21,584
Views
129
CrossRef citations to date
0
Altmetric
Reviews

Advances in therapeutic and managemental approaches of bovine mastitis: a comprehensive review

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , , ORCID Icon, , ORCID Icon, , , ORCID Icon, & show all
Pages 107-136 | Received 13 Apr 2020, Accepted 25 Jan 2021, Published online: 17 Feb 2021

References

  • 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(3):355–362.
  • Abebe R, Hatiya H, Abera M, Megersa B, Asmare K. 2016. Bovine mastitis: prevalence, risk factors and isolation of Staphylococcus aureus in dairy herds at Hawassa milk shed, South Ethiopia. BMC Vet Res. 12(1):270.
  • Aghamohammadi M, Haine D, Kelton DF, Barkema HW, Hogeveen H, Keefe GP, Dufour S. 2018. Herd-level mastitis-associated costs on Canadian dairy farms. Front Vet Sci. 5:100
  • Ahmad V, Khan MS, Jamal QMS, Alzohairy MA, Al Karaawi MA, Siddiqui MU. 2017. Antimicrobial potential of bacteriocins: in therapy, agriculture and food preservation. Int J Antimicrob Agents. 49(1):1–11.
  • Akers RM, Nickerson SC. 2011. Mastitis and its impact on structure and function in the ruminant mammary gland. J Mammary Gland Biol Neoplasia. 16(4):275–289.
  • Algharib SA, Dawood A, Xie S. 2020. Nanoparticles for treatment of bovine Staphylococcus aureus mastitis. Drug Deliv. 27(1):292–308.
  • Almeida RA, Kerro-Dego O, Prado ME, Headrick SI, Lewis MJ, Siebert LJ, Pighetti GM, Oliver SP. 2015. Protective effect of anti-SUAM antibodies on Streptococcus uberis mastitis. Vet Res. 46:133
  • Alva-Murillo N, Téllez-Pérez AD, Sagrero-Cisneros E, López-Meza JE, Ochoa-Zarzosa A. 2012. Expression of antimicrobial peptides by bovine endothelial cells. Cell Immunol. 280(1):108–112..
  • Alves A, Capra E, Morandi S, Cremonesi P, Pantoja J, Langoni H, de Vargas A, da Costa M, Jagielski T, Bolaños C, et al. 2017. In vitro algicidal effect of guanidine on Prototheca zopfii genotype 2 strains isolated from clinical and subclinical bovine mastitis. Lett Appl Microbiol. 64(6):419–423.
  • Amiri Fahliyani S, Beheshti-Maal K, Ghandehari F. 2018. Novel lytic bacteriophages of Klebsiella oxytoca ABG-IAUF-1 as the potential agents for mastitis phage therapy. FEMS Microbiol Lett. 365(20)p.fny223.
  • Andrews T, Neher DA, Weicht TR, Barlow JW. 2019. Mammary microbiome of lactating organic dairy cows varies by time, tissue site, and infection status. PLoS One. 14(11):e0225001
  • Anika TT, Al Noman Z, Ferdous MRA, Khan SH, Mukta MA, Islam MS, Hossain MT, Rafiq K. 2019. Time dependent screening of antibiotic residues in milk of antibiotics treated cows. J Adv Vet Anim Res. 6(4):516–520.
  • Ashraf A, Imran M. 2020. Causes, types, etiological agents, prevalence, diagnosis, treatment, prevention, effects on human health and future aspects of bovine mastitis. Anim Health Res Rev. 13:1–14.
  • Avancini C, Wiest JM, Dall’Agnol R, Haas JS, von Poser GL. 2008. Antimicrobial activity of plants used in the prevention and control of bovine mastitis in Southern Brazil. Latin Am J Pharm. 27(6):894–899.
  • Babra C, Tiwari JG, Pier G, Thein TH, Sunagar R, Sundareshan S, Isloor S, Hegde NR, de Wet S, Deighton M, et al. 2013. The persistence of biofilm-associated antibiotic resistance of Staphylococcus aureus isolated from clinical bovine mastitis cases in Australia. Folia Microbiol (Praha)). 58(6):469–474.
  • Bardhan D. 2013. Estimates of economic losses due to clinical mastitis in organized dairy farms. Indian J Dairy Sci. 66:168–172.
  • Barreiro JR, Gonçalves JL, Braga PA, Dibbern AG, Eberlin MN, Veiga Dos Santos M. 2017. Non-culture-based identification of mastitis-causing bacteria by MALDI-TOF mass spectrometry. J Dairy Sci. 100(4):2928–2934.
  • Bates AJ, Chambers G, Laven RA. 2016. Comparison of cephalonium alone and in combination with an internal teat sealant for dry cow therapy in seasonally calving dairy cows. N Z Vet J. 64(2):95–100.
  • Behera S, Rana R, Gupta PK, Kumar D, Rekha V, Arun TR, Jena D. 2018. Development of real-time PCR assay for the detection of Mycoplasma bovis. Trop Anim Health Prod. 50(4):875–882..
  • Benić M, Maćešić N, Cvetnić L, Habrun B, Cvetnić Ž, Turk R, Đuričić D, Lojkić M, Dobranić V, Valpotić H, Department for Bacteriology and Parasitology, Croatian Veterinary Institute, Zagreb, Croatia, et al. 2018. Bovine mastitis: a persistent and evolving problem requiring novel approaches for its control-a review. Vet Arhiv. 88(4):535–557.,
  • Bennett R, Christiansen K, Clifton-Hadley R. 1999. Preliminary estimates of the direct costs associated with endemic diseases of livestock in Great Britain. Prev Vet Med. 39(3):155–171.
  • Berry EA, Hillerton JE. 2002a. The effect of selective dry cow treatment on new intramammary infections. J Dairy Sci. 85(1):112–121.
  • Berry EA, Hillerton JE. 2002b. The effect of an intramammary teat seal on new intramammary infections. J Dairy Sci. 85(10):2512–2520.
  • Bhattarai D, Worku T, Dad R, Rehman ZU, Gong X, Zhang S. 2018. Mechanism of pattern recognition receptors (PRRs) and host pathogen interplay in bovine mastitis. Microb Pathog. 120:64–70.
  • Bobbo T, Roveglia C, Penasa M, Visentin G, Finocchiaro R, Cassandro M. 2019. Genetic relationships of alternative somatic cell count traits with milk yield, composition and udder type traits in Italian Jersey cows. Anim Sci J. 90(7):808–817.
  • Borm AA, Fox LK, Leslie KE, Hogan JS, Andrew SM, Moyes KM, Oliver SP, Schukken YH, Hancock DD, Gaskins CT, et al. 2006. Effects of prepartum intramammary antibiotic therapy on udder health, milk production, and reproductive performance in dairy heifers. J Dairy Sci. 89(6):2090–2098.
  • Bouchard DS, Rault L, Berkova N, Le Loir Y, Even S. 2013. Inhibition of Staphylococcus aureus invasion into bovine mammary epithelial cells by contact with live Lactobacillus casei. Appl Environ Microbiol. 79(3):877–885.
  • Bouchard DS, Seridan B, Saraoui T, Rault L, Germon P, Gonzalez-Moreno C, Nader-Macias FM, Baud D, François P, Chuat V, et al. 2015. Lactic acid bacteria isolated from bovine mammary microbiota: potential allies against bovine mastitis. PLoS One. 10(12):e0144831
  • Bradley AJ, Breen JE, Payne B, White V, Green MJ. 2015. An investigation of the efficacy of a polyvalent mastitis vaccine using different vaccination regimens under field conditions in the United Kingdom. J Dairy Sci. 98(3):1706–1720.
  • Breser ML, Felipe V, Bohl LP, Orellano MS, Isaac P, Conesa A, Rivero VE, Correa SG, Bianco ID, Porporatto C. 2018. Chitosan and cloxacillin combination improve antibiotic efficacy against different lifestyle of coagulase-negative Staphylococcus isolates from chronic bovine mastitis. Sci Rep. 8(1):5081
  • Breyne K, Honaker RW, Hobbs Z, Richter M, Żaczek M, Spangler T, Steenbrugge J, Lu R, Kinkhabwala A, Marchon B, et al. 2017. Efficacy and safety of a bovine-associated Staphylococcus aureus phage cocktail in a murine model of mastitis. Front Microbiol. 8:2348.
  • Buragohain R, Sar TK, Samanta I, Biswas U, Mandal TK. 2019. Disposition of ceftizoxime in staphylococcal mastitis in Indian crossbred cows. Vet J. 245:12–14.
  • Burović J. 2020. Isolation of bovine clinical mastitis bacterial pathogens and their antimicrobial susceptibility in the Zenica region in 2017. Vet Stn (Online)). 51(1):47–52.
