NE1748: Mastitis Resistance to Enhance Dairy Food Safety

(Multistate Research Project)

Status: Inactive/Terminating

SAES-422 Reports

Annual/Termination Reports:

[11/09/2017] [10/25/2018] [01/02/2020] [01/22/2021] [01/24/2022]

Date of Annual Report: 11/09/2017

Report Information

Annual Meeting Dates: 11/01/2017 - 11/03/2017
Period the Report Covers: 11/11/2016 - 11/03/2017

Participants

Brief Summary of Minutes

Accomplishments

<p><strong><span style="text-decoration: underline;">Accomplishments:</span></strong></p><br /> <p>Over the past 5-years of this project, we have studied three areas focused on identifying new means of detecting, preventing, and treating bovine mastitis.&nbsp; Our specific objectives were: (i) characterization of host mechanisms associated with mastitis susceptibility and resistance, (ii) characterization and manipulation of virulence factors of mastitis pathogens for enhancing host defense, and (iii) assessment and application of new technologies that advance mastitis control, milk quality and dairy food safety. Both joint research trials and individual studies have been conducted by NE-1048 and accomplishments (listed by objective) include:</p><br /> <p><strong>Objective 1: </strong>Characterization of host mechanisms associated with mastitis susceptibility and resistance.</p><br /> <p>&nbsp;</p><br /> <p>Achievements include the dietary supplementation of OmniGen &reg; (GA), 2,4-thiazolidinedione (OR), retinol-binding protein (RBP; ID) and vitamin E (MD) to improve the host immune response during mastitis. Other major achievements include the negative relationship between severity of negative energy balance and fat mobilization on important inflammatory mediators (MI), the ability of white blood cells to kill invading microorganisms (WA), the negative impact of antimicrobial resistance on the host immune response (NY), the response of peripheral tissues during mastitis (MD, OR), characterizing the nutrient utilization by leukocytes during mastitis (MD), identification of dermal fibroblasts as a model cell to investigate genetic and epigenetic differences between cows in their innate responses to mastitis causing pathogens (VT), and that CXCR1 may be a promising new candidate gene for mastitis susceptibility (TN). Whole genome SNP association studies also have led to a series of new candidate genes that will be evaluated (TN).</p><br /> <p>&nbsp;</p><br /> <p><strong>Objective 2:</strong> Characterization and manipulation of virulence factors of mastitis pathogens for enhancing host defenses.</p><br /> <p>&nbsp;</p><br /> <p>Major achievements for this objective are 1) the identification of iron-sulfur cluster metabolism as a virulence factor associated with <em>S. aureus </em>(NJ); 2) <em>S. uberis</em> adhesion molecule (SUAM) is a relevant virulence factor (TN) and 3) certain genes are involved with enhancing antimicrobial resistance of mastitis causing pathogens such as Klebsiella spp. (Quebec), <em>Escherichia coli</em> (<em>E. coli</em>; NY) and <em>Staphylococcus aureus</em> (<em>S. aureus</em>; NY).</p><br /> <p>&nbsp;</p><br /> <p><strong>Objective 3:</strong> Assessment and application of new technologies that advance mastitis control, milk quality, and dairy food safety.</p><br /> <p>&nbsp;</p><br /> <p>Achievements include controlling mastitis via the use of ultrasound scanning to monitor mastitis (CT), the use of an Automated Milk Leukocyte Differential (MLD) Test for detecting IMI (MN), teat dip efficiency trials to reduce the incidence of mastitis (WA),&nbsp; the development of multiple decision support tools aimed at improving milk quality, reducing mastitis and economics (KT), examining alternative therapeutics for the prevention or treatment of mastitis to reduce antibiotic usage (MO, MD, MA, Quebec), continuing outreach efforts to promote better stewardship of antibiotic use on dairy farms (MI, MO, MN, WI) and improving animal welfare via the development of behavioral monitors (KY, VA, MA).</p><br /> <p>In summary, the current and past five years work conducted within the framework of the NE-1048 (now NE-1748) has resulted in over 150 refereed publications and over 300 presentations at various scientific and stake-holder forums. International visitors and collaborators are often included in these presentations. In addition to the mastitis research workers conference, the NE-1048 members provide new management strategies to reduce antibiotic usage and technology transfer to the scientific community and industry stakeholders. In the last 4 years, members of the project have collectively published multiple book chapters, in excess of 192 peer-reviewed journal articles, over 300 abstracts and proceedings, and presented numerous oral and poster presentations related to mastitis, milk quality, and food safety. Venues for oral and poster presentations have included the National Mastitis Council regional and annual meetings (attendees include researchers, veterinarians, dairy producers, and representatives from industry), Conference for Research Workers in Animal Diseases, American Association of Bovine Practitioners annual meetings, International Dairy Federation meetings, American Dairy Science Association meetings, World Buiatrics Congress meetings, American Society of Microbiology meetings, Conference on Production Diseases in Farm Animals, Plant and Animal Genome Conference, Agriculture and Agri-Food Canada - Food Safety meetings, American College of Veterinary Internal Medicine annual forum meetings, and several regional extension and veterinary continuing education meetings.</p><br /> <p>We are continuing to build on our past findings to reduce the incidence of mastitis through additional research and extension activities. Mastitis is clearly a multi-faceted disease that will require continued efforts to not only ensure the production of safe, high quality food, but to do so in a sustainable fashion and with continued improvements in dairy animal welfare and reductions the use of antimicrobial drugs.</p>

