vet Anim Res 7(4): 597.

Erel O (2004) A novel automated direct measurement method for total antioxidant capacity using a new generation, more stable ABTS radical cation. Clinic Biochem 37: 277-285.

Erel O (2005) A new automated colorimetric method for measuring total oxidant status. Clinic Biochem 38: 1103-1111.

Farkhondeh T, Samarghandian S, Bafandeh F (2019) The cardiovascular protective effects of chrysin: a narrative review on experimental researches. Cardiovasc Hematol Agents Med Chem 17: 17-27.

Foye OT, Uni Z, Ferket PR (2006) Effect of in ovo feeding egg white protein, β-hydroxy-β-methylbutyrate, and carbohydrates on glycogen status and neonatal growth of turkeys. Poult Sci 85: 1185-1192.

Genc M, Kandemir FM, Coban O (2019) Effects of In-Ovo Rutin Injection to Fertile Japanese Quail (Coturnix Coturnix Japonica) Egg on Hatchability, Embryonic Death, Hatchling Weight, and Hatchling Liver Oxidative and Nitrosative Stress. Brazil J Poult Sci 21: 1-8.

Hajati H, Zaghari M, Noori O, Negarandeh R, de Oliveira HC (2021) Effects of in ovo injection of microalgae on hatchability, antioxidant and immunity-related genes expression, and post-hatch performance in broilers and Japanese quails. Ital J Anim Sci 20: 985-994.

Hanedan B, Ozkaraca M, Kirbas A, Kandemir FM, Aktas MS, Kilic K, Comakli S, Kucukler S, Bilgili A (2018) Investigation of the effects of hesperidin and chrysin on renal injury induced by colistin in rats. Biomed Pharmacother 108: 1607-1616.

Huss D, Poynter G, Lansford R (2008) Japanese quail (Coturnix japonica) as a laboratory animal model. Lab Anim 37: 513-519.

Ibrahim MI, Williams J, Botha CJ (2021) Dose-related morphological changes in the epididymal region of sexually active adult male Japanese quail treated with di-n-butyl phthalate (DBP) commencing during the pre-pubertal stage. Anim Reprod Sci 227: 106733.

Ileriturk M, Benzer F, Aksu EH, Yildirim S, Kandemir FM, Dogan T, Dortbudak MB, Genc A (2021) Chrysin protects against testicular toxicity caused by lead acetate in rats with its antioxidant, anti‐inflammatory, and antiapoptotic properties. J Food Biochem 45: e13593.

Kadam MM, Bhanja SK, Mandal AB, Thakur R, Vasan P, Bhattacharyya A, Tyagi JS (2008) Effect of in ovo threonine supplementation on early growth, immunological responses and digestive enzyme activities in broiler chickens. Brit Poult Sci 49: 736-741.

Kandemir FM, Kucukler S, Eldutar E, Caglayan C, Gülçin İ (2017) Chrysin protects rat kidney from paracetamol-induced oxidative stress, inflammation, apoptosis, and autophagy: a multi-biomarker approach. Sci Pharm 85: 4.

Kazaz IO, Demir S, Kerimoglu G, Colak F, Alemdar Türkmen N, Akman AU, Cekuc OC, Mentese A (2022) Effect of chrysin on endoplasmic reticulum stress in a rat model of testicular torsion. J Invest Surg 35: 1106-1111.

Kermanshahi H, Ghofrani Tabari D, Khodambashi Emami N, Daneshmand A, Ibrahim S (2017a) Effect of in ovo injection of threonine on immunoglobulin A gene expression in the intestine of Japanese quail at hatch. J Anim Physiol Anim Nutr 101: 10-14.

Kermanshahi H, Golian A, Khodambashi Emami N, Daneshmand A, Ghofrani Tabari D, Ibrahim SA (2017b) Effects of in ovo injection of threonine on hatchability, intestinal morphology, and somatic attributes in Japanese quail (Coturnix japonica) Journal of applied Anim Res 45: 437-441.

Kucukler S, Benzer F, Yildirim S, Gur C, Kandemir FM, Bengu AS, Ayna A, Caglayan C, Dortbudak MB (2021) Protective effects of chrysin against oxidative stress and inflammation induced by lead acetate in rat kidneys: a biochemical and histopathological approach. Biol Trace Elem Res 199: 1501-1514.

Mutlu M, Simsek UG, Iflazoglu S, Yilmaz A, Karabulut B, Incili CA, Cevik A, Incili GK, Seven Tatlı P, Mutlu SI (2024) Potential effect dietary supplementation of calcium tetraborate in quails exposed to cadmium: Its impact on productive performance, oxidative stress, cecal microflora, and histopathological changes. Ecotoxicol Environ Saf 270: 115883.

Salmanzadeh M, Bostanabad JG, Arva S (2014) The effects of in ovo injection of butyric acid into quails breeder eggs on hatchability, growth performance, development of the gastrointestinal tract, and carcass traits of Japanese quails. Bull Environ Pharmacol Life Sci 3(3): 126-130.

Sarabia Fragoso J, Pizarro Díaz M, Abad Moreno JC, Casanovas Infesta P, Rodriguez‐Bertos A, Barger K (2013) Relationships between fertility and some parameters in male broiler breeders (body and testicular weight, histology and immunohistochemistry of testes, spermatogenesis and hormonal levels) Reprod Domest Anim 48: 345-352.

Surai PF, Noble RC, Speake BK (1996) Tissue-specific differences in antioxidant distribution and susceptibility to lipid peroxidation during development of the chick embryo. Biochim Biophys Acta 1304: 1-10.

Taslimi P, Kandemir FM, Demir Y, İleritürk M, Temel Y, Caglayan C, Gulçin İ (2019) The antidiabetic and anticholinergic effects of chrysin on cyclophosphamide‐induced multiple organ toxicity in rats: Pharmacological evaluation of some metabolic enzyme activities. J Biochem Mol Tox 33: e22313.

Temel Y, Kucukler S, Yıldırım S, Caglayan C, Kandemir FM (2020) Protective effect of chrysin on cyclophosphamide-induced hepatotoxicity and nephrotoxicity via the inhibition of oxidative stress, inflammation, and apoptosis. Naunyn Schmiedebergs Archives Pharmacol 393: 325-337.

Uni Z, Ferket R (2004) Methods for early nutrition and their potential. World’s Poult Sci J 60: 101-111.

Uni Z, Yadgary L, Yair R (2012) Nutritional limitations during poultry embryonic development. J Appl Poult Res 21: 175-184.

Utsumi T, Yoshimura Y (2009) Sensitive embryonic endpoints with in ovo treatment for detecting androgenic and anti-androgenic effects of chemicals in Japanese quail (Coturnix japonica) Poult Sci 88: 1052-1059.

 

 

Go to article

Authors and Affiliations

U. Özentürk
1
M. Genç
1
A.D. Ömür
2
E. Erbaş
3
A. Uysal
1
A. Yörü
A. Genç
4
S.A. Akarsu
2

  1. Deparment of Animal Science, Faculty of Veterinary Medicine, Atatürk University, 25010 Erzurum, Turkey
  2. Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine,Atatürk University, 25010 Erzurum, Turkey
  3. Department of Histology and Embryology, Faculty of Veterinary Medicine,Atatürk University, 25010 Erzurum, Turkey
  4. Department of Biochemistry, Faculty of Veterinary Medicine, Bingöl University, 12010 Bingöl, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Intestinal ischemia-reperfusion (IR) injury is a major clinical challenge due to its high morbidity and mortality rates. This study aims to demonstrate the effect of honokiol, a natural antioxidant compound, on intestinal IR injury in rats using histochemical and biochemical methods. The protein-protein interaction (PPI) network construction and the honokiol-target network- -reactome pathway analysis were performed using Cytoscape v3.10.1 software to validate inclusion of focused proteins in the study. 1 hour/2 hours of IR was applied on intestinal (jejunum) tissues. The tissues were further processed for biochemical measurement of total oxidant status (TOS) and antioxidant status (TAS). 5 mg/kg honokiol treatment was administered to rats after ischemia protocol. The tissues were fixed in formaldehyde and embedded in paraffin protocol. Sections were stained with vascular endothelial growth factor (VEGF), a disintegrin and metalloproteinase with thrombospondin motifs 15 (ADAMTS-15) and caspase-3 antibodies. Analysis of the signaling network revealed that honokiol exerts a significant influence on the proposed mechanisms associated with IR through the VEGF, ADAMTS-15, and caspase-3 network. IR increased the TOS level and decreased the TAS level in ischemia and IR group, histopathologically damaged the intestinal tissues and led to epithelial degeneration, increased cell death, vascular dilatation and congestion. Honokiol treatment reduced the oxidant enzymes and supported the antioxidant system, and restored pathologies in the IR+honokiol group. Intestinal IR injury increased VEGF expression, ADAMTS-15 and caspase-3 expression in the ischemia and IR groups. Honokiol treatment after ischemia reduced the VEGF, ADAMTS15 and caspase-3 by restoring tissue integrity, preventing cell death and increasing cell matrix remodeling. The administration of honokiol provided protection against intestinal IR injury by modulating apoptosis, angiogenesis, extracellular matrix remodeling processes through regulation of the VEGF, ADAMTS-15, and caspase-3 expression.
Go to article

Bibliography

Ates B, Yilmaz I, Geckil H, Iraz M, Birincioglu M, Fiskin K (2004) Protective role of melatonin given either before ischemia or prior to reperfusion on intestinal ischemia‐reperfusion damage. J Pineal Res 37: 149-152.

Aycicek A, Ipek A (2008) Maternal active or passive smoking causes oxidative stress in cord blood. Eur J Pediatr 167: 81-85.

Boutin AT, Weidemann A, Fu Z, Mesropian L, Gradin K, Jamora C, Wiesener M, Eckardt K-U, Koch CJ, Ellies LG, Haddad G, Haase VH, Simon MC, Poellinger L, Powell FL, Johnson RS (2008) Epidermal Sensing of Oxygen Is Essential for Systemic Hypoxic Response. Cell 133: 223-234.

Buchman AL, Moukarzel AA, Bhuta S, Belle M, Ament ME, Eckhert CD, Hollander D, Gornbein J, Kopple JD, Vijayaroghavan SR (1995) Parenteral Nutrition Is Associated with Intestinal Morphologic and Functional Changes in Humans. JPEN J Parenter Enteral Nutr 19: 453-460.

Canillioglu YE, Senturk GE (2020) Alterations of IL-1 and VEGF After Ischemia-Reperfusion Injured Uterus and Ovary in Rats. Medeni Méd J 35: 106-115.

Chao LK, Liao PC, Ho CL, Wang EI, Chuang CC, Chiu HW, Hung LB, Hua KF (2010) Anti-Inflammatory Bioactivities of Honokiol through Inhibition of Protein Kinase C, Mitogen-Activated Protein Kinase, and the NF kappa B Pathway To Reduce LPS-Induced TNF alpha and NO Expression. J Agric Food Chem 58: 3472-3478.

Chen C, Zhang QW, Ye Y, Lin LG (2021) Honokiol: A naturally occurring lignan with pleiotropic bioactivities. Chin J Nat Med 19: 481-490.

Chen HY, Hung YC, Lee EJ, Chen TY, Chuang IC, Wu TS (2009) The protective efficacy of magnolol in hind limb ischemia-reperfusion injury. Phytomedicine 16: 976-981.

Cho SY, Lee JH, Bae KH, Kim YS, Jeong CS (2008) Anti-gastritic Effects of Magnolol and Honokiol from the Stem Bark of Magnolia obovata. Biomol Ther 16: 270-276.

Daniel RA, Cardoso VK, Gois E Jr, Parra RS, Garcia SB, da Rocha JJ, Féres O (2011) Effect of hyperbaric oxygen therapy on the intestinal ischemia reperfusion injury. Acta Cir Bras 26: 463-469.

Debsharma S, Pramanik S, Bindu S, Mazumder S, Das T, Saha D, De R, Nag S, Banerjee C, Siddiqui AA, Ghosh Z, Bandyopadhyay U (2023) Honokiol, an inducer of sirtuin‐3, protects against non‐steroidal anti‐inflammatory drug‐induced gastric mucosal mitochondrial pathology, apoptosis and inflammatory tissue injury. Br J Pharmacol 180: 2317-2340.

Demirpençe O, Sevim B, Yıldırım M, Nurlu NA, Mert D, Evliyaoğlu O (2014) Serum paraoxonase, TAS, TOS and ceruloplasmin in brucellosis. Int J Clin Exp Med 7: 1592-1597

Deng Y, Han X, Tang S, Li C, Xiao W, Tan Z (2018) Magnolol and Honokiol Attenuate Apoptosis of Enterotoxigenic Escherichia Coli-Induced Intestinal Epithelium by Maintaining Secretion and Absorption Homeostasis and Protecting Mucosal Integrity. Méd Sci Monit 24: 3348-3356.

Duffy MJ, McKiernan E, O’Donovan N, McGowan PM (2009) The role of ADAMs in disease pathophysiology. Clin Chim Acta 403: 31-36.

Edwards DR, Handsley MM, Pennington CJ (2008) The ADAM metalloproteinases. Mol Asp Med 29: 258-289.

Erel O (2005) A new automated colorimetric method for measuring total oxidant status. Clin Biochem 38: 1103-1111.

Ferrara N (2004) Vascular Endothelial Growth Factor: Basic Science and Clinical Progress. Endocr Rev 25: 581-611.

Ferrara N, Gerber H-P, LeCouter J (2003) The biology of VEGF and its receptors. Nat Med 9: 669-676.

Goldsmith JR, Perez-Chanona E, Yadav PN, Whistler J, Roth B, Jobin C (2013) Intestinal Epithelial Cell-Derived μ-Opioid Signaling Protects against Ischemia Reperfusion Injury through PI3K Signaling. Am J Pathol 182: 776-785.

Gonzalez LM, Moeser AJ, Blikslager AT (2015) Animal models of ischemia-reperfusion-induced intestinal injury: progress and promise for translational research. Am J Physiol-Gastrointest Liver Physiol 308: G63-G75.

Gordeeva AE, Sharapov MG, Tikhonova IV, Chemeris NK, Fesenko EE, Novoselov VI, Temnov AA (2017) Vascular Pathology of Ischemia/Reperfusion Injury of Rat Small Intestine. Cells Tissues Organs 203: 353-364.

He GZ, Zhou KG, Zhang R, Wang YK, Chen XF (2012) Impact of intestinal ischemia/reperfusion and lymph drainage on distant organs in rats. World J Gastroenterol 18: 7271-7278.

Hu H, Zhang X, Wang Y, Chen S (2005) Honokiol inhibits arterial thrombosis through endothelial cell protection and stimulation of prostacyclin. Acta Pharmacol Sin 26: 1063-1068.

Kelwick R, Desanlis I, Wheeler GN, Edwards DR (2015) The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol 16: 113.

Kuida K, Zheng TS, Na S, Kuan C-Y, Yang D, Karasuyama H, Rakic P, Flavell RA (1996) Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice. Nature 384: 368-372.

Kumar S, Rao N, Ge R (2012) Emerging Roles of ADAMTSs in Angiogenesis and Cancer. Cancers (Basel) 4:1252-1299.

Li J, Chen Y, Gao L, Li Y, Feng D (2023) Honokiol Prevents Intestinal Barrier Dysfunction in Mice with Severe Acute Pancreatitis and Inhibits JAK/STAT1 Pathway and Acetylation of HMGB1. Chin J Integr Med doi:10.1007/s11655-023-3562-y.

Liu KJ, Rosenberg GA (2005) Matrix metalloproteinases and free radicals in cerebral ischemia. Free Radic Biol Med 39: 71-80.

Makino Y, Cao R, Svensson K, Bertilsson G, Asman M, Tanaka H, Cao Y, Berkenstam A, Poellinger L (2001) Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature 414: 550-554.

McIlwain DR, Berger T, Mak TW (2013) Caspase Functions in Cell Death and Disease. Cold Spring Harb Perspect Biol 5: a008656.

Nagy JA, Dvorak AM, Dvorak HF (2007) VEGF-A and the Induction of Pathological Angiogenesis. Annu Rev Pathol 2: 251-275.

Niu L, Wang J, Shen F, Gao J, Jiang M, Bai G (2022) Magnolol and honokiol target TRPC4 to regulate extracellular calcium influx and relax intestinal smooth muscle. J Ethnopharmacol 290: 115105.

Ong CP, Lee WL, Tang YQ, Yap WH (2019) Honokiol: A Review of Its Anticancer Potential and Mechanisms. Cancers (Basel) 12: 48.

Qiang LQ, Wang CP, Wang FM, Pan Y, Yi LT, Zhang X, Kong LD (2009) Combined administration of the mixture of honokiol and magnolol and ginger oil evokes antidepressant-like synergism in rats. Arch Pharm Res 32: 1281-1292.

Rauf A, Olatunde A, Imran M, Alhumaydhi FA, Aljohani AS, Khan SA, Uddin MS, Mitra S, Emran TB, Khayrullin M, Rebezov M, Kamal MA, Shariati MA (2021) Honokiol: A review of its pharmacological potential and therapeutic insights. Phytomedicine 90: 153647.

Schweizer E, Gassel A, Deltz E, Schroeder P (1992) Morphologic and histologic alterations after small-bowel transplantation a comparison of different perfusion solutions. Transplant Proc 24(3): 1087.

Spanos CP, Papaconstantinou P, Spanos P, Karamouzis M, Lekkas G, Papaconstantinou C (2007) The Effect of L-arginine and Aprotinin on Intestinal Ischemia – reperfusion Injury. J Gastrointest Surg 11: 247-255.

Taha MO, Ferreira RM, Taha NS, Monteiro HP, Caricati-Neto A, Oliveira-Júnior IS, Fagundes DJ (2013) Ischemic preconditioning and the gene expression of enteric endothelial cell biology of rats submitted to intestinal ischemia and reperfusion. Acta Cir Bras 28:167-173.

Takeshita M, Tani T, Harada S, Hayashi H, Itoh H, Tajima H, Ohnishi I, Takamura H, Fushida S, Kayahara M (2010) Role of Transcription Factors in Small Intestinal IschemiaReperfusion Injury and Tolerance Induced by Ischemic Preconditioning. Transplant Proc 42: 3406-3413.

Tang X, Yao K, Zhang L, Yang Y, Yao H (2011) Honokiol inhibits H2O2-induced apoptosis in human lens epithelial cells via inhibition of the mitogen-activated protein kinase and Akt pathways. Eur J Pharmacol 650: 72-78.

Woo M, Hakem R, Furlonger C, Hakem A, Duncan GS, Sasaki T, Bouchard D, Lu L, Wu GE, Paige CJ, Mak TW (2003) Caspase-3 regulates cell cycle in B cells: a consequence of substrate specificity. Nat Immunol 4:1016-1022.

Woo M, Hakem R, Soengas MS, Duncan GS, Shahinian A, Kägi D, Hakem A, McCurrach M, Khoo W, Kaufman SA, Senaldi G, Howard T, Lowe SW, Mak TW (1998) Essential contribution of caspase 3/CPP32 to apoptosis and its associated nuclear changes. Genes Dev 12:806-819.

Yazici S, Demirtas S, Guclu O, Karahan O, Yavuz C, Caliskan A, Mavitas B (2014) Using oxidant and antioxidant levels to predict the duration of both acute peripheral and mesenteric ischemia. Perfusion 29: 450-455.

Zhai T, Wang J, Chen Y (2023) Honokiol affects the composition of gut microbiota and the metabolism of lipid and bile acid in methionine-choline deficiency diet-induced NASH mice. Sci Rep 13: 15203.

Zhang B, Wang PP, Hu KL, Li LN, Yu X, Lu Y, Chang H-S (2019) Antidepressant-Like Effect and Mechanism of Action of Honokiol on the Mouse Lipopolysaccharide (LPS) Depression Model. Molecules 24: 2035.

Zhang X, Chen S, Wang Y (2007) Honokiol up-regulates prostacyclin synthease protein expression and inhibits endothelial cell apoptosis. Eur J Pharmacol 554: 1-7.

Zhou YH, Han QF, Gao L, Sun Y, Tang ZW, Wang M, Wang W, Yao HC (2020) HMGB1 Protects the Heart Against Ischemia-Reperfusion Injury via PI3K/AkT Pathway-Mediated Upregulation of VEGF Expression. Front Physiol 10: 1595.

 

Go to article

Authors and Affiliations

E. Gökalp Özkorkmaz
1
F. Aşır
2
T. Korak
3
Y. Özay
4
E. Deveci
2

  1. Department of Nursery, Faculty of Health Science, Ankara Yıldırım Beyazıt University,Esenboğa Campus, 06760 Çubuk, Ankara, Türkiye
  2. Department of Histology and Embryology, Medical Faculty, Dicle University,Campus Area 21280 Sur, Diyarbakır, Türkiye
  3. Department of Medical Biology, Faculty of Medicine, Kocaeli University,Umuttepe Campus 41380 Kocaeli, Türkiye
  4. Department of Medical Biology, Medical Faculty, Adıyaman University,Campus Area 02040 Adıyaman, Türkiye
Download PDF Download RIS Download Bibtex

Abstract

Effective control of the cat flea (Ctenocephalides felis) infestation is essential for human and animal health. To date, several ectoparasitic drugs, such as carbamates, neonicotinoids, phenylpyrazoles, and pyrethroids have been used to control flea infestations; however their widespread use has led to resistance, hampering treatment success. In this study, the resistance potential of C. felis collected from cats to fipronil (FIP), imidacloprid (IMI), flumethrin (FLU), and propoxur (PRO), which are the commonly used compounds for flea control, was investigated by molecular analyses. Bioassays encompassed exposure of adult cat fleas to FIP (2%), IMI (6%), FLU (0.1%), and PRO (0.1%)-impregnated papers using an insecticide susceptibility test kit according to the WHO protocol. Afterward, PCR was performed to identify mutations indicating resistance to FIP and FLU. Based on molecular analyses, mutations in the para gene of C. felis were identified as L1014F and T929V, indicating FLU resistance. Frequencies of T929V and L1014F mutations in fleas were 2.5% and 10%, respectively. On the other hand, two fleas that survived after eight hours of exposure to FIP lacked the A302S mutation, which was partially associated with FIP resistance. Even though our results revealed no resistance to FIP, IMI, and PRO in fleas collected from the study’s cat population, the occurence of FLU resistance due to mutations in the para gene of the fleas was demonstrated at the molecular level.

Go to article

Bibliography

Akküçük Ş, Kaya ÖM, Karagöz M, Zerek A, Yaman M (2019) The louse and flea infestations in cats and dogs in Antakya animal shelter. Van Vet J 30: 37-40

Bass C, Schroeder I, Turberg A, Field LM, Williamson MS (2004) Identification of mutations associated with pyrethroid resistance in the para-type sodium channel of the cat flea, Ctenocephalides felis. Insect Biochem Mol Biol 34: 1305-1313.

Beugnet F, Franc M (2012) Insecticide and acaricide molecules and/or combinations to prevent pet infestation by ectoparasites. Trends Parasitol 28: 267-279.

Bossard RL (1997) Evaluation and use of bioassays for surveying insecticide susceptibility of cat fleas, Ctenocephalides felis felis Bouche, in relation to resistance. PhD Thesis. Kansas State Univ. Kansas, United States

Chandra S, Forsyth M, Lawrence AL, Emery D, Slapeta J (2017) Cat fleas (Ctenocephalides felis) from cats and dogs in New Zealand: Molecular characterisation, presence of Rickettsia felis and Bartonella clarridgeiae and comparison with Australia. Vet Parasitol 234: 25-30.

Coles TB, Dryden MW (2014) Insecticide/acaricide resistance in fleas and ticks infesting dogs and cats. Parasit Vectors 7: 8.

Costa LG, Hand H, Schwab BW, Murphy SD (1981) Tolerance to the carbamate insecticide propoxur. Toxicology 21: 267-278.

El-Gazzar LM, Patterson RS, Koehler PG (1988) Comparisons of cat flea (Sphonaptera: Pulicidae) adult and larval insecticide susceptibility. Florida Entomol 71: 359-363.

Erkunt Alak S, Köseoğlu AE, Kandemir Ç, Taşkın T, Demir S, Döşkaya M, Ün C, Can H (2020) High frequency of knockdown resistance mutations in the para gene of cat flea (Ctenocephalides felis) samples collected from goats. Parasitol Res 119: 2067-2073.

Ffrench-Constant RH, Anthony N, Aronstein K, Rocheleau T, Stilwell G (2000) Cyclodiene insecticide resistance: from molecular to population genetics. Annu Rev Entomol 45: 449-466.

Ffrench-Constant RH, Rocheleau TA, Steichen JC, Chalmers AE (1993) A point mutation in a Drosophila GABA receptor confers insecticide resistance. Nature 363: 449-451.

Genchi M, Vismarra A, Zanet S, Morelli S, Galuppi R, Cringolli G, Lia R, Diaferia M, di Regalbono A, Venegoni G, Solari Basano F, Varcasia A, Perrucci S, Musella V, Brianti E, Gazzonis A, Drigo M, Colombo L, Kramer L (2021) Prevalence and risk factors associated with cat parasites in Italy: a multicenter study. Parasit Vectors 14: 475.

Ghavami MB, Haghi FP, Alibabaei Z, Enayati AA, Vatandoost H (2018) First report of target site insensitivity to pyrethroids in human flea, Pulex irritans (Siphonaptera: Pulicidae). Pestic Biochem Physiol 146: 97-105.

González-Morales MA, DeVries ZC, Santangelo RG, Kakumanu ML, Schal C (2022) Multiple Mechanisms Confer Fipronil Resistance in the German Cockroach: Enhanced Detoxification and Rdl Mutation. J Med Entomol 59: 1721-1731.

Gracia MJ, Calvete C, Estrada R, Castillo JA, Peribanez MA, Lucientes J (2012) Survey of flea infestation in cats in Spain. Med Vet Entomol 27: 175-180.

Hosie AM, Baylis HA, Buckingham SD, Sattelle DB (1995) Actions of the insecticide fipronil, on dieldrin‐sensitive and resistant GABA receptors of Drosophila melanogaster. Br J Pharmacol 115: 909-912.

Karunaratne SH, De Silva WA, Weeraratne TC, Surendran, SN (2018) Insecticide resistance in mosquitoes: development, mechanisms and monitoring. Ceylon J Sci 47: 299-309.

Kopp S, Blagburn B, Coleman G, Davis W, Denholm I, Field C, Hostetler J, Mencke N, Rees R, Rust M, Williamson, M. S. (2013) Monitoring field susceptibility to imidacloprid in the cat flea: a world-first initiative twelve years on. Parasitol Res 112 (Suppl. 1): 47-56.

Lappin MR, Tasker S, Roura X (2020) Role of vector-borne pathogens in the development of fever in cats: 1. Flea-associated diseases. J Feline Med Surg 22: 31-39.

Leul B, Berihun A, Etsay K (2024) Epidemiology of Small Ruminant External Parasites: In the Case of Chemical Control Campaign in Welkait District, Tigray Region, Ethiopia. Asian J Res Anim Vet Sci 7: 159-174.

Rajamannar V, Govindarajan R, Kumar A, Samuel PP (2022) A review of public health important fleas (Insecta, Siphonaptera) and flea-borne diseases in India. J Vector Borne Dis 59: 12-21.

Rust MK (2016) Insecticide resistance in fleas. Insects 7: 10.

Rust MK, Vetter R, Denholm I, Blagburn B, Williamson MS, Kopp S, Coleman G, Hostetler J, Davis W, Mencke N, Rees R, Foit S, Böhm C, Tetzner K (2015) Susceptibility of Adult Cat Fleas (Siphonaptera: Pulicidae) to Insecticides and Status of Insecticide Resistance Mutations at the Rdl and Knockdown Resistance Loci. Parasitol Res 114 (Suppl. 1): 7-18.

Sakuma M (1998) Probit analysis of preference data. Appl Entomol Zool 33: 339-347.