  • Cahuascanco B, Bahamonde J, Huaman O, Jervis M, Cortez J, Palomino J, Escobar A, Retamal P, Torres CG, Peralta OA. 2019. Bovine fetal mesenchymal stem cells exert antiproliferative effect against mastitis causing pathogen Staphylococcus aureus. Vet Res. 50(1):25
  • Cameron M, Keefe GP, Roy JP, Stryhn H, Dohoo IR, McKenna SL. 2015. Evaluation of selective dry cow treatment following on-farm culture: milk yield and somatic cell count in the subsequent lactation. J Dairy Sci. 98(4):2427–2436.
  • Cao LT, Wu JQ, Xie F, Hu SH, Mo Y. 2007. Efficacy of nisin in treatment of clinical mastitis in lactating dairy cows. J Dairy Sci. 90(8):3980–3985.
  • Capuco AV, Choudhary RK, Daniels KM, Li RW, Evock-Clover CM. 2012. Bovine mammary stem cells: cell biology meets production agriculture. Animal. 6(3):382–393.
  • Carson L, Gorman SP, Gilmore BF. 2010. The use of lytic bacteriophages in the prevention and eradication of biofilms of Proteus mirabilis and Escherichia coli. FEMS Immunol Med Microbiol. 59(3):447–455.
  • Castelani L, Arcaro JRP, Braga JEP, Bosso AS, Moura Q, Esposito F, Sauter IP, Cortez M, Lincopan N. 2019. Short communication: Activity of nisin, lipid bilayer fragments and cationic nisin-lipid nanoparticles against multidrug-resistant Staphylococcus spp. isolated from bovine mastitis. J Dairy Sci. 102(1):678–683.
  • Ceballos-Marquez A, Barkema HW, Stryhn H, Wichtel JJ, Neumann J, Mella A, Kruze J, Espindola MS, Wittwer F. 2010. The effect of selenium supplementation before calving on early-lactation udder health in pastured dairy heifers. J Dairy Sci. 93(10):4602–4612.
  • Chaitanya Kumar TV, Muralidhar Y, Prasad PE, Prasad TN, Alpha Raj M. 2013. Evaluation of therapeutic potential of nanosilver particles synthesised using aloin in experimental murine mastitis model. IET Nanobiotechnol. 7(3):78–82.
  • Chakraborty S, Dhama K, Tiwari R, Yatoo MI, Khurana SK, Khandia R, Munjal A, Palanivelu M, Kumar MA, Singh M, et al. 2019. Technological interventions and advances in the diagnosis of intramammary infections in animals with emphasis on bovine population–a review. Vet Q. 39(1):76–94.
  • Chambers GP, Newton HT. 2019. Investigation of the association between the interval from internal teat sealant administration to calving and early lactation clinical mastitis in New Zealand dairy heifers. N Z Vet J. 67(2):93–100.
  • Chehabi CN, Nonnemann B, Astrup LB, Farre M, Pedersen K. 2019. In vitro antimicrobial resistance of causative agents to clinical mastitis in Danish dairy cows. Foodborne Pathog Dis.
  • Cheng WN, Jeong CH, Seo HG, Hau SG. 2019. Moringa extract attenuates inflammatory responses and increases gene expression of casein in bovine mammary epithelial cells. Animals. 7:391.
  • Chung PY, Khanum R. 2017. Antimicrobial peptides as potential anti-biofilm agents against multidrug-resistant bacteria. J Microbiol Immunol Infect. 50(4):405–410..
  • Collado R, Prenafeta A, González-González L, Pérez-Pons JA, Sitjà M. 2016. Probing vaccine antigens against bovine mastitis caused by Streptococcus uberis. Vaccine. 34(33):3848–3854.
  • Compton CW, Emslie FR, McDougall S. 2014. Randomised controlled trials demonstrate efficacy of a novel internal teat sealant to prevent new intramammary infections in dairy cows and heifers. N Z Vet J. 62(5):258–266.
  • Cornelissen JB, De Greeff A, Heuvelink AE, Swarts M, Smith HE, der Van Wal FJ, 1Health4Food—Dutch Mastitis Diagnostics Consortium. 2016. Rapid detection of Streptococcus uberis in raw milk by loop-mediated isothermal amplification. J Dairy Sci. 99(6):4270–4281.
  • Cortinhas CS, Tomazi T, Zoni MSF, Moro E, Veiga Dos Santos M. 2016. Randomized clinical trial comparing ceftiofur hydrochloride with a positive control protocol for intramammary treatment of nonsevere clinical mastitis in dairy cows. J Dairy Sci. 99(7):5619–5628.
  • Côté-Gravel J, Malouin F. 2019. Symposium review: features of Staphylococcus aureus mastitis pathogenesis that guide vaccine development strategies. J Dairy Sci. 102(5):4727–4740.
  • Cubeddu T, Cacciotto C, Pisanu S, Tedde V, Alberti A, Pittau M, Dore S, Cannas A, Uzzau S, Rocca S, et al. 2017. Cathelicidin production and release by mammary epithelial cells during infectious mastitis. Vet Immunol Immunopathol. 189:66–70..
  • Cvetnić L, Samardžija M, Habrun B, Kompes G, Benić M. 2016. Microbiological monitoring of mastitis pathogens in the control of udder health in dairy cows. Slov Vet Res. 53(3):131–140.
  • Das D, Panda SK, Jena B, Sahoo AK. 2018. Economic impact of subclinical and clinical mastitis in Odisha, India. IntJCurrMicrobiolAppSci. 7(03):3651–3654.
  • Dash JR, Sar TK, Samanta I, Mandal TK. 2016. Effects of herbal extract of Ocimum sanctum as supportive therapy with intravenous ceftriaxone in experimentally induced staphylococcal chronic mastitis in goat. Small Ruminant Res. 137:1–8.
  • de Magalhães Rodrigues Martins CM, Alves BG, Monteiro CP, Pinheiro ESC, Feckinghaus MA, Paranhos LG, Dos Santos MV. 2019. Noninferiority field trial for evaluation of efficacy of ciprofloxacin associated with internal teat sealant as dry-off protocol. Trop Anim Health Prod. 51(8):2547–2557.
  • De Vliegher S, Fox LK, Piepers S, McDougall S, Barkema HW. 2012. Invited review: mastitis in dairy heifers: nature of the disease, potential impact, prevention, and control. J Dairy Sci. 95(3):1025–1040.
  • Derakhshani H, Fehr KB, Sepehri S, Francoz D, De Buck J, Barkema HW, Plaizier JC, Khafipour E. 2018. Invited review: Microbiota of the bovine udder: Contributing factors and potential implications for udder health and mastitis susceptibility. J Dairy Sci. 101(12):10605–10625.
  • Derakhshani H, Plaizier JC, De Buck J, Barkema HW, Khafipour E. 2018. Composition of the teat canal and intramammary microbiota of dairy cows subjected to antimicrobial dry cow therapy and internal teat sealant. J Dairy Sci. 101(11):10191–10205.
  • Desriac F, Defer D, Bourgougnon N, Brillet B, Le Chevalier P, Fleury Y. 2010. Bacteriocin as weapons in the marine animal-associated bacteria warfare: inventory and potential applications as an aquaculture probiotic. Mar Drugs. 8(4):1153–1177.
  • Dhama K, Latheef SK, Munjal AK, Khandia R, Samad HA, Iqbal HMN, Joshi SK. 2017. Probiotics in curing allergic and inflammatory conditions - research progress and futuristic vision. Recent Pat Inflamm Allergy Drug Discov. 10(2):105–118.
  • Dias RS, Eller MR, Duarte VS, Pereira ÂL, Silva CC, Mantovani HC, Oliveira LL, de AM, Silva E, de Paula SQ. 2013. Use of phages against antibiotic-resistant Staphylococcus aureus isolated from bovine mastitis. J Anim Sci. 91(8):3930–3939.
  • Dimri U, Sharma MC, Singh SK, Kumar P, Jhambh R, Singh B, Bandhyopadhyay S, Verma MR. 2013. Amelioration of altered oxidant/antioxidant balance of Indian water buffaloes with subclinical mastitis by vitamins A, D3, E, and H supplementation. Trop Anim Health Prod. 45(4):971–978.
  • Dingwell RT, Duffield TF, Leslie KE, Keefe GP, DesCoteaux L, Kelton DF, Lissemore KD, Schukken YH, Dick P, Bagg R. 2002. The efficacy of intramammary tilmicosin at drying-off, and other risk factors for the prevention of new intramammary infections during the dry period. J Dairy Sci. 85(12):3250–3259.
  • Doehring C, Sundrum A. 2019. The informative value of an overview on antibiotic consumption, treatment efficacy and cost of clinical mastitis at farm level. Prev Vet Med. 165:63–70.