Publications

<p><strong>PUBLICATION LIST:</strong> use as much space as necessary to complete the publication list below.</p><br /> <p><strong>Peer-Reviewed Literature</strong></p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Skoulikas, S., S. Dufour, D. Haine, J. Y. Perreault, and J. P. Roy. Accepted. Early lactation extended pirlimycin therapy against naturally acquired Staphyloccoccus aureus intramammary infections in heifers: A randomized controlled trial. Journal of Dairy Science.</p><br /> <p>Gaudreau, A. M., J. Labrie, C. Goetz, S. Dufour, and M. Jacques. Accepted. Evaluation of MALDI-TOF MS for the identification of bacteria growing as biofilms. Journal of Microbiological Methods.</p><br /> <p>Haine, D., D. T. Scholl, I. R. Dohoo, and S. Dufour. Accepted. Diagnosing Intramammary Infection: Controlling Misclassification Bias in Longitudinal Udder Health Studies. Preventive Veterinary Medicine.</p><br /> <p>Francoz, D., V. Wellemans, J. P. Dupr&eacute;, J. P. Roy, F. Labelle, P. Lacasse, and S. Dufour. 2017. Invited review: A systematic review and qualitative analysis of treatments other than conventional antimicrobials for clinical mastitis in dairy cows. J Dairy Sci.</p><br /> <p>Francoz, D., V. Welllemans, J. P. Roy, P. Lacasse, A. Ordonez-Iturriaga, F. Labelle, and S. Dufour. 2017. Non-antibiotic approaches at drying off for treating and preventing intramammary infections: A protocol for a systematic review and meta-analysis. Animal Health Research Reviews:169-175.</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Troendle, J., L. W. Tauer, Y. T. Gr&ouml;hn. 2017. Optimally achieving milk bulk tank somatic cell count thresholds. J. Dairy Sci. 100:731-738.</p><br /> <p>Kaniyamattam, K., A. De Vries, L. W. Tauer, and Y. T. Gr&ouml;hn. 2017. Genetic, technical and financial performance of dairy herds achieving optimal milk bulk tank somatic cell count reductions through various voluntary culling strategies. J. Dairy Sci. (under review).</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Mississippi State University</span>:</p><br /> <p>Stone, A.E., B.A. Wadsworth, C.A. Becker, J.M. Bewley.&nbsp; 2017. Influence of breed, milk yield, and temperature humidity index on dairy cow lying time, neck activity, reticulorumen temperature, and rumination behavior.&nbsp; J. Dairy Sci.&nbsp; 100 (3) 2395&ndash;2403.</p><br /> <p>Borchers, M., Y.M. Chang, K. Proudfoot, B.A. Wadsworth, A.E. Stone, J.M. Bewley. 2017. Machine-learning based calving prediction from activity, lying, and ruminating behaviors in dairy cattle.&nbsp; J. Dairy Sci.&nbsp; 100 (7) 5664&ndash;5674.</p><br /> <p>Wadsworth, B.A., A. E. Stone, J. D. Clark, D. L. Ray, and J. M. Bewley.&nbsp; 2017.&nbsp; Characterization of milk yield, lying and rumination behavior, gait, cleanliness, and lesions between two different freestall bases.&nbsp; The Professional Animal Scientist 33 (1):140-149.&nbsp;</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Patel, K. , S. Godden, E. Royster, J. Timmerman, B. Crooker. N. McDonald. 2017. Pilot Study: Impact of using a Culture-Guided Selective Dry Cow Therapy Program Targeting Quarter-Level Treatment on Udder Health and Antibiotic Use. Bov Pract. 51:48-57</p><br /> <p>Godden, S., E. Royster, J. Timmerman, P. Rapnicki and H. Green. 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. 100:6527-6544</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Adkins PRF, Middleton JR, Calcutt MJ, Stewart GC, Fox LK.&nbsp; 2017.&nbsp; Speciation and strain-typing of Staphylococcus agnetis and Staphylococcus hyicus isolated from bovine milk using a novel multiplex PCR and pulsed-field gel electrophoresis.&nbsp; J Clin Microbiol.&nbsp; 55(6):1778-1788.&nbsp; doi: 10.1128/JCM.02239-16. [Epub ahead of print].</p><br /> <p>Condas LAZ, De Buck J, Nobrega DB, Carson DA, Roy JP, Keefe GP, DeVries T, Middleton JR, Dufour S, Barkema HW.&nbsp; 2017.&nbsp; Distribution of non-aureus staphylococci in quarters with low and high somatic cell counts and clinical mastitis.&nbsp; J Dairy Sci.&nbsp; 100(7):5613-5627.&nbsp; doi: 10.3168/jds.2016-12479. [Epub ahead of print 27 Apr 2017]</p><br /> <p>Condas LAZ, De Buck J, Nobrega DB, Carson DA, Naushad S, DeVliegher S, Zadoks RN, Middleton JR, Dufour S, Kastelic J, and Barkema HW.&nbsp; 2017.&nbsp; Prevalence of non-aureus staphylococci isolated from intramammary infection in Canadian dairy herds.&nbsp; J Dairy Sci.&nbsp; 100(7):5592-5612.&nbsp; doi: 10.3168/jds.2016-12478. [Epub ahead of print 17 May 2017]</p><br /> <p>Middleton JR, Fox LK.&nbsp; 2017.&nbsp; Contagious Mastitis: Staphylococcus aureus, Streptococcus agalactiae, and Mycoplasma spp.&nbsp; Large Dairy Herd Management eBook published by J Dairy Sci.&nbsp; Invited Review.</p><br /> <p>Rainard P, Foucras G, Fitzgerald JR, Watts JL Koop G, Middleton JR.&nbsp; 2017.&nbsp; Knowledge gaps and research priorities in Staphylococcus aureus mastitis control.&nbsp; Transbound Emerg Dis. Oct 6. doi: 10.1111/tbed.12698. [Epub ahead of print]&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>M.A. Crookenden, C.G. Walker, A. Heiser, A. Murray, V.S.R. Dukkipati, J.K. Kay, S. Meier, &nbsp;K.M. Moyes, M.D. Mitchell, J.J. Loor and J. R. Roche.&nbsp; 2017. Effects of precalving body condition and prepartum feeding level on gene expression in circulating neutrophils.&nbsp; J. Dairy Sci. 100:2310-2322.</p><br /> <p>&nbsp;</p><br /> <p>A.M. Abdeltty, M.E. Iwaniuk, M.Garcia, K.M. Moyes, B.B. Teter, P.&nbsp; Delmonte, A.K.G. Kadegowda, M.A. Tony, F.F. Mohamed and R.A. Erdman.&nbsp; 2017.&nbsp; Effect&nbsp; of short-term feed restriction on milk components and mammary lipogenic gene expression in mid-lactation Holstein dairy cows by extraction of total RNA from milk fat. J. Dairy Sci. 100:4000-4013.</p><br /> <p>&nbsp;</p><br /> <ol start="2017"><br /> <li>Garcia, Y. Qu, C.M. Scholte#, W. Rhoads, D. Connor and K.M. Moyes. 2017. Regulatory effect of dietary intake of chromium propionate on the response of monocyte-derived macrophages from Holstein cows in mid lactation. J. Dairy Sci. 100:6389-6399.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>K.M. Moyes, L. Ma, D. Johnson, S. Fultz and R.R. Peters.&nbsp; 2017. Estimating and quantifying the production outcomes and lifestyle changes for small&ndash;to medium sized dairy farms regarding the transition from conventional to automatic milking systems in the Northeast region: A case study report. Biomedical Journal of Scientific and Technical research. Accepted.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Kelly EJ, Wilson DJ: Pseudomonas aeruginosa mastitis in two goats associated with an essential oil-based teat dip. J Vet Diagn Invest 28:6:760-762, 2016</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Kayitsinga, J, Schewe RL, Contreras GA, and RJ Erskine. 2017. Antimicrobial treatment of clinical mastitis in the Eastern United States: The influence of dairy farmers&rsquo; mastitis management and treatment behavior and attitudes. J. Dairy Sci. 100:1388-1407.</p><br /> <p>Kuhn, M.J., V. Mavangira, J. C. Gandy, C. Zhang, A. D. Jones, and L. M. Sordillo.2017. Differences in the Oxylipid Profiles of Bovine Milk and Plasma at Different Stages of Lactation J. Agric. Food. Chem. 65:4980-4988.</p><br /> <p>Mavangira V, and L. M. Sordillo. 2017. Role of lipid mediators in the regulation of oxidative stress and inflammatory responses in dairy cattle. Res. Vet. Sci. doi: 10.1016/j.rvsc.2017.08.002. [Epub ahead of print].</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Rosa, F. T., J.S. Osorio, F.Y. Rivera, E. Trevisi, C.T. Estill, and M Bionaz. 2017. Role of Peroxisome Proliferator-Activated Receptor gamma on subclinical Mastitis in lactating dairy goats. PPAR Research vol. 2017, Article ID 7097450</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Siebert L, S Headrick, M Lewis, B Gillespie, C Young, L Wojakiewicz, O Kerro Dego, ME Prado, R Almeida, SP Oliver, GM Pighetti. 2017. Genetic variation in CXCR1 haplotypes linked to severity of Streptococcus uberis infection in an experimental challenge model. Vet Immunol Immunopathol. 190: 45-52</p><br /> <p>Pighetti, GM, L Wojakiewicz, SI Headrick, O Kerro Dego, SA Lockwood, HG Kattesh, MJ Lewis, CD Young, *LJ Siebert, BE Gillespie, ME Prado, RA Almeida, SP Oliver. 2017. Vaccination with recombinant Streptococcus uberis adhesion molecule alters immune response to experimental challenge. Int J Vet Dairy Sci. Published online 5/15/2017</p><br /> <p>Oudessa Kerro Dego, R. A. Almeida, Maria E. Prado, S. I. Headrick, M. J. Lewis, C. Young, B. E. Gillespie, L. J. Siebert, G. M. Pighetti, R. D. Abdi, D. B. Ensermu and S. P. Oliver. 2017. Functionally effective immune responses of dairy cows vaccinated with Streptococcus uberis adhesion molecule during the transition period. Annals of Vaccine and Vaccination, Accepted, 9/26/2017.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">State University of New Jersey</span></p><br /> <p>Mashruwala, A. A., Boyd J.M.&nbsp; The Staphylococcus aureus SrrAB regulatory system modulates hydrogen peroxide resistance factors, which imparts protection to aconitase during aerobic growth. PLoS One. 2017 Jan 18;12(1):e0170283. PMID: 28099473</p><br /> <p>Mashruwala A.A., Van de Guchte A., Boyd J.M. Impaired respiration elicits SrrAB-dependent programmed cell lysis and biofilm formation in Staphylococcus aureus. eLife. 2017 Feb 21;6. PMID: 28221135</p><br /> <p>Roberts C., Al-Tameemi H.M., Mashruwala A.A., Rosario-Cruz Z., Chauhan U., Sause, W., Torres V.J., and Boyd J.M. The Suf iron-sulfur cluster biosynthetic system is essential for Staphylococcus aureus viability and defective Fe-S cluster biosynthesis results in broad metabolic defects and decreased survival in neutrophils. Infect Immun. 2017 May 23;85(6). PMID: 28320837</p><br /> <p>Mashruwala A.A., Gries, C.M., Scherr T.D., Kielian, T., Boyd J.M. SaeRS is responsive to the cellular respiratory status and regulates fermentative biofilm formation in Staphylococcus aureus. Infect Immun. 2017 Jun. PMID: 28507069</p><br /> <p>Mashruwala A.A., Boyd J.M. Investigating the role(s) of SufT and the domain of unknown function 59 (DUF59) in the maturation of iron-sulfur proteins. Curr Genet. 2017 Jun. PMID: 28589301</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Washington State University</span></p><br /> <p>Adkins P.R.F, John R Middleton; M.J. Calcutt, G. C. Stewart, and L. K Fox. 2017.&nbsp; Species identification and stratin typing of Staphylococcus agnetis and Staphylococcus hyicus isolates from bovine milk by use of a novel multiplex PCR assay and Pulsed-field Gel Electrophoresis.&nbsp; J. of Clinical Micro. 55:1778-1788.</p><br /> <p>&nbsp;</p><br /> <p>Adkins P.R.F, John R Middleton; Lawrence K Fox. 2016. Staphylococcus aureus Strains Isolated from Cases of Subclinical Bovine Mastitis in the United States.&nbsp; Journal of Clinical Microbiology. 54: 1871-1876</p><br /> <p>&nbsp;</p><br /> <p>Nicholas, R.A.J., L.K. Fox, I. Lysnyansky. 2016.&nbsp; Mycoplasma mastitis in cattle: to cull or not to cull. &nbsp;The vet. J. 216:142-147.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Korkmaz, F.T., and D.E. Kerr. 2017. Genome-wide methylation analysis reveals differentially methylated loci that are associated with an age-dependent increase in bovine fibroblast response to LPS. BMC Genomics. May 25;18(1):405.</p><br /> <p>Carroll, J.A., N.C. Burdick-Sanchez, J.D. Arthingon, C.D. Nelson, A.L. Benjamin, F.T. Korkmaz, D.E. Kerr, and P.A. Lancaster. 2017. In utero exposure to lipopolysaccharide alters the postnatal acute phase response in beef heifers. Innate Immunity 23:97-108.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>Abstracts</strong></p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Kaniyamattam, K., A. De Vries, L. W. Tauer, and Y. T. Gr&ouml;hn. 2017. Impact of culling for SCC, milk revenue and estimated breeding values on herd performance. Proceedings of 2017 American Dairy Science Association annual meeting, Pittsburg, Pennsylvania.</p><br /> <p><span style="text-decoration: underline;">Mississippi State University</span>:</p><br /> <p>Keefer, R.D., J.G. Maples, A.E Stone. The economic feasibility of implementing center pivots on Mississippi dairy farms. 2017. Mississippi Academy of Sciences Summer Symposium. Stoneville, MS.</p><br /> <p>A.N. Rulewicz, K.B. Graves, R.D. Keefer, A.E Stone. Evaluating the effects of Vista Pre-T on feed efficiency in heat-stressed dairy. 2017. Mississippi Academy of Sciences Summer Symposium. Stoneville, MS.</p><br /> <p>B.L. Bowman and A.E Stone. Adoption of on-farm bacteriologic milk culturing:&nbsp; Evaluating farmer perception &amp; impact on management decisions. 2017. Mississippi Academy of Sciences Summer Symposium. Stoneville, MS.</p><br /> <p>Larson, J.E., G.R. Dunham, K.C. Yankey, M.P. Owen, M.M. Steichen, K. McCarty, A.E. Stone, C.O. Lemley. 2017. Additional exercise among grazing dairy cows and effects on uterine blood flow, milk production, or milk quality parameters. Mississippi Academy of Sciences Summer Symposium. Stoneville, MS.</p><br /> <p>Larson, J.E., G.R. Dunham, K.C. Yankey, M.P. Owen, M.M. Steichen, K. McCarty, A.E. Stone, C.O. Lemley. 2017. Additional exercise among grazing dairy cows and effects on uterine blood flow, milk production, or milk quality parameters. American Society of Animal Science Annual Meeting. Baltimore, MD.</p><br /> <p>A.E. Stone, B.W. Jones, L.M. Mayo, I.C. Tsai, Y.M. Chang, and J.M. Bewley. 2017. Evaluation of activity, feeding time, lying time, rumination time, reticulorumen temperature, and milk yield, conductivity, lactose, protein, and fat to detect subclinical mastitis. 2017. Abstract 71194. Dairy Science Association Annual Meeting. Pittsburgh, PA.</p><br /> <p>B.W. Jones, L.M. Mayo, I.C. Tsai, A.E. Stone, Y.M. Chang, and J.M. Bewley. 2017. Detection of lame cattle using behavioral and physiological changes as measured by precision dairy monitoring technologies. Abstract 355. American Dairy Science Association Annual Meeting. Pittsburgh, PA.</p><br /> <p>Mazon, G., J. Guinn, D. Nolan, P. Krawczel, C. Petersson-Wolfe, G. Pighetti, A. Stone, S. Ward, M. Marcondes, J. Bewley. 2017. Relationship between cow cleanliness, locomotion, and bulk tank somatic cell count in southeastern United States dairy farms. Abstract T187. Dairy Science Association Annual Meeting. Pittsburgh, PA.</p><br /> <p>Guinn, J.M., D.T. Nolan, P.D. Krawczel, C.S. Petersson-Wolfe, G.M. Pighetti, A.E. Stone, S.H. Ward, M. Marcondes, J.M. Bewley. 2017. Relationship between cow cleanliness, locomotion, and bulk tank somatic cell count in southeastern United States dairy farms. Abstract T187. Dairy Science Association Annual Meeting. Pittsburgh, PA.