Seidy S, Tavassoli, M, Malekifard F (2022) Pyrethroids resistance in Pulex irritans and Ctenocephalides canis in west and northwest Iran. Vet Res Forum 13: 529-535.

Slapeta J, King J, McDonell D, Malik R, Homer D, Hannan P, Emery D (2011) The cat flea (Ctenocephalides f. felis) is the dominant flea on domestic dogs and cats in Australian veterinary practices. Vet Parasitol 180: 383-388.

Thomas JE, Staubus L, Goolsby JL, Reichard MV (2016) Ectoparasites of free-roaming domestic cats in the central United States. Vet Parasitol 228: 17-22.

Van der Mescht L, Matthee S, Matthee CA (2021) New taxonomic and evolutionary insights relevant to the cat flea, Ctenocephalides felis: A geographic perspective. Mol Phylogenet Evol 155: 106990.

World Health Organization (1970) Instructions for determining the susceptibility or resistance of fleas to insecticides. 100-105. In Insecticide resistance and vector control. Technical Report Series No. 443. WHO, Geneva, Switzerland. Available at: https://iris.who.int/bitstream/handle/10665/
40771/WHO_TRS_443_%28part1%29.pdf?sequence=
1&isAllowed=y
Accessed: 10.07.2024.

World Health Organization (2016) Reflection paper on resistance in ectoparasites. Committee for Medicinal Products for Veterinary Use (CVMP). Available at: https://www.ema.europa.eu/en/documents/scientific-guideline/draft-reflection
-paper-resistance-ectoparasites_en.pdf
Accessed: 10.07.2024.

Xhaxhiu D, Kusi I, Rapti D, Visser M, Knaus M, Lindner T, Rehbein S (2009) Ectoparasites of dogs and cats in Albania. Parasitol Res 105: 1577-1587.

Zhang Y, Nie Y, Deng YP, Liu GH, Fu YT (2021) The complete mitochondrial genome sequences of the cat flea Ctenocephalides felis felis (Siphonaptera: Pulicidae) support the hypothesis that C. felis isolates from China and USA were the same C. f. felis subspecies. Acta Trop 217: 105880.

 

 

 

Go to article

Authors and Affiliations

T. Kuntuz
1
Y. Gunes
2
B. Diren Sigirci
3
C. Anlas
2
A.B. Sari
2
T. Bakirel
2
O. Ustuner
2

  1. Istanbul University-Cerrahpasa, Institute of Graduate Studies, 34320 Istanbul, Turkey
  2. Istanbul University-Cerrahpasa, Faculty of Veterinary Medicine,Department of Pharmacology and Toxicology, 34500 Istanbul, Turkey
  3. Istanbul University-Cerrahpasa, Faculty of Veterinary Medicine,Microbiology Department, 34500 Istanbul, Turkey
Download PDF Download RIS Download Bibtex

Abstract

In Northern Cyprus, around 51% of halloumi cheese is produced from sheep milk, and there­fore the livelihood of the farmers mostly depends on the sheep milk production. However masti­tis, an inflammation of the udder, significantly affects this production. Due to a lack of sufficient data concerning the prevalence, etiology, and antimicrobial resistance of sheep mastitis, there remains no effective method to control the disease. This study aims to estimate the prevalence of subclinical mastitis (SCM) and identify bacterial etiological agents and the antimicrobial sus­ceptibility profiles of bacterial isolates in sheep in Northern Cyprus. A total of 227 milk samples taken from sheep were analysed using somatic cell count (SCC), bacteriological isolation-identi­fication, and antimicrobial susceptibility procedures. Pathogens were isolated in 62 (27.3%) sheep milk samples. Somatic cell counts of more than 500000 cells/ml were found in 56 (24.6%). S. aureus (12.8%) was the most common isolate from the milk samples, followed by NAS (non-Aureus staphylococci) species (11.9%), Escherichia coli (0.9%), Streptococci (0.4%), Bacillus spp. (0.9%) and Staph spp. (0.4%). While a high resistance to sulphamethaxazole/trime­toprim (81.5%) was found, no resistance to gentamicin (10.6%) was found. The study findings indicate that subclinical mastitis is a serious problem in Cyprus. Therefore, continuous observa­tion of subclinical mastitis and application of antibiogram tests to combat mastitis and antibiotic resistance and reduce economic losses are needed.

Go to article

Bibliography

Abdalhamed AM, Zeedan GSG, Zeina HAAA (2018) Isolation and identification of bacteria causing mastitis in small ruminants and their susceptibility to antibiotics, honey, essential oils, and plant extracts. Vet World 11: 355-362.

Ahmed G, Timms LL, Morrical DG, Brackelsberg PO (1992) Dynamics and significance of ovine subclinical intramammary infections and their effects on lamb performance. Sheep Res J 8: 25-29.

Alba DF, da Rosa G, Hanauer D, Saldanha TF, Souza CF, Baldissera MD, Da Silva dos Santos D, Piovezan AP, Girardini LK, Da Silva AS (2019) Subclinical mastitis in Lacaune Sheep: causative agents, impacts on milk production, milk quality, oxidative profiles and treatment efficacy of Ceftiofur. Microb Pathog 137: 103732.

Alekish MO, Alshehabat MA, Abutarbush SM (2014) The prevalence and etiology of subclinical mastitis in Awassi sheep; emphasis on the relationship between the ısolated organisms and the somatic cell count. EJVM 8: 1-13.

Al-Majali AM, Jawabieh S (2003) Period prevalence and etiology of subclinical mastitis in Awassi sheep in Southern Jordan. Small Rumin Res 47: 243-248.

Attili AR, Preziuso S, Ngwa VN, Cantalamessa A, Moriconi M, Cuteri V (2016) Clinical evaluation of the use of Enrofloxacin against staphylococcus aureus clinical mastitis in sheep. Small Rumin Res 136: 72-77.

Bauer AW, Kirby WM, Sherris JC, Turck M (1996) Antibiotic susceptibility testing by standardised single disk method. Am J Clin Pathol 45: 493–6.

Bergonier D, Berthelot X (2003) New advances in epizootiology and control of ewe mastitis. Livest Prod Sci 79: 1-16.

Berthelot X, Lagriffoul G, Concordet D, Barillet F, Bergonier D (2006) Physiological and pathological thresholds of somatic cell counts in ewe milk. Small Rumin Res 62: 27-31.

Bradley AJ (2002) Bovine mastitis: an evolving disease. Vet J 164: 116-128.

Caboni P, Manis C, Ibba I, Contu M, Coroneo V, Scano P (2017) Compositional profile of ovine milk with a high somatic cell count: a metabolomics approach. Int Dairy J 69: 33-39.

Contreras GA, Rodríguez JM (2011) Mastitis: comparative etiology and epidemiology. J Mammary Gland Biol Neoplasia 16: 339-356.

da Silva ER, Siqueira AP, Martins JC, Ferreira WP, da Silva N (2004) Identification and in vitro antimicrobial susceptibility of Staphylococcus species isolated from goat mastitis in the Northeast of Brazil. Small Rumin Res 55: 45-49.

Daniel WW (1999) Biostatistics: a foundation for analysis in the health sciences. 7th ed., John Wiley & Sons, New York.

Dore S, Liciardi M, Amatiste S, Bergagnacy S, Bolzoni G, Caligiuri V, Cerrone A, Farina G, Montagna CO, Saletti MA, Scatassa ML, Sotgiu G, Cannas EA (2016) Survey on small ruminant bacterial mastitis in Italy, 2003-2014. Small Rum Res 141: 91-93.

El-Jakee J, Hableel HS, Kandil M, Hassan OF, Khairy EA, Marouf SA (2013) Antibiotic resistance patterns of Streptococcus agalactiae isolated from mastitic cows and ewes in Egypt. Glob Vet 10: 264-270.

Ergun Y, Aslantas O, Dogruer G, Kirecci E, Sarıbay MK, Ates CT, Ulku A, Demir C (2009) Prevalence and etiology of subclinical mastitis in Awassi dairy ewes in Southern Turkey. Turk J Vet Anim Sci 33: 5.

Fragkou IA, Boscos CM, Fthenakis GC (2014) Diagnosis of clinical or subclinical mastitis in ewes. Small Rumin Res 118: 86-92.

Gebrewahid TT, Abera BH, Menghistu HT (2012) Prevalence and etiology of subclinical mastitis in small ruminants of Tigray Regional State, North Ethiopia. Vet World 5: 103-109.

Gelasakis AI, Mavrogianni VS, Petridis IG, Vasileiou NG, Fthenakis GC (2015) Mastitis in sheep–the last 10 years and the future of research. Vet Microbiol 181: 136-146.

Gokhan M, Gulaydin O (2020) Prevalance and antimicrobial susceptibility of bacterial species isolated from ovine clinical mastitis cases in Van province. Etlik Vet Mikrobiyol Derg 31: 39-46.

Guler L, Ok U, Gunduz K, Gulcu Y, Hadimli HH (2005) Antimicrobial susceptibility and coagulase gene typing of Staphylococcus aureus ısolated from bovine clinical mastitis cases in Turkey. J Dairy Sci 88: 3149-3154.

Hariharan H, Donachie W, Macaldowie C, Keefe G (2004) Bacteriology and somatic cell counts in milk samples from ewes on a Scottish farm. Can J Vet Res 68: 188-192.

Harmon RJ (2001) Somatic cell count: A primer. In: Annual Meeting National Mastitis Council Reno, Proceedings. Madison: National Mastitis Council 40: 3-9.

Hawari AD, Obeidat M, Awaisheh SS, Al-Daghistani HI, Al-Abbadi AA, Omar SS, Qrunfleh IM, Al-Dmoor HM, El-Qudah J (2014) Prevalence of mastitis pathogens and their resistance against antimicrobial agents in Awassi sheep in Al-Balqa Province of Jordan. Am J Anim Vet Sci 9: 116-121.

Holko I, Tančin V, Tvarožková K, Supuka P, Supuková A, Mačuhová L (2019) Occurrence and antimicrobial resistance of common udder pathogens ısolated from sheep milk in Slovakia. Potr Slov J F and Sci 13: 258-261.

Jones JE (1984) An Investigation of Mastitis in Sheep in Cyprus. Report to the Veterinary Department, Ministry of Agriculture, Nicosia, p 47.

Kaskous S, Farschtschi S, Pfaffl MW (2022) Physiological aspects of milk somatic cell count in small ruminants – A Review. Dairy 4: 26-42.

Kern G, Traulsen I, Kemper N, Krieter J (2013) Analysis of somatic cell counts and risk factors associated with occurrence of bacteria in ewes of different primary purposes. Livest Sci 157: 597-604.

Kiossis E, Brozos CN, Petridou E, Boscos C (2007) Program for the control of subclinical mastitis in dairy Chios breed ewes during lactation. Small Rumin Res 73: 194-199.

Kunz F, Corti S, Giezendanner N, Stephan R, Wittenbrink MM, Zweifel C (2011) Antimicrobial resistance of Staphylococcus aureus and coagulase negative Staphylococci isolated from mastitis milk samples from sheep and goats. Schweiz Arch Tierheilkd 153: 63-69.

Leitner G, Silanikove N, Merin U (2008) Estimate of milk and curd yield loss of sheep and goat with intrammamary infection and its relation to somatic cell count. Small Rumin Res 74: 221-225.

Lollai SA, Ziccheddu M, Di Mauro C, Manunta D, Nudda A, Leori G (2008) Profile and evolution of antimicrobial resistance of ovine mastitis pathogens (1995-2004). Small Rumin Res 74: 249–254.

Martins HB, Selis ND, Nascimento FS, de Carvalho SP, Gusmão LD, Nascimento JD, Brito AK, de Souza SI, de Oliveira MV, Timenetsky J, Yatsuda R, Uetanabaro AP, Marques LM (2017) Anti-inflammatory activity of the essential oil citral in experimental infection with Staphylococcus aureus in a model air pouch. Evid Based Complement Alternat Med 2017: 2505610.

Maurer J, Schaeren W (2007) Udder health and somatic cell count in ewes. Agrarforschung 14: 162-167.

Mavrogenis AP, Koumas A, Kakoyiannis CK, Taliotis CH (1995) Use of somatic cell counts for the detection of subclinical mastitis in sheep. Small Rumin Res 17: 79-84.

McDougall S, Pankey W, Delaney C, Barlow J, Murdough PA, Scruton D (2002) Prevalence and incidence of subclinical mastitis in goats and dairy ewes in Vermont, USA. Small Rumin Res 46: 115-121.

Moawad AA, Osman SA (2005) Prevalence and etiology of subclinical mastitis in dairy ewes at Fayoum Governorate, Egypt. Assiut Vet Med J 51: 1-15.

Moroni P, Pisoni G, Varisco G, Boettcher P (2007) Effect of intramammary infection in Bergamasca meat sheep on milk parameters and lamb growth. J Dairy Res 74: 340-344.

Naccari F, Martino D, Giofrè F, Passantino A, De Montis P (2003) Therapeutic efficacy of Tilmicosin in ovine mammary infections. Small Rumin Res 47: 1-9.

Novick JR, William J (1989). Development of in vitro susceptibility testing criteria and quality control parameters. Clin. Microbiol. Newsl 11: 60-62.

Nunes GR, Blagitz MG, Freitas CB, Souza FN, Ricciardi M, Stricagnolo CR, Sanches BG, Azedo MR, Sucupira MC, Della Libera AM (2008) Evaluation of the indicators of inflammation in the diagnosis of ovine mastitis. Arq Inst Biol 75: 271-281.

Olivares-Pérez J, Kholif AE, Rojas-Hernández S, Elghandour MM, Salem AZM, Bastida AZ, Velázquez-Reynoso D, Cipriano-Salazar M, Camacho-Díaz LM, María Uxúa Alonso-Fresán MU, DiLorenzo N (2015) Prevalence of bovine subclinical mastitis, its etiology and diagnosis of antibiotic resistance of dairy farms in four municipalities of a tropical region of Mexico. Trop Anim Health Prod 47: 1497-1504.

Oliver SP, Murinda SE (2012) Antimicrobial resistance of mastitis pathogens. Vet Clin North Am Food Anim Prat 28: 165-185.

Ozenc E, Seker E, Baki Acar D, Birdane MK, Darbaz I, Dogan N (2011) The importance of Staphylococci and threshold value of somatic cell count for diagnosis of sub-clinical mastitis in Pirlak sheep at mid-lactation. Reprod Domest Anim 46: 970-974.

Paterna A, Contreras A, Gómez-Martín A, Amores J, Tatay-Dualde J, Prats-van der Ham M, Corrales JC, Sanchez A, De la Fe C (2014) The Diagnosis of mastitis and contagious agalactia in dairy goats. Small Rumin Res 121: 36-41.

Petersson-Wolfe CS, Mullarky IK, Jones GM (2010) Staphylococcus Aureus Mastitis: Cause, Detection, and Control; VirginiaTech: Blacksburg, VA, USA, 2010.

Podhorecka K, Borkova M, Sulc M, Seydlova R, Dragounova H, Svejcarova M, Peroutkova J, Elich O (2021) Somatic cell count in goat milk: an indirect quality indicator. FoodS 10: 1046.

Queiroga MC (2017) Prevalence and aetiology of sheep mastitis in Alentejo regions of Portugal. Small Rumin Res 153: 123-130.

Quinn PJ, Markey BK, Leonard FC, Fitzpatrick ES, Fanning S, Hartigan PJ (2011) Veterinary Microbiology and Microbial Disease, 2nd ed. West Sussex: Wiley-Blackwell, 837-850.

Quinn PJ, Markey BK, Carter ME, Donelly WJ, Leonard FC (2002) Veterinary Microbiology and Microbial Disease, Blackwell Science Ltd, a Blackwell Publishing Company, 465 475.

Rall VL, Miranda ES, Castilho IG, Camargo CH, Langoni H, Guimarães FF, Araújo Júnior JP, Fernandes Júnior A (2014) Diversity of Staphylococcus species and prevalence of enterotoxin genes isolated from milk of healthy cows and cows with subclinical mastitis. J Dairy Sci 97: 829-837.

Raynal-Ljutovac K, Pirisi A, De Crémoux R, Gonzalo C (2007) Somatic cells of goat and sheep milk: analytical sanitary, productive and technological aspects. Small Rumin Res 68: 126-144.

Riggio V, Pesce LL, Morreale S, Portolano B (2013) Receiveroperating characteristic curves for somatic cell scores and california mastitis test in Valle del Belice dairy sheep. Vet J 196: 528-532.

Sevi A, Albenzio M, Taibi L, Dantone D, Massa S, Annicchiarico G (1999) Changes of somatic cell count through lactation and their effects on nutritional, renneting and bacteriological characteristics of ewe’s milk. Adv Food Sci 21: 122-127.

Stocco G, Summer A, Cipolat-Gotet C, Zanini L, Vairani D, Dadousis C, Zecconi A (2020) Differential somatic cell count as a novel indicator of milk quality in dairy cows. Animals (Basel) 10: 753.

Świderek WP, Charon KM, Winnicka A, Gruszczyńska J, Pierzchała M (2016) Physiological threshold of somatic cell count in milk of polish heath sheep and polish lowland sheep. Ann Anim Sci 16: 155-170.

Tvarožková K, Tančin V, Holko I, Uhrinčať M, Mačuhová L (2019) Mastitis in ewes: somatic cell counts, pathogens and antibiotic resistance. J Microbiol Biotech Food Sci 9: 661-670.

Tvarožková K, Tančin V, Uhrincat M, Hleba L, Mačuhová L (2020) Mastitis pathogens and somatic cell count in ewes milk. Potravinarstvo Slovak J Food Sci 14: 164-169.

Tvarozkova K, Vasicek J, Uhrinca M, Macuhova L, Hleba L, Tancin V (2021) The presence of pathogens in milk of ewes in relation to the somatic cell count and subpopulations of leukocytes. Czech J Anim Sci 66: 315-322.

Vasıľ M, Elečko J, Farkašová Z, Zigo F (2018) Development of resistance to antibiotics in bacteria Staphylococcus sp. isolated from milk samples in the sheep breedings on East of Slovakia. Potravinarstvo Slovak J Food Sci 12: 273-278.

Vasileiou NG, Cripps PJ, Ioannidi KS, Chatzopoulos DC, Gougoulis DA, Sarrou S, Orfanou DC, Politis AP, Gonzalez-Valerio TC, Argyros S, Mavrogianni VS, Petinaki E, Fthenakis GC (2018) Extensive countrywide field investigation of subclinical mastitis in sheep in Greece. J Dairy Sci 101: 7297-7310.

Wald R, Hess C, Urbantke V, Wittek T, Baumgartner M (2019) Characterization of Staphylococcus species isolated from bovine quarter milk samples. Animals (Basel) 9: 200.

Zafalon LF, Santana RC, Pilon LE, Fim Júnior GA (2016) Diagnosis of subclinical mastitis in Santa Ines and Morada Nova sheep in Southeastern Brazil. Trop Anim Health Prod 48: 967-72.

Zdragas A, Tsakos P, Kotzamanidis C, Anatoliotis K, Tsaknakis I (2005) Outbreak of mastitis in ewes caused by Streptococcus Agalactiae. J Hellenic Vet Med Society 6: 114-121.

Zigo F, Vasiľ M, Kadáši M, Elečko J, Farkašová Z (2011) Bacteria Staphylococcus spp. izolated from mastitis of sheep and their enterotoxigenic properties. Potravinarstvo 5: 70-72.

 

Go to article

Authors and Affiliations

O. Ergene
1
H. Baloglu
2
V. Haciogullari
2
H.E. Çolakoğlu
3

  1. Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Near East University, 99158Nicosia, North Cyprus, Mersin-10, Turkey
  2. Directorate of Veterinary Department, Nicosia, Cyprus
  3. Ankara University, Faculty of Veterinary Medicine, Department of Obstetrics and Gynecology, 06110,Ankara, Turkey
Download PDF Download RIS Download Bibtex

Abstract

This study aimed to evaluate the effects of tideglusib and bone graft mixture on bone healing. Tideglusib is a drug used in the treatment of various neurological disorders such as Alzheimer’s disease. In a relevant study, the positive effect of tideglusib on the Wnt pathway, one of the pathways involved in bone regeneration and dentin tissue regeneration, was demonstrated. Dentin and bone tissues have structurally similar healing mechanisms. Therefore, tideglusib may have a similar effect on the bone tissue. The main goal of bone grafting is to provide bone regeneration and functional healing through remodeling. Bone graft materials are divided into four types based on their source: autogenous, allogenous, xenogenous, and alloplastic. Because these graft materials have various advantages and disadvantages, research continues to focus on alternative materials and applications. Sixteen New Zealand rabbits were included in this study. A unicortical 3.5 mm diameter defect was created in the tibia of rabbits under general anesthesia. The groups in the study were as follows: Group 1, left proximal tibia defect area was controlled (defect area was left empty); Group 2, left distal tibia defect area was treated with tideglusib + calcium sulfate; Group 3, right proximal tibia defect area was treated with calcium sulfate only; Group 4, right distal tibia defect area was treated with tideglusib only. Mediolateral (M/L) radiographs of the tibia were taken on the 30th and 60th postoperative days. On the 30th day, the first eight rabbits were sacrificed, and on the 60th day, the remaining eight were sacrificed for histopathological examination. New bone formation in the obtained samples was evaluated by radiological and histopathological analyses. The study concluded that the combination of tideglusib and calcium sulfate significantly enhanced bone healing compared with the other groups (p<0.005). This suggests that tideglusib, either alone or in combination with bone graft materials, could serve as a promising alternative for the repair of bone defects.
Go to article

Bibliography

Amini AR, Laurencin CT, Nukavarapu SP (2012) Bone tissue engineering: recent advances and challenges. Crit Rev Biomed Eng 40: 363-408.

Arioka M, Takahashi-Yanaga F, Sasaki M, Yoshihara T, Morimoto S, Hirata M, Mori Y, Sasaguri T (2014) Acceleration of bone regeneration by local application of lithium: Wnt signal-mediated osteoblastogenesis and Wnt signal-independent suppression of osteoclastogenesis. Biochem Pharmacol 90: 397-405.

Audisio SA, Vaquero PG, Torres PA, Verna EC, Ocampo LN, Cristofolini AL, Merkins CI (2015) Radiological evaluation of radial bone defects treated with demineralized bone matrix in an experimental rabbit model. Bulg J Vet Med 18: 132-139.

Aysan AL, Çalışır M, Kızıldağ A, Özdede M, Özmen Ö (2020) Effects of a glycogen synthase kinase 3 inhibitor tideglusib on bone regeneration with calvarial defects. J Craniofac Surg 31: 1477-1482.

Brinker MR, O’Conner DP (2008) Basic sciences. In: Miller MD (ed) Review of orthopaedics. 5th ed., Saunders Elsevier, Philadelphia, pp 1-132.

Bulut S, Durmuş AS, Köm M, Çobanoğlu B (2001) Use of the distal portion of ulna as a segmental cortical autograft for experimentally induced fractures with large bone defect on the diaphysis of the tibia and femur in dogs. Kafkas Univ Vet Fak Derg 7: 77-85.

Cavelier S, Tanzer M, Barthelat F (2020) Maximizing the strength of calcium sulfate for structural bone grafts. J Biomed Mater Res A 108: 963-971.

Chen H, Cui X, Yu X, Tian XZ, Zhang BX, Tang P, Wnag Y (2010) Effects of chitosan-coated pressed calcium sulfate pellets combined with recombinant human bone morphogenetic protein 2 on bone formation in femoral condyle-contained bone defects. J Craniofac Surg 21: 188-197.

Cho BC, Kim TG, Yang JD, Chung HY, Park JW, Kwon IC, Roh KH, Chung HS, Lee DS, Park NU, Kim IS (2005) Effect of calcium sulfate-chitosan composite: pellet on bone formation in bone defect. J Craniofac Surg 16: 213-224.

Clevers H, Nusse R (2012) Wnt/β-catenin signalling and disease. Cell 149: 1192-1205.

Comeau-Gauthier M, Tarchala M, Luna JL, Harvey E, Merle G (2020) Unleashing β-catenin with a new anti-Alzheimer drug for bone tissue regeneration. Injury 51: 2449-2459.

Cossu G, Borello U (1999) Wnt signalling and the activation of myogenesis in mammals. EMBO J 18: 6867-6872.

Domínguez JM, Fuertes A, Orozco L, del Monte-Millán M, Delgado E, Medina M (2012) Evidence for irreversible inhibition of glycogen synthase kinase-3β by tideglusib. J Biol Chem 287: 893-904.

Duan P, Bonewald LF (2016) The role of the Wnt/β-catenin signaling pathway in formation and maintenance of bone and teeth. Int J Biochem Cell Biol 77: 23-29.

Dumas A, Moreau MF, Gherardi RK, Basle MF, Chappard D (2009) Bone grafts cultured with bone marrow stromal cells for the repair of critical bone defects: an experimental study in mice. J Biomed Mater Res A 90: 1218-1229.

Durmuş AS, Çeribaşı AO, Can HN (2016) Effects of coral and demineralized bone matrix on bone healing. F U Sağ Bil Vet Derg 30: 131-136.

Efeoğlu C (2003) Experimental study of the application of autogenous platelet-rich plasma (PRP) and tricalcium phosphate in bone defects. Doctoral Dissertation, Ege Univ Inst Health Sci, Izmir. https://tez.yok.gov.tr/UlusalTez
Merkezi/tezDetay.jsp?id=9ZxgRBg5STYrbRsYsWg8AQ&no
=0gTrNRtQzPTOLuxz8Y5J6A

Emery SE, Fuller DA, Stevenson S (1996) Ceramic anterior spinal fusion: Biologic and biomechanical comparison in a canine model. Spine (Phila Pa 1976) 21: 2713-2719.

Ferguson JY, Dudareva M, Riley ND, Stubbs D, Atkins BL, McNally MA (2014) The use of a biodegradable antibiotic-loaded calcium sulfate carrier containing tobramycin for the treatment of chronic osteomyelitis: a series of 195 cases. Bone Joint J 96: 829-836.

Gual-Vaqués P, Polis-Yanes C, Estrugo-Devesa A, Ayuso-Montero R, Marí-Roig A, López-Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere MC, Bouxsein ML, Deng L, Clemens TL, Williams BO (2005) Essential role of beta-catenin in postnatal bone acquisition. J Biol Chem 280: 21162-21168.

İyilikci B (2020) Evaluation of the effects of bone graft mixed with tideglusib on new bone formation. Specialization Thesis, Pamukkale Univ Inst Health Sci, Denizli. https://gcris.pau.edu.tr/handle/11499/47881

Jiang Y, Qin H, Wan H, Yang J, Yu Q, Jiang M, Yu B (2020) Aspirin-loaded strontium-containing α-calcium sulfate hemihydrate/nano-hydroxyapatite composite promotes regeneration of critical bone defects. J Cell Mol Med 24: 13690-13702.

Köse İ (2012) Experimental investigation of the effects of synthetic bone grafts, calcium channel blockers, and β-2 receptor antagonists on the healing of bone defects. Doctoral Dissertation, Dicle Univ Inst Health Sci, Diyarbakır. https://tez.yok.gov.tr/UlusalTezMerkezi/tezDetay.jsp?id=5i52c1o
S7uzwWMSRpuugXg&no=uwl-RuK6yQev3PQGnkiK9Q

Lane JM, Sandhu HS (1987) Current approaches to experimental bone grafting. Orthop Clin North Am 18: 213-225.

Laurencin CT, Freeman JW (2005) Ligament tissue engineering: an evolutionary materials science approach. Biomaterials 26: 7530-7536.

López J (2018) Autogenous teeth used for bone grafting: A systematic review. Med Oral Patol Oral Cir Bucal 23: e112-e119.

Luna LG (1968) Manual of Histologic Staining Methods of the Armed Forces Institute of Pathology. 3rd ed., New York: Blakiston Division, McGraw-Hill.