  • Dos Anjos C, Sellera FP, Gargano RG, Lincopan N, Pogliani FC, Ribeiro MG, Jagielski T, Sabino CP. 2019. Algicidal effect of blue light on pathogenic Prototheca species. Photodiagnosis Photodyn Ther. 26:210–213.
  • Ðuričić D, Benić M, Maćešić N, Valpotić H, Turk R, Dobranić V, Cvetnić L, Gračner D, Vince S, Grizelj J, et al. 2017. Dietary zeolite clinoptilolite supplementation influences chemical composition of milk and udder health in dairy cows. Vet Stanica. 48(4):257–265.
  • Đuričić D, Sukalić T, Marković F, Kočila P, Žaja IŽ, Menčik S, Dobranić T, Benić M, Samardžija M. 2020. Effects of dietary vibroactivated clinoptilolite supplementation on the intramammary microbiological findings in dairy cows. Animals (Basel). 10(2):202.
  • Ebert F, Staufenbiel R, Simons J, Pieper L. 2017. Randomized, blinded, controlled clinical trial shows no benefit of homeopathic mastitis treatment in dairy cows. J Dairy Sci. 100(6):4857–4867.
  • Elbehiry A, Al-Dubaib M, Marzouk E, Moussa I. 2019. Antibacterial effects and resistance induction of silver and gold nanoparticles against Staphylococcus aureus-induced mastitis and the potential toxicity in rats. Microbiologyopen. 8(4):e00698.
  • Erskine RJ. 1993. Nutrition and mastitis. Vet Clin North Am Food Anim Pract. 9(3):551–561.
  • Falentin H, Rault L, Nicolas A, Bouchard DS, Lassalas J, Lamberton P, Aubry JM, Marnet PG, Le Loir Y, Even S. 2016. Bovine teat microbiome analysis revealed reduced alpha diversity and significant changes in taxonomic profiles in quarters with a history of mastitis. Front Microbiol. 7:480
  • Fenton M, Keary R, McAuliffe O, Ross RP, O'Mahony J, Coffey A. 2013. Bacteriophage-derived peptidase CHAP(K) Eliminates and prevents Staphylococcal biofilms. Int J Microbiol. 2013:625341
  • Francoz D, Wellemans V, Dupré JP, Roy JP, Labelle F, Lacasse P, Dufour S. 2017. Invited review: A systematic review and qualitative analysis of treatments other than conventional antimicrobials for clinical mastitis in dairy cows. J Dairy Sci. 100(10):7751–7770.
  • Frutis-Murillo M, Sandoval-Carrillo MA, Alva-Murillo N, Ochoa-Zarzosa A, López-Meza JE. 2019. Immunomodulatory molecules regulate adhesin gene expression in Staphylococcus aureus: effect on bacterial internalization into bovine mammary epithelial cells. Microb Pathog. 131:15–21.
  • Fuenzalida MJ, Ruegg PL. 2019. Negatively controlled, randomized clinical trial to evaluate use of intramammary ceftiofur for treatment of nonsevere culture-negative clinical mastitis. J Dairy Sci. 102(4):3321–3338.
  • Ganda EK, Bisinotto RS, Vasquez AK, Teixeira AGV, Machado VS, Foditsch C, Bicalho M, Lima FS, Stephens L, Gomes MS, et al. 2016. Effects of injectable trace mineral supplementation in lactating dairy cows with elevated somatic cell counts. J Dairy Sci. 99(9):7319–7329.
  • Garcia A. 2004. Contagious vs. environmental mastitis. Extension Extra. Paper 126. http://openprairie.sdstate.edu/extension_extra/126. [2020 March 4].
  • Geng H, Zou W, Zhang M, Xu L, Liu F, Li X, Wang L, Xu Y. 2020. Evaluation of phage therapy in the treatment of Staphylococcus aureus-induced mastitis in mice. Folia Microbiol (Praha). 65(2):339–351.
  • Gill JJ, Pacan JC, Carson ME, Leslie KE, Griffiths MW, Sabour PM. 2006. Efficacy and pharmacokinetics of bacteriophage therapy in treatment of subclinical Staphylococcus aureus mastitis in lactating dairy cattle. Antimicrob Agents Chemother. 50(9):2912–2918.
  • Godden SM, Royster E, Timmerman J, Rapnicki P, Green H. 2017. Evaluation of an automated milk leukocyte differential test and the California mastitis test for detecting intramammary infection in early- and late-lactation quarters and cows. J Dairy Sci. 100(8):6527–6544.
  • Godoy-Santos F, Pinto MS, Barbosa AA, Brito MA, Mantovani HC. 2019. Efficacy of a ruminal bacteriocin against pure and mixed cultures of bovine mastitis pathogens. Indian J Microbiol. 59(3):304–312.
  • Golder HM, Hodge A, Lean IJ. 2016. Effects of antibiotic dry-cow therapy and internal teat sealant on milk somatic cell counts and clinical and subclinical mastitis in early lactation. J Dairy Sci. 99(9):7370–7380.
  • Gomes F, Henriques M. 2016. Control of bovine mastitis: old and recent therapeutic approaches. Curr Microbiol. 72(4):377–382.
  • González RN, Wilson DJ. 2003. Mycoplasmal mastitis in dairy herds. Vet Clin North Am Food Anim Pract. 19(1):199–221.
  • Gröhn YT, Wilson DJ, González RN, Hertl JA, Schulte H, Bennett G, Schukken YH. 2004. Effect of pathogen-specific clinical mastitis on milk yield in dairy cows. J Dairy Sci. 87(10):3358–3374.
  • Gruet P, Maincent P, Berthelot X, Kaltsatos V. 2001. Bovine mastitis and intramammary drug delivery: review and perspectives. Adv Drug Deliv Rev. 50(3):245–259.
  • Gu B, Miao J, Fa Y, Lu J, Zou S. 2010. Retinoic acid attenuates lipopolysaccharide-induced inflammatory responses by suppressing TLR4/NF-kappaB expression in rat mammary tissue. Int Immunopharmacol. 10(7):799–805.
  • Gurao A, Kashyap SK, Singh R. 2017. β-defensins: An innate defense for bovine mastitis. Vet World. 10(8):990–998.
  • Gurjar A, Gioia G, Schukken Y, Welcome F, Zadoks R, Moroni P. 2012. Molecular diagnostics applied to mastitis problems on dairy farms. Vet Clin North Am Food Anim Pract. 28(3):565–576.
  • Gussmann M, Steeneveld W, Kirkeby C, Hogeveen H, Farre M, Halasa T. 2019. Economic and epidemiological impact of different intervention strategies for subclinical and clinical mastitis. Prev Vet Med. 166:78–85.
  • Hadrich JC, Wolf CA, Lombard J, Dolak TM. 2018. Estimating milk yield and value losses from increased somatic cell count on US dairy farms. J Dairy Sci. 101(4):3588–3596.
  • Haine D, Dohoo I, Scholl D, Dufour S. 2018. Diagnosing intramammary infection: controlling misclassification bias in longitudinal udder health studies. Prev Vet Med. 150:162–167.
  • Halasa T. 2012. Bioeconomic modeling of intervention against clinical mastitis caused by contagious pathogens. J Dairy Sci. 95(10):5740–5749.
  • Halasa T, Huijps K, Østerås O, Hogeveen H. 2007. Economic effects of bovine mastitis and mastitis management: a review. Vet Q. 29(1):18–31.
  • Halasa T, Nielen M, Whist AC, Østerås O. 2009. Meta-analysis of dry cow management for dairy cattle. Part 2. Cure of existing intramammary infections. J Dairy Sci. 92(7):3150–3157.
  • Hamann J. 2001. Mastitis notes from members countries. Germany Bullt IDF367: 18-21.IDF (1987) Bovine mastitis. Definition and guidelines for diagnosis. Bull IDF. 211:24.
  • Haq IU, Chaudhry WN, Akhtar MN, Andleeb S, Qadri I. 2012. Bacteriophages and their implications on future biotechnology: a review. Virol J. 9(1):9. https://doi.org/https://doi.org/10.1186/1743-422X-9-9.
  • Harjanti DW, Ciptaningtyas R, Wahyono F, Setiatin ET. 2018. Isolation and identification of bacterial pathogen from mastitis milk in Central Java Indonesia. International Symposium on Food and Agro-Biodiversity (ISFA). IOP Conf Ser: Earth Environ Sci. 102:012076.
  • Hase P, Digraskar S, Ravikanth K, Dandale M, Maini S. 2013. Management of subclinical mastitis with mastilep gel and herbal spray (AV/AMS/15). Int JPharm Pharmacol. 2:64–67.
  • Heinrichs AJ, Costello SS, Jones CM. 2009. Control of heifer mastitis by nutrition. Vet Microbiol. 134(1/2):172–176.