</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health and milk quality on dairy farms: preliminary results. American Association of Bovine Practitioners Annual conference, Omaha, NE, USA. September 14-16, 2017</p><br /> <p>Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health and milk quality on dairy farms: preliminary results. Minnesota Dairy Health Conference, Saint Paul, MN, USA. April 11-13, 2017</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Shwani A, Ekesi N, Zaki S, Rubaye AA, Adkins P, Calcutt M, Middleton JR, Rhoads D.&nbsp; 2017.&nbsp; A phylogenomic comparison of Staphylococcus agnetis isolated from dairy cattle mamamary glands and bacterial chondronecrosis with osteomyelitis in broilers.&nbsp; Plant and Animal Genome.&nbsp;</p><br /> <p>Middleton JR.&nbsp; 2017.&nbsp; Mastitis therapy: past successes, current challenges and vision for the future.&nbsp; American Dairy Science Association Annual Meeting. June.&nbsp; Pittsburgh, PA.</p><br /> <p>Arroyo GT, Adkins PRF, Middleton JR.&nbsp; 2017.&nbsp; Comparison of MALDI-TOF and PFGE for strain-typing Staphylococcus aureus isolated from cow&rsquo;s milk.&nbsp; Proceedings of the 40th Annual CVM Research Day (Phi Zeta).&nbsp; 5 May 2017.&nbsp; Abstract #1 &ndash; Veterinary Professional Student Poster Presentations.</p><br /> <p>Bernier Gosselin V, Middleton JR, Zhang M.&nbsp; 2017.&nbsp; Diagnostic performance of a competitive ELISA using frozen-thawed milk for identification of SRLV-infected goats.&nbsp; Proceedings of the 40th Annual CVM Research Day (Phi Zeta).&nbsp; 5 May 2017.&nbsp; Abstract #9 &ndash; 2nd and 3rd Year Resident &amp; Graduate Student Oral Presentations.</p><br /> <p>Bornheim H, Middleton JR, Adkins PRF, Calcutt M.&nbsp; 2017.&nbsp; Epidemiology of Mycoplasma arthritis in goat kids on a commercial goat dairy.&nbsp; Proceedings of the 40th Annual CVM Research Day (Phi Zeta).&nbsp; 5 May 2017.&nbsp; Abstract #6 &ndash; Interns, 1st Year Residents &amp; Graduate Students Oral Presentations.</p><br /> <p>Placheta L, Middleton JR, Bewley J, Borches M, Adkins PRF.&nbsp; 2017.&nbsp; The effect of housing environment on bedding, teat, and raw milk staphylococcal bacteria populations.&nbsp; VRSP Symposium, Bethesda, MD, August 3-4, 2017.</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Wilson DJ, Britten JE, Rood KA: Casein hydrolysate for involution of a single mastitic quarter in dairy cows.&nbsp; J Dy Sci Vol. 100, E-Suppl. 2:143, 2017.</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Tsai C. Y., F. Rosa, M. Bionaz, and P. Rezamand. 2017. The effect of 2,4- thiazolidinedione on lipid soluble vitamins in lactating goats induced with sub-clinical mastitis. Anim. Sci Vol. 95, E-Suppl.</p><br /> <p>Rosa F., Moridi, M., J. Osorio, Lohakare, J. Estill C., and M. Bionaz. Macrophage activation during subclinical mastitis in dairy goats treated with 2,4-thiazolidinedione 2017 ADSA Annual Meeting</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Abdi, R. A., J. M. Vaughn, Barbara E. Gillespie, C. Merrill and O. Kerro Dego. 2016. Antimicrobial Resistance Patterns of major mastitis pathogens. In: Proceedings of Conference of Research workers on Animal Diseases, December 4 &ndash; 6, Chicago, IL.</p><br /> <p>Couture VL, PD Krawczel, GM Pighetti, RA Almeida, SP Oliver. 2017. Relationship between the clinical and behavioural response to mastitis challenge with Streptococcus uberis from Holstein dairy cows. J Dairy Sci 100 (Suppl 2): 29.</p><br /> <p>Pighetti GM. 2017. Genetics, genomics, and improving mastitis resistance. J Dairy Sci 100 (Suppl 2): 188.</p><br /> <p>Siebert L, ME Staton, SP Oliver, GM Pighetti. 2017. Genome wide association analyses identify loci associated with mastitis phenotypes generated from Streptococcus uberis experimental challenge data. J Dairy Sci 100 (Suppl 2): 189.</p><br /> <p>Siebert, L. J.*, M. E. Staton, S. P. Oliver, and G. M. Pighetti.&nbsp; 2016.&nbsp; Genome wide association study identifies loci associated with intramammary infection (IMI) phenotypes following experimental challenge with Streptococcus uberis.&nbsp; Conference Research Workers in Animal Diseases, 097.</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Mugabi, R, D&rsquo;Amico S, Sensabaugh G, Barlow J. 2017. A Multilocus Sequence Typing Scheme Provides Insights into the Diversity of Staphylococcus chromogenes Isolated from Dairy Cattle Skin and Milk Samples. Microbe, American Society of Microbiology Joint Annual Meeting, June 1-5, New Orleans, LA.</p><br /> <p>Ecjstrom, K, Barlow J. 2017. Machine Learning Tools for Predicting Strain Type using&nbsp; MLST Isolate Databases. Microbe, American Society of Microbiology Joint Annual Meeting, June 1-5, New Orleans, LA.</p><br /> <p>&nbsp;</p><br /> <p><strong>Conference Proceedings </strong></p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Haine, D., I. R. Dohoo, D. T. Scholl, H. Stryhn, and S. Dufour. 2017. Sampling strategies to control misclassification bias in longitudinal udder health studies. in Proc. Society for Veterinary Epidemiology and Preventive Medicine 2017 annual meeting, Inverness, UK.</p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Dipastina, A., K. Kaniyamattam, and Y. T. Gr&ouml;hn. 2017. Optimal antimicrobial use for mastitis prevention and treatment on US dairy farms. Proceedings of National Veterinary Scholars Symposium, 2017. NIH. Betheseda, Maryland.</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Pilot Study: Impact of using a Culture-Guided Selective Dry Cow Therapy Program Targeting Quarter-Level Treatment on Udder Health and Antibiotic Use. National Mastitis Council 56th Annual Meeting, St. Pete Beach, FL, USA. January 29-31, 2017.</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Adkins PRF, Middleton JR, Dufour S.&nbsp; 2017.&nbsp; Cross-sectional study of teat and inguinal skin colonization of dairy heifers with various staphylococcal species.&nbsp; Proceedings of the 56th Annual Meeting of the National Mastitis Council, St. Pete Beach, FL.&nbsp; Jan 28-31.&nbsp; pp. 133-134.&nbsp;</p><br /> <p>Bernier Gosselin V, Placheta L, Quas E, Wilshusen J, Limberg E, Adkins PRF, Middleton JR.&nbsp; 2017.&nbsp; Persistence of coagulase negative staphylococcal intramammary infection in dairy goats. Proceedings of the 56th Annual Meeting of the National Mastitis Council, St. Pete Beach, FL.&nbsp; Jan 28-31.&nbsp; pp. 109-110.&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Wilson DJ, Middleton J, Adkins P, Goodell GM: Bovine milk bacterial diagnostic results - blind comparison between conventional culture, MALDI-TOF and 16S rRNA for test agreement. Proc Natl Mast Council:12-17, 2017.</p><br /> <p>Britten J, Wilson D, Rood K: Casein hydrolysate as a possible adjunct or replacement treatment to current antibiotic therapies used at dry-off in dairy cows. Proc Animal, Dairy and Veterinary Sciences Student Research Symposium:5, 2017.</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Erskine, R. J., M. Borek-Stine, and R. Moore. Engaged employees: The connection between protocols and performance. Short course presented at the 56th Annual Mtng National Mastitis Council, St. Petersburg, FL, February, 2017.</p><br /> <p>Martinez, R. O. How training could improve- The employee perspective. Seminar presented at the 1st Annual Michigan Dairy Health Symposium, Lansing, MI, February, 2017.</p><br /> <p>Moore-Foster, R. Do your cows like the milking protocols? Seminar presented at the 1st Annual Michigan Dairy Health Symposium, Lansing, MI, February, 2017.</p><br /> <p>Hovingh, E. P. and R. Moore-Foster. Milking efficiency- As defined by the cow. Workshop presented at the 1st Annual Michigan Dairy Health Symposium, Lansing, MI, February, 2017.</p><br /> <p>Durst, P., S. Moore, and F. San Emeterio. Engaged employees &ndash; What does that mean? Workshop presented at the 1st Annual Michigan Dairy Health Symposium, Lansing, MI, February, 2017.</p><br /> <p>Thomson, R., and R. J. Erskine. Five minute mastitis hot topics- Teat dips, antibiotics, somatic cell counts, selective dry cow therapy, and others&hellip;&hellip; Workshop presented at the 1st Annual Michigan Dairy Health Symposium, Lansing, MI, February, 2017.</p><br /> <p>Erskine, R. J., M. Borek-Stine, and R. Moore-Foster. Engaged employees: The connection between protocols and performance. Short course presented at the 56th Annual Mtng National Mastitis Council, St. Petersburg, FL February, 2017.</p><br /> <p>Erskine, R.J. Employee management to improve milk quality, seminar presented to the Western Canadian Dairy Seminar, Red Deer, AB, March, 2017.</p><br /> <p>Erskine, R.J.* and R. Moore-Foster. Rethinking milking efficiency. Oral presentation at the American Dairy Science Association, Pittsburgh, PA, June, 2017.</p><br /> <p>Moore-Foster, R. and R.J. Erskine*. Making the connection between employees and cows for milking protocols. Oral presentation at the American Diary Association, Pittsburgh, PA, June, 2017.</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Kerro Dego, O., R. D. Abdi, J. Vaughn, C. Merrill, S. M. Cantwell, B. E. Gillespie, R. A. Almeida, S. I. Headrick, G. M. Pighetti, P. Krawczel, J. Keflot, J. M. Bewley and S. P. Oliver. 2017. Antimicrobial Resistance Patterns of&nbsp;&nbsp; Staphylococcus aureus Isolates from Cases of Bovine Mastitis. Proceedings of National Mastitis Council, p 60, 56th meeting January 28 &ndash; 31, 2017, Trade winds Island Grand Resort, St. Pete Beach, Fl.</p><br /> <p>&nbsp;</p><br /> <p><strong>Poster Presentations</strong></p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Mass&eacute;, J., S. Dufour, and M. Archambault. 2016. Characterization of Klebsiella spp. isolates from bovine mammary gland infections. in Proc. Congr&egrave;s de bact&eacute;riologie int&eacute;grative: symbiose &amp; pathogen&egrave;se, Qu&eacute;bec, QC, Canada.</p><br /> <p>Kabera, F., S. Dufour, G. P. Keefe, and J. P. Roy. 2017. Quarter-based selective dry-cow therapy using on-farm diagnostics. in Proc. 2017 Canadian Bovine Mastitis and Milk Quality Research Network Annual Scientific Meeting, Montreal, Canada.</p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Kaniyamattam, K., A. De Vries, L. W. Tauer, and Y. T. Gr&ouml;hn. 2017. Impact of culling for SCC, milk revenue and estimated breeding values on herd performance. Proceedings of 2017 American Dairy Science Association annual meeting, Pittsburg, Pennslyvania.</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health and milk quality on dairy farms: preliminary results. Points of Pride Research Day. College of Veterinary Medicine, University of Minnesota, Saint Paul, MN. October 4, 2017.</p><br /> <p>Pilot Study: Evaluation of the Effect of Selective Dry Cow Therapy on Udder Health. Points of Pride Research Day. College of Veterinary Medicine, University of Minnesota, Saint Paul, MN. October 5, 2016.</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Couture VL, PD Krawczel, GM Pighetti, RA Almeida, SP Oliver. 2017. Relationship between the clinical and behavioural response to mastitis challenge with Streptococcus uberis from Holstein dairy cows. American Dairy Science Association Annual Meeting, Pittsburgh, PA, July 2017.</p><br /> <p>Kerro Dego, O., R. D. Abdi, J. Vaughn, C. Merrill, S. M. Cantwell, B. E. Gillespie, R. A. Almeida, S. I. Headrick, G. M. Pighetti, P. Krawczel, J. Keflot, J. M. Bewley and S. P. Oliver. 2017. Antimicrobial Resistance Patterns of&nbsp;&nbsp; Staphylococcus aureus Isolates from Cases of Bovine Mastitis. Proceedings of National Mastitis Council, p 60, 56th meeting January 28 &ndash; 31, 2017, Trade winds Island Grand Resort, St. Pete Beach, Fl.</p><br /> <p>Merrill C, Reta D. Abdi, Barbara E. Gillespie, Jacqueline Vaughn, and O. Kerro Dego. 2016. Evaluation of the Immune Response and Protection in Dairy Cows Vaccinated with Staphylococcus aureus Surface Proteins. UT Beef and Forage Center Annual Research and Recommendation Meeting, December 13, 2016. Poster presentation.</p><br /> <p>Vaughn J., R. D. Abdi, Barbara E. Gillespie, C. Merrill and O. Kerro Dego. 2016. Clonal diversity and enterotoxin production patterns of S. aureus isolates from cases of bovine mastitis. UT Beef and Forage Center Annual Research and Recommendation Meeting, December 13, 2016. Poster presentation.</p><br /> <p><span style="text-decoration: underline;">State University of New Jersey</span></p><br /> <p>Cerezo, J., Al-Tameemi, H.M., Boyd J.M. Screening the Library of FDA Approved Drugs for Inhibitors of Bacterial Iron-Sulfur Cluster Assembly. Rutgers Microbiology Symposium. Rutgers University, New Brunswick, NJ, 2017.</p><br /> <p>Mashruwala A.A., Earle C., van de Guchte A., Boyd J.M. Regulation of Clp proteases by SrrAB in Staphylococcus aureus. Rutgers Microbiology Symposium. Rutgers University, New Brunswick, NJ, 2017.</p><br /> <p>Mashruwala A.A., van de Guchte A., Boyd J.M. Cellular respiration as a trigger for multicellular behavior in Staphylococcus aureus.&nbsp; Rutgers Microbiology Symposium. Rutgers University, New Brunswick, NJ, 2017.</p><br /> <p>Al-Tameemi H.M., Boyd J.M. Copper Stress in Staphylococcus aureus involves Perturbing Iron Homeostasis. Rutgers Microbiology Symposium. Rutgers University, New Brunswick, NJ, 2017.</p><br /> <p>Purdy M., Mohammed N.*, Crane S., Boyd J.M. Effect of Blue Light on Propionibacterium acnes. Rutgers Microbiology Symposium. Rutgers University, New Brunswick, NJ, 2017.</p><br /> <p>Cerezo J., Al-Tameemi H.M., Boyd J.M. Screening the Library of FDA Approved Drugs for Inhibitors of Bacterial Iron-Sulfur Cluster Assembly. Aresty Research Symposium. Rutgers University, Piscataway, NJ, 2017.</p><br /> <p>Al&shy;Tameemi H.M., Roberts C., Mashruwala A.A., Rosario&shy;Cruz Z., Sause W., Torres V.J., Belden W.J., Boyd J.M. Iron Sulfur Protein Assembly: A viable Target for Antimicrobial Therapy in Staphylococcus aureus American Society of Microbiology General Meeting. New Orleans, LA, 2017.</p><br /> <p>Mashruwala A.A., Gries C.M., Scherr T.D., van de Guchte, A., Kielian T., Boyd J.M. Cellular respiration as a signal for programed cell lysis in Staphylococcus aureus. Gordon Research Conference on Staphylococcal Biology. Waterville Valley, NH, 2017.</p><br /> <p>Rosario-Cruz Z., Eletsky, Nourhan A., Daigham S., Swapna G.V.T., Szyperski T., Montelione G.T., Boyd J.M. NMR Studies of the CopB Protein of the Arginine Catabolic Mobile Element from Staphylococcus aureus and Bacillus subtilis. Center for Advanced Biology and Medicine Meeting. Rutgers University, Piscataway, NJ, 2017.</p>