Martinez A, Gil C, Perez DI (2011) Glycogen synthase kinase 3 inhibitors in the next horizon for Alzheimer’s disease treatment. Int J Alzheimers Dis 2011: 280502.

Morvan F, Boulukos K, Lacroix-Clement P, Roman SR, Suc-Royer I, Vayssiere B, Ammann P, Martin P, Pinho S, Pognonec P, Mollat P, Niehrs C, Baron R, Rawadi G (2006) Deletion of a single allele of the Dkk1 gene leads to an increase in bone formation and bone mass. J Bone Miner Res 21: 934-945.

Nandi SK, Roy S, Mukherjee P, Kundu B, De DK, Basu D (2010) Orthopaedic applications of bone graft and graft substitutes: a review. Indian J Med Res 132: 15-30.

Neves VC, Babb R, Chandrasekaran D, Sharpe PT (2017) Promotion of natural tooth repair by small molecule GSK3 antagonists. Sci Rep 7: 39654.

Özeç İ, Yeler H (2003) Bone morphogenetic proteins and osteoinduction. Cumhuriyet Univ Dent Fac J 6: 1. https://
acikbilim.yok.gov.tr/bitstream/handle/20.500.12812/
601075/yokAcikBilim_396389.pdf?sequence=-1

Özkurt B, Tabak AY (2011) Metallic biomaterials and metallosis. TOTBID Review Journal 10: 83-86.

Papadimitriou K, Karkavelas G, Vouros L, Kessopoulou E, Konstantinidis A (2015) Effects of local application of simvastatin on bone regeneration in femoral bone defects in rabbit. J Craniomaxillofac Surg 43: 232-237.

Santana RB, Trackman PC (2006) Controlled release of fibroblast growth factor 2 stimulates bone healing in an animal model of diabetes mellitus. Int J Oral Maxillofac Implants 21: 711-718.

Szponder T, Mytnik E, Jaegermann Z (2013) Use of calcium sulfate as a biomaterial in the treatment of bone fractures in rabbits – Preliminary studies. Bull Vet Inst Pulawy 57: 119-122.

Şimşek A, Çakmak G, Cila E (2004) Bone grafts and bone graft substitutes. TOTBID Review Journal 3: 3-4. chrome-extension:
//efaidnbmnnnibpcajpcglclefindmkaj/https://dergi.totbid.org.tr/uploads/pdf_75.pdf

Tolosa E, Litvan I, Höglinger GU, Burn D, Lees A, Andrés MV, Gómez-Carrillo B, León T, Del Ser T (2014) A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Mov Disord 29: 470-478.

Yahav A, Kurtzman GM, Katzap M, Dudek D, Baranes D (2020) Bone regeneration: properties and clinical applications of biphasic calcium sulfate. Dent Clin North Am 64: 453-472.

Yang X, Li Y, Huang Q, Yang J, Shen B, Pei F (2012) Evaluation of a biodegradable graft substitute in rabbit bone defect model. Indian J Orthop 46: 266- 273.

Zhao Z, Wang G, Zhang Y, Luo W, Liu S, Zeng Z, Liu Y, Zhou Y, Zhang Y (2020) Induced membrane technique combined with antibiotic-loaded calcium sulfate-calcium phosphate composite as bone graft expander for the treatment of large infected bone defects: preliminary results of 12 cases. Ann Transl Med 8: 1081.

 

Go to article

Authors and Affiliations

K. Yener
1
A. Hayat
1

  1. Department of Surgery, Faculty of Veterinary Medicine, Harran University,Akçakale Street Eyyübiye Campus 255 AN 63200 Eyyübiye/Şanlıurfa, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Foot and mouth disease (FMD) is an acute viral disease in animals. Inactive parapoxvirus ovis (IPPVO) strengthens humoral immunity. This study aimed to determine the effect of IPPVO application together with FMD vaccine on cattle immunity. It included 30 Holstein cattle randomly divided into two groups: one was administered only the FMD vaccine, and the other was administered the FMD vaccine and IPPVO simultaneously. Control blood was collected from all animals at 0 hours. Serum TNF-α, IL-1β, and IL-6 levels were measured in blood samples collected at 4, 8, 12, 16, and 24 hours post-vaccination, while serum IgG and IgM levels were measured in blood samples collected at 4, 8, 12, 16, and 20 days post-vaccination using ELISA kits. While no changes in serum TNF-α, IL-1β, and IL-6 levels were detected in the FMD group, IL-1β levels significantly increased (peaking at four hours) in the FMD + IPPVO group. In the FMD group, while IgG levels increased significantly (peaking at 16 days), IgM levels did not change. In the FMD + IPPVO group, IgG level on day 8 days was higher than the 16 days value. Also, the IgM level increased significantly on day 16. In conclusion, the application of FMD with IPPVO increases the primary immune response (IgM), but it does not effect the long-term immune response (IgM).
Go to article

Bibliography

Abdullah KMA (2016) The effect of immunstimulant drugs used together with fmd vaccine on immunoglobulins in sheep. (Phd thesis). Yüzüncü Yıl Univ., Graduate School of Health Sciences, Dep. of Internal Medicine: 49.

Akdoğan M, Yöntem M (2018) Cytokines. Online Turkish Journal of Health Sciences 3: 36-45.

Anziliero D, Weiblen R, Kreutz L, Spilki F, Flores E (2014) Inactivated Parapoxvirus ovis induces a transient increase in the expression of proinflammatory, Th1-related, and autoregulatory cytokines in mice. Braz J Med Biol Res 47: 110-118.

Arzt J, Juleff N, Zhang Z, Rodriguez L (2011) The pathogenesis of foot‐and‐mouth disease I: viral pathways in cattle. Transbound Emerg Dis 58: 291-304.

Avci O, Bulut O, Dik I (2016) Effects of inactive parapoxvirus ovis on cytokine levels in rats. J Vet Med Sci 78: 129-131.

Bent R, Moll L, Grabbe S, Bros M (2018) Interleukin-1 beta – a friend or foe in malignancies? Int J Mol Sci 19: 2155.

Büttner M, Czerny C-P, Lehner K-H, Wertz K (1995) Interferon induction in peripheral blood mononuclear leukocytes of man and farm animals by poxvirus vector candidates and some poxvirus constructs. Vet Immunol Immunopathol 46: 237-250.

Camcıoğlu Y, Aytaç E (2007) Immunopathogenesis of sepsis. Turkish J. Intensive Care 5: 81-85.

Carnet F, Paillot R, Fortier C, Hue E S, Briot L, de Geoffroy F, Vidalain P-O, Pronost S (2022) Immunostimulating Effect of Inactivated Parapoxvirus Ovis on the Serological Response to Equine Influenza Booster Vaccination. Vaccines 10: 2139.

Diaz-San Segundo F, Medina G N, Stenfeldt C, Arzt J, de Los Santos T (2017) Foot-and-mouth disease vaccines. Vet. Microbiol 206: 102-112.

Doel T (2003) FMD vaccines. Virus Res 91: 81-99.

El-Fadeel M, Mohamed MM, Khafagy HA, Kotb EE, El-Hawary RI (2019) Immunomodulating effect of zylexis on the immune response of cattle vaccinated with (Pneumo-4) vaccine. Int J Vet Sci 8(4): 380-384.

Erbasan S, Mamak N (2023) Investigation of the immunomodulatory effect of inactive parapoxvirus (iPPVO) on infectious bovine rhinotracheitis (IBR) vaccine in cattle. Medit Vet J 8: 183-189.

Friebe A, Siegling A, Friederichs S, Volk H-D, Weber O (2004) Immunomodulatory effects of inactivated parapoxvirus ovis (ORF virus) on human peripheral immune cells: induction of cytokine secretion in monocytes and Th1-like cells. J Virol 78: 9400-9411.

Grubman M J, Baxt B (2004) Foot-and-mouth disease. Clin Microbiol Rev 17: 465-493.

Gruys E, Toussaint M, Niewold T, Koopmans S (2005) Acute phase reaction and acute phase proteins. J Zhejiang Univ Sci 6: 1045-1056.

Guo P, Li R, Piao T H, Wang C L, Wu X L, Cai H Y (2022) Pathological mechanism and targeted drugs of COPD. Int J Chron Obstruct Pulmon Dis: 1565-1575.

Haig D M (1998) Poxvirus interference with the host cytokine response. Vet Immunol 63: 149-156.

Ibrahim M, Warda F F, Nermeen G S, Abdel-Baky M, Kalad M A (2017) Effect of Inactivated Sheep Pox Vaccine and Parapox Ovis Virus as Immunomodulators on Response of Equines to Inactivated Herpes Vaccine. J Egypt Vet Med Assoc 77: 61-72.

Janeway C, Travers P, Walport M, Shlomchik M J (2001) The immune system in health and disease Immunobiology 5. Garland Pub. New York, USA 20.05.2025 https://www.ncbi.nlm.nih.gov/books/NBK27162/

Kart A, Uzlu E, Karapehlivan M, Öğün M, Merhan O (2010) Effect of Immunomodulator Zylexis on Blood Glutathione, Malondialdehyde, Nitric Oxide, Total and Lipid Bound Sialic Acids Levels in Calves. Vet Sci Pract 5: 43-48.

Knight-Jones T J, Rushton J (2013) The economic impacts of foot and mouth disease–What are they, how big are they and where do they occur? Prev Vet Med 112: 161-173.

Koj A (1998) Termination of acute-phase response: role of some cytokines and anti-inflammatory drugs. Gen Pharmacol 31: 9-18.

Megha K, Mohanan P (2021) Role of immunoglobulin and antibodies in disease management. Int J Biol Macromol 169: 28-38.

Nowatzky J, Knorr A, Hirth‐Dietrich C, Siegling A, Volk HD, Limmer A, Knolle P, Weber O (2013) Inactivated Orf virus (Parapoxvirus ovis) elicits antifibrotic activity in models of liver fibrosis. Hepatol Res 43: 535-546.

Ons E, Van Brussel L, Lane S, King V, Cullinane A, Kenna R, Lyons P, Hammond T-A, Salt J, Raue R (2014) Efficacy of a Parapoxvirus ovis-based immunomodulator against equine herpesvirus type 1 and Streptococcus equi equi infections in horses. Vet Microbiol 173: 232-240.

Rashıd B M, Yüksek N (2019) The effects of immunostimulants (zinc, levamisole, vitamin AD3E) use together with enterotoxemia vaccine on immunoglobulins in sheep. Turkish J Vet Res 3: 57-65.

Rziha H-J, Henkel M, Cottone R, Meyer M, Dehio C, Büttner M (1999) Parapoxviruses: potential alternative vectors for directing the immune response in permissive and non-permissive hosts. Journal of biotechnology 73: 235-242.

Schütze N, Raue R, Büttner M, Alber G (2009) Inactivated parapoxvirus ovis activates canine blood phagocytes and T lymphocytes. Vet Microbiol 137: 260-267.

Smith R E, Strieter R M, Phan S H, Lukacs N, Kunkel S L (1998) TNF and IL-6 mediate MIP-1α expression in bleomycin-induced lung injury. J Leukoc Biol 64: 528-536.

Tizard I R (2017) Veterinary Immunology. 10: 480-482. Elsevier Health Sciences 032352348X 20.05.2025 https: //books.google.com.tr/books?hl=tr&lr=&id=JuM2DwAAQBA
J&oi=fnd&pg=PP1&dq=Veterinary+Immunology-E-Book: +Veterinary+Immunology-E-Book+2017&ots=ZIfAECT
tEq&sig=tXc2BcA7zQTEu4c6-q8P-bgm4pk&redir_esc
=y#v=onepage&q=Veterinary%20Immunology-E-Book
%3A%20Veterinary%20Immunology-E-Book%202017
&f=false

Traeder W, Brune A, Rehagen M (2005) Protective effect of the paramunity inducer Parapoxvirus ovis (Zylexis) in kittens experimentally infected with Calicivirus (strain FCV-KS 20). Prakt Tierarzt 86: 230-238.

Ülgenalp O, Koç B T, Oğuzoğlu T Ç (2018) A Review on Parapoxviruses Observed in Ruminants: History, Epidemiology, Molecular Characterization, Pathogenesis, Clinical Findings, Immunotherapy and Use as a Vector in Recombinant Vaccines. Anim Health Prod Hyg 7: 551-557.

Van Reeth K, Nauwynck H, Pensaert M (1998) Bronchoalveolar interferon-α, tumor necrosis factor-α, interleukin-1, and inflammation during acute influenza in pigs: a possible model for humans? J Infect Dis 177: 1076-1079.

Wang R, Wang Y, Liu F, Luo S (2019) Orf virus: A promising new therapeutic agent. Rev Med Virol 29: e2013.

Weber O, Siegling A, Friebe A, Limmer A, Schlapp T, Knolle P, Mercer A, Schaller H, Volk H-D (2003) Inactivated parapoxvirus ovis (Orf virus) has antiviral activity against hepatitis B virus and herpes simplex virus. J Gen Virol 84: 1843-1852.

Wojno E D T, Hunter C A, Stumhofer J S (2019) The immunobiology of the interleukin-12 family: room for discovery. Immunity 50: 851-870.

Wu Y, Zhang Z, Chen L, Sun S (2024) Immunoglobulin G glycosylation and its alterations in aging-related diseases: IgG glycosylation with aging-related diseases. Acta Biochim Biophys Sin 56: 1221.

Yılmaz N, Akgül Y (2014) Immunglobulins and Septicemia. Uludag Univ. J. Vet. Fac 33: 33-42.

Zhuan B, Yu Y, Yang Z, Zhao X, Li P (2017) Mechanisms of oxidative stress effects of the NADPH oxidase-ROS-NF-κB transduction pathway and VPO1 on patients with chronic obstructive pulmonary disease combined with pulmonary hypertension. Eur Rev Med Pharmacol Sci 21: 3459-3464.

 

 

Go to article

Authors and Affiliations

R. Canbar
1
M. Uslu
2

  1. Aksaray University, Faculty of Veterinary, Department of Pharmacology and Toxicology. Aksaray, Turkey
  2. Yozgat Bozok University, Department of Laboratory and Veterinary Health,Sefaatli Vocational College, Yozgat, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The objective of the present study was to investigate the impact of single or combined use of intermittent fasting and probiotics, which have been recommended as an alternative to anti-biotics in recent years due to their proven efficacy, against Campylobacter colonization in mice. For this purpose, mice infected with Campylobacter jejuni were divided into groups and exposed to intermittent fasting and probiotics, alone and together. At the end of the experimental study, ileum and cecum contents were obtained for microbiological analyzes, and stomach and intestinal tissue samples were collected for histopathological analyzes. It was determined that the level of C. jejuni colonization in the ileum and cecum of mice in the positive control group (PC group) was significantly higher than in the other groups (p<0.05). It was also determined that the level of C. jejuni colonization in the ileum of mice in the probiotic and intermittent fasting group (PB +IF group) was lower than in the other groups and the difference was statistically significant (p<0.05). As a result of the histological analyzes, mild inflammatory reaction was observed to occur in the stomach and intestinal tissues of the animals in the experimental groups, and the severity of the inflammation was lower in the PB +IF group than in the other groups. The findings of this study indicate that single or combined use of intermittent fasting and probiotics may represent a safe and feasible strategy for the control of Campylobacter infections.

Go to article

Bibliography

Açik MN, Çetinkaya B (2006) Heterogeneity of Campylobacter jejuni and Campylobacter coli strains from healthy sheep. Vet Microbiol 115: 370-375.

Ali I, Liu K, Long D, Faisal S, Hilal MG, Ali I, Huang X, Long R (2021) Ramadan fasting leads to shifts in human gut microbiota structured by dietary composition. Front Microbiol 12: 642999.

Altman DG, Gore SM, Gardner MJ, Pocock SJ (1983) Statistical guidelines for contributors to medical journals. Br Med J 286: 1489-1493.

Ayres JS, Schneider DS (2009) The role of anorexia in resistance and tolerance to infections in Drosophila. PLoS Biol 7: e1000150.

Bancroft JD (2008) Gamble, M. Theory and Practice of Histological Techniques. 6th ed.; Churchill Livingstone Elsevier: London, UK.

Bhatti SI, Mindikoglu AL (2022) The impact of dawn to sunset fasting on immune system and its clinical significance in COVID-19 pandemic. Metabol Open 13: 100162.

Bialkowska AB, Ghaleb AM, Nandan MO, Yang VW (2016) Improved swiss-rolling technique for intestinal tissue preparation for immunohistochemical and immunofluorescent analyses. J Vis Exp 113: 54161.

Brogi S, Tabanelli R, Puca S, Calderone V (2024) Intermittent Fasting: Myths, Fakes and Truth on this Dietary Regimen Approach. Foods 13: 1960.

Cabral LQ, Ximenez JA, Moreno KG, Fernandes R (2021) Probiotics have minimal effects on appetite-related hormones in overweight or obese individuals: a systematic review of randomized controlled trials. Clin Nutr 40: 1776-1787.

Cavalcante LP, da Rosa Lima T, de Almeida PC, Tolazzi GJ, Ávila ET, Navalta JW, Junior RC, Voltarelli FA (2021) Intermittent fasting compromises the performance of eutrophic rats submitted to resistance training. Nutrition 86: 111187.

Chang C, Miller JF (2006) Campylobacter jejuni colonization of mice with limited enteric flora. Infect Immun 74: 5261-5271.

Chen H, Zhang Y, Pan Y, Wu L, Wang W, Zhang H, Lou H (2024) Antibiotic-induced microbiome depletion promotes intestinal colonization by Campylobacter jejuni in mice. BMC Microbiol 24: 156.

Eaton KA, Danon SJ, Krakowka S, Weisbrode SE (2007) A reproducible scoring system for quantification of histologic lesions of inflammatory disease in mouse gastric epithelium. Comp Med 57: 57-65.

Elortegui Pascual P, Rolands MR, Eldridge AL, Kassis A, Mainardi F, Lê KA, Karagounis LG, Gut P, Varady KA (2023) A Meta‐Analysis comparing the effectiveness of alternate day fasting, the 5: 2 diet, and time‐restricted eating for weight loss. Obesity 31: 9-21.

Giallourou N, Medlock GL, Bolick DT, Medeiros PH, Ledwaba SE, Kolling GL, Tung K, Guerry P, Swann JR, Guerrant RL (2018) A novel mouse model of Campylobacter jejuni enteropathy and diarrhea. PLoS Pathog 14: e1007083.

Godínez VM, Campos RR, Rivera AV, Lara PE, Pacheco YJ, Jarillo LR, Drago SM (2014) Intermittent Fasting Promotes Bacterial Clearance and Intestinal Ig A Production in Salmonella typhimurium‐Infected Mice. Scand J Immunol 79: 315-324.

Grundler F, Mesnage R, Cerrada A, Wilhelmi de Toledo F (2023) Improvements during long-term fasting in patients with long COVID–a case series and literature review. Front Nutr 10: 1195270.

Hannan MA, Rahman MA, Rahman MS, Sohag AA, Dash R, Hossain KS, Farjana M, Uddin MJ (2020) Intermittent fasting, a possible priming tool for host defense against SARS-CoV-2 infection: Crosstalk among calorie restriction, autophagy and immune response. Immunol Lett 226: 38-45.

Hu X, Xia K, Dai M, Han X, Yuan P, Liu J, Liu S, Jia F, Chen J, Jiang F, Yu J (2023) Intermittent fasting modulates the intestinal microbiota and improves obesity and host energy metabolism. NPJ Biofilms Microbiomes 9(1): 19.

Janssen R, Krogfelt KA, Cawthraw SA, Van Pelt W, Wagenaar JA, Owen RJ (2008) Host-pathogen interactions in Campylobacter infections: the host perspective. Clin Microbiol Rev 21: 505-518.

Janssen H, Kahles F, Liu D, Downey J, Koekkoek LL, Roudko V, D’Souza D, McAlpine CS, Halle L, Poller WC, Chan CT (2023) Monocytes re-enter the bone marrow during fasting and alter the host response to infection. Immunity 56: 783-796.

Jaramillo AP, Castells J, Ibrahimli S, Jaramillo L, Andriuoli RR, Moncada D, Revilla JC (2023) Time-restricted feeding and intermittent fasting as preventive therapeutics: a systematic review of the literature. Cureus 15.

Katona P, Katona AJ (2008) The interaction between nutrition and infection. Clin Infect Dis 46: 1582-1588.

Khan MN, Khan SI, Rana MI, Ayyaz A, Khan MY, Imran M (2022) Intermittent fasting positively modulates human gut microbial diversity and ameliorates blood lipid profile. Front Microbiol 13: 922727.

Khattak F, Galgano S, Houdijk J (2022) Bacterial concentration and Campylobacter spp. quantification differ when fresh or ultra-frozen samples are analysed over time using molecular biology and culture-based methods. PLoS One 17: e0274682.

Liu F, Lee SA, Xue J, Riordan SM, Zhang L (2022) Global epidemiology of campylobacteriosis and the impact of COVID-19. Front Cell Infect Microbiol 12: 979055.

Longo VD, Mattson MP (2014) Fasting: molecular mechanisms and clinical applications. Cell Metab 19: 181-192.

Mazziotta C, Tognon M, Martini F, Torreggiani E, Rotondo JC (2023) Probiotics mechanism of action on immune cells and beneficial effects on human health. Cells 12: 184.

Morales SV, Collado SE, Peraita-Costa I, Llopis MA, Soriano JM (2021) Intermittent fasting and the possible benefits in obesity, diabetes, and multiple sclerosis: a systematic review of randomized clinical trials. Nutrients 13: 3179.

Mousavi S, Bereswill S, Heimesaat MM (2020) Novel clinical Campylobacter jejuni infection models based on sensitization of mice to lipooligosaccharide, a major bacterial factor triggering innate immune responses in human campylobacteriosis. Microorganisms 8: 482.

Ogden ID, Dallas JF, MacRae M, Rotariu O, Reay KW, Leitch M, Thomson AP, Sheppard SK, Maiden M, Forbes KJ, Strachan NJ (2009) Campylobacter excreted into the environment by animal sources: prevalence, concentration shed, and host association. Foodborne Pathog Dis 6: 1161-1170.

O’Loughlin JL, Samuelson DR, Braundmeier FA, White BA, Haldorson GJ, Stone JB, Lessmann, JJ, Eucker TP, Konkel, ME (2015) The intestinal microbiota influences Campylobacter jejuni colonization and extraintestinal dissemination in mice. Appl Environ Microbiol 8: 4642-4650.

Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Harmsen, HJ, Faber KN, Hermoso, MA (2019) Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol 10: 277.

Pickard JM, Zeng MY, Caruso R, Núñez G (2017) Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev 279: 70-89.

Popa AD, Niță O, Gherasim A, Enache AI, Caba L, Mihalache L, Arhire LI (2023) A Scoping Review of the Relationship between Intermittent Fasting and the Human Gut Microbiota: Current Knowledge and Future Directions. Nutrients 15: 2095.

Prestinaci F, Pezzotti P, Pantosti A (2015) Antimicrobial resistance: a global multifaceted phenomenon. Pathog Glob Health 109: 309-318.

Shane SM (1992) The significance of Campylobacter jejuni infection in poultry: a review. Avian Pathol 21: 189-213.

Sipponen P, Price AB (2011) The Sydney System for classification of gastritis 20 years ago. J Gastroenterol Hepatol 26: 31-34.

Stahl M, Ries J, Vermeulen J, Yang H, Sham HP, Crowley SM, Badayeva Y, Turvey SE, Gaynor EC, Li X, Vallance BA (2014) A novel mouse model of Campylobacter jejuni gastroenteritis reveals key pro-inflammatory and tissue protective roles for Toll-like receptor signaling during infection. PLoS Pathog 10: e1004264.

Sun ML, Yao W, Wang XY, Gao S, Varady KA, Forslund SK, Zhang M, Shi ZY, Cao F, Zou BJ, Sun MH (2024) Intermittent fasting and health outcomes: an umbrella review of systematic reviews and meta-analyses of randomised controlled trials. EClinicalMedicine 70: 102519.

Tay A, Pringle H, Penning E, Plank LD, Murphy R (2020) PROFAST: a randomized trial assessing the effects of intermittent fasting and Lacticaseibacillus rhamnosus probiotic among people with prediabetes. Nutrients 12: 3530.

Teker HT, Ceylani T, Keskin S, Samgane G, Allahverdi H, Acikgoz E, Gurbanov R (2024) Supplementing probiotics during intermittent fasting proves more effective in restoring ileum and colon tissues in aged rats. J Cell Mol Med 28: e18203.

Tiwari S, Sapkota N, Han Z (2022) Effect of fasting on cancer: a narrative review of scientific evidence. Cancer Sci 113: 3291-3302.

Wing EJ, Young JB (1980) Acute starvation protects mice against Listeria monocytogenes. Infect Immun 28: 771-776.

Zmora N, Suez J, Elinav E (2019) You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol 16: 35-56.

 

 

Go to article

Authors and Affiliations

M.N. Açık
Ü.G. Doğu
A. Geyik
M. Girgin
A. Ulucan
B. Karagülle
B. Çetinkaya
Download PDF Download RIS Download Bibtex

Abstract

This study was conducted to determine the presence of bacterial flora in the preputium samples of healthy rams in the province of Afyonkarahisar and identify the antibiotic sensitivity of the isolates. Preputial swab samples were collected from a total of 50 healthy 2-3-year-old rams in the center, districts, and villages of Afyonkarahisar, including 20 Merino, 10 Hampshire, and 20 Pirlak breed rams. Seventy-eight isolates obtained from the 50 clinically healthy rams were identified using standard microbiological and biochemical methods, as well as the VITEK-2 automated system device. Forty-four isolates were Gram-positive bacteria (56.4%), 29 isolates were Gram-negative bacteria (37.2%), and 5 isolates were yeasts (6.4%). Seventeen genera consisting of Acinetobacter lwoffii, Aerococcus viridans, Aeromonas hydrophila, Bacillus spp., Candida spp., Escherichia coli, Erysipelothrix rhusiopathiae, Kocuria kristinae, Kocuria rosea, Kytococcus sedentarius, Lactococcus lactis, Mannheimia haemolytica, Neisseria animaloris, Salmonella enterica ssp. diarizonae, Sphingobacterium thalpophilum, Sphingomonas paucimobilis, Staphylococcus capitis, Staphylococcus cohnii spp. urealyticus, Staphylococcus epidermidis, Staphylococcus lentus, Staphylococcus xylosus, Streptococcus ovis, and Streptococ­cus thoraltensis were isolated. The most frequently isolated species in the tested animals were Staphylococcus spp. (25.6%), E. coli (21.8%), Streptococcus spp. (7.7%), A. viridans (6.4%), Lactococcus lactis ssp. (6.4%), and Candida spp. (6.4%).

The susceptibility of the isolates to ampicillin, enrofloxacin, erythromycin, florfenicol, genta­micin, tetracycline, ceftiofur, and trimethoprim-sulfamethoxazole was tested using the Kirby- -Bauer disk diffusion method (Bauer et al. 1966). Most isolates were susceptible to ceftiofur, enrofloxacin, gentamicin, and florfenicol, while most were resistant to erythromycin, tetracy­cline, and ampicillin.

Go to article

Bibliography

Abdullah FF, Tijjani A, Adamu L, Abba Y, Mohammed K, Osman AY, Saharee A, Saad MZ, Haron AW (2014) Ulcerative balanitis in a bull. Int J Livest Res 4: 114-116.

Al-Katib WA, Dennis SM (2009) Ovine genital actinobacillosis: a review. N Z Vet J 57: 352-358.

Allen WE, Dagnall GJ (1982) Some observations on the aerobic bacterial flora of the genital tract of the dog and bitch. J Small Anim Pract 23: 325-335.

Aydin F, Abay M, Şahin O, Abay S, Karakaya E, Müştak İB, Müştak HK, Gümüşsoy KS, Kayman T (2020) Species distribution, genetic diversity and antimicrobial susceptibility of Campylobacter isolates recovered from the preputial cavity of healthy rams in Turkey. J App Microbiol 129: 1173-1184.