  • Hemingway RG. 1999. The influences of dietary selenium and vitamin E intakes on milk somatic cell counts and mastitis in cows. Vet Res Commun. 23(8):481–499.
  • Hogan JS, Weiss WP, Smith KL. 1993. Role of vitamin E and selenium in host defense against mastitis. J Dairy Sci. 76(9):2795–2803.
  • Hogeveen H, Pyorala S, Waller KP, Hogan JS, Lam TJGM, Oliver SP, Schukken YH, Barkema HW, Hillerton EJ. 2011. Current status and future challenges in mastitis research. In Proceedings of the 50th Annual Meeting of the National Mastitis Council, 23-26 January, 2011, Arlington, USA (pp. 36–48).
  • Hozyen HF, Ibrahim ES, Khairy EA, El-Dek SI. 2019. Enhanced antibacterial activity of capped zinc oxide nanoparticles: A step towards the control of clinical bovine mastitis. Vet World. 12(8):1225–1232.
  • Hu X, Li S, Fu Y, Zhang N. 2019. Targeting gut microbiota as a possible therapy for mastitis. Eur J Clin Microbiol Infect Dis. 38(8):1409–1423.
  • Hussein HA, Abd El-Razik KA, Gomaa AM, Elbayoumy MK, Abdelrahman KA, Hosein HI. 2018. Milk amyloid A as a biomarker for diagnosis of subclinical mastitis in cattle. Vet World. 11(1):34–41.
  • Izquierdo AC, Guerra Liera JE, Cervantes RE, Inzunza Castro JF, Abel Villa Mancera E, Crispín RH, Juárez Mosqueda ML, Vázquez AG, Pérez JO, Aparicio PS, et al. 2017. Production of milk and bovine mastitis. J Adv Dairy Res. 5(2):2.
  • Jenkins SN, Okello E, Rossitto PV, Lehenbauer TW, Champagne J, Penedo MC, Arruda AG, Godden S, Rapnicki P, Gorden PJ, et al. 2019. Molecular epidemiology of coagulase-negative Staphylococcus species isolated at different lactation stages from dairy cattle in the United States. Peer J. 7:e6749.
  • Kabera F, Dufour S, Keefe G, Roy JP. 2018. An observational cohort study on persistency of internal teat sealant residues in milk after calving in dairy cows. J Dairy Sci. 101(7):6399–6412.
  • Kalinska A, Gołębiewski M, Wójcik A. 2017. Mastitis pathogens in dairy cattle – a review. World Sci News. 89:22–31.
  • Kalińska A, Jaworski S, Wierzbicki M, Gołębiewski M. 2019. Silver and copper nanoparticles—an alternative in future mastitis treatment and prevention? Int J Mol Sci. 20(7):1672. doi: https://doi.org/10.3390/ijms20071672.
  • Kalmus P, Simojoki H, Pyörälä S, Taponen S, Holopainen J, Orro T. 2013. Milk haptoglobin, milk amyloid A, and N-acetyl-β-D-glucosaminidase activity in bovines with naturally occurring clinical mastitis diagnosed with a quantitative PCR test. J Dairy Sci. 96(6):3662–3670.
  • Kandeel SA, Morin DE, Calloway CD, Constable PD. 2018. Association of California mastitis test scores with intramammary infection status in lactating dairy cows admitted to a veterinary teaching hospital. J Vet Intern Med. 32(1):497–505.
  • Kazemi J, Ahmadi M, Saei HD, Adib Hesami M. 2014. Antibacterial effect of silver nanoparticles along with protein synthesis-inhibiting antibiotics on Staphylococcus aureus isolated from cattle mastitis. Biol J Microorg. 2(8):15–22.
  • Keefe G. 2012. Update on control of Staphylococcus aureus and Streptococcus agalactiae for management of mastitis. Vet Clin North Am Food Anim Pract. 28(2):203–216.
  • Keller D, Sundrum A. 2018. Comparative effectiveness of individualised homeopathy and antibiotics in the treatment of bovine clinical mastitis: randomised controlled trial. Vet Rec. 182(14):407. doi: https://doi.org/10.1136/vr.104555.
  • Kher MN, Sheth NR, Bhatt VD. 2019. In vitro antibacterial evaluation of Terminalia chebula as an alternative of antibiotics against bovine subclinical mastitis. Anim Biotechnol. 30(2):151–158.
  • Klaas IC, Zadoks RN. 2018. An update on environmental mastitis: challenging perceptions. Transbound Emerg Dis. 65(1):166–185.
  • Kleczkowski M, Kluciński W, Czerski M, Kudyba E. 2017. Association between acute phase response, oxidative status and mastitis in cows. Veterinarska Stanica. 48(3):177–186.
  • Kovačić M, Samardžija M, Đuričić D, Vince S, Flegar-Meštrić Z, Perkov S, Gračner D, Turk R. 2019. Paraoxonase-1 activity and lipid profile in dairy cows with subclinical and clinical mastitis. J Appl Animal Res. 47(1):1–4.
  • Kromker V, Friedrich J. 2009. Teat canal closure in non-lactating heifers and its association with udder health in the consecutive lactation. Vet Microbiol. 134(1-2):100–105.
  • Krömker V, Leimbach S. 2017. Mastitis treatment—reduction in antibiotic usage in dairy cows. Reprod Dom Anim. 52:21–29.
  • Krukowski H, Tietze M, Majewski T, Rózański P. 2001. Survey of yeast mastitis in dairy herds of small-type farms in the Lublin region, Poland. Poland Mycopathologia. 150(1):5–7.
  • Krut O, Bekeredjian-Ding I. 2018. Contribution of the immune response to phage therapy. J Immunol. 200(9):3037–3044.
  • Kumar GSN, Apannavar MM, Surnagi MD, Kotresh AM. 2010. Study on incidence and economics of clinical mastitis. Karnataka J Agric Sci. 23:407–408.
  • Kurjogi M, Issa Mohammad YH, Alghamdi S, Abdelrahman M, Satapute P, Jogaiah S. 2019. Detection and determination of stability of the antibiotic residues in cow's milk. PLoS One. 14(10):e0223475
  • Lacasse P, Lauzon K, Diarra MS, Petitclerc D. 2008. Utilization of lactoferrin to fight antibiotic-resistant mammary gland pathogens. J Anim Sci. 86(13 Suppl):66–71.
  • Lago A, Bruno DR, Lopez-Benavides M, Leibowitz S. 2016. Short communication: Efficacy of glycolic acid-based and iodine-based postmilking barrier teat disinfectants for prevention of new intramammary infections in dairy cattle. J Dairy Sci. 99(9):7467–7472.
  • Langer MN, Blodkamp S, Bayerbach M, Feßler AT, Buhr N, de Gutsmann T, Kreienbrock L, Schwarz S, Köckritz-Blickwede MV. 2017. Testing cathelicidin susceptibility of bacterial mastitis isolates: Technical challenges and data output for clinical isolates. Vet Microbiol. 210:107–115..
  • Laven RA. 2012. Effect of use of teat sealants at drying off on the uptake of colostral antibodies by calves, as estimated by measuring gamma-glutamyl transferase activity. N Z Vet J. 60(1):47–49.
  • Leite RF, Gonçalves JL, Peti APF, Figueiró FS, Moraes LAB, Santos MV. 2018. Antimicrobial activity of crude extracts from actinomycetes against mastitis pathogens. J Dairy Sci. 101(11):10116–10125.
  • Leitner G, Zilberman D, Papirov E, Shefy S. 2018. Assessment of acoustic pulse therapy (APT), a non-antibiotic treatment for dairy cows with clinical and subclinical mastitis. PLoS One. 13(7):e0199195.
  • Leitner G, Pinchasov Y, Morag E, Spanier Y, Jacoby S, Eliau D, Pitcovski J. 2013. Immunotherapy of mastitis. Vet Immunol Immunopathol. 153(3/4):209–216.
  • León-Galván M, Barboza-Corona JE, Lechuga-Arana AA, Valencia-Posadas M, Aguayo DD, Cedillo-Pelaez C, Martínez-Ortega EA, Gutierrez-Chavez AJ. 2015. Molecular detection and sensitivity to antibiotics and bacteriocins of pathogens isolated from bovine mastitis in family dairy herds of central Mexico. BioMed Res Int. 2015:1–9. https://doi.org/http://dx.doi.org/10.1155/2015/615153.
  • Levison LJ, Miller-Cushon EK, Tucker AL, Bergeron R, Leslie KE, Barkema HW, DeVries TJ. 2016. Incidence rate of pathogen-specific clinical mastitis on conventional and organic Canadian dairy farms. J Dairy Sci. 99(2):1341–1350.
  • Lim GH, Kelton DF, Leslie KE, Timms LL, Church C, Dingwell RT. 2007. Herd management factors that affect duration and variation of adherence of an external teat sealant. J Dairy Sci. 90(3):1301–1309.