Impact Statements

  1. Improved animal welfare via the development of behavioral monitors. (Obj 3)
Back to top

Date of Annual Report: 10/25/2018

Report Information

Annual Meeting Dates: 10/24/2018 - 10/26/2018
Period the Report Covers: 11/04/2017 - 10/24/2018

Participants

Brief Summary of Minutes

Accomplishments

<p><strong>NE-1748 ANNUAL REPORT WORKSHEET (FY18)</strong></p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 1:</strong> Characterize host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance to improve economic outcomes and animal welfare.</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Louisiana State University, Ag Center</span></p><br /> <p>Member: William E. Owens</p><br /> <p>Mastitis pathogens continue to be identified from bovine and goat milk samples submitted to this laboratory.&nbsp; Organisms were identified and mastitis reports generated to producers and field agents.&nbsp; Organisms were evaluated for antimicrobial susceptibility to a battery of antibiotics to determine therapeutic efficacy.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Member: Ronald Erskine, Lorraine Sordillo, Andres Contreras</p><br /> <p>Collaborators: Phil Durst, Ruben Martinez, Stan Moore</p><br /> <p>We continued to study the impact of negative energy balance and fat mobilization vascular endothelial and adipose inflammatory responses of in vitro by changing the expression of important inflammatory mediators. In particular, the role of n-3 fatty acid content in altering the profile of vasoactive eicosanoids and the role of poly-unsaturated fatty acids on attenuating endothelial cell inflammatory responses, as well as lymphocyte expression of pro-inflammatory cytokines was a central aim of our work.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>Collaborators: Gerd Bobe, Charles Estill, Shana Jaff, and Matteo Mezzetti and Erminio Trevisi (Universit&agrave; Cattolica del Sacro Cuore, Italy)</p><br /> <p>We performed an experiment to assess the effectiveness of feeding a relatively low amount of Se-fortified hay on improving the condition of Se and the effect on anti-oxidative status, immune condition, and transcriptome of liver and mammary macrophages in lactating primiparous. For the purpose we used 18 heifers (10 Jerseys and 8 Holsteins) that were fed ad libitum with TMR and, from approx. 40 days prior parturition to 2 weeks post-partum, were supplemented with 1 kg of Se-fortified Alfalfa hay (3.25 mg of Se/kg DM)/100 kg of body weight or the same amount of a control Alfalfa hay (0.4 mg of Se/kg DM). We collected blood samples for analysis of the immune status, metabolic and inflammatory profiling, antioxidative status, and level of microminerals, including Se. We collected also liver via biopsy and macrophages from milk somatic cells. On the liver we measured level of Se. In liver and macrophages we are measuring whole transcriptome. Our preliminary data indicated that the treatment was effective in increasing the Se in liver and blood, improving the antioxidative status, and improve milk production in Holsteins only. No effects were observed in the immune status or Se in milk.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Idaho</span></p><br /> <p>Member: Pedram Rezamand</p><br /> <p>Contributors: Massimo Bionaz (Oregan State University), Chia-Yu-Tsai (University of Idaho)</p><br /> <p>We have been collaborating with the Oregon State team on PPARg activator (2,4-thiazolidinedione or TZD) and its potential effect on mastitis. We have developed methods to detect various isomers of vitamin A in serum/milk and how they may response to TZD treatment during induced mastitis.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Member: Kasey M. Moyes</p><br /> <p>Contributors: Daniel Nelson, John Roche, Theodore Elasser</p><br /> <p>In collaboration with Dr. Richard Erdman in the Department of Animal and Avian Sciences (ANSC) at the University of Maryland (UMD), we investigated the use of milk instead of mammary tissue biopsies to investigate the response of animals due to treatment in lactating dairy cattle. International collaborations with Dr. John Roche, Dairy NZ in New Zealand, have led to the identification vof neutrophil function during the transition period. In collaboration with Dr. Theodore Elsasser at the Agricultural Research Service within the USDA, we have investigated the effect of dietary vitamin E on the neutrophil response in lactating dairy cattle.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, B. Crooker, R. Fink, T. Schoenfuss</p><br /> <p>Contributors: L. Fox (WSU), P. Gorden (ISU), D. Nydam (Cornell University), S. Sreevatsan (MSU)</p><br /> <ol start="2018"><br /> <li>Patel, S. Godden, S. Sreevatsan, E. Royster, L. Fox Molecular Epidemiology of Staphylococcus aureus on U.S. Dairy Farms. A prospective cross-sectional multi-state study is being conducted to Describe the prevalence, genotypic diversity, and antimicrobial susceptibility characteristics of Staphylococcus aureus isolates recovered from bulk tank milk from U.S. dairy farms and to identify herd characteristics and management factors associated with risk for presence of multidrug resistant MSSA and of MRSA on U.S. dairy farms. In winter and in summer 2016, bulk tank milk samples were collected from 188 dairy herds from 17 dairy states including. Approximately 150 S. aureus isolates were recovered from bulk tank milk for phenotypic characterization and whole genome sequencing, which was conducted in 2018. Molecular Evolutionary Genetics Analysis (MEGA) software was used to build a phylogenetic tree and to identify resistant isolates (MRSA &amp; MSSA), presence of genes responsible for toxin production such as Staphylococcal enterotoxins (SE a to SE i), Toxic shock syndrome (TSST) in addition to the Panton Valentine Leukocidin (PVL) toxin gene. Data analysis will be completed in fall 2018 with reporting in 2019.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Veronique Bernier Gosselin</p><br /> <p>Our group has been working to understand the impact of increased heat and humidity on the microbial populations of the milk, feces, rumen, skin, and environment. Understanding the impact of increased heat and humidity on bacterial populations will greatly increase our understanding of the effects of heat stress on the dairy cow as a whole as well as the mammary gland specifically.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Members: Gina Pighetti, Oudessa Kerro Dego</p><br /> <p>Contributors: Reta Duguma Abdi, Desta Beyene Enserum, Susan Headrick, Leszek Wojakiewicz, Caitlin Merill (MS), Jacqueline Vaughn (MS), Tori Couture (MS candidate), Catherine Donnell (BS candidate), Megan O&rsquo;Brien (visiting PhD scholar)</p><br /> <p>Regions in the bovine genome have been identified that are associated with phenotypes reflecting the strength of immune response to Streptococcus uberis infection and S. uberis concentrations in milk following intramammary challenge (TN-Pighetti). The first manuscript associated with this research was submitted and published (Siebert et al., 2018). The top candidate is currently being evaluated for its role in mammary epithelial cell defense mechanisms by an undergraduate honor&rsquo;s student. The second manuscript is nearing completion. Overall, these regions have provided several novel candidate genes that are being examined for their role in mastitis resistance and their potential as novel targets for mastitis control.</p><br /> <p>Staphylococcus aureus is an important zoonotic mastitis pathogen that has significant effects on animal and human health. The objectives of this study were: 1) evaluate clonal diversity of S. aureus isolates from cases of bovine mastitis 2) determine staphylococcal enterotoxin production patterns 3) evaluate in vitro adhesion and invasion ability of dominant strains on bovine mammary epithelial cell line (MAC-T cells) (TN-Kerro Dego). Of the 111 S. aureus strains isolated, 16 PFGE types (dominant clones) were identified (A &ndash; P), with PFGE type M being the most prevalent. Staphylococcal enterotoxin genes seb (11.7%), sec (2.7%), see (0.9%) and/or toxic shock syndrome toxin 1 (tsst-1) (7.2%) were expressed by a subset of isolates, whereas most strains (75.7%) were negative for enterotoxin genes. PFGE types O and M tended to cluster with beta-hemolysin, absence of enterotoxins and susceptibility to antimicrobials. Analysis of in vitro adhesion to and invasion into MAC-T cells showed relatively higher number of O strain adhered to and invaded into MAC-T cells followed by M and I strains, however this differences were not statistically significant.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Member: David Wilson</p><br /> <p>Contributors: Kerry Rood, Justine Britten, Zhongde Wang, Jacqueline LaRose Kurz, E Jane Kelly</p><br /> <p>Additional analysis of genome-wide association studies (GWAS) comparing bovine nucleotide differences (SNPs) between cows repeatedly mastitic vs. cows continually free of mastitis was performed. Twenty-seven quantitative trait loci (QTLs) associated with mastitis resistance were identified including 10 novel QTLs; 4 QTLs were for teat length. One QTL includes a guanyl releasing protein gene, a candidate gene for mastitis resistance.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Vermont</span></p><br /> <p>Members: John Barlow, David Kerr, Feng-Qi Zhao</p><br /> <p>The Kerr lab used a primary dermal fibroblast as a model to rank animals based on composite expression of the toll-like receptor 4 gene (TLR4) and lipopolysaccharide (LPS)-induced IL-8 and IL-6 protein production, and then challenged the mammary glands of these animals with the P4 strain of E. coli to determine how difference in rank would affect response to mastitis. They found that high responder animals have an early upregulation in their innate response that is beneficial for bacterial clearance; however, they are equally susceptible to tissue damage caused by an exuberant response to the infection.</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2:</strong> Characterization and manipulation of virulence factors of mastitis pathogens for enhancing host defenses.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Member: Kasey M. Moyes</p><br /> <p>Contributors: Daniel Nelson, John Roche, Theodore Elasser</p><br /> <p>In collaboration with Dr. Robert Peters, ANSC-UMD, and Mr. Dale Johnson, ANSC-UMD, we are currently following 4 New York dairy farms and to gather information regarding production outcomes, financial and lifestyle changes during their transition from conventional to automatic milking systems.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, B. Crooker, R. Fink, T. Schoenfuss</p><br /> <p>Contributors: L. Fox (WSU), P. Gorden (ISU), D. Nydam (Cornell University), S. Sreevatsan (MSU)</p><br /> <ol start="2019"><br /> <li>Godden, E. Royster, B. Crooker and S. Rowe. Investigation of the relationship between bedding characteristics and intramammary infection in late lactation dairy cows. The major objective of this study is to describe the relationship between bedding characteristics, bedding management and bedding bacteria counts (BBC) in different bedding materials. A secondary objective is to describe the relationship between bedding characteristics and herd-level measures of udder health for late lactation quarters and cows for specific pathogen groups of interest (e.g. Gram-negative organisms; Gram-positive organisms). Eighty herds were enrolled from 10 dairy states using one of four bedding types; new sand, reclaimed sand, manure solids or other organic materials.&nbsp; In summer, 2017 and winter 2018, sampling of each herd included collection of duplicate aseptic quarter milk samples from late lactation cows (&gt; 180 DCC) as well as new and used bedding samples.&nbsp; Milk and bedding samples are being analyzed by bacterial culture. Additionally bedding samples will undergo testing to measure pH, organic matter and dry matter.&nbsp; Data analysis is ongoing with final results and reports expected in early 2019.</li><br /> </ol><br /> <p>Patel, K., S. Godden, E. Royster, J. Timmerman, B. Crooker, and L. Fox.&nbsp; Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health and milk quality on dairy farms.&nbsp; The primary objective of this study is to conduct a multi-state, multi-herd cross-sectional observational study to describe the relationship between bedding bacteria counts and udder health and to identify goals (cutpoints) for interpreting BBC test results. A secondary objective is to identify bedding characteristics and bedding management strategies that are associated with lower BBC and improved udder health. One hundred-eighty eight herds were enrolled from 17 dairy states with the assistance of herd veterinarians or mastitis researchers. Herds used either new sand, reclaimed sand, manure solids or other organic bedding materials. New and used bedding samples, collected from the bedding storage area or from the back of stalls, respectively, and bulk tank milk samples were collected twice from each herd during summer and winter of 2016. Bedding samples were cultured and analyzed to measure pH, organic matter and dry matter. Herd level DHIA test day data describing udder health measures were recorded. Data analysis is ongoing with results expected to be reported in late 2018 and early 2019.</p><br /> <p>Godden, S., T. Schoenfus, R. Fink, J. Timmerman, C. Gebhart, E. Royster, S. Wells. Field validation of Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-ToF) for the identification of dairy microorganisms critical for safety and quality. The first objective is to develop and evaluate extraction methods to improve the MALDI-ToF method's ability to detect Mycoplasma bovis as well as individual species of Bacillus and Paenibacillus of importance to udder health, food safety and food quality. The second objective is to complete a field validation study using 2,500 bacterial isolates Report Date 02/17/2016 Page 1 of 3 Accession No. 233101 Project No. MIN-62-021 Multistate No. NE1048 derived from bovine mastitis samples and processed dairy foods. From this we will describe the diagnostic test characteristics of MALDI-ToF, as compared to 16S rDNA sequencing (gold standard) to identify 24 important dairy microorganisms critical to animal health, food safety and food quality. If the MALDI-ToF method proves to be accurate, then its adoption will strengthen the capacity, quality, and possibly the scope of diagnostic services performed by the VDL to support the MN dairy industry. Lab work and data analysis has been completed. A manuscript is in the final stages of preparation to be submitted in fall 2018.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Members: Gina Pighetti, Oudessa Kerro Dego</p><br /> <p>Contributors: Reta Duguma Abdi, Desta Beyene Enserum, Susan Headrick, Leszek Wojakiewicz, Caitlin Merill (MS), Jacqueline Vaughn (MS), Tori Couture (MS)</p><br /> <p>1) Assess the level of antimicrobial resistance by S. aureus isolates (n=239) collected in TN from 2004-2016. In total, 82 (34.3%) of the S. aureus isolates were found to be resistant at least to 1 of the 10 antimicrobials, whereas 157 (65.7%) of the 239 isolates were pan-susceptible. Overall, the prevalence of AMR among S. aureus varied from as low as 1.3% for ceftiofur to as high as 25% for sulphadimethoxine. The prevalence of AMR was widely distributed throughout the farms at a varying proportion for different antimicrobials tested. Out of all tested isolates, 25.5% were single drug resistant, 4.6% were double drug resistant, and 4.2% were multidrug resistant (resistant to more than three antimicrobials). Of the multidrug-resistant isolates, seven (70%) were also oxacillin-resistant isolates. This research was published earlier this year (Abdi et al., 2018).</p><br /> <p>2) Assess the level of antimicrobial resistance in isolates (n=197) collected with the Southeast Quality Milk Initiative (KY, MS, TN). The isolates evaluated were: 34% Staphylococcus aureus, 21.3% Streptococcus uberis, 18.3% Streptococcus dysgalactiae, 17.8% Escherichia coli, 6.6% Klebsiella pneumoniae and 2% Klebsiella oxytoca. Ceftiofur showed the highest efficacy on the most isolates followed by cephalothin, but K. pneumoniae isolates were resistant to most of the antimicrobials tested. Isolates from subclinical mastitis had higher MIC (P &gt; 0.05) compared with isolates from clinical mastitis for some antimicrobials. The 197 isolates showed 32 different AMR patterns, which varied with farms and states. Compared with S. aureus and Str. dysgalactiae, E. coli and Klebsiella spp. had widespread resistance to pirlmycin and higher MIC for most of the drugs tested. These results suggested that confirmatory diagnosis and subsequent sensitivity testing would be a prerequisite to treat these mastitis pathogens effectively</p><br /> <p>3) Assess antimicrobial resistant bacteria and genes associated with dairy cattle production systems. From 4 TN dairies, 80 samples were collected from different farm environments: bulk tank milk, pooled feces from the floor (feces), soil from dairy cattle manure fertilized land, and soil from adjacent land with no access of dairy cattle were collected in 5 replicates. In general, E. coli , Staphylococcus spp., Enterococcus spp., and Salmonella spp. had different patterns in these environments indicating adaptation and micro-ecological niches of each farm. Assessment of bulk tank milk identified different levels of antimicrobial resistant bacterial species on each farm: tetracycline- or cefotaxime-resistant (n=6, farm A), tetracycline- or nalidixic acid-resistant (n=3, farm B), tetracycline- or cefotaxime- or nalidixic acid-resistant (n=2, farm C) and cefotaxime- and nalidixic acid-resistant (n=1, farm D). The presence of resistance to a third generation cephalosporins (cefotaxime) indicates the need for further evaluation of dairy cattle production system to identify the resistome reservoir and determine critical control points to prevent potential spread among humans, animals and environment.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Vermont</span></p><br /> <p>Members: John Barlow, David Kerr, Feng-Qi Zhao</p><br /> <p>The Barlow lab has developed a multilocus sequence typing scheme for S. chromogenes. A manuscript is in preparation describing the characteristics of this strain typing scheme. Briefly, 120 isolates were examined during development of this scheme. We find the majority of the strain types (STs, n=33, 72 %) are connected in a core network consisting only of single and double locus variants; these 33 STs account for 96 of the 120 isolates in the sample population. The network pattern is indicative of a recent clonal expansion with a single strain as a founder, consistent with recent epidemiological evidence that S. chromogenes is a host adapted species of cattle. Members from Washington (Fox) and Belgium (De Vliegher) collaborated in this project.