Bath G, De Wet JMJ (2000) Sheep and goat diseases. 1st ed., Tafelberg, South Africa, pp:1-205.

Bauer AW, Kirby WM, Sherris JC, Turck M (1966) Antibiotic susceptibility testing by a standardized single disk method. Am J Clin Pathol 45: 493-496.

Bjurstrom L, Linde-Forsberg C (1992) Long-term study of aerobic bacteria of the genital tract in stud dogs. Am J Vet Res 53: 670-673.

Boukhliq R, Allali KE, Tibary A (2017) Gross anatomy and ultrasonographic examination of the reproductive organs in rams and bucks. Rev Mar Sci Agron Vet. 6: 226-240.

Brito EH, Fontenelle RO, Brilhante RS, Cordeiro RA, Monteiro AJ, Sidrim JJ, Rocha MF (2009) The anatomical distribution and antimicrobial susceptibility of yeast species isolated from healthy dogs. Vet J 182: 320-326.

Clinical and Laboratory Standards Institute (CLSI) (2015) Performance standards for antimicrobial susceptibility testing. 25th ed., Wayne, PA: CLSI, 2015. CLSI document M100-S25, pp: 1-231.

Daher NN, Ismaeel MA, Noomi BS (2018) Bacterial Contamination of Prepuce in Young and Adult Rams in Salahaddin Province. J Tikrit Univ For Agri Sci 18: 120-123.

Doig PA, Ruhnke HL, Bosu WT (1981) The genital Mycoplasma and Ureaplasma flora of healthy and diseased dogs. Can J Comp Med 45: 233-238.

Foster RA (2016) Male genital system. In: Maxie DG (ed) Jubb, Kennedy, Palmer’s Pathology of Domestic Animals. WB Saunders Co Ltd, pp: 465-510.

Freitas VM, Rabelo RE, Assis BM, Bao SN, Garcia Neto AF, Oliveira LP, Jesus LO, Helfenstein KK, Vulcani VAS (2022) Morphological and morphometric characterization of the preputial ostium, internal preputial leaflet, and free part of the penises of Aberdeen Angus and Nellore bulls. Arq Bras Med Vet Zootec 74: 1-10.

Gangwar C, Kumaresan G, Mishra AK, Kumar A, Pachoori A, Saraswat S, Singh PN, Kharche SD (2020) Molecular detection of important abortion‐causing microorganisms in preputial swab of breeding bucks using PCR‐based assays. Reprod Domest Anim 55: 1520-1525.

Gocmen H, Alçay S, Rosales RS, Ridley A (2020) Characterisation of genital Mycoplasma species from preputial swabs of bucks and rams. Kafkas Univ Vet Fak Derg 26: 305-308

Gouletsou PG, Fthenakis GC (2015) Microbial diseases of the genital system of rams or bucks. Vet Microbiol 181: 130-135.

Gouletsou PG, Fthenakis GC, Tzora A, Cripps PJ, Saratsis P (2006) Isolation of Arcanobacterium pyogenes from the scrotal skin and the prepuce of healthy rams or rams with testicular abnormalities. Small Rumin Res 63: 177-182.

Hernando-Amado S, Coque TM, Baquero F, Martínez JL (2019) Defining and combating antibiotic resistance from One Health and Global Health perspectives. Nat Microbiol 4: 1432-1442.

Holt JG, Krieg NR, Sneath PHA, Staley JT, Williams ST, (2000) Bergey’s manual of determinative bacteriology 9th ed., Williams and Wilkins, Baltimore, USA.

Jarvinen JA, Kinyon JM (2010) Preputial microflora of llamas (Lama glama) and alpacas (Vicugna pacos). Small Rumin Res 90: 156-160.

Koc Y, Alkan F (2001) Clinical evaluations of testes, penis and prepuce diseases in domestic animals. Vet Bil Derg 17: 67-74.

Larone DH (2002) Medically important fungi, A Guide to Identification, 4th ed., ASM Press, Washington DC.

Ling GV, Ruby AL (1978) Aerobic bacterial flora of the prepuce, urethra, and vagina of normal adult dogs. Am J Vet Res 39: 695-698.

McEntee K (1990) Reproductive Pathology of Domestic Mammals. Academic Press, San Diego, pp 359-383.

Rickwood AM (1999) Medical indications for circumcision. BJU int 83: 45-51.

Sarıtas ZK, Konak S, Pamuk K, Korkmaz M, Cevik-Demirkan A, Civelek T (2012) Identification and antimicrobial susceptibility of microorganisms isolated from the preputium of healthy dogs. J Anim Vet Adv 11: 553-555.

Scott PR, Sargison ND, Wilson DJ (2007) The potential for improving welfare standards and productivity in United Kingdom sheep flocks using veterinary flock health plans. Vet J 173: 522-531.

Stewart JL, Shipley CF (2021). Management of reproductive diseases in male small ruminants. Vet Clin North Am Food Anim 37: 105-123.

Stewart JL, Vieson MD, Shipley CF (2018) Corynebacterium pseudotuberculosis as a pathogen of the reproductive tract of male small ruminants: case study and review. Clin Theriogenology 10: 107-117.

Susan EA, Moses MA, Allen DG, Constable PD, Dart A, Davies PR, Quesenberry KE, Reeves PT, Sharma JM (2016) The Merck Veterinary Manual. 11 th ed., Merck Co Inc, Kenilworth, USA, pp: 1372-1402.

Quinn PJ, Markey BK, Leonard FC, Hartigan P, Fanning S, Fitzpatrick ES (2011) Veterinary Microbiology and Microbial Disease, 2nd ed, Blackwell Science Ltd, Wiley-Blackwell, UK.

Walther B, Tedin K, Lübke-Becker A (2017) Multidrug-resistant opportunistic pathogens challenging veterinary infection control. Vet Microbiol 200: 71-78.

 

Go to article

Authors and Affiliations

S. Konak
1
F. Avdatek
2

  1. Afyon Kocatepe University, Bayat Vocational School,Department of Laboratory and Veterinary Health, 03780 Afyonkarahisar, Turkey
  2. Afyon Kocatepe University, Faculty of Veterinary Medicine,Reproduction and Artificial Insemination Department, 03030, Afyonkarahisar, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to assess the effect of lactation number, lactation stage and somatic cell count (SCC) on the presence of pathogenic or opportunistic pathogens in cow milk. A total of 1712 milk samples were collected from the udder quarters of 428 lactating Holstein breed cows for bacteriological examination. Somatic cell count was taken from the controlled bovine records. The cows were divided into four groups according to the lactation number (viz. lactation numbers 1, 2, 3, 4 and above) and into three groups according to the lactation month (viz. 1-4, 5-8, 9 months and above). The statistical analysis was performed by SPSS 27.0 software (SPSS Inc., Chicago, Illinois, USA). Frequencies of microorganisms were calculated by determi-ning their confidence intervals (Wilson Confidence Interval 95%, CI). Various farm pathogens were identified: CNS (Coagulase negative staphylococci), S. aureus, Enterococcus spp., Str. agalactiae, E. coli. It was found that CNS and S. agalactiae increased with somatic cell count, lactation number and lactation stage. E. coli increased at the end of the lactation stage (p≤0.05). Enterococcus spp., count in milk differed significantly between cows in lactations 1 and 4 and older (p≤0.05). Pathogen number also increased with milk fat, but decreased with increased protein content (p≤0.01).

Go to article

Bibliography

Ágredo-Campos ÁS, Fernández-Silva JA, Ramírez-Vásquez NF (2023) Staphylococcus aureus, Escherichia coli, and Klebsiella spp. prevalence in bulk tank milk of Colombian herds and associated milking practices. Vet World 16: 869-881.

Antanaitis R, Juozaitiene V, Jonike V, Baumgartner W, Paulauskas A (2021) Milk Lactose as a Biomarker of Subclinical Mastitis in Dairy Cows. Animals (Basel) 11: 1736.

Bradley AJ, De Vliegher S, Green MJ, Larrosa P, Payne B, van de Leemput ES, Samson O, Valckenier D, Van Werven T, Waldeck HWF, White V, Goby L (2015) An investigation of the dynamics of intramammary infections acquired during the dry period on European dairy farms. J Dairy Sci 98: 6029-6047.

Breen JE, Green MJ, Bradley AJ (2009) Quarter and cow risk factors associated with the occurrence of clinical mastitis in dairy cows in the United Kingdom. J Dairy Sci 92: 2551-2561.

Cheng J Qu W, Barkema HW, Nobrega D B, Gao J, Gang L, De Buck J, Kastelic JP, Sun H, Han B (2019) Antimicrobial resistance profiles of 5 common bovine mastitis pathogens in large Chinese dairy herds. J Dairy Sci 102: 2416-2426.

Cheng WN, Han SG (2020) Bovine mastitis: risk factors, therapeutic strategies, and alternative treatments – A review. Asian-Australas J Anim Sci. 33: 1699-1713.

Cobirka M, Tancin V, Slama P (2020) Epidemiology and Classification of Mastitis. Animals (Basel) 10: 2212.

De Visscher A, Piepers S, Haesebrouck F, De Vliegher S (2016) Intramammary infection with coagulase-negative staphylococci at parturition: Species-specific prevalence, risk factors, and effect on udder health. J Dairy Sci 99: 6457-6469.

Duse A, Persson-Waller K, Pedersen K (2021) Microbial aetiology, antibiotic susceptibility and pathogen-specific risk factors for udder pathogens from clinical mastitis in dairy cows. Animals (Basel) 11: 2113.

Elmaghraby MM, El-Nahas AF, Fathala MM, Sahwan F, Tag EL-Dien MA (2017) Incidence of Clinical Mastitis and its Influence on Reproductive Performance of Dairy Cows. Alex J Vet Sci 54: 84-91.

Goli M, Ezzatpanah H, Ghavami M, Chamani M, Doosti A (2012) Prevalence assessment of Staphylococcus aureus and Streptococcus agalactiae by multiplex polymerase chain reaction (M-PCR) in bovine sub-clinical mastitis and their effect on somatic cell count (SCC) in Iranian dairy cows. Afr J Microbiol Res 6: 3005-10.

Hagi T, Kobayashi M, Nomura M (2010) Molecular-based analysis of changes in indigenous milk microflora during the grazing period. Biosci Biotechnol Biochem 74: 484-487.

Hussain R, Khan A, Javed MT, Rizvi F (2012) Possible risk factors associated with mastitis in indigenous cattle in Punjab, Pakistan. Pak Vet J 32: 605-608.

Jagielski T, Krukowski H, Bochniarz M, Piech T, Roeske K, Bakuła Z, Wlazło Ł, Woch P (2019) Prevalence of Prototheca spp. on dairy farms in Poland – a cross-country study. Microb Biotechnol 12: 556-566.

Kaczorek-Łukowska E, Małaczewska J, Wójcik R, Duk K, Blank A, Siwicki AK (2021) Streptococci as the new dominant aetiological factors of mastitis in dairy cows in north-eastern Poland: analysis of the results obtained in 2013-2019. Ir Vet J 74: 2.

Kayano M, Itoh M, Kusaba N, Hayashiguchi O, Kida K, Tanaka Y, Kawamoto K, Gröhn YT (2018) Associations of the first occurrence of pathogen-specific clinical mastitis with milk yield and milk composition in dairy cows. J Dairy Res 85: 309-316.

Kester HJ, Sorter DE, Hogan JS (2015) Activity and milk compositional changes following experimentally induced Streptococcus uberis bovine mastitis. J Dairy Sci 98: 999-1004.

Kibebew K (2017) Bovine Mastitis: A Review of Causes and Epidemiological Point of View. J Biol Agric Health 7: 1-14.

Kline KE, Flores S, Joyse F (2018) Factors affecting Somatic Cell Count in milk of dairy cows in Costa Rica. Int J Vet Sci Res 8: 1-8.

Król J, Brodziak A, Litwińczuk Z, Litwińczuk A (2013) Effect of age and stage of lactation on whey protein content in milk of cows of different breeds. Pol J Vet Sci 16: 395-397.

Law of the Republic of Lithuania on Animal Welfare and Protection No XI-2271 (2012) Official Gazette ‘Valstybės žinios’. https://e-seimas.lrs.lt/portal/legalAct/lt/TAD/TAIS.434660

Levison LJ, Miller-Cushon EK, Tucker AL, Bergeron R, Leslie KE, Barkema KW, DeVries TJ (2016) Incidence rate of pathogen-specific clinical mastitis on conventional and organic Canadian dairy farms. J Dairy Sci 99: 1341-1350.

Morales-Ubaldo AL, Rivero-Perez, N, Valladares-Carranza B, Velazquez-Ordoñez, V, Delgadillo-Ruiz L, Zaragoza-Bastida A (2023) Bovine mastitis, a worldwide impact disease: Prevalence, antimicrobial resistance, and viable alternative approaches. Vet Anim Sci 21: 100306.

Olde Riekerink RG, Barkema HW, Kelton DF, Scholl DT (2008) Incidence rate of clinical mastitis on Canadian dairy farms. J Dairy Sci 91: 1366-1377.

Petzer IM, Karzis, J, Donkin EF, Webb EC, Etter EM (2017) Somatic cell count thresholds in composite and quarter milk samples as indicator of bovine intramammary infection status. J Vet Res 84: e1-e10.

Quigley L, O’Sullivan O, Stanton C, Beresford TP, Ross RP, Fitzgerald GF, Cotter PD (2013) The complex microbiota of raw milk. FEMS Microbiol Rev 37: 664-698.

Rifatbegović M, Nicholas RAJ, Mutevelić T, Hadžiomerović M, Maksimović Z (2024) Pathogens Associated with Bovine Mastitis: The Experience of Bosnia and Herzegovina. Vet Sci 11: 63.

Souza FN, Cunha AF, Rosa DL, Brito MA, Guimarães AS, Mendonça LC, Souza GN, Lage AP, Blagitz MG, Libera AM, Heinemann MB, Cerqueira MM (2016) Somatic cell count and mastitis pathogen detection in composite and single or duplicate quarter milk samples. Pesq Vet Bras 36: 811-818.

Sumon SMMR, Parvin MS, Ehsan MA, Islam MT (2020) Dynamics of somatic cell count and intramammary infection in lactating dairy cows. J Adv Vet Anim Res 7: 314-319.

Taponen S, Liski E, Heikkilä AM, Pyörälä S (2017) Factors associated with intramammary infection in dairy cows caused by coagulase-negative staphylococci, Staphylococcus aureus, Streptococcus uberis, Streptococcus dysgalactiae, Corynebacterium bovis, or Escherichia coli. J Dairy Sci 100: 493-503.

Vangroenweghe F, Duchateau L, Burvenich C (2020) Short communication: J-5 Escherichia coli vaccination does not influence severity of an Escherichia coli intramammary challenge in primiparous cows. J Dairy Sci 103: 6692-6697.

Watts LJ (1988) Etiological agents of bovine mastitis. Vet Microbiol 16: 41-66.

Youssif NH, Hafiz NM, Halawa MA, Saad MF (2021) Association of selected risk factors with bovine subclinical mastitis. Acta Vet Bras 15: 153-160.

Zhang Z, Li XP, Yang F, Luo JY, Wang XR, Liu LH, Li HS (2016) Influences of season, parity, lactation, udder area, milk yield, and clinical symptoms on intramammary infection in dairy cows. J Dairy Sci 99: 6484-93.

 

Go to article

Authors and Affiliations

R. Mišeikienė
1
S. Tušas
1
J. Rudejevienė
2
M. Virgailis
3
B. Pilarczyk
4
A. Tomza-Marciniak
4

  1. Institute of Animal Rearing Technologies, Veterinary Academy,Lithuanian University of Health Sciences, Tilžės 18, LT-47181 Kaunas, Lithuania
  2. Dr. L.Kriauceliunas Small Animal Clinic, Veterinary Academy,Lithuanian University of Health Sciences, Tilžės 18, Kaunas, Lithuania
  3. Microbiology and Virology Institute,Lithuanian University of Health Sciences, Tilžės 18, LT-47181 Kaunas, Lithuania
  4. Department of Animal Reproduction Biotechnology and Environmental Hygiene,West Pomeranian University of Technology, Szczecin, Janickiego 29, 71-270 Szczecin, Poland
Download PDF Download RIS Download Bibtex

Abstract

The aim of the study was to determine the changes in the composition and physicochemical properties of cow’s colostrum and the levels of immunoglobulin G in the serum of their calves, depending on the inclusion of dried distiller’s grains with solubles (DDGS) as a substitute for concentrate feeds in the diet during the dry period. Sixty cows were divided into four groups: Group C – traditional TMR feed, Group I – modified feed with the inclusion of DDGS at 10% of dry matter, Group II – DDGS inclusion increased to 15% of dry matter, Group III – DDGS inclusion increased to 20% of dry matter. Colostrum samples were collected from all cows during the first full milking after calving. Blood samples were taken from all cows after calving and from their calves on days 3rd and 30th of life. No significant changes were observed in the basic composition of cows’ colostrum. The levels of amino acids were significantly lower in DDGS groups. There was an increase in a concentrations of IgG, IgM, IgA, lactoferrin, K, Na, and proportion of α-casein and κ-casein in the colostrum of cows receiving DDGS. The total serum protein content was lower in cows receiving DDGS, despite a higher content of IgG. On the 3rd day of life, significantly higher levels of total protein and IgG were found in the calves’ serum. Based on the results optained, it can be concluded that the use of DDGS in a cow’s diet during the dry period as a substitute for concentrated feeds does not reduce the colostrum quality. However, including DDGS at a level of 15% DM in the cows’ diet results in higher IgG content and, consequently, a higher level of passive immunity in their calves.
Go to article

Bibliography

Anderson J L, Schingoethe DJ, Kalscheur KF, Hippen AR (2006) Evaluation of dried and wet distillers grains included at two concentrations in the diets of lactating dairy cows. J Dairy Sci 89: 3133–3142.

AOAC. (2005) International Official Methods of Analysis, 18th ed., AOAC International: Gaithersburg, MD, USA. https://www.aoac.org/official-methods-of-analysis/

Bardsley CE, Lancaster JD (1960) Determination of reserve sulfur and soluble sulfates in soils. Soil Sci Soc Am J 24: 265-268.

Belyea RL, Rausch KD, Clevenger TE, Singh V, Johnston DB, Tumbleson ME (2010) Sources of variation in composition of DDGS. Anim Feed Sci Technol 159: 122-130.

Benchaar C, Hassanat F, Gervais R, Chouinard PY, Julien C, Petit HV, Masse DI (2013) Effects of increasing amounts of corn dried distillers grains with solubles in dairy cow diets on methane production, ruminal fermentation, digestion, N balance, and milk production. J. Dairy Sci 96: 2413-2427.

Britannica T, Editors of Encyclopaedia (2016). Kjeldahl Method. Encyclopedia Britannica. Available online: https://www.
britannica.com/science/Kjeldahl-method
.

Chesini RG, Takiya CS, Dias MS, Silva TB, Nunes AT, Grigoletto NT, da Silva GG, Vittorazzi PCJr, Rennó LN, Rennó FP (2023) Dietary Replacement of Soybean Meal with Heat-Treated Soybean Meal or High-Protein Corn Distillers Grains on Nutrient Digestibility and Milk Composition in Mid-Lactation. Cows. J Dairy Sci 106: 233-244.

Chibisa GE, Christensen DA, Mutsvangwa T (2012) Effects of replacing canola meal as the major protein source with wheat dried distillers grains with solubles on ruminal function, microbial protein synthesis, omasal flow, and milk production in cows. J Dairy Sci 95: 824-841.

Christen KA, Schingoethe DJ, Kalscheur KF, Hippen AR, Karges KK, Gibson ML (2010) Response of lactating dairy cows to high protein distillers grain or 3 other proteins supplements. J Dairy Sci 93: 2095-2104.

Clark KL, Park K, Lee C (2024) Exploring the cause of reduced production responses to feeding corn dried distillers’ grains in lactating dairy cows. J Dairy Sci 107: 6717-6731.

Contarini G, Povolo M, Pelizzola V, Monti L, Bruni A, Passolungo L, Abeni F, Degano L (2014) Bovine colostrum: changes in lipid constituents in the first 5 days after parturition. J Dairy Sci 97: 5065-5072.

Corassa A, da Silva LL, Souza C, Rodrigues LJ, Lemes GA, Kiefer C, Sbardella M, de Araújo CV, Rothmund VL, de Oliveira AS (2022) Dried distillers grains with solubles in diets during late gestation affects behaviour and maintains the performance of sows. R Bras Zootec 51: e20210172.

Costa A, Goi A, Penasa M, Nardino G, Posenato L, De Marchi M (2021) Variation of immunoglobulins G, A, and M and bovine serum albumin concentration in Holstein cow colostrum. Animal 15: 100299.

Dewettinck K, Rombaut R, Thienpont N, Le TT, Messens K, van Camp J (2008) Nutritional and technological aspects of milk fat globule membrane material. Int Dairy J 18: 436-457.

Dias MR, Moraes KA, Oliveira AS, Batista ED, Salomão AM, Zambenedetti A, Petrenko NB, Sousa JN, Ortelam JC, Ickert A, Chaves CS, Moraes EH (2024) Nutritional Performance of Grazing Beef Cattle Supplemented with High-Protein Distillers’ Dried Grain. Animals (Basel) 14: 1209.

Fatehi F, Parnian-khajehdizaj F, Tar M, Salem AZ (2022) Partial replacement of canola meal with dried dairy waste as a protein source in the diet of dairy cow: effects on lactation performance, ruminal measurements, nutrient digestibility, and nitrogen balance. Anim Feed Sci Tech 294: 115511.

Fick KR, McDowell LR, Miles PH, Wilkinson NS, Funk JD, Conrad JH (1979) Methods of mineral analysis for plant and animal tissues, 2nd ed., Department of Animal Science, University of Florida, Gainesville, p 74.

Fonseca NV, da Cardoso AS, Hoffmann A, Leite RG, Ferrari AC, Fernandes MH, Reis RA (2021) Characterization and Effects of DDG on the Intake and Digestibility of Finishing Bulls in Feedlots. Acta Sci 43: 1807-8672.

Gaillard C, Sørensen M, Vestergaard M, Weisbjerg M, Basar A, Larsen M, Martinussen H, Kidmose U, Sehested J (2017) Effect of substituting soybean meal and canola cake with dried distillers grains with solubles at 2 dietary crude protein levels on feed intake, milk production, and milk quality in dairy cows. J Dairy Sci 100: 8928-8938.

Garnsworthy PC, Marsden M, Goodman JR, Saunders N (2021) Inclusion of Wheat Dried Distillers’ Grains with Solubles from Bioethanol Plants in Diets for Dairy Cows. Animals (Basel) 11: 70.

Godden SM, Lombard JE, Woolums AR (2019) Colostrum management for dairy calves. Vet Clin North Am Food Anim Pract 35: 535-556.

Goi A, De Marchi M, Costa A (2023) Minerals and essential amino acids of bovine colostrum: phenotypic variability and predictive ability of mid- and near-infrared spectroscopy. J Dairy Sci 106: 8341–8356.

Hammon HM, Steinhoff-Wagner J, Schönhusen U, Metges CC, Blum JW (2012) Energy metabolism in the newborn farm animal with emphasis on the calf: endocrine changes and responses to milk-born and systemic hormones. Domest Anim Endocrinol 43: 171–185.

Holst DO (1973) Holst filtration apparatus for Van Soest detergent fiber analysis. J Assoc Off Agric Chem 56: 1352-1356.

Horst RL, Goff JP (1997) Milk fever and dietary potassium. In Proceedings of the Cornell Nutrition Conference for Feed Manufacturers; Cornell University: Ithaca, NY, USA, pp 181-189.

INRA (2007) Alimentation Minérale et Vitaminique des Ruminants: Actualisation des Connaissances. 16th ed. INRA. Paris, France. https://productions-animales.org/article/view/3444.

Kennedy VC, Gaspers JJ, Mordhorst BR, Stokka GL, Swanson KC, Bauer ML, Vonnahme KA (2019) Late gestation supplementation of corn dried distiller’s grains plus solubles to beef cows fed a low-quality forage: III. effects on mammary gland blood flow, colostrum and milk production, and calf body weights. J Anim Sci 97: 3337-3347.

Kume S, Tanabe S (1993) Effect of Parity on Colostral Mineral Concentrations of Holstein Cows and Value of Colostrum as a Mineral Source for Newborn Calves. J Dairy Sci 76: 1654-1660.

Li W, Chang Q, Liu G, Shan A, Li F (2014) Effect of distillers dried grains with solubles diet supplememted with rumenprotected lysine and methionine on milk production and milk amino acids in the lactating cow. Int J Dairy Tech 67: 47-54.

Liu C, Schingoethe DJ, Stegeman GA (2000) Corn distillers grains versus blend of protein supplements with or without ruminally protected amino acids for lactating cows. J Dairy Sci 83: 2075-2084.

Lopez AJ, Heinrichs AJ (2022) Invited review: the importance of colostrum in the newborn dairy calf. J Dairy Sci 105: 2733-2749.

Maxin G, Ouellet DR, Lapierre H (2013) Effect of substitution of soybean meal by canola meal or distillers grains in dairy rations on amino acid and glucose availability. J Dairy Sci 96: 7806-7817.

Mjoun K, Kalscheur KF, Hippen AR, Schingoethe DJ, Little DE (2010) Lactation performance and amino acid utilization of cows fed increasing amounts of reduced-fat dried distillers grains with solubles. J Dairy Sci 93: 288-303.

Morris DL, Kim SH, Kononoff PJ, Lee C (2018) Continuous 11-week feeding of reduced-fat distillers grains with and without monensin reduces lactation performance of dairy cows. J Dairy Sci 101: 5971-5983.

Murphy JM, Hagey JV, Chigerwe M (2014) Comparison of serum immunoglobulin G half-life in dairy calves fed colostrum, colostrum replacer or administered with intravenous bovine plasma. Vet Immunol Immunopathol 158: 233-237.

Naylor RL, Liska AJ, Burke MB, Falcon WP, Gaskell JC, Rozelle SD, Cassman KG (2007) The ripple effect: Biofuels, food security, and the environment. Environment Science and Policy for Sustainable Development 49: 30-43.

Nowak W, Mikuła R, Zachwieja A, Paczyńska K, Pecka E, Drzazga K, Ślósarz P (2012) The impact of cow nutrition in the dry period on colostrum quality and immune status of calves. Pol J Vet Sci 15: 77-82.

NRC (2001) Nutrients Requirements of Dairy Cattle. 7th rev. ed. National Academy Press, Washington, DC.

Pecka E, Dobrzański Z, Zachwieja A, Szulc T, Czyż K (2012) Studies of composition and major protein level in milk and colostrum of mares. Anim Sci J 83: 162–168.

Pecka-Kiełb E, Tumanowicz J, Zachwieja A, Miśta D, Kupczyński R, Króliczewska B, Kaszuba J, Zigo F, Suchocki T (2023) Changes in Fatty Acid Levels during In Vitro Ruminal Fluid Incubation with Different Proportions of Maize Distillers Dried Grains (DDGS). Agriculture 13: 763.

Pecka-Kiełb E, Zachwieja A, Kaszuba J, Ćwiertniewska M (2014) Suitability of colostrum for the production of probiotics. Przemysł Chemiczny 93: 1565-1569.

Penner GB, Tremblay GF, Dow T, Oba M (2008) Timothy hay with a low dietary cation-anion difference improves calcium homeostasis in periparturient Holstein cows. J Dairy Sci 91: 1959-1968.

Playford RJ, Weiser MJ (2021) Bovine Colostrum: Its Constituents and Uses. Nutrients 13: 265.

PN-81/R-64780 (1981) Determination of content of chlorides soluble in water. Polish Committee for Standardization, Measures and Quality, p 4.