  • Lima MGB, Blagitz MG, Souza FN, Sanchez EMR, Batista CF, Bertagnon HG, Diniz SA, Silva MX, Della Libera AMMP. 2018b. Profile of immunoglobulins, clinical and bacteriological cure after different treatment routes of clinical bovine mastitis. Arq Bras Med Vet Zootec. 70(4):1141–1149.
  • Lima SF, de Souza Bicalho ML, Bicalho RC. 2018a. Evaluation of milk sample fractions for characterization of milk microbiota from healthy and clinical mastitis cows. PLoS One. 13(3):e0193671.
  • Lind N, Hansson H, Lagerkvist CJ. 2019. Development and validation of a measurement scale for self-efficacy for farmers’ mastitis prevention in dairy cows. Prev Vet Med. 167:53–60.
  • Lisowska-Łysiak K, Dudko P, Kosecka-Strojek M, Walczak J, Wójcik P, Międzobrodzki J. 2018. Characteristics of advanced methods used for typing bacterial isolates from mastitis with particular reference to staphylococci. Polish J Vet Sci. 21(1):229–239.
  • Ma J, Zhu S, Guo Y, Hao M, Chen Y, Wang Y, Yang M, Chen J, Guo M. 2019. Selenium attenuates Staphylococcus aureus mastitis in mice by inhibiting the activation of the NALP3 inflammasome and NF-kappaB/MAPK pathway. Biol Trace Elem Res. 191(1):159–166.
  • Magro G, Rebolini M, Beretta D, Piccinini R. 2018. Methicillin-resistant Staphylococcus aureus CC22-MRSA-IV as an agent of dairy cow intramammary infections. Vet Microbiol. 227:29–33.
  • Malinowski E, Lassa H, Klossowska A, Markiewicz H, Kaczmarowski M, Smulski S. 2006. Relationship between mastitis agents and somatic cell counts in foremilk samples. Bull Vet Inst Pulawy. 50:349–352.
  • Mantovani HC, Kam DK, Ha JK, Russell JB. 2001. The antibacterial activity and sensitivity of Streptococcus bovis strains isolated from the rumen of cattle. FEMS Microbiol Ecol. 37(3):23–229.
  • Martin P, Barkema HW, Brito LF, Narayana SG, Miglior F. 2018. Symposium review: Novel strategies to genetically improve mastitis resistance in dairy cattle. J Dairy Sci. 101(3):2724–2736.
  • Martins CMMR, Pinheiro ESC, Gentilini M, Benavides ML, Santos MV. 2017. Efficacy of a high free iodine barrier teat disinfectant for the prevention of naturally occurring new intramammary infections and clinical mastitis in dairy cows. J Dairy Sci. 100(5):3930–3939.
  • McDougall S, Abbeloos E, Piepers S, Rao AS, Astiz S, van Werven T, Statham J, Pérez-Villalobos N. 2016. Addition of meloxicam to the treatment of clinical mastitis improves subsequent reproductive performance. J Dairy Sci. 99(3):2026–2042.
  • McDougall S, Agnew KE, Cursons R, Hou XX, Compton CR. 2007. Parenteral treatment of clinical mastitis with tylosin base or penethamate hydriodide in dairy cattle. J Dairy Sci. 90(2):779–789.
  • McDougall S, Bryan MA, Tiddy RM. 2009b. Effect of treatment with the nonsteroidal antiinflammatory meloxicam on milk production, somatic cell count, probability of re-treatment, and culling of dairy cows with mild clinical mastitis. J Dairy Sci. 92(9):4421–4431.
  • McDougall S, Clausen L, Hintukainen J, Hunnam J. 2019. Randomized, controlled, superiority study of extended duration of therapy with an intramammary antibiotic for treatment of clinical mastitis. J Dairy Sci. 102(5):4376–4386.
  • McDougall S, Parker KI, Heuer C, Compton CW. 2009a. A review of prevention and control of heifer mastitis via non-antibiotic strategies. Vet Microbiol. 134(1/2):177–185.
  • McDougall S, Parker KI, Weir AM, Compton CW. 2008. Effect of application of an external teat sealant and/or oral treatment with a monensin capsule pre-calving on the prevalence and incidence of subclinical and clinical mastitis in dairy heifers. N Z Vet J. 56(3):120–129.
  • McParland S, Dillon PG, Flynn J, Ryan N, Arkins S, Kennedy A. 2019. Effect of using internal teat sealant with or without antibiotic therapy at dry-off on subsequent somatic cell count and milk production. J Dairy Sci. 102(5):4464–4475.
  • McPhee CS, Anderson KL, Yeatts JL, Mason SE, Barlow BM, Baynes RE. 2011. Milk and plasma disposition of thymol following intramammary administration of a phytoceutical mastitis treatment. J Dairy Sci. 94(4):1738–1743.
  • Meisel R, Brockers S, Heseler K, Degistirici O, Bulle H, Woite C, Stuhlsatz S, Schwippert W, Jäger M, Sorg R, et al. 2011. Human but not murine multipotent mesenchymal stromal cells exhibit broad-spectrum antimicrobial effector function mediated by indoleamine 2,3-dioxygenase. Leukemia. 25(4):648–654.
  • Merriman KE, Poindexter MB, Kweh MF, Santos JEP, Nelson CD. 2017. Intramammary 1,25-dihydroxyvitamin D3 treatment increases expression of host-defense genes in mammary immune cells of lactating dairy cattle. J Steroid Biochem Mol Biol. 173:33–41.
  • Merriman KE, Powell JL, Santos JEP, Nelson CD. 2018. Intramammary 25-hydroxyvitamin D3 treatment modulates innate immune responses to endotoxin-induced mastitis. J Dairy Sci. 101(8):7593–7607.
  • Mohammadsadegh M. 2018. Impact of intramammary tilmicosin infusion as a dry cow therapy. J Vet Pharmacol Ther. 41(1):22–27.
  • Moravej H, Moravej Z, Yazdanparast M, Heiat M, Mirhosseini A, Moosazadeh Moghaddam M, Mirnejad R. 2018. Antimicrobial peptides: features, action, and their resistance mechanisms in bacteria. Microb Drug Resist. 24(6):747–767.
  • Mukherjee R. 2008. Selenium and vitamin E increases polymorphonuclear cell phagocytosis and antioxidant levels during acute mastitis in riverine buffaloes. Vet Res Commun. 32(4):305–313.
  • Muluye RA, Bian Y, Alemu PN. 2014. Anti-inflammatory and antimicrobial effects of heat-clearing Chinese herbs: a current review. J Tradit Complement Med. 4(2):93–98.
  • Mushtaq S, Shah AM, Shah A, Lone SA, Hussain A, Hassan QP, Ali MN. 2018. Bovine mastitis: an appraisal of its alternative herbal cure. Microb Pathog. 114:357–361.
  • Mutze K, Wolter W, Failing K, Kloppert B, Bernhardt H, Zschock M. 2012. The effect of dry cow antibiotic with and without an internal teat sealant on udder health during the first 100 d of lactation: a field study with matched pairs. J Dairy Res. 79(4):477–484.
  • Nabil MISK, Tarik MISK, EL-Khamary A, Semeika M. 2018. A retrospective study of surgical affections of mammary glands in cattle and buffaloes and their management in the field. J Veterin Med Sci. 80(10):1576–1583.
  • Nagasawa Y, Kiku Y, Sugawara K, Hirose A, Kai C, Kitano N, Takahashi T, Nochi T, Aso H, Sawada SI, et al. 2019. Staphylococcus aureus-specific IgA antibody in milk suppresses the multiplication of S. aureus in infected bovine udder. BMC Vet Res. 15(1):286.
  • Naqvi SA, De Buck J, Dufour S, Barkema HW. 2018. Udder health in Canadian dairy heifers during early lactation. J Dairy Sci. 101(4):3233–3247.
  • Ndlela M, Laing MD, Basdew IH. 2016. Biological control of Staphylococcus aureus-induced bovine mastitis in dairy cows using bacteriophages. Proceeding of 6th IDF Mastitis Conference, Nantes, France. p. 7–9. September 2016.
  • Nedić S, Vakanjac S, Samardžija M, Borozan S. 2019. Paraoxonase 1 in bovine milk and blood as marker of subclinical mastitis caused by Staphylococcus aureus. Res Vet Sci. 125:323–332.
  • Nelson CD, Merriman KE, Poindexter MB, Kweh MF, Blakely LP. 2018. Symposium review: Targeting antimicrobial defenses of the udder through an intrinsic cellular pathway. J Dairy Sci. 101(3):2753–2761.
  • Nickerson SC, Owens WE, Boddie RL. 1995. Mastitis in dairy heifers: initial studies on prevalence and control. J Dairy Sci. 78(7):1607–1618.