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>OBJECTIVE 3:</strong> Assessment and application of new technologies that advance mastitis control, milk quality, and dairy food safety.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Louisiana State University, Ag Center</span></p><br /> <p>Member: William E. Owens</p><br /> <p>Novel food grade products continue to be evaluated for antimicrobial activity.&nbsp; Novel products were evaluated using the AOAC in vitro test for germicidal activity.&nbsp; The products were also tested against mastitis pathogens in the MIC/MBC test.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Member: Ronald Erskine, Lorraine Sordillo, Andres Contreras</p><br /> <p>Collaborators: Phil Durst, Ruben Martinez, Stan Moore</p><br /> <p>As part of a multistate USDA-NIFA funded project, we developed an on-farm evaluation system for milk quality and reduction of antibiotic use.&nbsp; The evaluation system (Quality Milk Alliance) assesses traditional practices related to mastitis control as well as a novel integration with social and communication barriers on the part of dairy producers and employees. We are developing novel applications for VaDia vacuum analysis as a tool to determine milking efficiency in dairy herds.&nbsp; These standards will be used to evaluate milking protocols and equipment performance and have the potential to provide an on-farm education platform for producers and/or employees regarding milking protocols.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Mississippi State University</span></p><br /> <p>Member: Amanda Stone</p><br /> <p>Collaborators: Mauricio Xavier</p><br /> <p>This year we worked to evaluate the effects of Imrestor, an immunomodulatory vaccine, on mastitis incidence and milk quality. This year was also the last year for the Southeast Quality Milk Initiative project funded by the USDA to advance milk quality improvements in the southeastern United States.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Member: Kasey M. Moyes</p><br /> <p>Contributors: Daniel Nelson, John Roche, Theodore Elasser</p><br /> <p>In my laboratory at UMD, we are focused on the identification of new treatment strategies to reduce the use of antibiotics for treatment of bovine mastitis on farm to reduce the use of antibiotics commonly used to treat bovine mastitis.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, B. Crooker, R. Fink, T. Schoenfuss</p><br /> <p>Contributors: L. Fox (WSU), P. Gorden (ISU), D. Nydam (Cornell University), S. Sreevatsan (MSU)</p><br /> <ol start="2018"><br /> <li>Rowe, S. Godden, E. Royster, D. Nydam, A. Lago. Lockout study. Randomized equivalence study evaluating the efficacy of two commercial internal teat sealants in dairy cows. The objective of this study is to complete an equivalence study comparing the efficacy of LOCKOUT&trade; (Boehringer Ingelheim Vetmedica, Inc.) versus Orbeseal&reg; (Zoetis) when administered in conjunction with a long acting antibiotic (Ab) at dry off in lactating dairy cows. Dry-off and enrollment of approximately 840 cows (Lockout = 420; Orbeseal = 420) from 5 commercial dairy herds in NY, WI, MN and CA was completed during summer 2018. Collection of post-calving samples and follow-up of lactation health and performance for the first 100 DIM will continue into late fall, 2018.&nbsp; Data analysis and reporting will be completed in 2019.</li><br /> <li>Rowe, S. Godden, E. Royster, D. Nydam, P. Gorden, A. Lago, R. Bicalho. Selective Dry Cow Therapy on US Dairy Farms: Impact on Udder Health, Antimicrobial Use and Economics. The major objective is to complete a multi-location noninferiority randomized clinical trial to evaluate the effect of applying 2 different SDCT programs, a culture-guided program and an algorithm-guided program (vs BDCT) on measures of quarter health, cow health and performance, antibiotic use and economics. Dry-off and enrollment of approximately 1,200 cows (400 per treatmentgroup) from 7 commercial dairy herds in NY, MN, IA and CA was completed during summer 2018.&nbsp; Collection of post-calving samples and follow-up of lactation health and performance for the first 120 DIM will continue into late fall, 2018.&nbsp; Data analysis and reporting will be completed in 2019.</li><br /> </ol><br /> <p>Caixeta, L.S., Morley, P.S., Belk, K., Garry, F.B., Scheu, S., Parker, J, Noyes, N., Mijares, S., Hanes, A. Effect of selective dry cow therapy on the milk microbiome of dairy cattle with low somatic cell count. The purpose of this study was to determine the effects of SDCT on udder health and milk microbiome in separated milk cell pellets and cream in post-partum dairy cattle. 16S rRNA amplicon sequencing was performed and bioinformatics analysis of the sequence data is ongoing in order to identify differences between pellet and cream microbiomes for pre- and post-partum samples.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Veronique Bernier Gosselin</p><br /> <p>In the past year, our group has evaluated non-aureus staphylococcal (NAS) mastitis in dairy heifers and dairy goats.&nbsp; Results in heifers showed an association between skin colonization and intramammary infection (IMI) for some species of NAS and changes in NAS populations on heifer teat skin as animals age.&nbsp; In goats, we investigated the persistence of IMI with NAS during lactation and over the dry period.&nbsp; Results showed that IMI present at &le; 10 days in milk (DIM) are more likely to persist in lactation than IMI that occur &gt; 10 DIM.&nbsp; Some NAS species appear to be host-adapted in goats and NAS IMI can persist over the dry period.&nbsp; MALDI-TOF was validated for identifying NAS species isolated from goat&rsquo;s milk.&nbsp; We are also currently working on a project in collaboration with the University of Kentucky to understand the association between housing environment and staphylococcal species isolated from bulk tank milk, teat skin, and bedding.</p><br /> <p>We are working to understand the effect of intramammary pirlimycin treatment on the fecal microbiome of lactating dairy heifers. This study intends to further the understanding of the broader impacts of intramammary antimicrobial usage and any unintended effects on the microbial ecology on the dairy farm.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Members: Gina Pighetti, Oudessa Kerro Dego</p><br /> <p>Contributors: Reta Duguma Abdi, Desta Beyene Enserum, Susan Headrick, Leszek Wojakiewicz, Caitlin Merill (MS), Jacqueline Vaughn (MS)</p><br /> <p>1) Assessing the potential of Staphylococcal surface proteins as vaccine candidates to minimize risk and severity of S. aureus-based mastitis. Strain variation and limited knowledge of common immunogenic antigens are among major constraints to developing an effective vaccine. Surface proteins from nine genetically distinct S. aureus isolates from bovine mastitis cases were extracted and evaluated for immune-reactivity. Multiple immuno-reactive surface proteins were common across the nine strains, several of which reacted strongly. across the nine strains tested. The conserved immune-reactive surface proteins may serve as potential candidates for a vaccine to control S. aureus mastitis in dairy cows.</p><br /> <p>2) Evaluate the immune responses and protection against S. aureus IMI in dairy cows vaccinated with Staphylococcus aureus surface proteins (SASP) and Staphylococcus chromogenes surface proteins (SCSP) during early dry period. Vaccinated cows had increased milk and serum antibody titers and reduced bacterial shedding of S. aureus in milk following challenge. Interestingly, SCSP vaccine cross-protected against S. aureus clinical mastitis thus suggesting its potential as immunogenic antigens to control bovine S. aureus mastitis.</p><br /> <p>3) Evaluate the protective effects of Staphylococcus chromogenes surface proteins (SCSP) as vaccine antigens to control mastitis during early lactation. Our preliminary results showed that three series of&nbsp; consecutive vaccinations of dairy cows with SCSP at drying off (D0),&nbsp; 21 days after drying off (D+21) and 40 days after drying off&nbsp; (D+40) induced increased immune responses during&nbsp; early lactation in vaccinated cows compared to control cows. The subsequent experimental challenge of vaccinated cows with the heterologous strain of S. aureus resulted in protection from clinical mastitis, reduced number of bacterial shedding in milk, and lower SCC in milk compared to control cows. Overall conclusion from results of early dry period and early lactation studies: The SCSP vaccine cross-protected vaccinated cows from S. aureus clinical mastitis, reduced number of bacterial shedding in milk and somatic cell counts showing its promising immunogenic potential to control mastitis in dairy cows.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Member: David Wilson</p><br /> <p>Contributors: Kerry Rood, Justine Britten, Zhongde Wang, Jacqueline LaRose Kurz, E Jane Kelly</p><br /> <p>Cows with a single high SCC quarter were treated IMM with casein hydrolysate (CH), non-hydrolyzed casein (NHC), or cessation of milking only (negative; N). Following treatment and involution of the mastitic quarter for remainder of lactation, decreases in cow-level SCC (-966,000/ml) and milk production (-14%) with 3 remaining lactating quarters were significant.&nbsp; Among treatments, decrease in cow-level SCC (-1,150,000/ml) for the CH group was significant.&nbsp; All treated quarters returned to milk production after calving; proportion of total-cow milk production (24%) was not different from pre-treatment or from 25% of total milk.&nbsp; After calving, treated quarters&rsquo; decrease in SCC was significant for CH (-2,763,000/ml) and N (-5,324,000/ml). Quarters with positive milk culture before treatment had 88% (14/16) bacterial cure (no isolation of the same bacteria for 3 weeks following calving).</p><br /> <p>Data analysis nearly complete for study of casein hydrolysate (CH) intramammary infusion for cessation of lactation in all 4 quarters at time of dry-off at the end of lactation.&nbsp; Udder halves were compared between control (dry cow antibiotic treatment plus teat sealant) with 4 treatment groups, each including CH, one being CH alone.&nbsp; All quarters of all cows showed no signs of clinical mastitis or cow discomfort and all returned to milk production following calving.&nbsp; All udder halves were not significantly different from 50% of total-cow milk production regardless of treatment group.&nbsp; Involution, SCC and bacteriological results between treatment groups are being analyzed.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">Vermont</span></p><br /> <p>Members: John Barlow, David Kerr, Feng-Qi Zhao</p><br /> <p>The Barlow lab contributed to a study examining the transmission dynamics of Corynebacterium species causing persistent intramammary infections (IMI) in dairy herds. Using a mathematical model and empirical data from 2 dairy herds, we demonstrated that the rate of new infections was significantly related to preexisting IMI in both farms, underscoring the importance of preexisting Corynebacterium spp. IMI for the transmission of Corynebacterium spp. within groups of lactating dairy cattle. NE1748 members from New York (Schukken and Grohn) collaborated in this research.</p><br /> <p>&nbsp;</p><br /> <p><strong>WORK PLANNED FOR THE COMING YEAR, LISTED BY OBJECTIVE:</strong></p><br /> <p><strong>OBJECTIVE 1</strong></p><br /> <p>&nbsp;</p><br /> <p>LSU:</p><br /> <p>Organisms isolated from cases of mastitis from goats and cows will continue to be identified and tested for antimicrobial susceptibility.</p><br /> <p>&nbsp;</p><br /> <p>University of Idaho:</p><br /> <p>We are collecting samples form periparturient cows (-28 d through +21 d, relative to expected parturition date), their dry off, close-up and lactation ration, and their calves (1-3d old) along with udder health, milk SCC and similar data from a large dairy unit (10000+ milking cows). One of the goals is to establish relations among mammary health measures, serum metabolites (lipid soluble vitamins, including vitamin A isomers), and calves passive immunity.</p><br /> <p>&nbsp;</p><br /> <p>University of Maryland:</p><br /> <p>In my laboratory at UMD, we are currently fast beginning the use of new treatment strategies to reduce the use of antibiotics for treatment of bovine mastitis on farm to reduce the use of antibiotics commonly used to treat bovine mastitis. Furthermore, we are caught industry professionals regarding improving the immune response during a mastitis challenge in lactating dairy cows.&nbsp; We are currently collaborating with others internationally to investigate the role of neutrophils during the transition period. We are currently forwarding with others at the USDA to investigate the mammary gland response during dietary vitamin E E in lactating dairy cattle.</p><br /> <p>&nbsp;</p><br /> <p>University of Minnesota:</p><br /> <ol start="2019"><br /> <li>Patel, S. Godden, S. Sreevatsan, E. Royster, L. Fox Molecular Epidemiology of Staphylococcus aureus on U.S. Dairy Farms. Data analysis will be completed in fall 2018 with reporting in 2019.</li><br /> <li>Rowe, S. Godden, E. Royster, D. Nydam, A. Lago. Lockout study. Randomized equivalence study evaluating the efficacy of two commercial internal teat sealants in dairy cows. Collection of post-calving samples and follow-up of lactation health and performance for the first 100 DIM will continue into late fall, 2018. Data analysis and reporting will be completed in 2019.</li><br /> <li>Rowe, S. Godden, E. Royster, D. Nydam, P. Gorden, A. Lago, R. Bicalho. Selective Dry Cow Therapy on US Dairy Farms: Impact on Udder Health, Antimicrobial Use and Economics. Collection of post-calving samples and follow-up of lactation health and performance for the first 120 DIM will continue into late fall, 2018. Data analysis and reporting will be completed in 2019.</li><br /> <li>Godden, E. Royster, B. Crooker and S. Rowe. Investigation of the relationship between bedding characteristics and intramammary infection in late lactation dairy cows. Data analysis is ongoing with final results and reports expected in early 2019.</li><br /> </ol><br /> <p>Patel, K., S. Godden, E. Royster, J. Timmerman, B. Crooker, and L. Fox.&nbsp; Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health and milk quality on dairy farms.&nbsp; Data analysis is ongoing with results expected to be reported in late 2018 and early 2019.</p><br /> <p>Godden, S., T. Schoenfus, R. Fink, J. Timmerman, C. Gebhart, E. Royster, S. Wells. Field validation of Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-ToF) for the identification of dairy microorganisms critical for safety and quality. A manuscript is in the final stages of preparation to be submitted in fall 2018.</p><br /> <p>&nbsp;</p><br /> <p>University of Missouri:</p><br /> <p>Over the next year, we plan to finish two projects related to the effects of heat and humidity on microbial populations of the dairy cow.</p><br /> <p>&nbsp;</p><br /> <p>University of Tennessee:</p><br /> <p>Regions in the bovine genome have been identified that are associated with phenotypes reflecting the strength of immune response to Streptococcus uberis infection and S. uberis concentrations in milk following intramammary challenge. We will submit a manuscript evaluating novel phenotypes. Target the top 1-3 candidates and assess their role in mastitis resistance and potential as novel targets for mastitis control.</p><br /> <p>&nbsp;</p><br /> <p>Utah:</p><br /> <p>One publication accepted, others submitted for publication regarding the genomic study described above.</p><br /> <p>&nbsp;</p><br /> <p>Vermont:</p><br /> <p>The Zhao lab will investigate the effects of bacterial toxins [lipopolysaccharide (LPS) from E. coli and lipoteichoic (LTA) acid from S. aureus] and inflammatory cytokines (TNF-&alpha;, IL-1&beta;, and IL-6) on i) apoptosis and viability and ii) milk protein gene expression in primary bovine mammary epithelial cells (bMECs). Primary bMECs have been isolated from five lactating cows. These cells will be treated with i) different doses of LPS or LTA and ii) different does of TNF-&alpha;, IL-1&beta;, or IL-6 for various times. Next, we will treat the cells with the combinations of cytokines and bacterial toxins (LPS+TNF-&alpha;, LTA+TNF-&alpha;, LPS+IL-1&beta;, LTA+IL-1&beta;, LPS+IL-6, and LTA+IL-6) using the lowest effective dose of each cytokine and toxin. The viability of the cells will be analyzed using the Abcam Cell Viability Assay kit. The cell apoptosis rate will be determined by the terminal dUTP nick end-labeling (TUNEL) assay. mRNA expression of &alpha;-s1-, &alpha;-s2-, &beta;- and &kappa;- caseins, &alpha;-LA, and &beta;-LB will be analyzed by qRT-PCR.</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p>Vermont:</p><br /> <p>The Barlow lab will initiate a study comparing milk quality and mastitis prevalence on organic dairy herds utilizing different bedding management practices. NE-1748 members from Minnesota (Godden), Missouri (Adkins), and Montreal (Dufour) will collaborate in this research. The Barlow lab will quantify potential associations between S. chromogenes strain types and intramammary infection phenotypes. We will use whole genome sequencing to characterize S. aureus and S. chromogenes strains with different in vitro phenotypes.</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 3</strong></p><br /> <p>&nbsp;</p><br /> <p>LSU:</p><br /> <p>Plant based products will continue to be evaluated for antibacterial activity and tested for their potential activity as mastitis therapeutics.</p><br /> <p>&nbsp;</p><br /> <p>University of Missouri:</p><br /> <p>In the coming year, work will continue to be focused on the epidemiology of staphylococcal mastitis in dairy cows.&nbsp; Several publications will be completed and submitted relative to our work on NAS mastitis in goats. We also plan to continue working on the effects of intramammary antimicrobials on fecal bacterial populations.</p><br /> <p>&nbsp;</p><br /> <p>University of Tennessee:</p><br /> <p>1) Evaluate protective effects of Staphylococcus chromogenes surface proteins (SCSP) Vaccine against Staphylococcal and other mastitis pathogens in dairy cows through natural exposure 2) Identification and management of critical control points for antimicrobial resistant bacteria in dairy farms aimed at reducing urgent and serious level threat antibiotic resistant bacteria, 3) Evaluate the potential of a nutritional supplement to enhance the effectiveness of selective dry cow therapy when used in conjunction with an internal teat sealant.</p><br /> <p>&nbsp;</p><br /> <p>Utah:</p><br /> <p>One publication, others in preparation regarding the casein hydrolysate studies described above.</p>