Ranathunga SD, Kalscheur KF, Anderson JL, Herrick KJ (2018) Production of dairy cows fed distillers dried grains with solubles in low- and high-forage diets. J Dairy Sci 101: 10886-10898.

Santos JE, Lean IJ, Golder H, Block E (2019) Meta-analysis of the effects of prepartum dietary cation-anion difference on performance and health of dairy cows. J Dairy Sci 102: 2134-2154.

Silva FG, Silva SR, Pereira AM, Cerqueira JL, Conceição C (2024) A Comprehensive Review of Bovine Colostrum Components and Selected Aspects Regarding Their Impact on Neonatal Calf Physiology. Animals (Basel) 14: 1130.

Thanh LP, Suksombat W (2015) Milk Production and Income over Feed Costs in Dairy Cows Fed Medium-roasted Soybean Meal and Corn Dried Distiller’s Grains with Solubles. Asian-Australas J Anim Sci 28: 519–529.

Westreicher-Kristen E, Kaiser R, Steingass H, Rodehutscord M (2014) Effect of feeding dried distillers’ grains with solubles on milk yield and milk composition of cows in mid-lactation and digestibility in sheep. J Anim Physiol Anim Nutr (Berl) 98: 347-56.

Wilm J, Costa JHC, Neave HW, Weary DM, von Keyserlingk MAG (2018) Technical note: Serum total protein and immunoglobulin G concentrations in neonatal dairy calves over the first 10 days of age. J Dairy Sci 101: 6430-6436.

Xu X, Tian QY, Ma XK, Long SF, Piao XS (2020) The effects of applying ME values for high or low-oil corn distillers dried grains with solubles originating from prediction equations on the reproductive performance, colostrum and blood composition as well as diet digestibility for sows in late gestation. Anim Feed Sci Technol 259: 114317.

Zachwieja A, Bodarski R, Pecka-Kiełb E, Wojtas E, Zielak-Steciwko A, Król B, Tumanowicz J (2022) The influence of dcad value in feed ration on hypocalcaemia during transition period in cows – Review. Pak J Agri Sci 59: 181-186.

 

 

Go to article

Authors and Affiliations

E. Pecka-Kiełb
1
A. Zachwieja
2
K. Humienna
2
B. Króliczewska
1
A.E. Zielak-Steciwko
2
J. Kaszuba
2
M. Adamski
2

  1. Wroclaw University of Environmental and Life Sciences, Department of Biostructure and Animal Physiology, Norwida 31, Wroclaw 50-375, Poland
  2. Wroclaw University of Environmental and Life Sciences,Department of Cattle Breeding and Milk Production, Chełmońskiego 38c, Wroclaw 51-630, Poland
Download PDF Download RIS Download Bibtex

Abstract

Feline mammary gland tumors are a serious health concern, resulting in a significant reduction in the animal’s lifespan and a decrease in the overall quality of life. Malignant tumors often lead to recurrences and metastases. Among endogenous factors that may influence the development or progression of mammary neoplasia, prolactin (PRL) and vascular endothelial growth factor (VEGF) appear to be of crucial importance. This study involved 60 queens with surgically removed mammary gland tumors, which were subsequently stained with hematoxylin and eosin (HE) and immunofluorescence to assess the expression of PRL and VEGF. Variables considered during analyses included the time of ovariohysterectomy, inflammation severity and clinical tumor behavior. The VEGF expression in tumors exhibited an increase in malignant cases, providing evidence of heightened angiogenesis. A lack of differences in the overall expression of PRL receptor was found between tumor types. However, the lower expression of PRL receptor in tumors with increased inflammation may suggest PRL’s immunomodulating functions in feline malignant neoplastic tumors. Interestingly, the absence of positive influence of gonadectomy on tumor behavior highlights the need for further research regarding this form of prevention. High expression of PRL receptor and VEGF only in distant metastases may prompt future research on the proangiogenic function of PRL in feline mammary gland tumors.
Go to article

Bibliography

Badowska-Kozakiewicz AM (2012) Biological role of prolactin. Menopause Rev 4: 305-308.

Bole-Feysot C, Goffin V, Edery M, Binart N, Kelly PA (1998) Prolactin (PRL) and Its Receptor: Actions, Signal Transduction Pathways and Phenotypes Observed in PRL Receptor Knockout Mice. Endocr Rev 19: 225-268.

Carpini JD, Karam AK, Montgomery L (2010) Vascular endothelial growth factor and its relationship to the prognosis and treatment of breast, ovarian, and cervical cancer. Angiogenesis 13: 43-58.

Clapp C, Thebault S, Martínez de la Escalera G (2008) Role of Prolactin and Vasoinhibins in the Regulation of Vascular Function in Mammary Gland. J Mammary Gland Biol Neoplasia 13: 55-67.

Dorn CR, Taylor DO, Schneider R, Hibbard HH, Klauber MR (1968) Survey of animal neoplasms in Alameda and Contra Costa Counties, California. II. Cancer morbidity in dogs and cats from Alameda County. J Natl Cancer Inst 40: 307-318

Estrela-Lima A, Araújo MS, Costa-Neto JM, Teixeira-Carvalho A, Barrouin-Melo SM, Cardoso SV, Martins-Filho OA, Serakides R, Cassali GD (2010) Immunophenotypic features of tumor infiltrating lymphocytes from mammary carcinomas in female dogs associated with prognostic factors and survival rates. BMC Cancer 10: 256.

Fernandez I, Touraine P, Goffin V (2010) Prolactin and Human Tumourogenesis. J Neuroendocrinol 22: 771-777.

Ferreira M, Mesquita M, Quaresma M, André S (2008) Prolactin receptor expression in gynaecomastia and male breast carcinoma. Histopathology 53: 56-61.

Gill S, Peston D, Vonderhaar BK, Shousha S (2001) Expression of prolactin receptors in normal, benign, and malignant breast tissue: an immunohistological study. J Clin Pathol 54: 956-960.

Giménez F, Hecht S, Craig LE, Legendre AM (2010) Early detection, aggressive therapy: optimizing the management of feline mammary masses. J Feline Med Surg 123: 214-224

Ginsburg E, Vonderhaar BK (1995) Prolactin synthesis and secretion by human breast cancer cells. Cancer Res 55: 2591-2595.

Giziński S, Boryczko Z, Katkiewicz M, Bostedt H (2004) Investigations on the peripheral steroid hormone concentration and on the estrogen receptor density and antigen Ki-67 distribution in mammary gland tumors of different classifications in bitches. Tierarztl Prax 32: 214-220.

Hanahan D, Weinberg RA (2011) Hallmarks of Cancer: The Next Generation. Cell 144: 646-674.

Harvey S, Martínez-Moreno CG, Luna M, Arámburo C (2015) Autocrine/paracrine roles of extrapituitary growth hormone and prolactin in health and disease: An overview. Gen Comp Endocrinol 220: 103-111.

Islam MS, Matsumoto M, Hidaka R, Miyoshi N, Yasuda N (2012) Expression of NOS and VEGF in feline mammary tumours and their correlation with angiogenesis. Vet J 192: 338-344.

Jensen HM, Chen I, DeVault MR, Lewis AE (1982) Angiogenesis Induced by “Normal” Human Breast Tissue: A Probable Marker for Precancer. Science 218: 293-295.

Karten HJ (1998) Information Management of Confocal Microscopy Images: Traditional Text-Based Databases and Image Gallery Databases. In: Confocal Microscopy Methods and Protocols. Humana Press, New Jersey, pp 403-420

Kim JH, Chon S-K, Im KS, Kim NH, Sur JH (2013) Correlation of tumor-infiltrating lymphocytes to histopathological features and molecular phenotypes in canine mammary carcinoma: A morphologic and immunohistochemical morphometric study. Can J Vet Res 77: 142-149

Klopfleisch R, von Euler H, Sarli G, Pinho SS, Gärtner F, Gruber AD (2011) Molecular Carcinogenesis of Canine Mammary Tumors: news from an old disease. Vet Pathol 48(1): 98-116.

Köllermann J, Helpap B (2001) Expression of Vascular Endothelial Growth Factor (VEGF) and VEGF Receptor Flk-1 in Benign, Premalignant, and Malignant Prostate Tissue. Am J Clin Pathol 116: 115-121.

Lana SE, Rutterman GR, Wthrow SJ (2007) Tumors of the mammary gland. In: Withrow SJ, Vail DM (eds) Withrow & MacEwen’s small animal clinical oncology. Saunders Elsevier, St. Louis, pp 619-636

Leav I, Merk FB, Lee KF, Loda M, Mandoki M, McNeal JE, Ho SM (1999) Prolactin Receptor Expression in the Developing Human Prostate and in Hyperplastic, Dysplastic, and Neoplastic Lesions. Am J Pathol 154: 863-870.

Levina VV, Nolen B, Su Y, Godwin AK, Fishman D, Liu J, Mor G, Maxwell LG, Herberman RB, Szczepanski MJ, Szajnik ME, Gorelik E, Lokshin AE (2009) Biological Significance of Prolactin in Gynecologic Cancers. Cancer Res 69: 5226-5233.

Li Y, Clevenger CV, Minkovsky N, Kumar KGS, Raghunath PN, Tomaszewski JE, Spiegelman VS, Fuchs SY (2006) Stabilization of prolactin receptor in breast cancer cells. Oncogene 25: 1896-1902.

Marano RJ, Ben-Jonathan N (2014) Minireview: Extrapituitary Prolactin: An Update on the Distribution, Regulation, and Functions. Mol Endocrinol 28: 622-633.

Meuten DJ (2017) Tumors in Domestic Animals. 5th ed., Wiley, Hoboken.

Michel E, Feldmann SK, Kowalewski MP, Bley CR, Boos A, Guscetti F, Reichler IM (2012) Expression of prolactin receptors in normal canine mammary tissue, canine mammary adenomas and mammary adenocarcinomas. BMC Vet Res 8: 72.

Michel E, Rohrer Bley C, Kowalewski MP, Feldmann SK, Reichler IM (2014) Prolactin – to be reconsidered in canine mammary tumourigenesis? Vet Comp Oncol 12: 93-105.

Millanta F, Lazzeri G, Vannozzi I, Viacava P, Poli A (2002) Correlation of Vascular Endothelial Growth Factor Expression to Overall Survival in Feline Invasive Mammary Carcinomas. Vet Pathol 39: 690-696

Mills SW, Musil KM, Davies JL, Hendrick S, Duncan C, Jackson ML, Kidney B, Philibert H, Wobeser BK, Simko E (2015) Prognostic Value of Histologic Grading for Feline Mammary Carcinoma: a retrospective survival analysis. Vet Pathol 52: 238-249

Misdorp W, Romijin A, Hart A (1991) Feline mammary tumors: a case-control study of hormonal factors. Anticancer Res 11: 1793-1797

Nieto A, Peña L, Pérez-Alenza MD, Sánchez MA, Flores JM, Castaño M (2000) Immunohistologic Detection of Estrogen Receptor Alpha in Canine Mammary Tumors: Clinical and Pathologic Associations and Prognostic Significance. Vet Pathol 37: 239-247

Overley B, Shofer FS, Goldschmidt MH (2005) Association between Ovarihysterectomy and Feline Mammary Carcinoma. J Vet Intern Med 19: 560-563.

Peirce S, Chen WY (2001) Quantification of prolactin receptor mRNA in multiple human tissues and cancer cell lines by real time RT-PCR. J Endocrinol 171: R1-R4.

Philips N, McFadden K (2004) Inhibition of transforming growth factor-beta and matrix metalloproteinases by estrogen and prolactin in breast cancer cells. Cancer Lett 206: 63-68.

Plotnikov A, Varghese B, Tran TH, Liu C, Rui H, Fuchs SY (2009) Impaired Turnover of Prolactin Receptor Contributes to Transformation of Human Breast Cells. Cancer Res 69: 3165-3172.

Queiroga FL, Pérez‐Alenza MD, González Gil A, Silvan G, Peña L, Illera JC (2014) Clinical and prognostic implications of serum and tissue prolactin levels in canine mammary tumours. Vet Rec 175: 403-403.

Reynolds C, Montone KT, Powell CM, Tomaszewski JE, Clevenger CV (1997) Expression of Prolactin and Its Receptor in Human Breast Carcinoma. Endocrinology 138: 5555-5560.

Rutteman GR, Bevers MM, Misdorp W, Van den Brom WE (1989) Anterior pituitary function in female dogs with mammary tumors: II. Prolactin. Anticancer Res 9: 241-245

Skałba W, Lemm M, Witek A (2016) The role of pituitary and extrapituitary prolactin in reproduction and oncology. Ann Acad Med Siles 70: 46-50.

Sorenmo K (2003) Canine mammary gland tumors. Vet Clin North Am Small Anim Pract 33: 573-596.

Spoerri M, Guscetti F, Hartnack S, Boos A, Oei C, Balogh O, Nowaczyk RM, Michel E, Reichler IM, Kowalewski MP (2015) Endocrine control of canine mammary neoplasms: serum reproductive hormone levels and tissue expression of steroid hormone, prolactin and growth hormone receptors. BMC Vet Res 11: 235.

Strum JM (1983) Angiogenic responses elicited from chorioallantoic membrane vessels by neoplastic, preneoplastic, and normal mammary tissues from GR mice. Am J Pathol 111: 282-287

van Garderen E, van der Poel HJ, Swennenhuis JF, Wissink EH, Rutteman GR, Hellmén E, Mol JA, Schalken JA (1999) Expression and Molecular Characterization of the Growth Hormone Receptor in Canine Mammary Tissue and Mammary Tumors. Endocrinology 140: 5907-5914.

Viste JR, Myers SL, Singh B, Simko E (2002) Feline mammary adenocarcinoma: tumor size as a prognostic indicator. Can Vet J 43: 33-37

Zappulli V, Rasotto R, Caliari D, Mainenti M, Peña L, Goldschmidt MH, Kiupel M (2015) Prognostic Evaluation of Feline Mammary Carcinomas: a review of the literature. Vet Pathol 52: 46-60.

 

 

Go to article

Authors and Affiliations

S. Giziński
1
Ł. Zdrojkowski
1
J. Olszewski
2
K. Malin
3
A. Niwińska
1
E. Kautz-Wasilewska
1
A. Rodo
4
A. Jaeckel
5
M. Domino
1

  1. Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
  2. Center of Translational Medicine, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
  3. Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
  4. Department of Pathology and Veterinary Diagnostics, Institute of Veterinary Medicine, Warsaw University of Life Science, Warsaw, Poland 5 Private Veterinary Practice, Leeds, UK
  5. Private Veterinary Practice, Leeds, UK
Download PDF Download RIS Download Bibtex

Abstract

This study aims to estimate the heritability and genetic trend for herd life (HL) and productive life (PL) on an organic dairy cattle farm in the Aegean Region of Turkey. The data from 1093 cows culled from the farm were evaluated. Ancestors were collected by going 6 generations back. Thus, the pedigree file consisted of 4212 animals. Variance components and breeding values were estimated using the MTDFREML package program. The heritability for both HL and PL was found to be equal and 0.16. Standard errors of the heritabilities were 0.06 and 0.08, respectively. Genetic progress per year for HL and PL was 1.60 and 0.60 days, respectively. Estimations for HL and PL were generally consistent with other studies. Heritabilities indicate that breeding for HL and PL is possible; however, response to selection would be slow. Despite the extended length of the selection process, genetic variation could be sufficient for a successful breeding. Indirect selection based on particular traits that have a strong relationship with HL and PL may be beneficial. Our study showed a phenotypic decline in HL and PL, confirming the fluctuation in genetic trends. Therefore, the importance of life traits in the selection index should be emphasised.
Go to article

Bibliography

Alıç D (2007) Investigations on the conformation traits, herd life and milk yield in Holstein cows. PhD thesis, Ankara University, Ankara, Turkey, 96 p. [in Turkish] https://tez.yok.gov.tr/UlusalTezMerkezi/TezGoster?key=wBmNpkQC9Nhi90NLW7E7-ViGVZtfgumgpmIkiplOqPha6LMprr4tfrBxWqCpdAsy

Barański W, Nowicki A, Zduńczyk S (2021) Effect of an integrated veterinary herd health program on fertility performance and incidence of reproductive disorders in five dairy herds. Pol J Vet Sci 24: 433-437.

Barański W, Nowicki A, Zduńczyk S, Polak Z (2023) Effect of repeated low doses of GnRH analogue (buserelin) on fertility performance of dairy cows with anovulation type I. Pol J Vet Sci 26: 223-229.

Boğokaşayan H, Bakır G (2013) Determination of Lifetime Yield Performance of Holstein Cattle Raised in Şanlıurfa Ceylanpınar Farm. Atatürk Üniversitesi Ziraat Fakültesi Dergisi 44: 75-81. [in Turkish] https://www.cabidigitalli
brary.org/doi/full/10.5555/20143110711

Boldman KG, Freeman AE, Harris BL, Kuck AL (1992) Prediction of sire transmitting abilities for herd life from transmitting abilities for linear type traits. J Dairy Sci 75: 552-563.

Boldman KG, Kriese LA, Van Vleck LD, Van Tassel CP, Kachman SD (1995) A manual for use of MTDFREML. A set of programs to obtain estimates of variances and covariances [DRAFT]. Rev. ed. [Washington, D.C.]: U.S. Department of Agriculture, Agricultural Research Service, p 116.

Brickell JS, Wathes DC (2011) A descriptive study of the survival of Holstein-Friesian heifers through to third calving on English dairy farms. J Dairy Sci 94: 1831-1838.

Buenger A, Ducrocq V, Swalve HH (2001) Analysis of survival in dairy cows with supplementary data on type scores and housing systems from a region of northwest Germany. J Dairy Sci 84: 1531-1541.

Cassandro M, Ghiroldi S, Bagnato A, Gallo L, Carnier P, Santus E (1999) Genetic evaluation of longevity in Italian Brown cattle breed. Interbull bulletin 21: 73.

Chirinos Z, Carabaño MJ, Hernández D (2007) Genetic evaluation of length of productive life in the Spanish Holstein-Friesian population. Model validation and genetic parameters estimation. Livest Sci 106: 120-131.

Daliri Z, Hafezian SH, Parvar AS, Rahimi G (2008) Genetic relationships among longevity, milk production and linear type traits in Iranian Holstein Cattle. J Anim Vet Adv 7: 512-515.

Djedović R, Vukašinović N, Stanojević D, Bogdanović V, Ismael H, Janković D, Gligović N, Brka M, Štrbac L (2023) Genetic Parameters for Functional Longevity, Type Traits, and Production in the Serbian Holstein. Animals 13: 534.

Egger-Danner C, Cole JB, Pryce JE, Gengler N, Heringstad B, Bradley A, Stock KF (2015) Invited review: overview of new traits and phenotyping strategies in dairy cattle with a focus on functional traits. Animal 9: 191-207.

Hare E, Norman HD, Wright JR (2006) Survival rates and productive herd life of dairy cattle in the United States. J Dairy Sci 89: 3713-3720.

Hu H, Mu T, Ma Y, Wang X, Ma Y (2021) Analysis of longevity traits in Holstein cattle: A review. Front Genet 12: 695543.

Imbayarwo-Chikosi VE, Ducrocq V, Banga CB, Halimani TE, Van Wyk JB, Maiwashe A, Dzama K. (2016) Impact of conformation traits on functional longevity in South African Holstein cattle. Anim Prod Sci 58: 481-488.

Koç A (2017) A research on herd life of Holstein-Friesian, Red-Holstein and Simmental cows. Adnan Menderes Üniversitesi Ziraat Fakültesi Dergisi. 14(2): 63-68. (in Turkish) https://doi.org/10.25308/aduziraat.321914

Lactanet (2021) Canadian Dairy Network. https://lactanetgen.ca/query/query-individual.php

Martinez GE, Koch RM, Cundiff LV, Gregory KE, Van Vleck LD (2004) Genetic parameters for six measures of length of productive life and three measures of lifetime production by 6 yr after first calving for Hereford cows. J Anim Sci 82: 1912-1918.

M‘hamdi N, Aloulou R, Bouallegue M, Brar SK, Hamouda MB (2010) Study on functional longevity of Tunisian Holstein dairy cattle using a Weibull proportional hazard model. Livest Sci 132: 173-176.

Microsoft Excel (2016) Release 2016. Microsoft Corporation One Microsoft Way Redmond, WA 98052-7329 USA. https://office.microsoft.com/excel

Miglior F, Fleming A, Malchiodi F, Brito LF, Martin P, Baes CF (2017) A 100-Year Review: Identification and genetic selection of economically important traits in dairy cattle. J Dairy Sci 100: 10251-10271.

Minitab (2019) Minitab® 19 Statistical Software. https://www.minitab.com/en-us/

Mirhabibi S, Kashan N, Gharahveysi S (2018) Genetic evaluation of survival traits in the Holstein dairy cows of Iran. Egyp J Vet Sci 49: 71-74.

Nienartowicz-Zdrojewska A, Dymarski I, Sobek Z, Wolc A (2009) Culling reasons as related to lifetime dairy performance in Polish Friesian (Black-and-White) cows on Pawłowice farm in the years 1909-2006. Anim Sci Pap Rep 27: 173-180.

Pritchard T, Coffey M, Mrode R, Wall E (2013) Understanding the genetics of survival in dairy cows. J Dairy Sci 96: 3296-3309.

Sargolzaei M, Iwaisaki H, Colleau JJ (2006) CFC: a tool for monitoring genetic diversity. Proceedings of the 8th World Congress on Genetics Applied to Livestock Production, 13-18 August 2006, Belo Horizonte, Minas Gerais, Brazil, pp 27-28.

Short TH, Lawlor TJ (1992) Genetic parameters of conformation traits, milk yield, and herd life in Holsteins. J Dairy Sci 75: 1987-1998.

Stock KF, Cole J, Pryce J, Gengler N, Bradley A, Andrews L, Egger-Danner C (2012) Survey on the recording and use of functional traits in dairy management and breeding. In: Proceedings ICAR Annual Meeting. Vol. 30. Cork, Ireland.

Vollema AR, Groen AF (1998) A comparison of breeding value predictors for longevity using a linear model and survival analysis. J Dairy Sci 81: 3315-3320.

Yaylak E (2003) Reasons for Culling, Herd Life and Productive Life in Holstein Cows. Akdeniz Üniversitesi Ziraat Fakültesi Dergisi 16: 179-185. [in Turkish] https://www.cabidigitalli
brary.org/doi/full/10.5555/20043055900

Zavadilová L, Štípková M (2012) Genetic correlations between longevity and conformation traits in the Czech Holstein population. Czech J Anim Sci 57: 125-136.

Zavadilová L, Zink V (2013) Genetic relationship of functional longevity with female fertility and milk production traits in Czech Holsteins. Czech J Anim Sci 58: 554-565.

 

Go to article

Authors and Affiliations

S.C. Baycan
1
S. Duru
1

  1. Department of Animal Science, Faculty of Agriculture, Bursa Uludağ University, Gorukle Campus,1st University Street, No:949-1, PBX:16059, Nilufer District, Bursa Province, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The rise of antibiotic resistance is a global health crisis affecting human and veterinary med­icine, highlighted within the One Health framework. Horses, classified as both meat and compan­ion animals, play a crucial role in facilitating resistant bacteria spread to humans. Despite increased awareness and reduced antibiotic use in livestock, data on equine antibiotic use and resistance remain limited. This retrospective study examines antimicrobial resistance in 978 bacterial isolates from horses in Poland over 12 years (2010-2022), utilizing data from the Micro­biological Diagnostic Laboratory at the Warsaw University of Life Sciences. The most common isolates were Streptococcus spp. (45.60%) and Staphylococcus spp. (19.22%). Clinical isolates resistance pattern observed in this study, particularly to gentamicin, tetracyclines, enrofloxacin, third-generation cephalosporins, and chloramphenicol, strongly suggests the overuse and misuse of these antibiotics, a trend that urgently needs to be addressed to preserve their efficacy. The detection of high-resistance Rhodococcus equi isolates resistant to erythromycin, rifampicin, and doxycycline may pose challenges for foal rhodococcosis treatment soon, especially conside-ring the lack of alternative treatment.

The outcomes of this study show the urgent and critical need to collect and analyze local data for improved antimicrobial stewardship. They also emphasize the importance of continuous monitoring of resistance patterns in equine pathogens, as this is crucial for developing effective treatment strategies and staying ahead of potential threats.

Go to article

Bibliography

Adams R, Smith J, Locke S, Phillips E, Erol E, Carter C, Odoi A (2018) An epidemiologic study of antimicrobial resistance of Staphylococcus species isolated from equine samples submitted to a diagnostic laboratory. BMC Vet Res 14: 42.

Albert E, Biksi I, Német Z, Csuka E, Kelemen B, Morvay F, Bakos Z, Bodó G, Tóth B, Collaud A, Rossano A, Perreten V (2019) Outbreaks of a Methicillin-Resistant Staphylococcus aureus Clone ST398-t011 in a Hungarian Equine Clinic: Emergence of Rifampicin and Chloramphenicol Resistance After Treatment with These Antibiotics. Microb Drug Resist 25: 1219-1226.

Alvarez-Narvaez S, Giguere S, Cohen N, Slovis N, Vazquez-Boland JA (2021) Spread of Multidrug-Resistant Rhodococcus equi, United States. Emerg Infect Dis 27: 529-537.

Alvarez-Narvaez S, Berghaus LJ, Morris ER, Willingham-Lane JM, Slovis NM, Giguere S, Cohen ND (2020) A Common Practice of Widespread Antimicrobial Use in Horse Production Promotes Multi-Drug Resistance. Sci Rep 10: 911.

Alvarez-Narvaez S, Huber L, Giguere S, Hart KA, Berghaus RD, Sanchez S, Cohen ND (2021) Epidemiology and Molecular Basis of Multidrug Resistance in Rhodococcus equi. Microbiol Mol Biol Rev 85(2):e00011-21.

Ammazzalorso A, Granese A, De Filippis B (2024) Recent trends and challenges to overcome Pseudomonas aeruginosa infections. Expert Opin Ther Pat 34: 493-509.

Anwaar F, Ijaz M, Rasheed H, Shah SF, Haider SA, Sabir MJ (2023) Evidence and Molecular Characterization of Multidrug Resistant Staphylococcus aureus Isolated From Equines in Pakistan. J Equine Vet Sci 126: 104498.

Arafa AA, Hedia RH, Ata NS, Ibrahim ES (2021) Vancomycin resistant Streptococcus equi subsp. equi isolated from equines suffering from respiratory manifestation in Egypt. Vet World 14: 1808-1814.

Awosile BB, Heider LC, Saab ME, McClure JT (2018) Antimicrobial resistance in bacteria isolated from horses from the Atlantic Provinces, Canada (1994 to 2013). Can Vet J 59: 951-957.

Barba M, Stewart AJ, Passler T, Wooldridge AA, van Santen E, Chamorro MF, Cattley RC, Hathcock T, Hogsette JA, Hu XP (2015) Experimental Transmission of Corynebacterium pseudotuberculosis Biovar equi in Horses by House Flies. J Vet Intern Med 29: 636-643.

Billington EO, Phang SH, Gregson DB, Pitout JD, Ross T, Church DL, Laupland KB, Parkins MD (2014) Incidence, risk factors, and outcomes for Enterococcus spp. blood stream infections: a population-based study. Int J Infect Dis 26: 76-82.

Bollig ER, Granick JL, Webb TL, Ward C, Beaudoin AL (2022) A quarterly Survey of antibiotic prescribing in small animal and equine practices-Minnesota and North Dakota, 2020. Zoonoses Public Health 69: 864-874.

Bookbinder LC, Mani R, Carr EA (2023) Antibiograms of field and hospital acquired equine neonatal bacterial fluid cultures in the Midwestern United States: 149 samples (2007-2018). J Vet Intern Med 37: 1193-1200.

Bordin AI, Huber L, Sanz MG, Cohen ND (2022) Rhodococcus equi foal pneumonia: Update on epidemiology, immunity, treatment and prevention. Equine Vet J 54: 481-494.