  • Nirala NR, Harel Y, Lellouche JP, Shtenberg G. 2020. Ultrasensitive haptoglobin biomarker detection based on amplified chemiluminescence of magnetite nanoparticles. J Nanobiotechnology. 18(1):6
  • Oliver SP, Almeida RA, Gillespie BE, Headrick SJ, Dowlen HH, Johnson DL, Lamar KC, Chester ST, Moseley WM. 2004. Extended ceftiofur therapy for treatment of experimentally-induced Streptococcus uberis mastitis in lactating dairy cattle. J Dairy Sci. 87(10):3322–3329.
  • Oliver SP, Murinda SE. 2012. Antimicrobial resistance of mastitis pathogens. Vet Clin North Am Food Anim Pract. 28(2):165–185.
  • Omara ST. 2017. MIC and MBC of honey and gold nanoparticles against methicillin-resistant (MRSA) and vancomycin-resistant (VRSA) coagulase-positive S. aureus isolated from contagious bovine clinical mastitis. J Genet Eng Biotechnol. 15(1):219–230.
  • Orellano MS, Isaac P, Breser ML, Bohl LP, Conesa A, Falcone RD, Porporatto C. 2019. Chitosan nanoparticles enhance the antibacterial activity of the native polymer against bovine mastitis pathogens. Carbohydr Polym. 213:1–9.
  • O'Rourke D. 2009. Nutrition and udder health in dairy cows: a review. Ir Vet J. 62(S4)S15-S20.
  • Oultram JW, Ganda EK, Boulding SC, Bicalho RC, Oikonomou G. 2017. A metataxonomic approach could be considered for cattle clinical mastitis diagnostics. Front Vet Sci. 4:36
  • Park YK, Fox LK, Hancock DD, McMahan W, Park YH. 2012. Prevalence and antibiotic resistance of mastitis pathogens isolated from dairy herds transitioning to organic management. J Vet Sci. 13(1):103–105.
  • Parker KI, Compton CW, Anniss FM, Heuer C, McDougall S. 2008. Quarter-level analysis of subclinical and clinical mastitis in primiparous heifers following the use of a teat sealant or an injectable antibiotic, or both, precalving. J Dairy Sci. 91(1):169–181.
  • Parker KI, Compton C, Anniss FM, Weir A, Heuer C, McDougall S. 2007. Subclinical and clinical mastitis in heifers following the use of a teat sealant precalving. J Dairy Sci. 90(1):207–218.
  • Pellegrino M, Berardo N, Giraudo J, Nader-Macias MEF, Bogni C. 2017. Bovine mastitis prevention: humoral and cellular response of dairy cows inoculated with lactic acid bacteria at the dry-off period. Benef Microbes. 8(4):589–596.
  • Pellegrino MS, Frola ID, Natanael B, Gobelli D, Nader-Macias MEF, Bogni CI. 2019. In vitro characterization of lactic acid bacteria isolated from bovine milk as potential probiotic strains to prevent bovine mastitis. Probiotics Antimicrob Proteins. 11(1):74–84.
  • Peralta OA, Carrasco C, Vieytes C, Tamayo MJ, Muñoz I, Sepulveda S, Tadich T, Duchens M, Melendez P, Mella A, et al. 2020. Safety and efficacy of a mesenchymal stem cell intramammary therapy in dairy cows with experimentally induced Staphylococcus aureus clinical mastitis. Sci Rep. 10(1):2843
  • Petersson-Wolfe CS, Mullarky IK, Jones GM. 2010. Staphylococcus aureus mastitis: cause, detection, and control. Virginia Tech. Blacksburg, VA.
  • Petitclerc D, Lauzon K, Cochu A, Ster C, Diarra MS, Lacasse P. 2007. Efficacy of a lactoferrin-penicillin combination to treat {beta}-lactam-resistant Staphylococcus aureus mastitis. J Dairy Sci. 90(6):2778–2787.
  • Petzl W, Zerbe H, Günther J, Seyfert HM, Hussen J, Schuberth HJ. 2018. Pathogen-specific responses in the bovine udder. Models and immunoprophylactic concepts. Res Vet Sci. 116:55–61.
  • Pieterse R, Todorov SD. 2010. Bacteriocins - exploring alternatives to antibiotics in mastitis treatment. Braz J Microbiol. 41(3):542–562.
  • Pinedo P, Karreman H, Bothe H, Velez J, Risco C. 2013. Efficacy of a botanical preparation for the intramammary treatment of clinical mastitis on an organic dairy farm. Can Vet J. 54(5):479–484.
  • Pinheiro Machado GG, Veleirinho MB, Mazzarino LL, Machado Filho LCP, Maraschin M, Cerri R, Kuhnen S. 2019. Development of propolis nanoparticles for the treatment of bovine mastitis: in vitro studies on antimicrobial and cytotoxic activities. Canadian J Animl Sci.
  • Porter J, Anderson J, Carter J, Donjacour E, Paros M. 2016. In vitro evaluation of a novel bacteriophage cocktail as a preventative for bovine coliform mastitis. J Dairy Sci. 99(3):2053–2062.
  • Pyörälä S. 2009. Treatment of mastitis during lactation. Irish Vet J. 62 (Suppl 4):S40–S44.
  • Qiao J, Kwok L, Zhang J, Gao P, Zheng Y, Guo Z, Hou Q, Huo D, Huang W, Zhang H. 2015. Reduction of Lactobacillus in the milks of cows with subclinical mastitis. Benef Microbes. 6(4):485–490.
  • Rainard P. 2017. Mammary microbiota of dairy ruminants: fact or fiction? Vet Res. 48(1):25
  • Rainard P, Foucras G. 2018. A critical appraisal of probiotics for mastitis control. Front Vet Sci. 5:251
  • Rainard P, Foucras G, Fitzgerald JR, Watts JL, Koop G, Middleton JR. 2018. Knowledge gaps and research priorities in Staphylococcus aureus mastitis control. Transbound Emerg Dis. 65:149–165.
  • Rajala-Schultz P, Grohn Y, McCulloch CE, Guard CL. 1999. Effects of clinical mastitis on milk yield in dairy cows. J Dairy Sci. 82(6):1213–1220.
  • Rambabu K, Sreenu M, Suresh RVK, Rao TSC. 2011. Incidence of udder and teat affections in buffaloes. Tamilnadu J Vet Anim Sci. 7(6):309–311.
  • Ranjith D, Nisha AR, Nair SN, Litty M, Rahman M, Juliet S. 2018. Evaluation of analgesic and anti-inflammatory activity of herbal formulation used for mastitis in animals. Int J Appl Sci Eng. 6(1):37.
  • Ren X, Wang L, Chen W. 2020. Oxytropis glabra DC. Inhibits biofilm formation of Staphylococcus epidermidis by down-regulating ica operon expression. Curr Microbiol. 77(7):1167–1173. https://doi.org/10.1007/s00284-019-01847-w.
  • Ribeiro MG, Risseti RM, Bolaños CA, Caffaro KA, de Morais AC, Lara GH, Zamprogna TO, Paes AC, Listoni FJ, Franco MM. 2015. Trueperella pyogenes multispecies infections in domestic animals: a retrospective study of 144 cases (2002 to 2012). Vet Q. 35(2):82–87.
  • Rossi RS, Amarante AF, Guerra ST, Latosinski GS, Rossi BF, Rall VL, de Figueiredo Pantoja JC. 2019. Efficacy of cefquinome and a combination of cloxacillin and ampicillin for treatment of dairy cows with Streptococcus agalactiae subclinical mastitis. PLoS One. 14(4):e0216091.
  • Ruegg PL. 2009. Management of mastitis on organic and conventional dairy farms. J Anim Sci. 87(13 Suppl):43–55.
  • Ruegg PL. 2017a. A 100-year review: mastitis detection, management, and prevention. J Dairy Sci. 100(12):10381–10397..
  • Ruegg PL. 2017b. Practical approaches to mastitis therapy on large dairy herds. In: Large Dairy Herd Management. American Dairy Science Association, Champaign, IL. p. 933–948.
  • Runciman DJ, Malmo J, Deighton M. 2010. The use of an internal teat sealant in combination with cloxacillin dry cow therapy for the prevention of clinical and subclinical mastitis in seasonal calving dairy cows. J Dairy Sci. 93(10):4582–4591.
  • Sang Y, Blecha F. 2008. Antimicrobial peptides and bacteriocins: alternatives to traditional antibiotics. Anim Health Res Rev. 9(2):227–235.
  • Scaletti RW, Trammell DS, Smith BA, Harmon RJ. 2003. Role of dietary copper in enhancing resistance to Escherichia coli mastitis. J Dairy Sci. 86(4):1240–1249.
  • Seegers H, Fourichon C, Beaudeau F. 2003. Production effects related to mastitis and mastitis economics in dairy cattle herds. Vet Res. 34(5):475–491.