Publications

<p><strong>PUBLICATION LIST</strong></p><br /> <p><strong>Peer-Reviewed Literature</strong></p><br /> <p>Michigan State University:</p><br /> <p>Putman,&nbsp; A. K., J. L. Brown, J. C. Gandy, L. Wisnieski, and L. M. Sordillo. 2018. Changes in biomarkers of nutrient metabolism, inflammation, and oxidative stress in dairy cows during the transition into the early dry period. J Dairy Sci. 2018 Aug 1. pii: S0022-0302(18)30689-1. doi: 10.3168/jds.2018-14591. [Epub ahead of print]</p><br /> <p>&nbsp;</p><br /> <p>University of Idaho:</p><br /> <p>Scholte, C.M., P. Rezamand*, Z. M. Amiri, K.C. Ramsey, C. Tsai, and M.A. McGuire. 2017. The effects of elevated subcutaneous fat stores on fatty acid composition and gene expression of pro-inflammatory markers in periparturient dairy cows. J Dairy Sci 100:2104-2118.</p><br /> <p>&nbsp;</p><br /> <p>University of Maryland:</p><br /> <ol start="2018"><br /> <li>Qu, T. H. Elsasser, C.M. Scholte, M. Garcia and K.M. Moyes. 2018. The effects of feeding mixed tocopherol oil on whole-blood respiratory burst and neutrophil immunometabolic-related gene expression in lactating dairy cows. J. Dairy Sci. 101:4332-4342.</li><br /> </ol><br /> <p>K.M. Moyes, L. Ma, D. Johnson, S. Fultz and R.R. Peters.&nbsp; 2017. Estimating and quantifying the production outcomes and lifestyle changes for small&ndash;to medium sized dairy farms regarding the transition from conventional to automatic milking systems in the Northeast region: A case study report. Biomedical Journal of Scientific and Technical Research. DOI: 10.26717/BJSTR.2017.01.000610.</p><br /> <p>K.M. Moyes, L. Ma, D. Johnson, S. Fultz and R.R. Peters.&nbsp; 2018. Estimating and quantifying the production outcomes and lifestyle changes for small&ndash;to medium sized dairy farms regarding the transition from conventional to automatic milking systems in the mid-Atlantic region: A case study report. In Natl. Mastitis Counc. Ann.&nbsp; Mtg. Proc., Glendale, AZ.&nbsp; Natl. Mastitis Counc., Inc., Madison, WI.</p><br /> <p>C.M. Scholte, D.C. Nelson, S. Linden, T.H. Elsasser, M. Garcia, Y. Qu and K.M Moyes. 2018. Short Communication:&nbsp; Recombinant bacteriophage endolysin, PlyC, is non-toxic and does not alter blood neutrophil response in lactating dairy cows. J. Dairy Sci. DOI: 10.3168/jds.2017-13908.</p><br /> <p>&nbsp;</p><br /> <p>University of Minnesota:</p><br /> <p>Afifi, M., F. Kabera, H. Stryhn, J. Roy, L. Heider, S. Godden, W. Montelpare, J. Sanchez and S. Dufour. 2018. Antimicrobial-based dry cow therapy approaches for cure and prevention of intramammary infections: a protocol for a systematic review and meta-analysis.&nbsp; Animal Health Research Reviews. 19:74-48. https://doi.org/10.1017/S1466252318000051</p><br /> <p>Patel, K., S. Godden, E. Royster, J. Timmerman, B. Crooker. N. McDonald. 2017. Pilot Study: Impact of using a Culture-Guided Selective Dry Cow Therapy Program Targeting Quarter-Level Treatment on Udder Health and Antibiotic Use. Bov Pract. 51:48-57</p><br /> <p>Godden, S., E. Royster, J. Timmerman, P. Rapnicki and H. Green. 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. 100:6527-6544.</p><br /> <p>&nbsp;</p><br /> <p>University of Missouri:</p><br /> <p>Rainard P, Foucras G, Fitzgerald JR, Watts JL, Koop G, Middleton JR.&nbsp; 2018.&nbsp; Knowledge gaps and research priorities in Staphylococcus aureus mastitis control.&nbsp; Transbound Emerg Dis.&nbsp; 65(Suppl 1):149-165.&nbsp;</p><br /> <p>Cameron M, Perry J, Middleton JR, Chaffer M, Lewis PJ, Keefe G.&nbsp; 2018.&nbsp; Short Communication: Evaluation of MALDI-TOF mass spectrometry and a custom reference spectra expanded database for identification of bovine-associated coagulase-negative staphylococci.&nbsp; J Dairy Sci.&nbsp; 101(1):590-595.</p><br /> <p>Adkins PRF1, Dufour S, Spain J, Calcutt M, Reilly T, Stewart G, Middleton JR*.&nbsp; 2018.&nbsp; Cross-sectional study to identify staphylococcal species isolated from teat and inguinal skin of different aged dairy heifers.&nbsp; J Dairy Sci.&nbsp; 101(4):3213-3225.&nbsp;</p><br /> <p>Adkins PRF1, Dufour S, Spain J, Calcutt M, Reilly T, Stewart G, Middleton JR*.&nbsp; 2018.&nbsp; Molecular characterization of non-aureus Staphylococcus spp. from heifer intramammary infections and body sites.&nbsp; J Dairy Sci.&nbsp; 101(6):5388-5403.&nbsp;</p><br /> <p>Bernier Gosselin V1,4, Lovstad J4, Dufour S, Adkins PRF, Middleton JR*.&nbsp; 2018.&nbsp; Use of MALDI-TOF to characterize staphylococcal intramammary infections in dairy goats.&nbsp; J Dairy Sci.&nbsp; 101(7):6262-6270.&nbsp;</p><br /> <p>Bernier Gosselin V1, Dufour S, Zhang M, Middleton JR*. 2018. Sensitivity and specificity of a competitive ELISA using frozen-thawed milk or serum for the diagnosis of small ruminant lentivirus infection in goats using a Bayesian latent class model.&nbsp; Small Ruminant Research.&nbsp; [Epub ahead of print 6 Aug 18]&nbsp; https://doi.org/10.1016/j.smallrumres.2018.08.004</p><br /> <p>Adkins PRF, Middleton JR.&nbsp; 2018.&nbsp; Methods for diagnosing mastitis.&nbsp; Vet Clin North Am Food Anim Pract.&nbsp; Invited article.&nbsp; In Press.</p><br /> <p>&nbsp;</p><br /> <p>University of Tennessee:</p><br /> <p>Abdi R. D., B. E. Gillespie, J. Vaughn, C. Merrill, S. I. Headrick, D. Ensermu, D. H. D&rsquo;Souza, R. A. Almeida, S. P. Oliver, Getahun E. Agga and O. Kerro Dego. 2018. Antimicrobial Resistance of Staphylococcus aureus Isolates from Dairy Cows and Genetic Diversity of Resistant Isolates. Foodborne Pathogens and Disease, 15(7): https://doi.org/10.1089/fpd.2017.2362.</p><br /> <p>Almeida RA, O. Kerro Dego, and A. G. Rius. Effect of heat stress on adherence to and internalization of Streptococcus uberis into bovine mammary epithelial cells. 2018. J Dairy Res, 85: 53&ndash;56. https://doi.org/10.1017/S0022029917000875.</p><br /> <p>Couture, V.L., GM Pighetti, LG Schneider, RA Almeida, SP Oliver, PD Krawczel. Behavioral response to the clinical and subclinical effects of a Streptococcus uberis mastitis challenge in Holstein dairy cows. J Dairy Sci (submitted, 8/31/2018)</p><br /> <p>Kull JA, PD Krawczel, and GM Pighetti. 2018. Short communication: Evaluation of an automated method for assessing white blood cell concentration in Holstein dairy cows. Vet Immunol Immunopathol. 197:21-23. doi: 10.1016/j.vetimm.2018.01.002</p><br /> <p>Oudessa Kerro Dego, R. A. Almeida, Maria E. Prado, S. I. Headrick, M. J. Lewis, C. Young, B. E. Gillespie, L. J. Siebert, G. M. Pighetti, R. D. Abdi, D. B. Ensermu and S. P. Oliver. 2017. Functionally effective immune responses of dairy cows vaccinated with Streptococcus uberis adhesion molecule during the transition period. Ann Vaccines Immunization 3(1): 1015.</p><br /> <p>Siebert L, ME Staton, S Headrick, M Lewis, B Gillespie, C Young, RA Almeida, SP Oliver, and GM Pighetti. 2018. Genome wide association study identifies loci associated with milk leukocyte phenotypes following experimental challenge with Streptococcus uberis. Immunogenetics 70(9):553-562. doi: 10.1007/s00251-018-1065-3</p><br /> <p>&nbsp;</p><br /> <p>Utah:</p><br /> <p>Britten JE, Rood KA, Wilson DJ: Intramammary infusion of casein hydrolysate for involution of single mastitic mammary quarters elevating cow-level somatic cell count. Adv Dairy Res 6:3:1-6, 2018.</p><br /> <p>&nbsp;</p><br /> <p><strong>Abstracts</strong></p><br /> <p>Mississippi State University:</p><br /> <p>Xavier, M.X.S., D.D. McGee, J.A. Brett, A.E. Stone. Evaluation of mammary and uterine health and production parameters of dairy cows treated with pegbovigrastim. 2018. American Dairy Science Association Discover Conference. Chicago, IL.</p><br /> <p>Stone, A., B. Bowman, M. Denny. On-farm bacteriologic milk culturing: Producer perception and decision impact. 2018. American Dairy Science Association Annual Meeting. Knoxville, TN.</p><br /> <p>Bowman, B.L., M.D. Denny, A.E. Stone. Adoption of on-farm bacteriologic milk culturing: Evaluating farmer perception and impact on management decisions. 2018. National Mastitis Council Annual Meeting. Tucson, AZ.</p><br /> <p>Enger, K., C. Petersson-Wolfe, R. A. Almeida, D. T. Nolan, P. D. Krawczel, J. Bewley, A. E. Stone, S. H. Ward, S. P. Oliver, and G. M. Pighetti. 2018. Microorganisms isolated from subclinical intramammary infections present in dairy cattle from the southeast United States. #T201. J Dairy Sci Vol 101 Suppl 2. p285.</p><br /> <p>&nbsp;</p><br /> <p>University of Idaho:</p><br /> <ol start="2018"><br /> <li>Y. Tsai, F. Rosa, M. Bionaz, and P. Rezamand. 2018. Effects of 2, 4- thiazolidinedione on milk fatty acid profile and vitamins in dairy goats with subclinical mastitis. In: Proceedings of the 51 st Annual Pacific Northwest Animal Nutrition Conference: p. 7.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>University of Maryland:</p><br /> <ol><br /> <li>Qu, T.H. Elsasser, S. Kahl, M. Garcia, C.M. Scholte, E.E. Conner, G.F. Schroeder</li><br /> </ol><br /> <p>and K.M. Moyes.&nbsp; 2017. The determination of concentrations of tocopherol isoforms in whole tissues and mitochondria via high-performance liquid chromatography after short-term supplementation in dairy cows. J. Dairy Sci. 100(Suppl 1):1.</p><br /> <p>M.A. Crookenden, C.G. Walker, A. Heiser, J.J. Loor, K.M. Moyes, J.K. Kay, S. Meier,</p><br /> <ol start="2016"><br /> <li>Murray, V.S.R. Dukkipati, M. Mitchell and J.R. Roche. 2016. miRNAseq from whole blood over the transition period. J. Dairy Sci. 99(Suppl 1):85.</li><br /> </ol><br /> <p>Cynthia M. Scholte, T.H. Elsasser, S. Kahl, D.C. Nelson, D. Biswas and K.M. Moyes.&nbsp;</p><br /> <ol start="2018"><br /> <li>Effects of citrus oil fractions on Escherichia coli P4 growth and on bovine neutrophils. American Dairy Science Association (ADSA) Annual Meetings, Knoxville, TN.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>University of Minnesota:</p><br /> <p>Scheu S., Huebner, K., Parker, J., Mijares, S., Hanes, A., Garry, F., Belk, K., Morley, P., Caixeta, L.S. &ldquo;Effect of selective dry cow therapy on udder health and milk microbiome in dairy cattle with low somatic cell count.&rdquo; 51st Annual Conference of the American Association of Bovine Practitioners, 2018.</p><br /> <ol start="2018"><br /> <li>Patel, S. Godden, E. Royster, J. Timmermen, B. Crooker and L. Fox. 2018. Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health on dairy farms using manure solids bedding: preliminary results. 51st American Association of Bovine Practitioners Annual conference, Phoenix, AZ, USA. September 12-15, 2018.</li><br /> <li>Patel, S. Godden, E. Royster, J. Timmermen, B. Crooker and L. Fox. 2017. Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health on U.S. dairy farms: preliminary results.&rdquo; Mastitis Research Workers Conference, Chicago, IL, USA. November 1 - 3, 2017.</li><br /> <li>Patel, S. Godden, E. Royster, J. Timmermen, B. Crooker and L. Fox. 2017. Investigation of the relationship between bacteria counts, bedding characteristics and bedding management practices with udder health and milk quality on dairy farms: preliminary results. Points of Pride Research Day. College of Veterinary Medicine, University of Minnesota, Saint Paul, MN. October 4, 2017.</li><br /> </ol><br /> <p>Rowe, S. Investigation of the relationship between bedding bacteria count and intramammary infection in late lactation dairy cows.&nbsp; American Association of Bovine Practitioners, Phoenix, AZ, September 2018</p><br /> <p>Rowe, S. Investigation of the relationships between towel laundering practices, towel bacteria count and intramammary infection in late lactation dairy cows.&nbsp; American Association of Bovine Practitioners, Phoenix, AZ, September 2018</p><br /> <p>Rowe, S. Investigation of the relationship between bedding materials, bedding characteristics, and intramammary infection in late lactation dairy cows.&nbsp; Mastitis Research Workers Conference, Chicago, IL, November 2017</p><br /> <p>&nbsp;</p><br /> <p>University of Missouri:</p><br /> <p>McKinnon E, Bernier Gosselin V, Royal A, Rindt H, Middleton JR, Adkins PRF.&nbsp; 2018.&nbsp; Evaluation of milk leukocyte populations in relation to caprine arthritis-encephalitis virus in dairy goats.&nbsp; Proceedings of the 41st Annual CVM Research Day (Phi Zeta).&nbsp; 25 May 2018.&nbsp; Abstract #23.</p><br /> <p>&nbsp;</p><br /> <p>Placheta L, Middleton JR (Co-Sponsor), Borchers M, Bewley J, Adkins P (Co-Sponsor).&nbsp; 2018.&nbsp; The effect of housing environment on teat skin staphylococcal populations.&nbsp; Proceedings of the 41st Annual CVM Research Day (Phi Zeta).&nbsp; 25 May 2018.&nbsp; Abstract #16.</p><br /> <p>&nbsp;</p><br /> <p>Bernier Gosselin V, Middleton JR (Sponsor), Dufour S.&nbsp; 2018.&nbsp; Infection dynamics of staphylococcal intramammary infections in dairy goats.&nbsp; Proceedings of the 41st Annual CVM Research Day (Phi Zeta).&nbsp; 25 May 2018.&nbsp; Abstract #36.</p><br /> <p>&nbsp;</p><br /> <p>Schmidt R, Ierardi R, Placheta L, Adkins PRF, Middleton JR.&nbsp; 2018.&nbsp; Epidemiol0gy of mastitis pathogens on Amish Dairy Farms in Missouri.&nbsp; National Veterinary Scholars Symposium.&nbsp; Texas A&amp;M University, College Station, Texas.&nbsp; August 2-5, 2018.</p><br /> <p>&nbsp;</p><br /> <p>Wattenburger K, Schmidt R, Placheta L, Middleton JR, Adkins PRF.&nbsp; 2018.&nbsp; Evaluation of aseptic techniques used to collect bovine milk samples.&nbsp; National Veterinary Scholars Symposium.&nbsp; Texas A&amp;M University, College Station, Texas.&nbsp; August 2-5, 2018.</p><br /> <p>&nbsp;</p><br /> <p>University of Tennessee:</p><br /> <p>Abdi R. D., D. Ensermu, J. Vaughn, C. Merrill, B. Gillespie, O. Kerro Dego. Staphylococcus aureus cell surface proteins extraction and evaluation of immunogenicity. The 98th Annual Conference of Research Workers in Animal Diseases 2017 in Chicago, Illinois, p81, December 3 &ndash; 5. http://crwad.org/wp-content/uploads/2017/11/CRWAD-2017-Author-Index-and-Abstracts.pdf</p><br /> <p>&nbsp;</p><br /> <p>Abdi R. D., B. E. Gillespie, S. I. Headrick, G. M. Pighetti, R. A. Almeida, S. P. Oliver, and O. Kerro Dego. Antimicrobial resistance patterns of bacterial isolates from cases of mastitis in dairy cows. American Dairy Science Association Annual Meeting, June 24 &ndash; 27, 2018, Knoxville, TN.</p><br /> <p>&nbsp;</p><br /> <p>Kerro Dego O., R. D. Abdi and R. A. Almeida. Experimental S. aureus mastitis teat-dip Infection model for evaluation of efficacy of vaccine against S. aureus IMI. American Dairy Science Association Annual Meeting, June 24 &ndash; 27, 2018, Knoxville, TN.</p><br /> <p>&nbsp;</p><br /> <p>Utah:</p><br /> <p>&nbsp;</p><br /> <p>Britten JE, Wilson DJ, Rood KA: Intramammary casein hydrolysate alone or combined with other treatments when drying off dairy cows.&nbsp; J Dy Sci Vol. 101, E-Suppl. 2:177, 2018.</p><br /> <p>&nbsp;</p><br /> <p>Vermont:</p><br /> <p>&nbsp;</p><br /> <p>Korkmaz FT, Elsasser TH, Kerr DE. 2018. Variation in fibroblast expression of toll-like receptor 4 and lipopolysaccharide-induced cytokine production between animals predicts control of bacterial growth but not severity of Escherichia coli mastitis. J Dairy Sci. Aug 29. pii: S0022-0302(18)30784-7. doi: 10.3168/jds.2017-14372. [Epub ahead of print] PMID: 30172411.</p><br /> <p>&nbsp;</p><br /> <p>Dalen G, Rachah A, N&oslash;rsteb&oslash; H, Schukken YH, Gr&ouml;hn YT, Barlow JW, Reksen O. 2018. Transmission dynamics of intramammary infections caused by Corynebacterium species. J Dairy Sci. 101(1):472-479. doi: 10.3168/jds.2017-13162.</p><br /> <p>&nbsp;</p><br /> <p><strong>Conference Proceedings </strong></p><br /> <p>Michigan State University:</p><br /> <p>Erskine, R.J. Dumpster Diving, Somatic Cell Counts and Antimicrobials: What Do They Have in Common? Seminar presented to the Annual Meeting of the Academy of Dairy Veterinary Consultants. Reno, NV, October, 2017.</p><br /> <p>Erskine, R.J. Selective Dry Cow Therapy: It&rsquo;s Complicated. DAIReXNET Webinar, October 18, 2017.</p><br /> <p>Erskine, R. J. Random Thoughts on Parlor Efficiency. Seminar presented to the North Florida/South Georgia Dairy Management Meeting, Quitman, GA, November 16, 2017.</p><br /> <p>Erskine, R. J. Barn Yard Pharmacology and Milking Efficiency. Discussions and workshops presented to the Dairy Production Medicine Certificate Program for Veterinarians, Ontario Veterinary College, Guelph, ONT, December 5-6, 2017.</p><br /> <p>Erskine, R. J. BLV: The Silent Thief Seminar presented at the Michigan Veterinary Medical Conference, Lansing, MI, January, 2018</p><br /> <p>Erskine, R. J., M. Borek-Stine, R. Moore-Foster. Engaged Employees: The Key to Quality Milk. Shortcourse presented at the 58th Annual Meeting of the National Mastitis Council, Tucson, AZ, February 2, 2018.</p><br /> <p>Erskine, R. J. Bi-modal Milk Ejection and Milk Yield-Is There a Connection? Seminar presented at the Meeting of the National Mastitis Council, Milan, Italy, June 12, 2018.</p><br /> <p>Erskine, R. J. and D. Thompson. The Cows Speak to Us About Milking- Are You Listening? Shortcourse presented at the Regional Meeting of the National Mastitis Council, Guelph, ONT, June 20, 2018.</p><br /> <p>Erskine, R. J. Selective Dry Therapy-A Good Tool For The Mastitis Toolbox, If&hellip;&hellip;.. Seminar presented at the Regional Meeting of the National Mastitis Council, Guelph, ONT, June 21, 2018.</p><br /> <p>&nbsp;</p><br /> <p>University of Maryland:</p><br /> <p>Invited: C.M Scholte#.&nbsp; 2017.&nbsp; LACTATION BIOLOGY SYMPOSIUM: Alternatives to antibiotic treatment for bovine mastitis.&nbsp; American Animal Science Association Annual Meeting, July 8-12, Baltimore, MD.</p><br /> <p>&nbsp;</p><br /> <p>University of Minnesota:</p><br /> <ol start="2018"><br /> <li>Godden, K. Patel, E. Royster, J. Timmerman, B. Crooker and L. Fox. 2018. Relationships between bedding bacteria counts, bedding characteristics and udder health: We have a lot more to learn. 2018 International Bovine Mastitis Conference. Milano, Italy. June 11-13, 2018.</li><br /> <li>Godden, K. Patel, E. Royster, J. Timmerman, B. Crooker and L. Fox. 2018. Relationships between bedding bacteria counts, bedding characteristics and udder health. National Mastitis Council Regional Meeting. Guelph Ont. Canada. June 20-22, 2018. Pp. 18-27.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>University of Missouri:</p><br /> <p>Bernier Gosselin V, Adkins PRF, Middleton JR.&nbsp; 2018.&nbsp; Use of strain-typing of staphylococci to determine persistence of intramammary infection in dairy goats.&nbsp; Proceedings of the 57th Annual Meeting of the National Mastitis Council, Tucson, AZ.&nbsp;</p><br /> <p>Placheta L, Middleton JR, Borchers M, Bewley JM, Adkins PRF.&nbsp; 2018.&nbsp; Association of housing environment and staphylococcal species isolated from teat skin of dairy cattle.&nbsp; Proceedings of the 57th Annual Meeting of the National Mastitis Council, Tucson, AZ.</p><br /> <p>&nbsp;</p><br /> <p>University of Tennessee:</p><br /> <p>Abdi, R. D., J. Vaughn, C. Merrill, S. M. Cantwell, B. E. Gillespie, R. A. Almeida, S. I. Headrick, G. M. Pighetti, P. Krawczel, J. Keflot, J. M. Bewley, S. P. Oliver and O. Kerro Dego. 2017. Antimicrobial Resistance Patterns of Staphylococcus aureus Isolates from Cases of Bovine Mastitis. Proceedings of National Mastitis Council, p 60, 56th meeting January 28 &ndash; 31, 2017, Trade winds Island Grand Resort, St. Pete Beach, Fl.</p><br /> <p>&nbsp;</p><br /> <p>Utah:</p><br /> <p>Wilson DJ: J5 mastitis vaccinates&rsquo; milk production change and reproductive performance following clinical mastitis.&nbsp; Proc 10th World Congress of Vaccine: 45, 2017.</p><br /> <p>Britten JE, Wilson DJ, Rood KA: Casein hydrolysate as a possible adjunct to or replacement treatment for current antibiotic therapies used at dry-off in dairy cows.&nbsp; Proc 57th Ann Mtg Natl Mast Council:238-239, 2018.</p><br /> <p>Britten JE, Wilson DJ, Rood KA: Effect of casein hydrolysate on bovine mammary involution: changes to milk composition and preliminary histological evaluation. Proc Animal, Dairy and Veterinary Sciences Student Research Symposium:35, 2018.</p><br /> <p>&nbsp;</p><br /> <p><strong>Poster Presentations</strong></p><br /> <p>University of Idaho:</p><br /> <ol start="2018"><br /> <li>Y. Tsai, F. Rosa, M. Bionaz, and P. Rezamand. 2018. Effects of 2, 4- thiazolidinedione on milk fatty acid profile and vitamins in dairy goats with subclinical mastitis. In: Proceedings of the 51 st Annual Pacific Northwest Animal Nutrition Conference: graduate poster competition (January 2018, Boise, Idaho).</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>University of Minnesota:</p><br /> <p>Rowe, S. Selective dry cow therapy on U.S. dairy farms: impact on udder health, antimicrobial use and economics.&nbsp; Minnesota Dairy Health Conference, Saint Paul, MN, April,18-19, 2018.</p><br /> <p>&nbsp;</p><br /> <p>University of Tennessee:</p><br /> <p>Abdi R. D., J. R. Dunlap, D. B. Ensermu and O. Kerro Dego. Staphylococcus aureus surface proteins extraction methods with immunoproteomics and electron microscopic study. American Dairy Science Association Annual Meeting, June 24 &ndash; 27, 2018, Knoxville, TN.</p><br /> <p>Merrill C., D. B. Ensermu, R. D. Abdi, B. E. Gillespie, J. Vaughn, S. I. Headrick, K.&nbsp; Hash, T. B. Walker and O. Kerro Dego. Protective effects of staphylococcal surface proteins as vaccine antigens to control mastitis in Dairy cows. American Dairy Science Association&nbsp; Annual Meeting, June 24 &ndash; 27, 2018, Knoxville, TN.</p><br /> <p>Merrill C., Desta B. Ensermu, Reta Duguma Abdi, Barbara E. Gillespie, Jacqueline Vaughn, Raul A. Almeida, S. P. Oliver, Susan I. Headrick, Kody Hash, Tate Bradley Walker and O. Kerro Dego. Evaluation of the immune response and protection in dairy cows vaccinated with immunodominant Staphylococcal surface proteins. The 98th Annual Conference of Research Workers in Animal Diseases, p187, December 1-5, 2017, Chicago Marriott, Downtown Magnificent Mile, Chicago, Illinois. http://crwad.org/wp-content/uploads/2017/11/CRWAD-2017-PRESENTATION-SCHEDULE.pdf</p><br /> <p>Merrill C., D. B. Ensermu, R. D. Abdi, B. E. Gillespie, J. Vaughn, R. A. Almeida, S. P. Oliver, S. I. Headrick, K. Hash, T. B. Walker and O. Kerro Dego. Protective Effects of a new Staphylococcal vaccine against mastitis in dairy cows. UT Beef and Forage Center annual Meeting, Dec 19, 2017 at the University of Tennessee, Knoxville, TN.</p><br /> <p>Vaughn J., R. D. Abdi, B. E. Gillespie, C. Merrill, D. B. Ensermu and O. Kerro Dego. Genetic Diversity and Associated Enterotoxin Production Patterns of S. aureus Isolates from Cases of Bovine Mastitis. American Dairy Science Association&nbsp; Annual Meeting, June 24 &ndash; 27, 2018, Knoxville, TN.</p><br /> <p>Vaughn J., R. D. Abdi1, B. E. Gillespie1, C. Merrill1 and O. Kerro Dego. Genetic Diversity of Staphylococcus aureus from Cases of Bovine Mastitis. UT Beef and Forage Center annual Meeting, Dec 19, 2017 at the University of Tennessee, Knoxville, TN.</p><br /> <p>Vaughn J., R. Duguma Abdi, B. Gillespie, C. Merrill, O. Kerro Dego.&nbsp; 2017. Genetic diversity and enterotoxin production profiles of Staphylococcus aureus strains from cases of bovine mastitis. The 98th Annual Conference of Research Workers in Animal Diseases, P144, December 1-5, 2017, Chicago Marriott, Downtown Magnificent Mile, Chicago, Illinois.http://crwad.org/wp-content/uploads/2017/11/CRWAD-2017-Author-Index-Abstracts.pdf</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Web articles</strong></p><br /> <p>Michigan State University:</p><br /> <p>Launstein E., and R. J. Erskine. Revisited article: You are here on the somatic cell count map</p><br /> <p>http://qualitymilkalliance.com/2018/07/25/revisited-article-you-are-here-on-the-somatic-cell-count-map/</p><br /> <p>Erskine, R. J. How is Your Milking Efficiency? Part 1.</p><br /> <p>http://qualitymilkalliance.com/2018/05/01/how-is-your-milking-efficiency-part-1/</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Mississippi State University:</strong></p><br /> <p>Becker, C. and A. Stone. 2018. Dry cow therapy: Aseptic infusion. Mississippi State University Extension Factsheet. P3291.</p><br /> <p>Becker, C. and A. Stone. 2018. Dry cow therapy: Choosing the best protocol for your dairy. Mississippi State University Extension Factsheet. P3290.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Public outreach</strong></p><br /> <p>Pighetti, GM. High quality milk: Farm to table. Knoxville Academy of Nutrition and Dietetics, 9/26/2018, 24 attendees.</p>

Impact Statements

Back to top

Date of Annual Report: 01/02/2020

Report Information

Annual Meeting Dates: 11/06/2019 - 11/08/2019
Period the Report Covers: 11/08/2018 - 11/08/2019

Participants

Pamela Adkins University of Missouri
John Middleton University of Missouri
Chris Luby University of Saskatchewan
Feng-Qi Zhao University of Vermont
John Barlow University of Vermont
Pamela Ruegg Michigan State University
Bill Owens Louisiana State University
Sandra Godden University of Minnesota
Benjamin Enger Ohio State University
Massimo Bionaz Oregon State University
Simon Dufour University of Montreal
Paolo Moroni Cornell University