Bourely C, Cazeau G, Jarrige N, Haenni M, Gay E, Leblond A (2020) Antimicrobial resistance in bacteria isolated from diseased horses in France. Equine Vet J 52: 112-119.

Bowen IM, Clegg PD (2015) Antimicrobial resistance in the horse - EVJ’s online collection. Equine Vet J 47: 745-746.

Britz E, Spier SJ, Kass PH, Edman JM, Foley JE (2014) The relationship between Corynebacterium pseudotuberculosis biovar equi phenotype with location and extent of lesions in horses. Vet J 200: 282-286.

Chalder RH, Knott T, Rushton JO, Nikolic-Pollard D (2020) Changes in antimicrobial resistance patterns of ocular surface bacteria isolated from horses in the UK: An eight-year surveillance study (2012-2019). Vet Ophthalmol 23: 950-956.

Chipangura JK, Chetty T, Kgoete M, Naidoo V (2017) Prevalence of antimicrobial resistance from bacterial culture and susceptibility records from horse samples in South Africa. Prev Vet Med 148: 37-43.

Cummings KJ, Perkins GA, Khatibzadeh SM, Warnick LD, Aprea VA, Altier C (2016) Antimicrobial resistance trends among Salmonella isolates obtained from horses in the northeastern United States (2001-2013). Am J Vet Res 77: 505-513.

Duchesne R, Castagnet S, Maillard K, Petry S, Cattoir V, Giard JC, Leon A (2019) In vitro antimicrobial susceptibility of equine clinical isolates from France, 2006-2016. J Glob Antimicrob Resist 19: 144-153.

Dziubinski N, Mählmann K, Lübke-Becker A, Lischer C (2020) Retrospective Identification of Bacterial Isolates From Emergency Laparotomy Surgical Site Infections in Horses. J Equine Vet Sci 87: 102927.

EFSA Panel on Animal Health and Welfare (AHAW), Nielsen SS, Bicout DJ, Calistri P, Canali E, Drewe JA, Garin-Bastuji B, Rojas JL, Gortázar C, Herskin M, Michel V, Chueca MA, Padalino B, Pasquali P, Roberts HC, Spoolder H, Ståhl K, Velarde A, Viltrop A, Winckler C, Baldinelli F, Broglia A, Kohnle L, Alvarez J (2022) Assessment of listing and categorisation of animal diseases within the framework of the Animal Health Law (Regulation (EU) No 2016/429): antimicrobial-resistant Staphylococcus aureus in cattle and horses. EFSA J 20: e07312.

Erol E, Locke S, Saied A, Cruz Penn MJ, Smith J, Fortner J, Carter C (2020) Antimicrobial susceptibility patterns of Rhodococcus equi from necropsied foals with rhodococcosis. Vet Microbiol 242: 108568.

Estell KE, Young A, Kozikowski T, Swain EA, Byrne BA, Reilly CM, Kass PH, Aleman M (2016) Pneumonia Caused by Klebsiella spp. in 46 Horses. J Vet Intern Med 30: 314-321.

European Medicines Agency ESoVAC. (2022). Sales of veterinary antimicrobial agents in 31 European countries in 2022 (EMA/299538/2023). https://www.ema.europa.eu/en/
documents/report/sales-veterinary-antimicrobial-agents31-european-countries-2022-trends-2010-2022-thirteenth-esvac-report_en.pdf

Fonseca JD, Mavrides DE, Morgan AL, Na JG, Graham PA, McHugh TD (2020) Antibiotic resistance in bacteria associated with equine respiratory disease in the United Kingdom. Vet Rec 187: 189.

Gehring R, Mochel JP, Schmerold I (2023) Understanding the background and clinical significance of the WHO, WOAH, and EMA classifications of antimicrobials to mitigate antimicrobial resistance. Front Vet Sci 10: 1153048.

Gravey F, Sévin C, Castagnet S, Foucher N, Maillard K, Tapprest J, Léon A, Langlois B, Le Hello S, Petry S (2024) Antimicrobial resistance and genetic diversity of Klebsiella pneumoniae strains from different clinical sources in horses. Front Microbiol 14:1334555.

Groves MD, Crouch B, Coombs GW, Jordan D, Pang S, Barton MD, Giffard P, Abraham S, Trott DJ (2016) Molecular Epidemiology of Methicillin-Resistant Staphylococcus aureus Isolated from Australian Veterinarians. Plos One 11(1): e0146034.

Guérin F, Fines-Guyon M, Meignen P, Delente G, Fondrinier C, Bourdon N, Cattoir V, Léon A (2017) Nationwide molecular epidemiology of methicillin-resistant Staphylococcus aureus responsible for horse infections in France. Bmc Microbiol 17: 104.

Higgins C, Huber L (2023) Rhodococcus Equi: Challenges to Treat Infections and to Mitigate Antimicrobial Resistance. J Equine Vet Sci 127: 104845.

Higgins C, Cohen ND, Slovis N, Boersma M, Gaonkar PP, Golden DR, Huber L (2024) Antimicrobial Residue Accumulation Contributes to Higher Levels of Rhodococcus equi Carrying Resistance Genes in the Environment of Horse-Breeding Farms. Vet Sci 11: 92.

Huber L, Giguere S, Cohen ND, Slovis NM, Hanafi A, Schuckert A, Berghaus L, Greiter M, Hart KA (2019) Prevalence and risk factors associated with emergence of Rhodococcus equi resistance to macrolides and rifampicin in horse-breeding farms in Kentucky, USA. Vet Microbiol 235: 243-247.

Isgren CM, Williams NJ, Fletcher OD, Timofte D, Newton RJ, Maddox TW, Clegg PD, Pinchbeck GL (2022) Antimicrobial resistance in clinical bacterial isolates from horses in the UK. Equine Vet J 54: 390-414.

Jacobsen ABJE, Damborg P, Hopster-Iversen C (2023) Usage of Antimicrobials in Equine Veterinary Practice in Denmark – A Case-Based Survey. J Equine Vet Sci 126: 104267.

Kalinowski M, Jarosz L, Gradzki Z (2020) Assessment of Antimicrobial Susceptibility of Virulent Strains of Rhodococcus equi Isolated From Foals and Soil of Horse Breeding Farms With and Without Endemic Infections. J Equine Vet Sci 91: 103114.

Kim DH, Chung YS, Park YK, Yang SJ, Lim SK, Park YH, Park KT (2016) Antimicrobial resistance and virulence profiles of Enterococcus spp. isolated from horses in korea. Comp Immunol Microb Infect Dis. 48: 6-13.

Knych HK, Magdesian KG (2021) Equine antimicrobial therapy: Current and past issues facing practitioners. J Vet Pharmacol Ther 44: 270-279.

Leon A, Castagnet S, Maillard K, Paillot R, Giard JC (2020) Evolution of In Vitro Antimicrobial Susceptibility of Equine Clinical Isolates in France between 2016 and 2019. Animals (Basel) 10: 812.

Leonard F (2020) Monitoring antimicrobial resistance in bacteria isolated from horses. Vet Rec 187: 186-188.

Lord J, Carter C, Smith J, Locke S, Phillips E, Odoi A (2022) Antimicrobial resistance among Streptococcus equi subspecies zooepidemicus and Rhodococcus equi isolated from equine specimens submitted to a diagnostic laboratory in Kentucky, USA. PeerJ 10: e13682.

Maddox TW, Clegg PD, Williams NJ, Pinchbeck GL (2015) Antimicrobial resistance in bacteria from horses: Epidemiology of antimicrobial resistance. Equine Vet J 47: 756-765.

Malaluang P, Wilen E, Frosth S, Lindahl J, Hansson I, Morrell JM (2022) Vaginal Bacteria in Mares and the Occurrence of Antimicrobial Resistance. Microorganisms 10: 2204.

Malaluang P, Wilén E, Frosth S, Lindahl JF, Hansson I, Morrell JM (2023) Antimicrobial Resistance in Vaginal Bacteria in Inseminated Mares. Pathogens 12: 375.

Malo A, Cluzel C, Labrecque O, Beauchamp G, Lavoie JP, Leclere M (2016) Evolution of in vitro antimicrobial resistance in an equine hospital over 3 decades. Can Vet J 57: 747-751.

Mitchell C, Steward KF, Charbonneau AR, Walsh S, Wilson H, Timoney JF, Wernery U, Joseph M, Craig D, van Maanen K, Hoogkamer-van Gennep A, Leon A, Witkowski L, Rzewuska M, Stefanska I, Zychska M, van Loon G, Cursons R, Patty O, Acke E, Gilkerson JR, El-Hage C, Allen J, Bannai H, Kinoshita Y, Niwa H, Becú T, Pringle J, Guss B, Böse R, Abbott Y, Katz L, Leggett B, Buckley TC, Blum SE, Cruz López F, Fernández Ros A, Marotti Campi MC, Preziuso S, Robinson C, Newton JR, Schofield E, Brooke B, Boursnell M, de Brauwere N, Kirton R, Barton CK, Abudahab K, Taylor B, Yeats CA, Goater R, Aanensen DM, Harris SR, Parkhill J, Holden MT, Waller AS (2021) Globetrotting strangles: the unbridled national and international transmission of Streptococcus equi between horses. Microb Genom 7: mgen000528.

Mustikka MP, Grönthal TS, Pietilä EM (2020) Equine infectious keratitis in Finland: Associated microbial isolates and susceptibility profiles. Vet Ophthalmol 23: 148-159.

Nocera FP, D’Eletto E, Ambrosio M, Fiorito F, Pagnini U, De Martino L (2022) Occurrence and Antimicrobial Susceptibility Profiles of Streptococcus equi subsp. zooepidemicus Strains Isolated from Mares with Fertility Problems. Antibiotics (Basel) 11: 25.

Nwobi OC, Anyanwu MU, Jaja IF, Nwankwo IO, Okolo CC, Nwobi CA, Ezenduka EV, Oguttu JW (2023) Staphylococcus aureus in Horses in Nigeria: Occurrence, Antimicrobial, Methicillin and Heavy Metal Resistance and Virulence Potentials. Antibiotics (Basel) 12: 242.

Oguttu JW, Qekwana DN, Odoi A (2017) An Exploratory Descriptive Study of Antimicrobial Resistance Patterns of Staphylococcus Spp. Isolated from Horses Presented at a Veterinary Teaching Hospital. BMC Vet Res 13: 269.

Prouillac C (2021) Use of Antimicrobials in a French Veterinary Teaching Hospital: A Retrospective Study. Antibiotics (Basel) 10: 1369.

Rakowska A, Cywinska A, Witkowski L (2020) Current Trends in Understanding and Managing Equine Rhodococcosis. Animals (Basel) 10: 1910.

Rakowska A, Marciniak-Karcz A, Bereznowski A, Cywinska A, Zychska M, Witkowski L (2022) Less Typical Courses of Rhodococcus equi Infections in Foals. Vet Sci 9: 605.

Rasheed H, Ijaz M, Muzammil I, Ahmed A, Anwaar F, Javed MU, Ghumman NZ, Raza A (2023) Molecular evidence of β-lactam resistant Staphylococcus aureus in equids with respiratory tract infections: Frequency and resistance modulation strategy. Acta Trop 245: 106967.

Rathbone P, Arango-Sabogal JC, De Mestre AM, Scott CJ (2023) Antimicrobial resistance of endometrial bacterial isolates collected from UK Thoroughbred mares between 2014 and 2020. Vet Rec 192: e2591.

Redpath A, Hallowell GD, Bowen IM (2021) Use of aminoglycoside antibiotics in equine clinical practice; a questionnaire-based study of current use. Vet Med Sci 7: 279-288.

Rhodes DM, Magdesian KG, Byrne BA, Kass PH, Edman J, Spier SJ (2015) Minimum inhibitory concentrations of equine Corynebacterium pseudotuberculosis isolates (1996-2012). J Vet Intern Med 29: 327-332.

Ribeiro MG, de Morais AB, Alves AC, Bolaños CA, de Paula CL, Portilho FV, de Nardi GJr, Lara GH, Martins LD, Moraes LS, Risseti RM, Guerra ST, Bello TS, Siqueira AK, Bertolini AB, Rodrigues CA, Paschoal NR, de Almeida BO, Listoni FJ, Sánchez LF, Paes AC (2022) Klebsiella‑induced infections in domestic species: a case-series study in 697 animals (1997–2019). Braz J of Microbiol 53: 455-464.

Robinson CS, Timofte D, Singer ER, Rimmington L, Rubio-Martínez LM (2016) Prevalence and antimicrobial susceptibility of bacterial isolates from horses with synovial sepsis: A cross-sectional study of 95 cases. Vet J 216: 117-121.

Roudaud M, Allano M, Fairbrother JH, Sauvé F (2020) A retrospective study on methicillin-resistant Staphylococcus spp. isolated from horses admitted to a Canadian veterinary teaching hospital between 2008 and 2018. Can Vet J 61: 1197-1202.

Sanchez LC, Giguere S, Lester GD (2008) Factors associated with survival of neonatal foals with bacteremia and racing performance of surviving Thoroughbreds: 423 cases (1982-2007). J Am Vet Med Assoc 233: 1446-1452.

Schnepf A, Bienert-Zeit A, Ertugrul H, Wagels R, Werner N, Hartmann M, Feige K, Kreienbrock L (2020) Antimicrobial Usage in Horses: The Use of Electronic Data, Data Curation, and First Results. Front Vet Sci 7: 216.

Schoster A, Amsler M, Kirchgaessner C, Saleh L, Schwarzwald C, Schmitt S (2021) Gentamicin plasma concentrations in hospitalized horses and retrospective minimal inhibitory concentrations of gram-negative equine pathogens. J Vet Emerg Crit Care (San Antonio) 31: 323-330.

Slater JD (2015) Antimicrobial resistance, equine practitioners and human health: A true One Health issue or political interference? Equine Vet J 47: 750-752.

Steinman A, Navon-Venezia S (2020) Antimicrobial Resistance in Horses. Animals (Basel) 10: 1161.

Sukmawinata E, Sato W, Uemura R, Kanda T, Kusano K, Kambayashi Y, Sato T, Ishikawa Y, Toya R, Sueyoshi M (2020) Antimicrobial-Resistant Enterococcus faecium and Enterococcus faecalis Isolated From Healthy Thoroughbred Racehorses in Japan. J Equine Vet Sci 94: 103232.

Takai S, Suzuki Y, Sasaki Y, Kakuda T, Ribeiro MG, Makrai L, Witkowski L, Cohen N, Sekizaki T (2023) Short review: Geographical distribution of equine-associated pVAPA plasmids in Rhodococcus equi in the world. Vet Microbiol 287: 109919.

Tallon RE, Whitt B, Bladon BM (2024) Antibiotic usage in 14 equine practices over a 10-year period (2012-2021). Equine Vet J 56: 544-551.

Tirosh-Levy S, Steinman A, Carmeli Y, Klement E, Navon-Venezia S (2015) Prevalence and risk factors for colonization with methicillin resistant Staphylococcus aureus and other Staphylococci species in hospitalized and farm horses in Israel. Prev Vet Med 122: 135-144.

Toombs-Ruane LJ, Riley CB, Kendall AT, Bolwell CF, Benschop J, Rosanowski SM (2015) Antimicrobial Susceptibilities of Aerobic Isolates from Respiratory Samples of Young New Zealand Horses. J Vet Intern Med 29: 1700-1706.

Toombs-Ruane LJ, Riley CB, Kendall AT, Hill KE, Benschop J, Rosanowski SM (2016) Antimicrobial susceptibility of bacteria isolated from neonatal foal samples submitted to a New Zealand veterinary pathology laboratory (2004 to 2013). N Z Vet J 64: 107-111.

Tyrnenopoulou P, Fthenakis GC (2023) Clinical Aspects of Bacterial Distribution and Antibiotic Resistance in the Reproductive System of Equids. Antibiotics (Basel) 12: 664.

Van den Eede A, Hermans K, Van den Abeele A, Floré K, Dewulf J, Vanderhaeghen W, Nemeghaire S, Butaye P, Gasthuys F, Haesebrouck F, Martens A (2013) The nasal vestibulum is the optimal sampling site for MRSA screening in hospitalised horses. Vet J 197: 415-419.

Vazquez-Boland J, Val-Calvo J, Scortti M (2024) Update on emerging multidrug-resistant Rhodococcus equi. Vet Rec 194: e4071.

Vercelli C, Della Ricca M, Re M, Gambino G, Re G (2021) Antibiotic Stewardship for Canine and Feline Acute Urinary Tract Infection: An Observational Study in a Small Animal Hospital in Northwest Italy. Antibiotics (Basel) 10: 562.

Vercruysse EM, Narinx FP, Rives AC, Sauvage AC, Grauwels MF, Monclin SJ (2022) Equine ulcerative keratitis in Belgium: Associated bacterial isolates and in vitro antimicrobial resistance in 200 eyes. Vet Ophthalmol 25: 326-337.

Waqar N, Amin Q, Munir T, Ikram MS, Shahzad N, Mirza A, Ali A, Arshad MI (2019) A cross-sectional study of methicillin-resistant Staphylococcus aureus at the equine-human interface. Trop Anim Health Prod 51: 1927-1933.

Weese JS (2015) Antimicrobial use and antimicrobial resistance in horses. Equine Vet J 47: 747-749.

White A, Hughes JM (2019) Critical Importance of a One Health Approach to Antimicrobial Resistance. Ecohealth 16: 404-409.

WHO (2024) WHO List of Medically Important Antimicrobials: a risk management tool for mitigating antimicrobial resistance due to non-human use. World Health Organization, Licence: CC BY-NC-SA 3.0 IGO. https://cdn.who.int/
media/docs/default-source/gcp/who-mia-list-2024-lv.pdf

Williams NJ, Slovis NM, Browne NS, Troedsson MH, Giguere S, Hernandez JA (2022) Enterococcus durans infection and diarrhea in Thoroughbred foals. J Vet Intern Med 36: 2224-2229.

Willis AT, Magdesian KG, Byrne BA, Edman JM (2019) Enterococcus infections in foals. Vet J 248: 42-47.

Wilson A, Mair T, Williams N, McGowan C, Pinchbeck G (2023) Antimicrobial prescribing and antimicrobial resistance surveillance in equine practice. Equine Vet J 55: 494-505.

Wilson WD, Magdesian KG (2021) Antimicrobial Selection for the Equine Practitioner. Vet Clin North Am Equine Pract 37: 461-494.

Witkowski L, Rzewuska M, Takai S, Kizerwetter-Swida M, Kita J (2016) Molecular epidemiology of Rhodococcus equi in slaughtered swine, cattle and horses in Poland. BMC Microbiol 16: 98.

Witkowski L, Rzewuska M, Cisek AA, Chrobak-Chmiel D, Kizerwetter-Swida M, Czopowicz M, Welz M, Kita J (2015) Prevalence and genetic diversity of Rhodococcus equi in wild boars (Sus scrofa), roe deer (Capreolus capreolus) and red deer (Cervus elaphus) in Poland. BMC Microbiol 15: 110.

Wolny-Koladka K, Lenart-Boron A (2018) Antimicrobial resistance and the presence of extended-spectrum beta-lactamase genes in Escherichia coli isolated from the environment of horse riding centers. Environ Sci Pollut Res Int 25: 21789-21800.

Yuen KY, Gibson JS, Hinrichsen S, Medina-Torres CE, Bertin FR, Stewart AJ (2021) Cumulative antibiogram and multidrug-resistant organisms in a regional equine referral hospital. J Vet Diagn Invest 33: 149-155.

Zuniga MP, Badillo E, Abalos P, Valencia ED, Marin P, Escudero E, Galecio JS (2023) Antimicrobial susceptibility of Rhodococcus equi strains isolated from foals in Chile. World J Microbiol Biotechnol 39: 231.

Zychska M, Witkowski L, Klementowska A, Rzewuska M, Kwiecien E, Stefanska I, Czopowicz M, Szalus-Jordanow O, Mickiewicz M, Moroz A, Bonecka J, Kaba J (2021) Rhodococcus equi-Occurrence in Goats and Clinical Case Report. Pathogens 10: 1141.

 

Go to article

Authors and Affiliations

M. Żychska
1
M. Rzewuska
2
M. Kizerwetter-Świda
2
D. Chrobak-Chmiel
2
I. Stefańska
2
E. Kwiecień
2
L. Witkowski
1

  1. Division of Veterinary Epidemiology and Economics, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776 Warsaw, Poland
  2. Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
Download PDF Download RIS Download Bibtex

Abstract

being the most common. A1 β-casein may release β-casomorphin-7 (BCM-7) during digestion, which is associated with adverse health effects, whereas A2 β-casein does not. This study investigated the A2 allele frequency and its potential influence on milk performance in Holstein cattle in Taiwan. A total of 1,050 cows from five herds were genotyped using the GeneSeek 50K SNP chip. The A2 allele frequency ranged from 0.58 to 0.75 among herds, with an average of 0.66. Hardy-Weinberg equilibrium tests indicated no significant deviation within herds. Pedigree validation confirmed Mendelian inheritance of A1 and A2 alleles. Furthermore, 348 cows with complete lactation records were evaluated for 305-day mature equivalent milk and fat yields across genotypes. Although cows with the A1/A1 genotype showed numerically higher milk and fat yields, one-way ANOVA and Tukey’s HSD tests revealed no statistically significant differences. These results suggest that while A2 allele selection is increasing in Taiwan, β-casein genotype does not significantly influence milk or fat production in the studied population.
Go to article

Bibliography

Ardicli S, Aldevir O, Aksu E, Gumen A (2023) The variation in the beta-casein genotypes and its effect on milk yield and genomic values in Holstein-Friesian cows. Anim Biotechnol 34: 4116-4125.

Ariton A, Neculai-Valeanu AS, Poroșnicu I (2023) Impact of A2 milk on human health and the dairy industry – a review. Sci Pap J Vet Ser 66: 96-99.

Bors A, Bors SI, Floriștean VC (2024) Health-related outcomes and molecular methods for the characterization of A1 and A2 cow’s milk: review and update. Vet Sci 11: 172.

Caroli AM, Chessa S, Erhardt GJ (2009) Invited review: milk protein polymorphisms in cattle: effect on animal breeding and human nutrition. J Dairy Sci 92: 5335-5352.

Cattaneo S, Masotti F, Stuknyte M, De Noni I (2023) Impact of in vitro static digestion method on the release of beta-casomorphin-7 from bovine milk and cheeses with A1 or A2 beta-casein phenotypes. Food Chem 404: 134617.

Chao CH, Yeh YH, Chen YM, Lee KH, Wang SH, Lin TY (2022) Sire pedigree error estimation and sire verification of the Taiwan dairy cattle population by using SNP markers. Pol J Vet Sci 25: 61-65.

Choi Y, Kim N, Song CH, Kim S, Lee DH (2024) The effect of A2 milk on gastrointestinal symptoms in comparison to A1/A2 milk: a single-center, randomized, double-blind, cross-over study. J Cancer Prev 29: 45-53.

Dantas A, Kumar H, Prudencio ES, de Avila LB Junior, Orellana-Palma P, Dosoky NS, Nepovimova E, Kuca K, Cruz-Martins N, Verma R (2023) An approach on detection, quantification, technological properties, and trends market of A2 cow milk. Food Res Int 167: 112690.

European Food Safety Authority (2009) Review of the potential health impact of β-casomorphins and related peptides. EFSA J 7: 231.

Faggion S, Degano L, Carnier P, Bonfatti V (2025) β-casein A2 affects milk renneting properties, cheese yield before and after ripening, and alters the texture of Caciotta cheese produced in field conditions. J Dairy Sci 108: 3199-3213.

Fernández-Rico S, Mondragón ADC, López-Santamarina A, Cardelle-Cobas A, Regal P, Lamas A, Ibarra IS, Cepeda A, Miranda JM (2022) A2 milk: new perspectives for food technology and human health. Foods 11: 2387.

He M, Sun J, Jiang ZQ, Yang YX (2017) Effects of cow’s milk beta-casein variants on symptoms of milk intolerance in Chinese adults: a multicentre, randomised controlled study. Nutr J 16: 72.

Ho S, Woodford K, Kukuljan S, Pal S (2014) Comparative effects of A1 versus A2 beta-casein on gastrointestinal measures: a blinded randomized cross-over pilot study. Eur J Clin Nutr 68: 994-1000.

Hohmann LG, Yin T, Schweizer H, Giambra IJ, König S, Scholz AM (2021) Comparative effects of milk containing A1 versus A2 β-casein on health, growth and β-casomorphin-7 level in plasma of neonatal dairy calves. Animals (Basel) 11: 55.

Illumina (2017) Infinium XT Assay. Document # 1000000016440v01. https://support.illumina.com/content/dam/illumina-support/documents/documentation//chemistry
_documentation/infinium_assays/infinium-xt/infinium-xtht-reference-guide-1000000016440-01.pdf

Jeong H, Park YS, Yoon SS (2024) A2 milk consumption and its health benefits: an update. Food Sci Biotechnol 33: 491-503.

Jiménez-Montenegro L, Alfonso L, Mendizabal JA, Urrutia O (2022) Worldwide research trends on milk containing only A2 β-casein: a bibliometric study. Animals (Basel) 12: 1909.

Kamiński S, Cieślińska A, Kostyra E (2007) Polymorphism of bovine beta-casein and its potential effect on human health. J Appl Genet 48: 189-198.

Kappes R, Schneider V, Schweizer H, Nüske S, Knob DA, Thaler Neto A, Scholz AM (2024) Effect of β-casein A1 or A2 milk on body composition, milk intake, and growth in Holstein, Simmental, and crossbred dairy calves of both sexes. J Dairy Sci 107: 4033-4044.

Miluchová M, Gábor M, Candrák J (2023) The effect of the genotypes of the CSN2 gene on test-day milk yields in the Slovak Holstein cow. Agriculture 13: 154.

Park YW, Haenlein GFW (2021) A2 bovine milk and caprine milk as a means of remedy for milk protein allergy. Dairy 2: 191-201.

Ramakrishnan M, Eaton TK, Sermet OM, Savaiano DA (2020) Milk containing A2 β-casein only, as a single meal, causes fewer symptoms of lactose intolerance than milk containing A1 and A2 β-caseins in subjects with lactose maldigestion and intolerance: a randomized, double-blind, crossover trial. Nutrients 12: 3855.

Ramakrishnan M, Mysore Saiprasad S, Savaiano DA (2024) Prolonged consumption of A2 β-casein milk reduces symptoms compared to A1 and A2 β-casein milk in lactose maldigesters: a two-week adaptation study. Nutrients 16: 1963.

Sebastiani C, Arcangeli C, Ciullo M, Torricelli M, Cinti G, Fisichella S, Biagetti M. (2020) Frequencies evaluation of β-casein gene polymorphisms in dairy cows reared in central Italy. Animals (Basel) 10: 252.

Sheng X, Li Z, Ni J, Yelland G (2019) Effects of conventional milk versus milk containing only A2 β-casein on digestion in Chinese children: a randomized study. J Pediatr Gastroenterol Nutr 69: 375-382.

Smolenski G, Armstrong K, Trivedi M, Clarke A (2025) BCM-7 release from processed dairy products containing measured amounts of beta-casein variants. Int Dairy J 161: 106136.

Summer A, Di Frangia F, Ajmone Marsan P, De Noni I, Malacarne M (2020) Occurrence, biological properties and potential effects on human health of beta-casomorphin 7: Current knowledge and concerns. Crit Rev Food Sci Nutr 60: 3705-3723.

Ul Haq MR, Kapila R, Kapila S (2015) Release of β-casomorphin-7/5 during simulated gastrointestinal digestion of milk β-casein variants from Indian crossbred cattle (Karan Fries). Food Chem 168: 70-79.

Yamada A, Sugimura M, Kuramoto T (2021) Genetic polymorphism of bovine beta-casein gene in Japanese dairy farm herds. Anim Sci J 92: e13644.