  • Serna-Cock L, Pabon-Rodriguez OV. 2016. Development of a teat bio-sealant and evaluation of its technological and functional properties. Probiotics Antimicrob Proteins. 8(2):111–119.
  • Seroussi E, Blum SE, Krifucks O, Lavon Y, Leitner G. 2018. Application of pancreatic phospholipase A2 for treatment of bovine mastitis. PLoS One. 13(8):e0203132
  • Shah MS, Qureshi S, Kashoo Z, Farooq S, Wani SA, Hussain MI, Banday MS, Khan AA, Gull B, Habib A, et al. 2019. Methicillin resistance genes and in vitro biofilm formation among Staphylococcus aureus isolates from bovine mastitis in India. Comp Immunol Microbiol Infect Dis. 64:117–124.
  • Sharifi S, Pakdel A, Ebrahimie E, Aryan Y, Zefrehee MG, Reecy JM. 2019. Prediction of key regulators and downstream targets of E. coli induced mastitis. J Appl Genet. :1–7.
  • Sharma N, Huynh DL, Kim SW, Ghosh M, Sodhi SS, Singh AK, Kim NE, Lee SJ, Hussain K, Oh SJ, Jeong DK. 2017. A PiggyBac mediated approach for lactoferricin gene transfer in bovine mammary epithelial stem cells for management of bovine mastitis. Oncotarget. 8(61):104272–104285.
  • Sharma N, Jeong DK. 2013. Stem cell research: a novel boulevard towards improved bovine mastitis management. Int J Biol Sci. 9(8):818–829.
  • Sheet OH, Grabowski NT, Klein G, Abdulmawjood A. 2016. Development and validation of a loop mediated isothermal amplification (LAMP) assay for the detection of Staphylococcus aureus in bovine mastitis milk samples. Mol Cell Probes. 30(5):320–325.
  • Shibata Y, Tien LHT, Nomoto R, Osawa R. 2014. Development of a multilocus sequence typing scheme for Streptococcus gallolyticus. Microbiology (Reading)). 160(Pt 1):113–122.
  • Shinozuka Y, Kawai K, Takeda A, Yamada M, Kayasaki F, Kondo N, Sasaki Y, Kanai N, Mukai T, Sawaguchi M, et al. 2019. Influence of oxytetracycline susceptibility as a first-line antibiotic on the clinical outcome in dairy cattle with acute Escherichia coli mastitis. J Vet Med Sci. 81(6):863–868.
  • Shinozuka Y, Morita T, Watanabe A, Kawai K. 2018. Live bacteria in clots from bovine clinical mastitis milk with no growth in conventional culturing. Asian J Anim Vet Adv. 13(2):197–200.
  • Singh M, Rai RB, Dhama K, Saminathan M, Tiwari R, Chakraborty S, Damodaran T, Malik YPS, Singh B. 2013. Bovine mastitis: the disease. Diagnosis, Prevention, Treatment and Control. Izatnagar, Bareilly, India: Indian Veterinary Research Institute. p. 1–44.
  • Singh PJ, Singh KB. 1994. A study of economic losses due to mastitis in India. Indian J Dairy Sci. 47:265–272.
  • Sinha MK, Thombare NN, Mondal B. 2014. Subclinical mastitis in dairy animals: incidence, economics, and predisposing factors. Sci World J. 523984.
  • Skoulikas S, Dufour S, Haine D, Perreault JY, Roy JP. 2018. Early-lactation extended pirlimycin therapy against naturally acquired Staphylococcus aureus intramammary infections in heifers: a randomized controlled trial. J Dairy Sci. 101(2):1419–1427.
  • Skowron K, Sękowska A, Kaczmarek A, Grudlewska K, Budzyńska A, Białucha A, Gospodarek-Komkowska E. 2019. Comparison of the effectiveness of dipping agents on bacteria causing mastitis in cattle. Ann Agric Environ Med. 26(1):39–45.
  • Skurnik M, Pajunen M, Kiljunen S. 2007. Biotechnological challenges of phage therapy. Biotechnol Lett. 29(7):995–1003.
  • Smith KL, Hogan JS, Weiss WP. 1997. Dietary vitamin E and selenium affect mastitis and milk quality. J Anim Sci. 75(6):1659–1665.
  • Sordillo LM. 2011. New concepts in the causes and control of mastitis. J Mammary Gland Biol Neoplasia. 16(4):271–273.
  • Sordillo LM. 2016. Nutritional strategies to optimize dairy cattle immunity. J Dairy Sci. 99(6):4967–4982.
  • Souza RFS, Rault L, Seyffert N, Azevedo V, Le Loir Y, Even S. 2018. Lactobacillus casei BL23 modulates the innate immune response in Staphylococcus aureus-stimulated bovine mammary epithelial cells. Benef Microbes. 9(6):985–995.
  • Steele N, McDougall S. 2014. Effect of prolonged duration therapy of subclinical mastitis in lactating dairy cows using penethamate hydriodide. N Z Vet J. 62(1):38–46.
  • Steeneveld W, van Werven T, Barkema HW, Hogeveen H. 2011. Cow-specific treatment of clinical mastitis: an economic approach. J Dairy Sci. 94(1):174–188.
  • Su Y, Yu CY, Tsai Y, Wang SH, Lee C, Chu C. 2016. Fluoroquinolone-resistant and extended-spectrum β-lactamase-producing Escherichia coli from the milk of cows with clinical mastitis in Southern Taiwan. J Microbiol Immunol Infect. 49(6):892–901.
  • Sun W, Wang Q, Guo Y, Zhao Y, Wang X, Zhang Z, Deng G, Guo M. 2017. Selenium suppresses inflammation by inducing microRNA-146a in Staphylococcus aureus-infected mouse mastitis model. Oncotarget. 8(67):110949–110964.
  • Suojala L, Kaartinen L, Pyörälä S. 2013. Treatment for bovine Escherichia coli mastitis - an evidence-based approach. J Vet Pharmacol Ther. 36(6):521–531.
  • Suresh S, Sankar P, Telang AG, Kesavan M, Sarkar SN. 2018. Nanocurcumin ameliorates Staphylococcus aureus-induced mastitis in mouse by suppressing NF-κB signaling and inflammation . Int Immunopharmacol. 65:408–412.
  • Swarup D, Kumar PN, Singh R. 1989. Evaluation of milk conductivity test in detecting subclinical udder infection. Indian J Anim Sci. 59:1227–1229.
  • Szweda P, Schielmann M, Kotlowski R, Gorczyca G, Zalewska M, Milewski S. 2012. Peptidoglycan hydrolases-potential weapons against Staphylococcus aureus. Appl Microbiol Biotechnol. 96(5):1157–1174.
  • Texasdairymatters.org [Internet]. June, 2009 [cited on 23 August 2020]. Available from: http://agrilife.org/texasdairymatters/files/2012/09/6-09-MastitisImmunotherapy-Updated-01-17-1.pdf
  • Titze I, Lehnherr T, Lehnherr H, Krömker V. 2020. Efficacy of bacteriophages against Staphylococcus aureus isolates from bovine mastitis. Pharmaceuticals (Basel). 13(3):35.
  • Tiwari R, Chakraborty S, Dhama K, Rajagunalan S, Singh SV. 2013. Antibiotic resistance - an emerging health problem: causes, worries, challenges and solutions – a review. Int J Curr Res. 5(07):1880–1892.
  • Tiwari R, Dhama K, Chakraborty S, Kumar A, Rahal A, Kapoor S. 2014. Bacteriophage therapy for safeguarding animal and human health: A review. Pak J Biol Sci. 17(3):301–315.
  • Tiwari R, Latheef SK, Ahmed I, Iqbal HMN, Bule MH, Dhama K, Samad HA, Karthik K, Alagawany M, El-Hack MEA, et al. 2018. Herbal immunomodulators – a remedial panacea for designing and developing effective drugs and medicines: Current scenario and future prospects. Curr Drug Metab. 19(3):264–301.
  • Turk R, Koledić M, Maćešić N, Benić M, Dobranić V, Ðuričić D, Cvetnić LC, Samardžija M. 2017. The role of oxidative stress and inflammatory response in the pathogenesis of mastitis in dairy cows. Mljekarstvo. 67(2):91–101.
  • Turk R, Piras C, Kovačić M, Samardžija M, Ahmed H, De Canio M, Urbani A, Meštrić ZF, Soggiu A, Bonizzi L, et al. 2012. Proteomics of inflammatory and oxidative stress response in cows with subclinical and clinical mastitis. J Proteomics. 75(14):4412–4428.