Brief Summary of Minutes

Accomplishments

<p><strong>OBJECTIVE 1: Characterize host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance to improve economic outcomes and animal welfare.</strong></p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Member: William E. Owens</p><br /> <p>Identify and evaluate bacteria causing bovine mastitis in Louisiana. Test bacteria for antimicrobial resistance to mastitis therapeutics.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Members: Ronald Erskine, Lorraine Sordillo, Pamela Ruegg</p><br /> <p>We continued to study the impact of negative energy balance and fat mobilization on vascular endothelial and adipose inflammatory responses. The role of n-3 fatty acid content in altering the profile of vasoactive eicosanoids and the role of poly-unsaturated fatty acids on attenuating endothelial cell inflammatory responses, as well as lymphocyte expression of pro-inflammatory cytokines was a central aim of our work.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Member: Mulumebet Worku</p><br /> <p>Expression of LGALS and secretion of Galectins is responsive to modulation in a cow dependent manner. Exogenous Galectin-8 had differential effects in the neutrophil transcriptome.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Member: Benjamin D. Enger</p><br /> <p>Collagen localization and abundance was determined in uninfected and <em>Staphylococcus aureus </em>infected mammary tissues from nonlactating cows to determine the impact of <em>Staphylococcus aureus </em>intramammary infection on collagen deposition. Immunohistochemistry of progesterone receptor and estradiol receptor was also completed in these same tissues to determine if mastitis impacts expression of these pivotal hormone receptors that are involved in directing mammary growth and development.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Member: Jeffrey M. Boyd</p><br /> <p>We have found that the activity of the <em>Staphylococcus aureus </em>ClpCP protease changes upon switches between aerobic and fermentative growth. These activity changes are governed by the SrrAB regulatory system. Changes in ClpCP activity correlates with changes in virulence factor expression.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Member: Kasey Moyes</p><br /> <p>In collaboration with industry personnel, we are currently determining the immune response in milk of dairy cows after intramammary infection (IMI) with <em>Streptococcus uberis</em> (<em>S. uberis)</em> for cows fed Omnigen-AF&reg; during mid-lactation.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, B. Crooker, C. Gebhart, S. Wells, T. Schoenfus, B. Heins.</p><br /> <p>Contributors: P. Gorden (Iowa State), D. Nydam (Cornell), S. Sreevatsan (Michigan State), R. Fink (St. Cloud State), V. Machado (Texas Tech), P. Pinedo (Colorado State), A. Formiga (Oregon)</p><br /> <p>A. Crooker, W. J. Weber, J. D. Lippolis and J. B. Cole. Impact of Holstein genotype on innate immune response to lipopolysaccharide and <em>Escherichia coli</em>. A greater understanding of how the bovine innate immune system is regulated and how this regulation has changed through selection could help reduce disease and would benefit the industry. The University of Minnesota has a unique herd of Holsteins that has not been subjected to selection since 1964 - our unselected Holsteins (UH). Our experimental model of UH vs. contemporary Holsteins (CH) provides opportunities to determine effects of 50+ years of selection. We administered an intermammary <em>E. coli</em> challenge (250 cfu of E. coli strain P4) to primiparous UH (n=5) and CH (n = 7) cows. Increases in body temperature, milk somatic cell count and milk bovine serum albumin, IL-1b and IL-6 concentrations were greater in CH than in UH cows. The prolonged increase in bacterial counts and more severe mastitis in the CH cows indicate selection has reduced the ability of the innate immune system to clear <em>E. coli </em>from the mammary gland and has increased susceptibility to infection by this pathogen. A manuscript is expected in 2020.</p><br /> <p>Patel*, S. Godden, E. Royster, B. Crooker, T. Johnson, E. Smith, S. Sreevatsan. Prevalence, antibiotic resistance, virulence and genetic diversity of <em>Staphylococcus aureus</em> isolated from bulk tank milk samples of U.S. dairy herds. The objective of this study was to investigate the prevalence, antibiotic resistance gene content, virulence determinants and genomic diversity of <em>S. aureus</em> in bulk tank milk samples (BTM) from U.S. dairy herds. Of 365 pooled BTM samples collected from 189 herds across the United States, <em>S</em><em>. aureus</em> was cultured from 170 samples, indicating a sample prevalence of 46.6% in the BTM and a herd prevalence of 62.4% (118 out of 189 herds). Among a subset of 138 <em>S. aureus</em> isolates that were stored for further analysis, 124 were genome sequenced after being confirmed as <em>S. aureus</em> using phenotypic tests. The most commonly identified antimicrobial resistance-associated gene was <em>norA</em> (99.2%) and the <em>mecA</em> gene responsible for methicillin resistance (MRSA) was identified in one isolate (0.8%). The most frequently detected putative virulence genes were <em>aur</em> (100%), <em>hlgB</em> (100%), <em>hlgA, hlgC, hlb</em> (99.2%), <em>lukE</em> (95.9%) and <em>lukD</em> (94.3%). In the 53 staphylococcal enterotoxin (SE) gene positive isolates, <em>sen</em> (37.9%), <em>sem</em> (xx%)<em>, sei</em> (35.5%) and <em>seg</em> (33.1%) were the most frequently detected enterotoxin genes. Among the 14 different sequence types (ST) and 18 <em>spa</em> types identified, the most common was ST2187 (20.9%) and t529 (28.2%), respectively. The most predominant clone was CC5 (50.8%) followed by CC151 (26.6%). The single MRSA isolate belonged to ST72-CC5, <em>spa</em> type t126 and was negative for the <em>tst</em> gene but harbored all the other virulence genes. Our findings indicated a high prevalence of <em>S. aureus</em> in BTM, with little evidence of antibiotic resistance but often carrying genes for the various enterotoxins. This study identified predominant genetic clones and provides continued surveillance of <em>S. aureus</em> strains found in U.S. dairy herds. Manuscript in preparation.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Monica Witzke, Thomas McFadden</p><br /> <p>A series of studies have been conducted to evaluate the impact of heat stress on dairy cattle microbiomes to determine how changes in these microbial populations may influence dairy cattle susceptibility to mastitis. Preliminary results indicate heat stress does results in a change in fecal and rumen microbiomes of dairy cattle.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Members: Jean-Philippe Roy, Mario Jacques, Simon Dufour, Marie Archambault, David Francoz</p><br /> <p>Our recent work highlighted the presence of bacteria in a biofilm <em>in vivo</em> in the mammary gland of dairy cow (Jacques). We also described Klebsiella and Raoultella isolates from clinical mastitis cases (Archambault). Finally, we investigated on the presence of superantigens in coagulase-negative staphylococci retrieved from intramammary infections and concluded that they were uncommon in this population of isolates (Jacques).&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>In collaboration with Tom McFadden&rsquo;s group at the University of Missouri, Zhao Lab at VT investigated the timing and magnitude of acute local and systemic mammary responses to intramammary LPS challenge in lactating cows and revealed distinct local and systemic effects of LPS on milk yield and composition of milk and blood. In addition, Zhao&rsquo;s lab found differential effects of LPS on milk protein expression in mouse mammary gland and mammary epithelial cells.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2: Assess and apply new technologies that advance mastitis control, milk quality and/or dairy food safety.</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Member: Mulumebet Worku</p><br /> <p>Genes involved in innate and adaptive immunity in bovine neutrophils are differentially modulated by galectins</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Member: Jeffrey M. Boyd</p><br /> <p>We have spent much effort examining how <em>S. aureus</em> detoxifies copper and the mechanism by which copper poisons <em>S. aureus</em>. Copper has been shown to be effective at decreasing disease burden when used on touch surfaces. We have examined novel benzalkonium-templated mesoporous silica nanoparticles that are effective antimicrobial agents. These nanoparticles are a novel drug delivery systems that could be used to coat surfaces to decrease bacterial burden or used in wound healing.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Member: Kasey Moyes</p><br /> <p>In collaboration with Dr. Robert Peters, University of Maryland (UMD), and Mr. Dale Johnson, UMD, we are currently following 4 New York dairy farms and to gather information regarding production outcomes, financial and lifestyle changes during their transition from conventional to automatic milking systems.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Illinois</span></p><br /> <p>Member: Peter Constable</p><br /> <p>We characterized the analytical performance of a relatively inexpensive ($300) point-of-care sodium ion selective electrode (ISE) meter (Horiba, Japan) in measuring milk Na concentration. As milk Na concentration increases with glandular inflammation due to increased permeability of the blood-milk barrier, this rapid and low-cost test has promise for the cow-side detection of abnormal glandular secretions.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, B. Crooker, C. Gebhart, S. Wells, T. Schoenfus, B. Heins.</p><br /> <p>Contributors: P. Gorden (Iowa State), D. Nydam (Cornell), S. Sreevatsan (Michigan State), R. Fink (St. Cloud State), V. Machado (Texas Tech), P. Pinedo (Colorado State), A. Formiga (Oregon)</p><br /> <p>Noyes, L. Caixeta, B. Heins, V. Machado, P. Pinedo, A. Formiga, D. Van Nydam, J. Velez, H. Karremann. Catalyzing an open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case.</p><br /> <p>The long-term goal of this proposal is to ignite robust, systematic and application-driven microbiome research activity within the livestock community, thus producing a panoply of new, evidence-based tools to combat livestock disease and production challenges.&nbsp;In order to provide this ignition, we plan to use the udder microbiome and mastitis as a test case to demonstrate the power and applied benefit of well-conducted, comprehensive microbiome research. In executing this demonstration, we will simultaneously address the microbiome data shortage within the organic and dairy communities, while also piloting the applied use of promising microbiome-based treatments for mastitis. Underlying this effort is an outreach program that will serve as the foundation for a sustained, innovative and productive microbiome program within the livestock community.&nbsp; Specific Aims: 1) investigation of epidemiologic associations between the udder microbiome, host/farm risk factors, and development of pathogen-specific mastitis, specifically on representative organic dairy farms 2)&nbsp;demonstration of&nbsp;<em>in vitro&nbsp;</em>proof-of-concept for a novel microbiome-based mastitis intervention, using data generated in Activity #1&nbsp; 3)&nbsp;development of comprehensive, targeted, and multimodal educational tools for improving stakeholders&rsquo; information literacy regarding the microbiome and microbiome-based research 4)&nbsp;to initiate and sustain &ldquo;open-ROAMER&rdquo; (<strong>open</strong>-community&nbsp;<strong>r</strong>esource for&nbsp;<strong>o</strong>rganic&nbsp;<strong>a</strong>nimal&nbsp;<strong>m</strong>icrobiome&nbsp;<strong>e</strong>ducation and&nbsp;<strong>r</strong>esearch) -- a user-friendly, comprehensive online repository for organic-specific microbiome data, pipelines and tutorials. Field work in progress.</p><br /> <p>&nbsp;</p><br /> <p>S.M. Rowe,* S.M. Godden, E. Royster, J. Timmerman, B.A. Crooker, M. Boyle. Cross-sectional study of the relationship among bedding materials, bedding bacteria counts and intramammary infection in late lactation dairy cows. Objectives of this study were to; 1) describe the intramammary infection (IMI) prevalence and pathogen profiles in quarters of cows approaching dry-off in U.S. dairy herds, 2) compare IMI prevalence in quarters of cows exposed to different bedding material types, and 3) identify associations between bedding bacteria count (<strong>BBC</strong>) and IMI in cows approaching dry-off.&nbsp; Eighty herds using one of four common bedding materials (manure solids; <strong>MS</strong>, organic non-manure; <strong>ON</strong>, new sand; <strong>NS</strong> and recycled sand: <strong>RS</strong>) were recruited in a multi-site cross-sectional study. Each herd was visited twice for sampling. At each visit, aseptic quarter-milk samples were collected from 20 cows approaching dry-off (&gt;180 days pregnant). Samples of unused and used bedding were also collected. Aerobic culture was used to determine the IMI status of 10,448 quarters and to enumerate counts (log<sub>10</sub> CFU / cc) of all bacteria, <em>Staphylococcus spp., Streptococcus spp.</em> and Strep-like organisms (<strong>SSLO</strong>), Coliforms, <em>Klebsiella spp.,</em> non-coliform Gram-negatives, <em>Bacillus spp. </em>and <em>Prototheca spp.</em> in unused (n = 148) and used (n = 150) bedding. Quarter-level prevalence of IMI was 21.1%, which was primarily caused by non-aureus <em>Staphylococcus spp.</em> (<strong>NAS; </strong>11.4%) and SSLO (5.6%). Only modest differences in IMI prevalence were observed between the four common bedding material types. Counts of all bacteria in unused bedding was positively associated with odds of IMI caused by any pathogen (<strong>ALL-IMI</strong>; OR = 1.08). A positive association was also observed for counts of SSLO in unused bedding and SSLO-IMI (OR = 1.09). These patterns of association were generally consistent across the four common bedding materials. In contrast, the association between counts of all bacteria in used bedding and ALL-IMI varied by bedding type, with positive associations observed in quarters exposed to MS (OR = 2.29) and ON (OR = 1.51) and a negative association in quarters exposed to NS (OR = 0.47). Findings from this study suggest that quarter-level IMI prevalence in late lactation cows is low in U.S. dairy herds. Furthermore, bedding material type may not be an important risk factor for IMI in late lactation.&nbsp; Higher levels of bacteria in bedding may increase IMI prevalence at dry-off in general, but this relationship is likely to vary according to bedding material type.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>S.M. Rowe,* S.M. Godden, E. Royster, J. Timmerman, M. Boyle. Cross-sectional study of the relationship between cloth udder towel management, towel bacteria counts and intramammary infection in late lactation dairy cows. The objectives of this cross-sectional study were to: 1) Describe associations between herd-level measures of towel bacteria count (<strong>ToBC</strong>) and quarter-level intramammary infection (<strong>IMI</strong>) status in late lactation cows; 2) Establish pathogen-specific target levels of bacteria in CUT to aid the interpretation of towel culture reports and 3) Identify laundering-related risk factors for high ToBC. The study was conducted in 67 herds, from 10 dairy states in the U.S. that used CUT. These 67 herds were originally recruited as part of a larger (80 herd) cross-sectional study of bedding management. Each herd was visited once during December 2017 to April 2018 and quarter-milk samples (n = 4,656) were collected from late gestation (&gt; 180 d pregnant) cows (n = 1,313). Two recently laundered CUT were collected and a questionnaire was used to collect information about pre-milking teat preparation and CUT management practices. Quarter-level IMI status was determined using standard bacteriologic methods. In addition, colony forming units of all bacteria (total bacteria), <em>Staphylococcus spp., Streptococcus spp.</em> or Strep-like organisms (<strong>SSLO</strong>), Coliforms, non-coliform Gram-negatives and <em>Bacillus spp. </em>were determined for each pair of CUT (log<sub>10</sub> CFU / in<sup>2</sup>). Results showed that counts of <em>Staphylococcus spp.</em> and SSLO on towels were positively associated with odds of IMI caused by NAS (OR = 1.40) and SSLO (OR = 1.45), respectively. Of 12 CUT management practices evaluated, only the failure to use a dryer was identified as a clear predictor of risk for a high ToBC (Risk ratio of high coliform count = 8.17). Our study findings suggest that CUT may act as a fomite for NAS and SSLO. We recommend that herds aim to keep counts of <em>Staphylococcus spp. </em>and SSLO in cloth udder towels below 5 CFU / in<sup>2,</sup>, and that laundered towels be completely dried in a hot air dryer.</p><br /> <p>&nbsp;</p><br /> <p>Godden, S., P. Raynor, R. Singer, E. Royster, J. Hadrich, K. Janni, B. Crooker, J. Bender, B. Alexander. Investigation of the Relationship between Method of Processing Recycled Manure Solids Bedding, Udder Health, Milk Quality, Air Quality, Antimicrobial Resistance and Economics on Midwest Dairy Farms.&nbsp; Objectives of this study are to investigate if method of processing recycled manure solids (RMS) is associated with i) air quality measures of importance to dairy worker health and ii) the amount of antimicrobial resistance genes in the bedding material. A convenience sample of 30 MN and WI dairy systems using RMS as bedding were recruited in summer 2019. Systems were selected to represent the following four methods for processing RMS: i) processing methods: i) Green RMS (<strong>GRN</strong>), ii) Anaerobic digester (<strong>DIG</strong>), iii) Composted, and iv) Mechanical hot-air drum dryer.&nbsp; All herds were visited once in August 2019 to collect bedding samples, air quality (dust) samples, bulk tank milk samples, herd DHIA test day and cow health records, a herd management questionnaire, and an economic survey of expenses and revenues associated with the manure processing system in use. Bedding samples are undergoing culture, nutrient analysis, and antimicrobial resistance genes. Bulk tank milk samples underwent routine culture. We plan to resample all herds again in winter 2020 specifically for the udder health and milk quality objectives. Results will be forthcoming in 2020.