Żbik K, Onopiuk A, Górska-Horczyczak E, Wierzbicka A (2024) Trends and opportunities in the dairy industry: A2 milk and processing methods. Appl Sci 14: 6513.

 

Go to article

Authors and Affiliations

J.W. Shiau
1
C.H. Chao
1

  1. Northern Region Branch, Taiwan Livestock Research Institute, Ministry of Agriculture, 207-5, Bi-tou-mian, Wu-hoo village, Si-hoo Township, Miaoli County, Taiwan
Download PDF Download RIS Download Bibtex

Abstract

The objective of this study was to investigate the effect of mannose oligosaccharides (MOS) against cadmium (Cd)-induced hepatic oxidative damage and analyze its underlying antioxidant mechanism. Thirty male Sprague-Dawley (SD) rats were randomly divided into five groups: control group and four others treated with cadmium chloride (CdCl2)(2 mg/kg body weight (b.w.)) and different MOS levels at 0, 100, 300, 500 mg/kg b.w.. The results demonstrated that adminis­tration of MOS at a dose of 500 mg/kg significantly reduced Cd-induced oxidative damage in rat livers. This was evidenced by an increase in body weight gain (BWG) and thymus index. Addi­tionally, liver superoxide dismutase (SOD), catalase (CAT) and total antioxidant capacity (T-AOC) activities were significantly improved compared to the group exposed to Cd alone. Conversely, MOS resulted in significant reductions in the liver index, liver malondialdehyde (MDA), hydrogen peroxide (H2O2), glutathione (GSH), and serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels. Morphological analysis showed that MOS ameliorated Cd-induced histopathology of the rat liver. Notably, Nrf2 gene expression levels increased, while heme oxygenase 1 (HO-1) and quinone oxidoreductase 1 (NQO1) mRNA levels decreased in the MOS group. In conclusion, MOS effectively attenuate Cd-induced oxida­tive damage in rat liver and the Nrf2 signaling pathway is involved in this process. This study provides valuable insights for the implementation of MOS applications in livestock and poultry production.

Go to article

Bibliography

Ahmad S, Yousaf MS, Tahir SK, Rashid MA, Majeed KA, Naseem M, Raza M, Hayat Z, Khalid A, Zaneb H, Rehman H (2023) Effects of co-supplementation of β-galacto-oligosaccharides and methionine on breast meat quality, meat oxidative stability, and selected meat quality genes in broilers. Pak Vet J 43: 428-434.

Ahmed SM, Luo L, Namani A, Wang XJ, Tang XW (2017) Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim Biophys Acta Mol Basis Dis 1863: 585-597.

Al-Suwailem NK, Kamel NN, Abbas AO, Nassar FS, Mohamed HS, Gouda GF, Safaa HM (2024) The impact of dietary Moringa oleifera leaf supplementation on stress markers, immune responses, and productivity in heat-stressed broilers. Int J Vet Sci 13: 980-987.

Attia YA, Al-Khalaifah H, Ibrahim MS, Al-Hamid AE, Al-Harthi MA, El-Naggar A (2017) Blood Hematological and Biochemical Constituents, Antioxidant Enzymes, Immunity and Lymphoid Organs of Broiler Chicks Supplemented with Propolis, Bee Pollen and Mannan Oligosaccharides Continuously or Intermittently. Poult Sci 96: 4182-4192.

Bautista CJ, Arango N, Plata C, Mitre-Aguilar IB, Trujillo J, Ramírez V (2024) Mechanism of cadmium-induced nephrotoxicity. Toxicology 502: 153726.

Bhattacharjee B, Pal PK, Ghosh AK, Mishra S, Chattopadhyay A, Bandyopadhyay D (2019) Aqueous bark extract of Terminalia arjuna protects against cadmium-induced hepatic and cardiac injuries in male Wistar rats through antioxidative mechanisms. Food Chem Toxicol 124: 249-264.

Cheng Y, Chen Y, Chen R, Su Y, Zhang R, He Q, Wang K, Wen C, Zhou Y (2019) Dietary mannan oligosaccharide ameliorates cyclic heat stress-induced damages on intestinal oxidative status and barrier integrity of broilers. Poult Sci 98: 4767-4776.

Cram DL, Blount JD, Young AJ (2015) Oxidative status and social dominance in a wild cooperative breeder. Funct Ecol 29: 229-238.

Dai Y, Zhang H, Zhang J, Yan M (2018) Isoquercetin attenuates oxidative stress and neuronal apoptosis after ischemia/reperfusion injury via Nrf2-mediated inhibition of the NOX4/ROS/NF-κB pathway. Chem Biol Interact 284: 32-40.

Dik B, Parlak TM, Tufan O, Ozgur FB, Er A (2023) Comparative evaluation of single and combined efficacy of dipyridamole, ketotifen and quercetin on cyclosporine induced hepatorenal toxicity. Pol J Vet Sci 26: 549-557.

Gao W, Wang J, Tang G, Su L, Qiao H, Zhang Y, Li L (2021) Establishment of Liver Oxidative Damage Model Induced by Cadmium in Rats. Chinese Journal of Animal Science (in Chinese) 57: 213-216+222. DOI: 10.19556/j.0258-7033.
20210115-02.

Ito F, Sono Y, Ito T (2019) Measurement and Clinical Significance of Lipid Peroxidation as a Biomarker of Oxidative Stress: Oxidative Stress in Diabetes, Atherosclerosis, and Chronic Inflammation. Antioxidants (Basel) 8: 72.

Janero DR (1990) Malondialdehyde and thiobarbituric acid-reactivity as diagnostic indices of lipid peroxidation and peroxidative tissue injury. Free Radic Biol Med 9: 515-540.

Karabekir SC, Gultekın B, Ayan IC, Savas HB, Cuce G, Kalkan S (2024) Protective effect of astaxanthin on histopathologic changes induced by bisphenol a in the liver of rats. Pak Vet J 44: 244-251.

Khatoon A, Khan MZ, Abidin ZU, Saleemi MK, Oguz H, Gul ST, Abbas RZ, Ali A, Bhatti SA (2024) Aflatoxin B1-associated oxidative stress along with toxicopathological and immunological alterations is efficiently counteracted by dietary supplementation of distillery yeast sludge in broilers. Mycotoxin Res 40: 615-629.

Leone F, Ferrante V (2023) Effects of prebiotics and precision biotics on performance, animal welfare and environmental impact. A review. Sci Total Environ 901: 165951.

Levonen AL, Inkala M, Heikura T, Jauhiainen S, Jyrkkänen HK, Kansanen E, Määttä K, Romppanen E, Turunen P, Rutanen J, Ylä-Herttuala S (2007) Nrf2 gene transfer induces antioxidant enzymes and suppresses smooth muscle cell growth in vitro and reduces oxidative stress in rabbit aorta in vivo. Arterioscler Thromb Vasc Biol 27: 741-747.

Lu J, Qi C, Limbu SM, Han F, Yang L, Wang X, Qin J, Chen L (2019) Dietary mannan oligosaccharide (MOS) improves growth performance, antioxidant capacity, non-specific immunity and intestinal histology of juvenile Chinese mitten crabs (Eriocheir sinensis). Aquaculture 510: 337-346.

Lu Z, Feng L, Jiang W, Wu P, Liu Y, Jiang J, Kuang S, Tang L, Li S, Zhong C, Zhou X (2023) Mannan oligosaccharides alleviate oxidative injury in the head kidney and spleen in grass carp (Ctenopharyngodon idella) via the Nrf2 signaling pathway after Aeromonas hydrophila infection. J Anim Sci Biotechnol 14: 58.

Mohammad LM, Kamil AM, Tawfeeq RK, Ahmed SJ (2023) Ameliorating effects of herbal mixture for dexamethasone induced histological changes in mice. Int J Vet Sci 12: 126-131.

Nemmiche S, Chabane-Sari D, Guiraud P (2007) Role of alpha-tocopherol in cadmium-induced oxidative stress in Wistar rat’s blood, liver and brain. Chem Biol Interact 170: 221-230.

AI Olayan EM, Aloufi AS, AIAmri OD, El-Habit OH, Abdel Moneim AE (2020) Protocatechuic acid mitigates cadmium-induced neurotoxicity in rats: role of oxidative stress, inflammation and apoptosis. Sci Total Environ 723: 137969.

Qiao H, Chen Y, Yang K, Wang J, Chou Y, Chen L, Zhang Y, Huang J, Duan E, Su L (2024) Treatment with mannose oligosaccharides reverses the intestinal injury in the acetylsalicylic acid-treated rat model. Pol J Vet Sci 27: 249-259

Rani A, Kumar A, Lal A, Pant M (2014) Cellular mechanisms of cadmium-induced toxicity: a review. Int J Environ Health Res 24: 378-399.

Sabbioni E, Marafante E (1975) Heavy metals in rat liver cadmium binding protein. Environ Physiol Biochem 5: 132-141.

Saha S, Buttari B, Panieri E, Profumo E, Saso L (2020) An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules 25: 5474.

Scott MD (2006) H2O2 injury in beta thalassemic erythrocytes: protective role of catalase and the prooxidant effects of GSH. Free Radic Biol Med 40: 1264-1272.

Souza-Arroyo V, Fabián JJ, Bucio-Ortiz L, Miranda-Labra RU, Gomez-Quiroz LE, Gutiérrez-Ruiz MC (2022) The mechanism of the cadmium-induced toxicity and cellular response in the liver. Toxicology 480: 153339.

Theocharis S, Margeli A, Fasitsas C, Loizidou M, Deliconstantinos G (1991) Acute exposure to cadmium causes time-dependent liver injury in rats. Comp Biochem Physiol C Comp Pharmacol Toxicol 99: 127-130.

Torrecillas S, Makol A, Betancor MB, Montero D, Caballero MJ, Sweetman J, Izquierdo M (2013) Enhanced intestinal epithelial barrier health status on European sea bass (Dicentrarchus labrax) fed mannan oligosaccharides. Fish Shellfish Immunol 34: 1485-1495.

Ullah A, Al-Saeed FA, Abduallah AM, Ahmed AE, Shahzad A, Amjad N, Ali A, Mostafa MS, Hussain R (2023) Calcium Nanoparticles Induce Oxidative Stress in Erythrocytes, Neurotoxicity and Testicular Toxicity in Albino Rats (Rattus norvegicus). Pak Vet J 43: 241-247.

Vemula S, Mylaram J, Yadala R, Alla G, Banothu A, Veera HD (2024) Protective effects of naringenin on 5-Fluorouracil induced pulmonary toxicity via modulation of NF-κB and Nrf2 pathway. Pak Vet J 44: 63-70.

Widjiati W, Hestianah EP, Luqman EM, Taqwa SF, Caesar J, Shabira Z, Mega RL (2024) Effect of heat shock through MDA and 8-OHdG levels of post-thawing goat oocytes. Int J Vet Sci 13: 611-616.

Xue S, Xia B, Zhang B, Li L, Zou Y, Shen Z, Xiang X, Han Y, Chen W (2022) Mannan oligosaccharide (MOS) on growth performance, immunity, inflammatory and antioxidant responses of the common carp (Cyprinus carpio) under ammonia stress. Front Mar Sci 9: 1062597.

Yan LJ, Allen DC (2021) Cadmium-Induced Kidney Injury: Oxidative Damage as a Unifying Mechanism. Biomolecules 11: 1575.

Yuan C, Ren L, Sun R, Yun X, Zang X, Zhang A, Wu M (2023) Mannan oligosaccharides improve the fur quality of raccoon dogs by regulating the gut microbiota. Front Microbiol 14: 1324277.

Zhao Z, Hyun JS, Satsu H, Kakuta S, Shimizu M (2006) Oral exposure to cadmium chloride triggers an acute inflammatory response in the intestines of mice, initiated by the over-expression of tissue macrophage inflammatory protein-2 mRNA. Toxicol lett 164: 144-154.

 

 

Go to article

Authors and Affiliations

E. Duan
1
W. Gao
1
C. Liu
1
J. Wang
1
L. Su
1
H. Qiao
1
P. Wang
1
J. Huang
1
L. Gan
1

  1. College of Biological Engineering, Henan University of Technology, 100 Lianhua Street, Hi-tech Zone, Zhengzhou 450001, Henan, China
Download PDF Download RIS Download Bibtex

Abstract

The aim of this study was to carry out a follow-up evaluation of cryopreserved semen of native breed cattle collected in the National Bank of Biological Material (NBBM). The oldest material, from 2004–2010, was included in the study. A total of 70 ejaculates from 62 bulls (5 Polish Red-and-White, 7 Polish Black-and-White, 11 Whitebacked, 39 Polish Red) were used in the study. After thawing sperm concentration and viability (plasma membrane intactness) were determined using a fluorescence-based instrument – NucleoCounter SP-100. Sperm concentration was higher (p≤0.05) for the semen of Polish Red-and-White and Polish Red bulls compared to the semen of Polish Black-and-White and Whitebacked bulls. For the viability, no significant differences were observed between breeds. The number of viable spermatozoa per straw in the examined semen ranged from 4.09 to 18.29 × 106 . This study has shown large differences in the quality of bull semen frozen between 2004 and 2010, from the collection of the National Bank of Biological Material while its quality parameters allow it to be used for insemination.
Go to article

Bibliography

Bodmer M, Janett F, Hässig M, den Daas N, Reichert P, Thun R (2005) Fertility in heifers and cows after low dose insemination with sex-sorted and non-sorted sperm under field conditions. Theriogenology 64: 1647-1655.

Christensen P, Boelling D, Pedersen KM, Korsgaard IR, Jensen J (2005) Relationship between sperm viability as determined by flow cytometry and nonreturn rate of dairy bulls. J Androl 26: 98-106.

DeJarnette JM, Leach MA, Nebel RL, Marshall CE, McCleary CR, Moreno JF (2011) Effects of sex sorting and sperm dosage on conception rates in Holstein heifers. Is comparable fertility of sex-sorted and conventional semen plausible? J Dairy Sci 94: 3477-3483.

FAO (2012) Cryoconservation of animal genetic resources. FAO Animal Production and Health Guidelines No. 12. Rome. https://www.fao.org/4/i3017e/i3017e00.pdf

Holden SA, Fernandez-Fuertes B, Murphy C, Whelan H, O’Gorman A, Brennan L, Butler ST, Lonergan P, Fair S (2017) Relationship between in vitro sperm functional assessments, seminal plasma composition, and field fertility after AI with either non-sorted or sex-sorted bull semen. Theriogenology 87: 221-228.

Karoui S, Díaz C, Serrano M, Cue R, Celorrio I, Carabaño MJ (2011) Time trends, environmental factors and genetic basis of semen traits collected in Holstein bulls under commercial conditions. Anim Reprod Sci 124: 28-38.

 

Go to article

Authors and Affiliations

P. Gogol
1
J. Warchoł
1

  1. National Bank of Biological Material,National Research Institute of Animal Production, Krakowska 1, 32-083 Balice/Kraków, Poland
Download PDF Download RIS Download Bibtex

Abstract

The genus Teschovirus consists of two species, Teschovirus A and Teschovirus B, with over 19 genotypes. This study sequenced the near-complete genomes of the PTV YC2 strain, previously isolated. Comparative analyses revealed nucleotide and amino acid homologies between PTV-YC2 and other PTV strains ranging from 69.7% to 93.2% and 75.6% to 99.0%, respectively. Genetic divergence analysis of the P1, polyprotein and 2C-3CD genes confirmed that PTV-YC2 belonged to the Teschovirus B species. Phylogenetic analyses showed a close evo­lutionary relationship between YC2 and the HuN42 strain, the prototype of the TV-B3 genotype, leading to PTV-YC2’s categorization as TV-B3. This study is the first to document the prevalence of the TV-B3 genotype in Jiangxi Province, China.

Go to article

Bibliography

Boros Á, Nemes C, Pankovics P, Kapusinszky B, Delwart E, Reuter G (2012) Porcine teschovirus in wild boars in Hungary. Arch Virol 157: 1573-1578.

Burland TG (2000) DNASTAR’s Lasergene sequence analysis software. Methods Mol Biol 132: 71-91.

Cano-Gomez C, Fernandez-Pinero J, Garcia-Casado MA, Zell R, Jimenez-Clavero MA (2017) Characterization of PTV-12, a newly described porcine teschovirus serotype: in vivo infection and cross-protection studies. J Gen Virol 98: 1636-1645.

ICTV (2022) International Committee on Taxonomy of Viruses. https://ictv.global/taxonomy. Accessed 18 June 2024.

Katoh K, Toh H (2010) Parallelization of the MAFFT multiple sequence alignment program. Bioinformatics 26: 1899-1900.

Letunic I, Bork P (2024) Interactive Tree of Life (iTOL) v6: recent updates to the phylogenetic tree display and annotation tool. Nucleic Acids Res 52: W78-W82.

Oba M, Naoi Y, Ito M, Masuda T, Katayama Y, Sakaguchi S, Omatsu T, Furuya T, Yamasato H, Sunaga F, Makino S, Mizutani T, Nagai M (2018) Metagenomic identification and sequence analysis of a Teschovirus A-related virus in porcine feces in Japan, 2014-2016. Infect Genet Evol 66: 210-216.

Sun H, Gao H, Chen M, Lan D, Hua X, Wang C, Yuan C, Yang Z, Cui L (2015) New serotypes of porcine teschovirus identified in Shanghai, China. Arch Virol 160: 831-835.

Tamura K, Stecher G, Peterson D, Filipski A, Kumar S (2013) MEGA6: Molecular Evolutionary Genetics Analysis version 6.0. Mol Biol Evol 30: 2725-2729.

Yang T, Lu Y, Zhang L, Li X, Chang Y (2020a) Novel species of Teschovirus B comprises at least three distinct evolutionary genotypes. Transbound Emerg Dis 67: 1015-1018.

Yang T, Lu Y, Zhang L, Li X (2020b) Identification of novel genotypes belonging to the species Teschovirus A from indigenous pigs in western Jiangxi, China. Arch Virol 165: 993-1001.

Yang T, Li R, Yao Q, Zhou X, Liao H, Ge M, Yu X (2018a) Prevalence of Porcine teschovirus genotypes in Hunan, China: identification of novel viral species and genotypes. J Gen Virol 99: 1261-1267.

Yang T, Yu X, Luo B, Yan M, Li R, Qu T, Ren X (2018b) Epidemiology and molecular characterization of Porcine teschovirus in Hunan, China. Transbound Emerg Dis 65: 480-490.

Yang T, Lin A, Cui S, Chen J, Wan H, Lu Y, Li P, Zhang Z, Li K, Guo M (2023) Identification of a Novel Porcine Teschovirus Subtype 19 within the Species Teschovirus A. Transbound Emerg Dis doi: 10.1155/2023/9977581.

Zell R, Dauber M, Krumbholz A, Henke A, Birch-Hirschfeld E, Stelzner A, Prager D, Wurm R (2001) Porcine teschoviruses comprise at least eleven distinct serotypes: molecular and evolutionary aspects. J Virol 75: 1620-1631.

Zell R, Delwart E, Gorbalenya AE, Hovi T, King AMQ, Knowles NJ, Lindberg AM, Pallansch MA, Palmenberg AC, Reuter G, Simmonds P, Skern T, Stanway G, Yamashita T, Ictv RC (2017) ICTV Virus Taxonomy Profile: Picornaviridae. J Gen Virol 98: 2421-2422.

 

 

 

Go to article

Authors and Affiliations

W. Liu
1
D. Zhao
2
Y. Peng
3
Z. Zhou
4
X. Tang
5
T. Yang
2

  1. Changde Center for Animal Disease Prevention and Control, Changde, Hunan, China
  2. Laboratory of Animal Pathogenic Microbiology, Yichun University, Yichun, Jiangxi, China
  3. Longshan Center for Animal Disease Prevention and Control, Xiangxi, Hunan, China
  4. Longhui Center for Animal Disease Prevention and Control, Shaoyang, Hunan, China
  5. Hunan Center for Animal Disease Prevention and Control, Changsha, Hunan, China
Download PDF Download RIS Download Bibtex

Abstract

Metabolic diseases in cows have a significant influence on their health status, milk production, and the health and immune parameters of newborn calves. One of the most critical periods in the lactation cycle of dairy cows is the transition period, which covers the last three weeks before and the first three weeks after calving. It is characterized by the occurrence of metabolic and endocrine adaptations resulting from the increased demand for nutrients to support future lactation and avoid metabolic dysfunction. Metabolic stress is a hypermetabolic and catabolic response to homeostatic imbalance which affects the development and maturation of foetus. Lipid mobilization is a typical feature of a negative energy balance (NEB), whereby non-esterified fatty acids (NEFA) are released into the bloodstream and used as a source of energy. The aim of this review was to present results of our research and research of other authors on the impact of metabolic disorders in dairy cows on the immune and health status of calves.
Go to article

Bibliography

Abuelo A (2020) Symposium review: Late-gestation maternal factors affecting the health and development of dairy calves. J Dairy Sci 103: 3882-3893.

Alharthi AS, Batistel F, Abdelmegeid MK, Lascano G, Parys C, Helmbrecht A, Trevisi E, Loor JJ (2018) Maternal supply of methionine during late-pregnancy enhances rate of Holstein calf development in utero and postnatal growth to a greater extent than colostrum source. J Anim Sci Biotechnol 9: 83.

Alharthi AS, Coleman DN, Alhidary IA, Abdelrahman MM, Trevisi E, Loor JJ (2021) Maternal body condition during late-pregnancy is associated with in utero development and neonatal growth of Holstein calves. J Anim Sci Biotechnol 12: 44.

Alharthi AS, Lopreiato V, Dai H, Bucktrout R, Abdelmegeid M, Batistel F, Parys C, Shen X, Ballou MA, Trevisi E, Alhidary IA, Abdelrahman MM, Loor JJ (2019) Short communication: Supply of methionine during late pregnancy enhances whole-blood innate immune response of Holstein calves partly through changes in mRNA abundance in polymorphonuclear leukocytes. J Dairy Sci 102: 10599-10605.

Alomari M, Dec M, Puchalski A, Stęgierska D, Urban-Chmiel R (2016) Screening of probiotic lactobacillus strains to reduce shiga-toxigenic e. coli in cattle. Proceedings of XV Congress PTNW, 22-24 Sept 2016r, Lublin, pp. 257

Arthur JR, McKenzie RC, Beckett GJ (2003) Selenium in the immune system. J Nutr 133 (5 Suppl 1):1457S-9S.

Avery JC, Hoffmann PR ( 2018) Selenium, selenoproteins, and immunity. Nutrients 10: 1203.

Batistel F, Alharthi AS, Wang L, Parys C, Pan YX, Cardoso FC, Loor JJ (2017) Placentome Nutrient Transporters and Mammalian Target of Rapamycin Signaling Proteins Are Altered by the Methionine Supply during Late Gestation in Dairy Cows and Are Associated with Newborn Birth Weight. J Nutr 147: 1640-1647,

Berge AC, Vertenten G (2018) The importance of preventive health and vaccination programmes in ruminant production. 017. [Accessed in: July 7th. 2019]. Available from https://www.timetovaccinate.com/

Berry DP, Lee JM, Macdonald KA, Roche JR (2007) Body Condition Score and Body Weight Effects on Dystocia and Stillbirths and Consequent Effects on Postcalving Performance. J Dairy Sci 90: 4201-4211.

Cao YZ, Maddox JF, Mastro AM, Scholz RW, Hildenbrandt G, Reddy CC (1992) Selenium deficiency alters the lipoxygenase pathway and mitogenic response in bovine lymphocytes. J Nutr 122: 2121-2127

Carlson BA, Yoo MH, Shrimali RK, Irons R., Gladyshev VN, Hatfield DL, Park JM (2010) Role of selenium-containing proteins in T-cell and macrophage function. Proc Nutr Soc 69: 300-310

Carvalho MR, Aboujaoude C, Peñagaricano F, Santos JEP, DeVries TJ, McBride BW, Ribeiro E S (2020) Associations between maternal characteristics and health, survival, and performance of dairy heifers from birth through first lactation. J Dairy Sci 103: 823–839.

Chigerwe M, Tyler JW, Summers MK, Middleton JR, Schultz LG, Nagy DW (2009) Evaluation of factors affecting serum IgG concentrations in bottle-fed calves. J Am Vet Med Assoc 234: 785-789.

Çolakoğlu HE, Yazlık MO, Çolakoğlu EC, Kaya U, Bayramoğlu R, Kurt S, Vural R, Küplülü S (2021) Oxidative stress in cows according to calving season: passive calf immunity and its relationship with colostrum quality. Pol J Vet Sci 24: 225-233.

Corcionivoschi N, Drinceanu D, Pop IM, Stack D, Ştef L, Julean C, Bourke B (2010) The Effect of Probiotics on Animal Health. Anim Sci Biotechnol 43: 35-41.

Curtis CR, Erb HN, Sniffen CJ, Smith RD (1984) Epidemiology of parturient paresis: predisposing factors with emphasis on dry cow feeding and management. J Dairy Sci 67: 817-825.

Dade-Senn B, Laporta J, Dahl GE (2020) Carry over effects of late-gestational heat stress on dairy cattle progeny. Theriogenology 154: 17-23.

Filteau V, Bouchard E, Fecteau G, Dutil L, DuTremblay D (2003) Health status and risk factors associated with failure of passive transfer of immunity in newborn beef calves in Québec. Can Vet J 44: 907-913.

Gao H, Wu G, Spencer TE, Johnson GA, Bazer FW (2009) Select nutrients in the ovine uterine lumen. II. Glucose transporters in the uterus and peri-implantation conceptuses. Biol Reprod 80: 94-104.

Garcia M, Greco LF, Favoreto MG, Marsola RS, Martins LT, Besinotto RS, Shin JH, Lock AL, Block E, Thatcher WW, Santos JE, Staples CR (2014) Effect of supplementing fat to pregnant nonlactating cows on colostral fatty acid profile and passive immunity of the newborn calf. J Dairy Sci 97: 392-405.

Girard J (1990) Metabolic adaptations to change of nutrition at birth. Biol Neonate 58 (Suppl 1): 3-15.

Godden S (2008) Colostrum management for dairy calves. Vet Clin North Ame Food Anim Pract 24: 19-39.

Graugnard DE, Bionaz M, Trevisi E, Moyes KM, Salak-Johnson JL, Wallace RL, Drackley J K, Bertoni G, Loor JJ (2012) Blood immunometabolic indices and polymorphonuclear neutrophil function in peripartum dairy cows are altered by level of dietary energy prepartum. J Dairy Sci 95: 1749-1758.

Guo JR, Monteiro AP, Weng XS, Ahmed BM, Laporta J, Hayen MJ, Dahl GE, Bernard JK, Tao S (2016) Short communication: Effect of maternal heat stress in late gestation on blood hormones and metabolites of newborn calves. J Dairy Sci 99: 6804-6807.

Haddad B, McCluskie K, Birrell MA, Dabrowski D, Pecoraro M, Underwood S, Chen B, De Sanctis GT, Webber SE, Foster ML, Belvisi MG (2002) Differential effects of ebselen on neutrophil recruitment, chemokine, and inflammatory mediator expression in a rat model of lipopolysaccharide-induced pulmonary inflammation. J Immunol 169: 974-982.

Hall JA, Bobe G, Vorachek VR, Estill CT, Mosher WD, Pirelli GJ, Gamroth M (2014) Effect of supranutritional maternal and colostral selenium supplementation on passive absorption of immunoglobulin G in selenium-replete dairy calves. J Dairy Sci 97: 4379-4391.

Hefnawy EG, Tórtora-Pérez JL (2010) The importance of selenium and the effects of its deficiency in animal health. Small Ruminant Res 89: 185-192.

Horst EA, Kvidera SK, Baumgard LH (2021) Invited review: The influence of immune activation on transition cow health and performance – A critical evaluation of traditional dogmas. J Dairy Sci 104: 8380-8410

Hulbert LE, Moisá SJ (2016) Stress, immunity, and the management of calves. J Dairy Sci 99: 3199-3216.