  • Vakkamäki J, Taponen S, Heikkilä AM, Pyörälä S. 2017. Bacteriological etiology and treatment of mastitis in Finnish dairy herds. Acta Vet Scand. 59(1):33
  • van den Borne BH, Halasa T, van Schaik G, Hogeveen H, Nielen M. 2010. Bioeconomic modeling of lactational antimicrobial treatment of new bovine subclinical intramammary infections caused by contagious pathogens. J Dairy Sci. 93(9):4034–4044.
  • van den Borne BHP, van Schaik G, Lam TJGM, Nielen M, Frankena K. 2019. Intramammary antimicrobial treatment of subclinical mastitis and cow performance later in lactation. J Dairy Sci. 102(5):4441–4451.
  • Varela-Ortiz DF, Barboza-Corona JE, González-Marrero J, León-Galván MF, Posadas M, Lechuga-Arana AA, Sánchez-Felipe CG, Ledezma-García F, Gutiérrez-Chávez AJ. 2018. Antibiotic susceptibility of Staphylococcus aureus isolated from subclinical bovine mastitis cases and in vitro efficacy of bacteriophage. Vet Res Commun. 42(3):243–250.
  • Vasquez AK, Nydam DV, Foditsch C, Wieland M, Lynch R, Eicker S, Virkler PD. 2018. Use of a culture-independent on-farm algorithm to guide the use of selective dry-cow antibiotic therapy. J Dairy Sci. 101(6):5345–5361.
  • Vyas S, Shukla V, Doshi N. 2019. FMD and mastitis disease detection in cows using Internet of Things (IOT). Procedia Comput Sci. 160:728–733.
  • Wald R, Hess C, Urbantke V, Wittek T, Baumgartner M. 2019. Characterization of Staphylococcus species isolated from bovine quarter milk samples. Animals (Basel.) 9(5):200.doi: https://doi.org/10.3390/ani9050200.
  • Wallis JK, Kromker V, Paduch JH. 2018. Biofilm formation and adhesion to bovine udder epithelium of potentially probiotic lactic acid bacteria. AIMS Microbiol. 4(2):209–224.
  • Wang H, Bi C, Wang Y, Sun J, Meng X, Li J. 2018. Selenium ameliorates Staphylococcus aureus-induced inflammation in bovine mammary epithelial cells by inhibiting activation of TLR2, NF-κB and MAPK signaling pathways . BMC Vet Res. 14(1):197
  • Wang L-H, Li X-Y, Jin L-J, You J-S, Zhou Y, Li S-Y, Xu Y-P. 2011. Characterization of chicken egg yolk immunoglobulins (IgYs) specific for the most prevalent capsular serotypes of mastitis-causing Staphylococcus aureus. Vet Microbiol. 149(3/4):415–421..
  • Wang X, Teng D, Wang X, Hao Y, Chen H, Mao R, Wang J. 2019. Internalization, distribution, and activity of peptide H2 against the intracellular multidrug-resistant bovine mastitis-causing bacterium Staphylococcus aureus. Sci Rep. 9(1):7968.
  • Wawron W, Bochniarz M, Piech T. 2010. Yeast mastitis in dairy cows in the middle-eastern part of Poland. Bull Vet Inst Pulawy. 54:201–204.
  • Weigel KA, Shook GE. 2018. Genetic selection for mastitis resistance. Vet Clinics: Food Anim Pract. 34(3):457–472.
  • Werner C, Sobiraj A, Sundrum A. 2010. Efficacy of homeopathic and antibiotic treatment strategies in cases of mild and moderate bovine clinical mastitis. J Dairy Res. 77(4):460–467.
  • Wernicki A, Puchalski A, Urban-Chmiel R, Dec M, Stegierska D, Dudzic A, Wojcik A. 2014. Antimicrobial properties of gold, silver, copper and platinum nanoparticles against selected microorganisms isolated from cases of mastitis in cattle. Med Weter. 70(9):564–657.
  • Williamson JH, Lacy-Hulbert SJ. 2014. Lack of efficacy of homeopathic therapy against post-calving clinical mastitis in dairy herds in the Waikato region of New Zealand. N Z Vet J. 62(1):8–14.
  • Wilson DJ, Gonzalez RN, Das HH. 1997. Bovine mastitis pathogens in New York and Pennsylvania: prevalence and effects on somatic cell count and milk production. J Dairy Sci. 80(10):2592–2598.
  • Wilson DJ, Mallard BA, Burton JL, Schukken YH, Grohn YT. 2009. Association of Escherichia coli J5-specific serum antibody responses with clinical mastitis outcome for J5 vaccinate and control dairy cattle. Clin Vaccine Immunol. 16(2):209–217.
  • Wu J, Hu S, Cao L. 2007. Therapeutic effect of nisin Z on subclinical mastitis in lactating cows. Antimicrob Agents Chemother. 51(9):3131–3135.
  • Xu W, Guan R, Lu Y, Su X, Xu Y, Du A, Hu S. 2015. Therapeutic effect of polysaccharide fraction of Atractylodis macrocephalae Koidz in bovine subclinical mastitis. BMC Vet Res. 11:165.
  • Yadav RK, Singh M, Roy S, Gautam S, Rawat JK, Singh L, Ansari MN, Saeedan AS, Kaithwas G. 2020. Short communication: Evaluation of α-linolenic acid-based intramammary nanosuspension for treatment of subclinical mastitis. J Dairy Sci. 103(3):2701–2706.
  • Yang WT, Ke CY, Wu WT, Lee RP, Tseng YH. 2019. Effective treatment of bovine mastitis with intramammary infusion of Angelica dahurica and Rheum officinale extracts. Evid Based Complement Alternat Med. 2019:7242705.
  • Yang X, Ouyang W, Sun J, Li X. 2009. Post-antibiotic effect of amoxicillin nanoparticles against main pathogenic bacteria of bovine mastitis in vitro. J Northwest A & F Univ–Nat Sci Ed. 37(6):1–6.
  • Young-Speirs M, Drouin D, Cavalcante PA, Barkema HW, Cobo ER. 2018. Host defense cathelicidins in cattle: types, production, bioactive functions and potential therapeutic and diagnostic applications. Int J Antimicrob Agents. 51(6):813–821..
  • Younus M, AhmadT Sharif A, Bilal MQ, Nadeem M, Ashfaq K. 2018. Comparative therapeutic efficacy of homeopathic complex, herbal extract and antibiotic in the treatment of subclinical mastitis in dairy buffaloes. Buffalo Bull. 37(2):221–234.
  • Yu J, Ren Y, Xi X, Huang W, Zhang H. 2017. A Novel lactobacilli-based teat disinfectant for improving bacterial communities in the milks of cow teats with subclinical mastitis. Front Microbiol. 8:1782
  • Yu L, Shang F, Chen X, Ni J, Yu L, Zhang M, Sun D, Xue T. 2018. The anti-biofilm effect of silver-nanoparticle-decorated quercetin nanoparticles on a multi-drug resistant Escherichia coli strain isolated from a dairy cow with mastitis. Peer J. 6:e5711
  • Yuan Y, Lin S, Guo N, Zhao C, Shen S, Bu X, Ye H. 2014. Marrow mesenchymal stromal cells reduce methicillin-resistant Staphylococcus aureus infection in rat models. Cytotherapy. 16(1):56–63.
  • Zadoks RN, Middleton JR, McDougall S, Katholm J, Schukken YH. 2011. Molecular epidemiology of mastitis pathogens of dairy cattle and comparative relevance to humans. J Mammary Gland Biol Neoplasia. 16(4):357–372.
  • Zecconi A, Piccinini R, Fiorina S, Cabrini L, Daprà V, Amadori M. 2009. Evaluation of interleukin-2 treatment for prevention of intramammary infections in cows after calving. Comp Immunol Microbiol Infect Dis. 32(5):439–451.
  • Zecconi A, Sesana G, Vairani D, Cipolla M, Rizzi N, Zanini L. 2019. Somatic cell count as a decision tool for selective dry cow therapy in Italy. Italian J Anim Sci. 18(1):435–440.
  • Zhang L, Gao J, Barkema HW, Ali T, Liu G, Deng Y, Naushad S, Kastelic JP, Han B. 2018b. Virulence gene profiles: alpha-hemolysin and clonal diversity in Staphylococcus aureus isolates from bovine clinical mastitis in China. BMC Vet Res. 14(1):63
  • Zhen Y-H, Jin L-J, Guo J, Li X-Y, Lu Y-N, Chen J, Xu Y-P. 2008. Characterization of specific egg yolk immunoglobulin (IgY) against mastitis-causing Escherichia coli. Vet Microbiol. 130(1/2):126–133..
  • Zhu L, Cao X, Xu Q, Su J, Li X, Zhou W. 2018. Evaluation of the antibacterial activity of tilmicosin-SLN against Streptococcus agalactiae: in vitro and in vivo studies. Int J Nanomedicine. 13:4747–4755.