Hunter AL (2015) Association of serum calcium status at calving on survival, health, and performance of post-partum holstein cows and calves. Doctoral Thesis The Ohio State University, 2015 pp.81 https://etd.ohiolink.edu/acprod/odb_
etd/ws/send_file/send?accession=osu1429743557&disposi
tion=inline

Hyrslova I, Krausova G, Michlova T, Kana A, Curda L. (2020) Fermentation Ability of Bovine Colostrum by Different Probiotic Strains. Fermentation 6: 93.

Ingvartsen KL, Moyes K (2013) Nutrition, immune function and health of dairy cattle. Animal 7: Suppl 1: 112-122.

Itle AJ, Huzzey JM, Weary DM, von Keyserlingk MA (2015) Clinical ketosis and standing behavior in transition cows. J Dairy Sci 98: 128-134.

Jovanovic-Peterson L, Fuhrmann K, Hedden K, Walker L, Peterson CM (1989) Maternal milk and plasma glucose and insulin levels: Studies in normal and diabetic subjects. J Am Coll Nutr 8: 125-131.

József L, Filep JG (2003) Selenium-containing compounds attenuate peroxynitrite-mediated NF-kappaB and AP-1 activation and interleukin-8 gene and protein expression in human leukocytes. Free Radic Biol Med 35: 1018-1027.

Kamada H, Nonaka I, Ueda Y, Murai M (2007) Selenium addition to colostrum increases immunoglobulin G absorption by newborn calves. J. Dairy Sci 90: 5665-5670.

Khan MZ, Ma Y, Xiao J, Chen T, Ma J, Liu S, Wang Y, Khan A, Alugongo GM, Cao Z (2022) Role of Selenium and Vitamins E and B9 in the Alleviation of Bovine Mastitis during the Periparturient Period. Antioxidants (Basel) 11: 657.

Kushibiki S, Hodate K, Shingu H, Obara Y, Touno E, Shinoda M, Yokomizo Y (2003) Metabolic and lactational responses during recombinant bovine tumor necrosis factor-alpha treatment in lactating cows. J Dairy Sci 86: 819-827.

LeBlanc SJ (2020) Review: Relationships between metabolism and neutrophil function in dairy cows in the peripartum period. Animal 14: (S1) s44-s54.

Ling T, Hernandez-Jover M, Sordillo LM, Abuelo A (2018) Maternal late-gestation metabolic stress is associated with changes in immune and metabolic responses of dairy calves. J Dairy Sci 101: 6568-6580.

Lopes MG, Alharthi AS, Lopreiato V, Abdel-Hamied E, Liang Y, Coleman DN, Dai H, Corrêa MN, Fernandez C, Loor JJ (2021) Maternal body condition influences neonatal calf whole-blood innate immune molecular responses to ex vivo lipopolysaccharide challenge. J Dairy Sci 104: 2266-2279.

Lopez AJ, Heinrichs AJ (2022) Invited review: The importance of colostrum in the newborn dairy calf. J Dairy Sci 105: 2733-2749

Lorenz I, Mee JF, Earley B, More SJ (2011) Calf health from birth to weaning. I. General aspects of disease prevention. Ir Vet J 64: 10.

Lucy MC, Green JC , Meyer JP , Williams AM , Newsom EM, Keisler DH (2012) Short communication: glucose and fructose concentrations and expression of glucose transporters in 4- to 6-week pregnancies collected from Holstein cows that were either lactating or not lactating. J Dairy Sci 95: 5095-5101

Mann S, Yepes FA, Overton TR, Lock AL, Lamb SV, Wakshlag JJ, Nydam DV (2016) Effect of dry period dietary energy level in dairy cattle on volume, concentrations of immunoglobulin G, insulin, and fatty acid composition of colostrum. J Dairy Sci 99: 1515-1526.

Martel CA, Mamedova LK, Minton JE, Garcia M, Legallet C, Bradford BJ (2018) Effects of TNF receptor blockade on in vitro cell survival and response to negative energy balance in dairy cattle. J Anim Sci Biotechnol 9: 6.

McGee M, Drennan MJ, Caffrey PJ (2006) Effect of age and nutrient restriction pre partum on beef suckler cow serum immunoglobulin concentrations, colostrum yield, composition and immunoglobulin concentration and immune status of their progeny. Ir J Agriculture Food Res 45: 157-171.

Mikuła R, Pruszyńska-Oszmałek E, Pszczola M. Rząsińska J, Sassek M, Nowak KW, Nogowski L, Kołodziejski PA (2021) Changes in metabolic and hormonal profiles during transition period in dairy cattle – the role of spexin. BMC Vet Res 17: 359.

Monteiro AP, Guo JR, Weng XS, Ahmed BM, Hayen MJ, Dahl GE, Bernard JK, Tao S (2016) Effect of maternal heat stress during the dry period on growth and metabolism of calves. J Dairy Sci 99: 3896–3907.

Mordak R, Nicpoń J, Illek J (2017) Metabolic and mineral conditions of retained placenta in highly productive dairy cows: pathogenesis, diagnostics and prevention – a review. Acta Vet Brno 86: 239-248.

Nardone A, Lacetera N, Bernabucci U, Ronchi B (1997) Composition of Colostrum from Dairy Heifers Exposed to High Air Temperatures During Late Pregnancy and the Early Postpartum Period. J Dairy Sci 80: 838 844

Nowak W, Mikuła R, Zachwieja A, Paczyńska K, Pecka E, Drzazga K, Ślósarz P (2012) The impact of cow nutrition in the dry period on colostrum quality and immune status of calves. Pol J Vet Sci 15: 77-82.

Osorio JS, Trevisi E, Ballou MA, Bertoni G, Drackley JK, Loor JJ (2013) Effect of the level of maternal energy intake prepartum on immunometabolic markers, polymorphonuclear leukocyte function, and neutrophil gene network expression in neonatal Holstein heifer calves. J Dairy Sci 96: 3573-3587.

Osorio JS (2020) Gut health, stress, and immunity in neonatal dairy calves: the host side of host-pathogen interactions. J Anim Sci Biotechnol 11: 105.

Paul SS, Dey A (2015) Nutrition in health and immune function of ruminants. Indian J Anim Sci 85: 103-112.

Piñeiro JM, Menichetti BT, Barragan AA, Relling AE, Weiss WP, Bas A, Schuenemann GM (2019) Associations of pre- and postpartum lying time with metabolic, inflammation, and health status of lactating dairy cows. J Dairy Sci 102: 3348-3361.

Planski B, Abrashev N (1987) Dynamic aspects of mineral metabolism in dry cows, puerperants and calves. Vet-Med Nauki 24: 48-57.

Podhorsky A, Pechová A, Dvořák R, Pavlata L (2007) Metabolic Disorders in Dairy Calves in Postpartum Period. Acta Vet Brno 76: S45-S53.

Puppel K, Gołebiewski M, Grodkowski G, Slósarz J, Kunowska-Slósarz M, Solarczyk P, Łukasiewicz M, Balcerak M, Przysucha T (2019) Composition and Factors Affecting Quality of Bovine Colostrum: A Review. Animals (Basel) 9: 1070.

Quigley JD, Drewry JJ (1998) Nutrient and Immunity Transfer from Cow to Calf Pre- and Postcalving. J Dairy Sci 81: 2779-2790.

Reinhardt TA, Lippolis JD, McCluskey BJ, Goff JP, Horst RL (2011) Prevalence of subclinical hypocalcemia in dairy herds. Vet J 188: 122-124.

Roberts T, Chapinal N, LeBlanc SJ, Kelton DF, Dubuc J, Duffield TF (2012) Metabolic parameters in transition cows as indicators for early-lactation culling risk. J Dairy Sci 95: 3057-3063.

Sheldon IM, Cronin JG, Bromfield JJ (2019) Tolerance and innate immunity shape the development of postpartum uterine disease and the impact of endometritis in dairy cattle. Annu Rev Anim Biosci 7: 361-384.

Shrimali RK, Irons RD, Carlson BA, Sano Y, Gladyshev VN, Park JM, Hatfield DL (2008) Selenoproteins mediate T cell immunity through an antioxidant mechanism. J Biol Chem 283: 20181-20185

Singh V, Singh VK, Singh SP, Sahoo B (2015) The effect of body condition score at calving on milk yield, milk composition and udder health status of dairy animals. J Dairy Vet Anim Res 2: 47-50.

Skibiel AL, Fabris TF, Corrá FN, Torres YM, McLean DJ, Chapman JD, Kirk DJ, Dahl GE, Laporta J (2017) Effects of feeding an immunomodulatory supplement to heat-stressed or actively cooled cows during late gestation on postnatal immunity, health, and growth of calves J Dairy Sci 100: 7659-7668.

Sordillo LM, Aitken SL (2009) Impact of oxidative stress on the health and immune function of dairy cattle. Vet Immunol Immunopathol 128: 104-109.

Sordillo LM, Mavangira V (2014) The nexus between nutrient metabolism, oxidative stress and inflammation in transition cows. Anim Prod Sci 54: 1204-1214.

Sordillo LM, Raphael W (2013) Significance of metabolic stress, lipid mobilization, and inflammation on transition cow disorders. Vet Clin North Am Food Anim Pract 29: 267-278.

Stanek S, Zink V, Doležal O, Stolc L (2014) Survey of preweaning dairy calf-rearing practices in Czech dairy herds. J Dairy Sci 97: 3973-3981.

Ster C, Loiselle MC, Lacasse P (2012) Effect of postcalving serum nonesterified fatty acids concentration on the functionality of bovine immune cells. J Dairy Sci 95: 708-717.

Sundrum A (2015) Metabolic Disorders in the Transition Period Indicate that the Dairy Cows’ Ability to Adapt is Overstressed. Animals (Basel) 5: 978-1020.

Tao S, Monteiro AP, Thompson IM, Hayen MJ, Dahl GE (2012) Effect of late -gestation maternal heat stress on growth and immune function of dairy calves. J Dairy Sci 95: 7128-7136.

Trimboli F, Ragusa M, Piras C, Lopreiato V, Britti D (2020) Outcomes from experimental testing of nonsteroidal anti-inflammatory drug (NSAID) administration during the transition period of dairy cows. Animals (Basel) 10: 1832.

Tufarelli V, Colonna MA, Losacco C, Puvača N (2023) Biological health markers associated with oxidative stress in dairy cows during lactation period. Metabolites 13: 405.

Urban-Chmiel R (2019) Metabolic disorders of cows and the level of immunity of calves. Monograph CCXXIV entitled: The newborn and the environment. Copyright by Uniwersytet Przyrodniczy we Wrocławiu, pp 43-51

Urban-Chmiel R, Hola P, Lisiecka U, Wernicki A, Puchalski A, Dec M, Wysocka M (2011) An evaluation of the effects of α-tocopherol and ascorbic acid in bovine respiratory disease complex occurring in feedlot calves after transport. Livest Sci 141: 53-58.

Vlasova AN, Saif LJ (2021) Bovine Immunology: Implications for Dairy Cattle. Front Immunol 12: 643206.

Waldner CL, Rosengren LB (2009) Factors associated with serum immunoglobulin levels in beef calves from Alberta and Saskatchewan and association between passive transfer and health outcomes. Can Vet J 50: 275-281.

Walker WL, Epperson WB, Wittum TE, Lord LK, Rajala-Schultz PJ, Lakritz J (2012) Characteristics of dairy calf ranches: morbidity, mortality antibiotic use practices and biosecurity and biocontainment practices. J Dairy Sci 95: 2204-2214.

Wang B, Wang Ch, Guan R, Shi K, Wei Z, Liu J, Liu H (2019) Effects of Dietary Rumen-Protected Betaine Supplementation on Performance of Postpartum Dairy Cows and Immunity of Newborn Calves. Animals (Basel) 9: 167.

Wankhade PR, Manimaran A, Kumaresan A, Jeyakumar S, Ramesha KP, Sejian V, Rajendran D, Varghese MR (2017) Metabolic and immunological changes in transition dairy cows: A review. Vet World 10: 1367-1377.

Wilhelm AL, Maquivar MG, Bas S, Brick TA, Weiss WP, Bothe H, Velez JS, Schuenemann GM (2017) Effect of serum calcium status at calving on survival, health, and performance of postpartum Holstein cows and calves under certified organic management. J Dairy Sci 100: 3059-3067.

Windeyer MC, Leslie KE, Godden SM, Hodgins DC, Lissemore KD, LeBlanc SJ (2014) Factors associated with morbidity, mortality, and growth of dairy heifer calves up to 3 months of age. Prev Vet Med 113: 231-240.

Wintergerst ES, Maggini S, Hornig DH (2007) Contribution of selected vitamins and trace elements to immune function. Ann. Nutr. Metab 51: 301-323.

Wnorowska K, Młynek K, Pupel K (2024) The impact of negative energy balance in Holstein-Friesian cows on the blood concentrations of interleukin-6 and plasminogen. Metabolites 14: 548.

Yang DY, Chang C J, Peh HC, Chen MT (2004) Anti-peroxidation effects of vitamin E on low density lipoprotein and milk fat globule membrane of lactating goats: in vivo versus metal ion challenge in vitro. Comp. Biochem. Physiol. A Mol Integr Physiol 139: 11-20

Yang M, Zou Y, Wu ZH, Li SL, Cao ZJ (2015) Colostrum quality affects immune system establishment and intestinal development of neonatal calves. J Dairy Sci 98: 7153-7163.

Žekić-Stošić M, Kanački Z, Stojanović D, Bugarski D, Lazarević M, Milovanović A, Masic A (2018) Neutrophil phagocytosis and respiratory burst activity of dairy cows during the transition period and early lactation. J Infect Dev Ctries 12: 894-903.

Zhao W, Chen X,, Xiao J, Chen XH, Zhang XF, Wang T, Zhen YG, Qin GX (2019) Prepartum body condition score affects milk yield, lipid metabolism, and oxidation status of Holstein cows. Asian-Australas J Anim Sci 32: 1889-1896

Zou Y, Wang Y, Deng Y. Cao Z, Li S, Wang J. (2017) Effects of feeding untreated, pasteurized and acidified waste milk and bunk tank milk on the performance, serum metabolic profiles, immunity, and intestinal development in Holstein calves. J Anim Sci Biotechnol 8: 53

Go to article

Authors and Affiliations

R. Urban-Chmiel
1
P. Sobiech
2

  1. Department of Veterinary Prevention and Avian Diseases,Faculty of Veterinary Medicine, Głęboka 30, 20-612 Lublin, Poland
  2. Department of Internal Diseases with Clinic, Faculty of Veterinary Medicine,University of Warmia-Mazury in Olsztyn, Oczapowskiego 14, 10-911 Olsztyn, Poland

Abstract

The global consumption of poultry is expected to increase by 2032, especially in Asian and European countries. Proper nutrition, including feed additives, plays a role in meeting the global demand for livestock products. In addition, the use of antibiotics as growth promoters resulted in antibiotic resistance issues, leading to the need to investigate alternative methods for replacing the role of antibiotics. The use of probiotic bacteria has proved to improve the growth performance of poultry and suppress pathogenic bacteria growth in the digestive tract. However, microenvironment conditions in the digestive tract affect the survival of probiotic bacteria. Modification of bacteria by encapsulation shows promising potential to protect bacteria from the harsh conditions in the poultry digestive tract. In addition, probiotic encapsulation also showed improvement in performance in poultry. This review will discuss the current encapsulation technology in probiotics applied to poultry and its effects. It also will explore the prospect of encapsulated probiotics, especially in the poultry industry, and its challenge.
Go to article

Bibliography

Alkushi AG, Elazab ST, Abdelfattah-Hassan A, Mahfouz H, Salem GA, Sheraiba NI, Mohamed EAA, Attia MS, El-Shetry ES, Saleh AA, Elsawy NA, Ibrahim D (2022) Multi-Strain-Probiotic-Loaded Nanoparticles Reduced Colon Inflammation and Orchestrated the Expressions of Tight Junction, NLRP3 Inflammasome and Caspase-1 Genes in DSS-Induced Colitis Model. Pharmaceutics 14.

Anselmo AC, McHugh KJ, Webster J, Langer R, Jaklenec A (2016) Layer‐by‐Layer Encapsulation of Probiotics for Delivery to the Microbiome. Advanced Materials 28: 9486-9490.

Asgari S, Pourjavadi A, Licht TR, Boisen A, Ajalloueian F (2020) Polymeric carriers for enhanced delivery of probiotics. Adv Drug Deliv Rev 161-162: 1-21.

Babot JD, Argañaraz-Martínez E, Apella MC, Perez Chaia A (2023) Microencapsulation of Probiotics with Soy Protein Isolate and Alginate for the Poultry Industry. Food Bioproc Tech 16: 1478-1487.

Baindara P, Chakraborty R, Holliday ZM, Mandal SM, Schrum AG (2021) Oral probiotics in coronavirus disease 2019: connecting the gut-lung axis to viral pathogenesis, inflammation, secondary infection and clinical trials. New Microbes New Infect 40: 100837.

Barszcz M, Tuśnio A, Taciak M (2024) Poultry nutrition. Physical Sciences Reviews 9: 611-650.

Bilal M, Si W, Barbe F, Chevaux E, Sienkiewicz O, Zhao X (2021) Effects of novel probiotic strains of Bacillus pumilus and Bacillus subtilis on production, gut health, and immunity of broiler chickens raised under suboptimal conditions. Poult Sci 100: 100871.

Cao J, Cheng J, Xi S, Qi X, Shen S, Ge Y (2019) Alginate/
/chitosan microcapsules for in-situ delivery of the protein, interleukin-1 receptor antagonist (IL-1Ra), for the treatment of dextran sulfate sodium (DSS)-induced colitis in a mouse model. European Journal of Pharmaceutics and Biopharmaceutics 137: 112-121.

Chan ES, Zhang Z (2005) Bioencapsulation by compression coating of probiotic bacteria for their protection in an acidic medium. Process Biochemistry 40: 3346-3351.

Chen S, Yong Y, Ju X (2021) Effect of heat stress on growth and production performance of livestock and poultry: Mechanism to prevention. J Therm Biol 99: 103019.

de Oliveira GLV, Oliveira CNS, Pinzan CF, de Salis LVV, Cardoso CR de B (2021) Microbiota Modulation of the Gut-Lung Axis in COVID-19. Front Immunol 12: 635471.

Dibner JJ, Richards JD (2005) Antibiotic growth promoters in agriculture: history and mode of action. Poult Sci 84: 634-643.

Dong ZL, Wang YW, Song D, Hou YJ, Wang WW, Qi WT, Yun TT, Li AK (2016) The effects of dietary supplementation of pre-microencapsulated Enterococcus fecalis and the extract of Camellia oleifera seed on growth performance, intestinal morphology, and intestinal mucosal immune functions in broiler chickens. Anim Feed Sci Technol 212: 42-51.

Dong ZL, Wang YW, Song D, Wang WW, Liu KB, Wang L, Li AK (2019) Effects of microencapsulated probiotics and plant extract on antioxidant ability, immune status and caecal microflora in Escherichia coli K88-challenged broiler chickens. Food Agric Immunol 30: 1123-1134.

Fesseha H, Demlie T, Mathewos M, Eshetu E (2021) Effect of Lactobacillus Species Probiotics on Growth Performance of Dual-Purpose Chicken. Veterinary Medicine: Research and Reports Volume 12: 75-83.

Gharib-Naseri K, Dorigam JCP, Doranalli K, Morgan N, Swick RA, Choct M, Wu S-B (2021) Bacillus amyloliquefaciens CECT 5940 improves performance and gut function in broilers fed different levels of protein and/or under necrotic enteritis challenge. Animal Nutrition 7: 185-197.

Gyawali I, Zeng Y, Zhou J, Li J, Wu T, Shu G, Jiang Q, Zhu C (2022) Effect of novel Lactobacillus paracaesi microcapsule on growth performance, gut health and microbiome community of broiler chickens. Poult Sci 101.

Han W, Zhang XL, Wang DW, Li LY, Liu GL, Li AK, Zhao YX (2013) Effects of microencapsulated Enterococcus fecalis CG1.0007 on growth performance, antioxidation activity, and intestinal microbiota in broiler chickens1. J Anim Sci 91: 4374-4382.

Hashem NM, Hosny NS, El-Desoky N, Soltan YA, Elolimy AA, Sallam SMA, Abu-Tor ESM (2023) Alginate Nanoencapsulated Synbiotic Composite of Pomegranate Peel Phytogenics and Multi-Probiotic Species as a Potential Feed Additive: Physicochemical, Antioxidant, and Antimicrobial Activities. Animals 13: 2432.

Hernández-Granados MJ, Ortiz-Basurto RI, Jiménez-Fernández M, García-Munguía CA, Franco-Robles E (2022) Dietary encapsulated Bifidobacterium animalis and Agave fructans improve growth performance, health parameters, and immune response in broiler chickens. Anim Biosci 35: 587-595.

Hill JM, Clement C, Pogue AI, Bhattacharjee S, Zhao Y, Lukiw WJ (2014) Pathogenic microbes, the microbiome, and Alzheimer’s disease (AD). Front Aging Neurosci 6: 127.

Ismail H, Ibrahim D, El Sayed S, Wahdan A, El-Tarabili RM, Rizk El-Ghareeb W, Abdullah Alhawas B, Alahmad BAHY, Abdel-Raheem SM, El-Hamid MIA (2023) Prospective Application of Nanoencapsulated Bacillus amyloliquefaciens on Broiler Chickens’ Performance and Gut Health with Efficacy against Campylobacter jejuni Colonization. Animals 13: 775.

Jiang T, Li H-S, Han GG, Singh B, Kang S-K, Bok J-D, Kim D-D, Hong Z-S, Choi Y-J, Cho C-S (2017) Oral Delivery of Probiotics Using pH-Sensitive Tablets. J Microbiol Biotechnol 27: 739-746.

Kamwa R, Khurajog B, Muangsin N, Pupa P, Hampson DJ, Prapasarakul N (2024) Water-soluble microencapsulation using gum Arabic and skim milk enhances viability and efficacy of Pediococcus acidilactici probiotic strains for application in broiler chickens. Anim Biosci 37:1440-1451.

Krasaekoopt W, Bhandari B, Deeth H (2003) Evaluation of encapsulation techniques of probiotics for yoghurt. Int Dairy J 13: 3-13.

Lorenzo-Rebenaque L, Malik DJ, Catalá-Gregori P, Marin C, Sevilla-Navarro S (2022) Gastrointestinal Dynamics of Non-Encapsulated and Microencapsulated Salmonella Bacteriophages in Broiler Production. Animals 12.

Neveling DP, Dicks LMT (2021) Probiotics: an Antibiotic Replacement Strategy for Healthy Broilers and Productive Rearing. Probiotics Antimicrob Proteins 13: 1-11.

OECD-FAO Agricultural Outlook 2023-2032 (2023) OECD. https: //www.oecd.org/en/publications/oecd-fao-agriculturaloutlook-2023-2032_08801ab7-en.html

Plaza-Diaz J, Ruiz-Ojeda FJ, Gil-Campos M, Gil A (2019) Mechanisms of Action of Probiotics. Adv Nutri 10: S49-S66.

Poorbaghi SL, Gheisari H, Dadras H, Sepehrimanesh M, Zolfaghari A (2016) Effects of Simple and Microencapsulated Lactobacillus acidophilus With or Without Inulin on the Broiler Meat Quality Infected by Avian Influenza Virus (H9N2). Probiotics Antimicrob Proteins 8: 221-228.

Pradipta MS, Harimurti S, Widodo W (2019) Feed Supplementation with Encapsulated Indigenous Probiotic Lactic Acid Bacteria Increased Broiler Chicken Performance. ASEAN J Sci Techno Dev 36: 29-34.

Qi W, Liang X, Yun T, Guo W (2019) Growth and survival of microencapsulated probiotics prepared by emulsion and internal gelation. J Food Sci Technol 56: 1398-1404.

Rajendran D, Ezhuthupurakkal PB, Lakshman R, Gowda NK, Manimaran A, Rao SB (2022) Application of encapsulated nano materials as feed additive in livestock and poultry: a review. Vet Res Commun 46: 315-328.

Rocha-Ramírez LM, Hernández-Ochoa B, Gómez-Manzo S, Marcial-Quino J, Cárdenas-Rodríguez N, Centeno-Leija S, García-Garibay M (2020) Evaluation of Immunomodulatory Activities of the Heat-Killed Probiotic Strain Lactobacillus casei IMAU60214 on Macrophages In Vitro. Microorganisms 8: 79.

Rodklongtan A, La-ongkham O, Nitisinprasert S, Chitprasert P (2014) Enhancement of Lactobacillus reuteri KUB-AC5 survival in broiler gastrointestinal tract by microencapsulation with alginate-chitosan semi-interpenetrating polymer networks. J Appl Microbiol 117: 227-238.

Shahbazi R, Yasavoli-Sharahi H, Alsadi N, Ismail N, Matar C (2020) Probiotics in Treatment of Viral Respiratory Infections and Neuroinflammatory Disorders. Molecules 25: 4891.

Talani G, Biggio F, Mostallino MC, Locci V, Porcedda C, Boi L, Saolini E, Piras R, Sanna E, Biggio G (2020) Treatment with gut bifidobacteria improves hippocampal plasticity and cognitive behavior in adult healthy rats. Neuropharmacology 165: 107909.

Trabelsi I, Ktari N, Ben Slima S, Bouchaala K, Ben Salah R (2016) Effects of supplementation with L. plantarum TN8 encapsulated in alginate-chitosan in broiler chickens. Int J Biol Macromol 89: 677-681.

Vimon S, Angkanaporn K, Nuengjamnong C (2023a) Microencapsulation of Lactobacillus plantarum MB001 and its probiotic effect on growth performance, cecal microbiome and gut integrity of broiler chickens in a tropical climate. Anim Biosci 36: 1252-1262.

Vimon S, Kertsomboon T, Chirachanchai S, Angkanaporn K, Nuengjamnong C (2023b) Matrices-charges of agar-alginate crosslinked microcapsules via o/w microemulsion: A non-spore forming probiotic bacteria encapsulation system for extensive viability. Carbohydr Polym 321: 121302.

Wen P, Zong MH, Linhardt RJ, Feng K, Wu H (2017) Electrospinning: A novel nano-encapsulation approach for bioactive compounds. Trends Food Sci Technol 70: 56-68.

Wu H, Ding C, Ma X, Gao Z, Liu S, Liu B, Song S (2024) Microencapsulate Probiotics (MP) Promote Growth Performance and Inhibit Inflammatory Response in Broilers Challenged with Salmonella typhimurium. Probiotics Antimicrob Proteins 16: 623-635.

Yao M, Wu J, Li B, Xiao H, McClements DJ, Li L (2017) Microencapsulation of Lactobacillus salivarious Li01 for enhanced storage viability and targeted delivery to gut microbiota. Food Hydrocoll 72: 228-236.

Yazhini P, Visha P, Selvaraj P, Vasanthakumar P, Chandran V (2018) Dietary encapsulated probiotic effect on broiler serum biochemical parameters. Vet World 11: 1344-1348.

Yousefi M, Khanniri E, Shadnoush M, Khorshidian N, Mortazavian AM (2020) Development, characterization and in vitro antioxidant activity of chitosan-coated alginate microcapsules entrapping Viola odorata Linn. extract. Int J Biol Macromol 163: 44-54.

Zhang L, Li J, Yun TT, Qi WT, Liang XX, Wang YW, Li AK (2015) Effects of pre-encapsulated and pro-encapsulated Enterococcus faecalis on growth performance, blood characteristics, and cecal microflora in broiler chickens. Poult Sci 94: 2821-2830.

Zhang Y, Liu Y, Jiao S, Wang Y, Sa R, Zhao F, Xie J (2024) Short-term supplementation with uncoated and encapsulated Enterococcus faecium affected growth performance, gut microbiome and intestinal barrier integrity in broiler chickens. Poult Sci 103: 103808.

 

 

Go to article

This page uses 'cookies'. Learn more