&nbsp;&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>Nusrat Annie Jahan<sup>*</sup>, S. Godden, T. Schoenfus, C. Gebhart, E. Royster, S. Wells, J. Timmerman, and R. Fink. Evaluation of the Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS) system in the detection of Mastitis pathogens from Bovine milk samples. In the present study, we prospectively compared MALDI-TOF MS (Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry or MALDI-ToF) to the conventional 16S rDNA sequencing method for the identification of&nbsp; mastitis isolates (481). Among the 481 milk isolates, 445 (93%) were putatively identified to the genus level by MALDI-TOF MS and 355 (74%) were identified to the species level, but no reliable identification was obtained for 16 (3.3%), and 20 (4.2%) discordant results were identified. Future studies can help to overcome the limitation of MALDI database and additional sample preparation steps might help to reduce the number of discordances in identification. In conclusion, our results show that MALDI-TOF MS is a fast and reliable technique which has the potential to replace conventional identification methods for most dairy pathogens, routinely isolated from the milk and dairy products. Thus it&rsquo;s adoption will strengthen the capacity, quality, and possibly the scope of diagnostic services to support the dairy industry. Manuscript in preparation.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: V&eacute;ronique Bernier Gosselin, Monica Witzke, Thomas McFadden</p><br /> <p>A series of studies were completed to evaluate the epidemiology of non-aureus staphylococcal (NAS) mastitis during lactation and over the dry period in dairy goats in Missouri.&nbsp; Overall, these studies showed that MALDI-TOF mass spectrometry had good typeability and accuracy for speciating NAS isolated from goat&rsquo;s milk. <em>Staphylococcus caprae</em>, <em>Staphylococcus chromogenes</em>, <em>Staphylococcus epidermidis</em>, <em>Staphylococcus simulans, </em>and<em> Staphylococcus xylosus</em> tended to be associated with elevated milk somatic cell count (SCC).&nbsp; <em>Staphylococcus caprae</em> and <em>S. simulans</em> tended to be associated with persistent intramammary infection (IMI).&nbsp; Udder skin colonization with some species of NAS during the non-lactating period increased the risk of IMI at kidding.</p><br /> <p>A study was conducted to compare two techniques for collecting milk samples to diagnose IMI in dairy cattle.&nbsp; Results indicated that the microbiological status of milk samples aseptically collected via the teat orifice had high agreement with the microbiological status of milk collected directly from the teat cistern by aseptic aspiration, indicating that most bacteria isolated from uncontaminated milk samples collected via the teat orifice tend to be associated with IMI.</p><br /> <p>A study was completed to evaluate the impact of intramammary pirlimycin on the fecal microbiome of dairy heifers. Results indicated that the treatment did results in changes in the fecal microbiome.&nbsp; To evaluate this further, fecal samples collected from heifers both pre and post treatment were cultured, specifically to recover Gram-positive cocci.&nbsp; All isolates were speciated and evaluated for antimicrobial resistance. To date, resistance is minimal among staphylococcal and streptococcal isolates recovered from the feces of heifers both post and pretreatment with intramammary pirlimycin.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Members: Jean-Philippe Roy, Mario Jacques, Simon Dufour, Marie Archambault, David Francoz</p><br /> <p>We investigated impact of recycled manure bedding on bacterial and parasite load in cows and bulk milk.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab has completed a study of the mammary microbiome of lactating organic dairy cattle and we have submitted this work for publication (in revision). The Barlow lab has completed a study applying machine learning to identify key predictors of pathogen strain type from multilocus sequence typing databases for <em>Staphylococcus aureus</em>, <em>Streptococcus agalactiae</em>, and <em>Streptococcus uberis</em>. The Barlow lab has initiated a study comparing milk quality and mastitis prevalence on organic dairy herds utilizing different bedding management practices. NE-1748 members from Minnesota (Godden), Missouri (Adkins) will collaborate in this research.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Members: David Wilson</p><br /> <p>Contributors: Kerry Rood, Justine Britten, Zhongde Wang, Jacqueline LaRose Kurz, Robert Weiss, George Liu, Zhou Yang, John Middleton, Pamela Adkins, Gregory Goodell</p><br /> <p>Bacterial culture isolation and biochemical testing (Cx), Matrix-assisted laser desorption ionization&ndash;time of flight (MALDI-TOF), and 16S rRNA partial genome sequencing (16S) were compared for microbial identification from bovine milk.&nbsp; 181 bacterial colonies isolated from a commercial herd were tested in blind comparison by each method.&nbsp; Positive agreement among all 3 methods was 94%, with 95% to 98% between each pair of methods. Overall (including negative agreement) agreement among all 3 methods ranged from 97% to 100%, depending on the mastitis pathogen.&nbsp; Any of the 3 methods is a useful tool for identification of bacteria isolated from dairy cow milk. (Collaboration with MO)</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 3: Identify and apply new strategies associated with the control of mastitis that can reduce the use of antibiotics in dairy herds.</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Member: William E. Owens</p><br /> <p>Continue evaluation of botanical formulations from plants for antimicrobial activity against mastitis pathogens.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Members: Ronald Erskine, Lorraine Sordillo, Pamela Ruegg</p><br /> <p>As part of a multistate USDA-NIFA funded project, we developed an on-farm evaluation system for milk quality and reduction of antibiotic use.&nbsp; The evaluation system (Quality Milk Alliance) assesses traditional practices related to mastitis control and communication barriers on the part of dairy producers and employees. We are developing novel applications of vacuum analysis to determine milking efficiency in dairy herds.&nbsp; This will provide an on-farm education platform for producers and/or employees regarding milking protocols.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Member: Mulumebet Worku</p><br /> <p>We have identified phytochemicals that may be used in therapeutics that target galectins to control inflammatory diseases</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>We concluded an experiment where we tested the role of a relatively low amount of selenium biofortified alfalfa hay on the immune system of primiparous dairy cows during the transition from pregnancy to lactation. For the purpose we enrolled 18 pregnant heifers (10 Jersey and 8 Holstein) and randomly assigned to be supplemented with 1 kg/kg of BW of Se biofortified hay (3.2 mg Se/kg DM) or a control hay (0.4 mg Se/kg BW) from -40 day from expected parturition to 14 days post-partum. We collected blood, milk, and liver tissue for analysis of large panels of parameters, including phagocytosis, WBC count and differential, selenium level, and metabolic/inflammatory/oxidative parameters. Our results indicated a relatively positive effect on the antioxidative status of the animals but no effects on the immune system or performance, including SCC. However, data indicated a better liver status in cows receiving the Se biofortified hay.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Member: Jeffrey M. Boyd</p><br /> <p>We have examined the mechanisms used by <em>Staphylococcus aureus</em> to prevent intoxication by copper. These studies will help aid in the correct usage of ionic copper to prevent S. aureus spread and infections. We examined inhibitors of the Agr system which result in decreased virulence factor production. We characterized benzalkonium-templated mesoporous silica antimicrobial agents and showed that they effectively kill S. aureus.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Illinois</span></p><br /> <p>Member: Peter Constable</p><br /> <p>We characterized the clinical utility of a relatively inexpensive ($300) point-of-care pH meter and pH strips (Multistix 10 SG Reagent urine dipstick, pH Hydrion paper) in measuring milk pH as a predictor of milk SCC and the degree of inflammation in dairy cows at dry-off and freshening. Glandular inflammation at dry off and freshening was accompanied by an increase in milk pH; however, milk pH did not provide the required level of test sensitivity or specificity to identify quarters with increased SCC or presence of microbial growth. Test accuracy was inferior to that of the California Mastitis Test.</p><br /> <p>We characterized the clinical utility of three relatively inexpensive ($300) point-of-care sodium, potassium, and calcium ion selective electrode (ISE) meters and an electrical conductivity meter (Horiba, Japan) in measuring milk Na, K, and Ca concentrations and conductivity as predictors of milk SCC and the degree of inflammation in dairy cows at dry-off and freshening. Glandular inflammation at dry off increased milk Na concentration and conductivity, decreased milk K concentration, and had a variable effect on milk ionized Ca concentration; the latter was primarily an indirect effect of the change in milk pH. Cisternal milk Na concentration at dry off was the most accurate of the four tests for identifying quarters with increased SCC; however, test accuracy was not better than that of the California Mastitis Test. Diagnostic test performance was reduced in milk from fresh cows.</p><br /> <p>We characterized the clinical utility of three commercially available esterase tests (PortaCheck; PortaSCC color test; PortaSCC reader; PortaSCC quick test) in identifying the presence of inflammation in milk from dairy cows at dry-off and freshening. Glandular inflammation at dry off and freshening was accompanied by an increase in milk esterase activity as detected by the esterase test. The PortaSCC color test was the best performing PortaSCC test at dry-off and freshening, but required 45 minutes to produce a result. Based on the test sensitivity, cost, and analysis time, there does not appear to be a persuasive reason to select the PortaSCC color test over the traditional California Mastitis Test for diagnosing inflammation or the presence of microbial growth in glandular secretions from lactating dairy cows.</p><br /> <p>Values for pharmacokinetic variables are usually obtained in healthy animals, whereas drugs are frequently administered to diseased animals. We investigated cefquinome pharmacokinetics in healthy goats and goats with experimentally-induced mastitis. Five adult lactating goats received 75 mg of cefquinome intramammary infusion using a commercially available product into one udder half in healthy goats and goats with clinical mastitis that was induced by intracisternal infusion of 100 cfu of Staphylococcus aureus ATCC 29213 suspended in 5 ml of sterile culture broth. Experimentally induced mastitis significantly increased the California Mastitis Test score and pH, and decreased the maximal cefquinome concentration and shortened the half‐life in milk when compared to healthy goats. In conclusion, mastitis facilitated the absorption of cefquinome from the mammary gland of lactating goats and induced marked changes in milk pH, emphasizing the importance of performing pharmacokinetic studies of antimicrobial agents to treat mastitis in animals with clinical or subclinical mastitis. Note that cefquinome is not permitted for the treatment of mastitis in the United States.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Member: Kasey Moyes</p><br /> <p>In collaboration with Drs. Daniel Nelson (UMD) and Ronald Erskine (Michigan State University), we are examining the use of PlyC, a bacteriophage endolysin, as a new intramammary (IM) therapeutic for <em>S. uberis</em> mastitis in lactating dairy cows</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, B. Crooker, C. Gebhart, S. Wells, T. Schoenfus, B. Heins.</p><br /> <p>Contributors: P. Gorden (Iowa State), D. Nydam (Cornell), S. Sreevatsan (Michigan State), R. Fink (St. Cloud State), V. Machado (Texas Tech), P. Pinedo (Colorado State), A. Formiga (Oregon)</p><br /> <p>S.M. Rowe,* S.M. Godden, D.V. Nydam, A. Lago, A.K. Vasquez, E. Royster, J. Timmerman. Randomized Equivalence Study Evaluating the Efficacy of Two Commercial Internal Teat Sealants in Dairy Cows. The objective of this study was to evaluate a new ITS product (Lockout&reg;, Boehringer-Ingelheim Animal Health), by comparing it directly to Orbeseal&reg; in a multi-site, multi-herd, randomized, positively-controlled equivalence trial for health indicators during the dry period and during the first 100 days of lactation. At dry-off, cows were randomly allocated to be treated with Orbeseal&reg; or Lockout&reg; after blanket administration of a cloxacillin dry cow therapy (<strong>DCT</strong>) product. Cows were then followed from dry-off until 100 DIM. Intramammary infection (<strong>IMI</strong>) status at enrollment and at 1-13 DIM was determined using standard bacteriological methods, allowing for the measurement of IMI dynamics during the dry period (i.e. IMI cures and new IMI). Results showed that measures of quarter-level IMI dynamics were similar between ITS groups. Furthermore, Lockout&reg; was found to be equivalent to Orbeseal&reg; for dry period new IMI risk using an equivalence hypothesis test. There was no effect of treatment on risk for clinical mastitis and culling or death, or on SCC and milk yield during the first 100 DIM. One manuscript submitted in November, 2019 (in review).</p><br /> <p><strong>&nbsp;</strong></p><br /> <p>S.M. Rowe,<sup>1</sup> S.M. Godden, D.V. Nydam, P.J. Gorden, A. Lago, A.K. Vasquez, E. Royster, J. Timmerman, M.J. Thomas. Randomized controlled non-inferiority trial investigating the effect of two selective dry cow therapy protocols on antibiotic use and udder health: quarter-level outcomes. The objective of this study was to compare culture- and algorithm-guided SDCT programs to blanket dry cow therapy (BDCT) in a multi-site, randomized, natural exposure, non-inferiority trial on quarter-level infection dynamics during the dry period and on cow health and performance during the first 120 DIM. Two days before planned dry-off, cows in each of 7 herds were randomly allocated to BDCT (&ldquo;Blanket&rdquo;), culture-guided SDCT (&ldquo;Culture&rdquo;) and algorithm-guided SDCT (&ldquo;Algorithm&rdquo;). At dry-off, Blanket cows received an intramammary antibiotic (500mg ceftiofur hydrochloride) in all four quarters. Antibiotic treatments were selectively allocated to quarters of Culture cows by only treating quarters from which aseptically collected milk samples tested positive on the Minnesota Easy&reg; 4Cast&reg; plate after 30-40 hours of incubation. For Algorithm cows, antibiotic treatments were selectively allocated at the cow-level, with all quarters receiving antibiotic treatment if the cow met at least one of the following criteria: 1) any Dairy Herd Improvement Association (DHIA) test with a somatic cell count &gt; 200,000 cells / ml during the current lactation; 2) two or more clinical mastitis cases during the current lactation; and 3) one or more clinical mastitis cases in the 14 days period before enrollment. All quarters of all cows were treated with an internal teat sealant (ITS). Intramammary infection status at enrollment and at 1-13 DIM was determined using standard bacteriological methods. Results showed that the two SDCT approaches (culture- and algorithm-guided) each reduced antibiotic use at dry-off by 55%, without causing any negative impacts on intramammary infection dynamics during the dry period. We conclude that SDCT can be used in appropriate U.S. dairy herds to reduce antibiotic use while maintaining udder health. Two manuscripts submitted (in review).<br /> </p><br /> <p>Caixeta, L.S., Morley, P.S., Belk, K., Garry, F.B., Scheu, S., Parker, J, Noyes, N., Mijares, S., Hanes, A. Effect of selective dry cow therapy on the milk microbiome of dairy cattle with low somatic cell count. The purpose of this study was to determine the effects of SDCT on udder health and milk microbiome in separated milk cell pellets and cream in post-partum dairy cattle. 16S rRNA amplicon sequencing was performed and bioinformatics analysis of the sequence data is ongoing in order to identify differences between pellet and cream microbiomes for pre- and post-partum samples. Project ongoing.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Members: Jean-Philippe Roy, Mario Jacques, Simon Dufour, Marie Archambault, David Francoz</p><br /> <p>We synthesized and re-analyzed the available literature on use of an internal teat sealant (ITS) alone, without concomitant use of an antimicrobial for preventing new infections during the dry period and clinical mastitis in the subsequent lactation (Dufour). We concluded that an ITS performed better than an antimicrobial for preventing new infections.</p><br /> <p>&nbsp;</p><br /> <p>We also completed our investigations on a quarter-level selective dry cow treatment approach supported by milk culture (Roy). We concluded that a reduction in use of antibiotic of 50% could be achieved, without negative udder health or production impacts.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab has completed collecting data from a field study exploring <em>Staphylococcus aureus </em>strain variation among dairy cattle and farm workers on 21 dairy farms producing farmstead of artisan cheeses. Strain typing and characterization of antimicrobial susceptibility phenotypes and genotypes has been completed for approximately 160 isolates and cross species (zoonotic) transmission dynamics of <em>S. aureus</em> will be explored using these data.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Members: David Wilson</p><br /> <p>Contributors: Kerry Rood, Justine Britten, Zhongde Wang, Jacqueline LaRose Kurz, Robert Weiss, George Liu, Zhou Yang, John Middleton, Pamela Adkins, Gregory Goodell</p><br /> <p>Casein hydrolysate (CH) intramammary infusion was evaluated as a dry treatment.&nbsp; Udder halves were compared between control (dry cow antibiotic plus teat sealant) and 4 treatment groups including CH, one being CH alone.&nbsp; All halves returned to milking, statistically 50% of total-cow milk, following calving.&nbsp; At 7 or 10 days dry, milk indicators of involution were higher in CH cows; SCC was not different among treatment groups.&nbsp; Dry period new IMI post-calving for CH cows (88%) were significantly higher than for CH + teat sealant cows (25%).&nbsp; Use of casein hydrolysate combined with internal teat sealant may be an alternative to antibiotic dry treatment.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>WORK PLANNED FOR THE COMING YEAR, LISTED BY OBJECTIVE:</strong></p><br /> <p><strong>OBJECTIVE 1</strong></p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Identify and evaluate bacteria causing bovine mastitis in Louisiana. Test bacteria for antimicrobial resistance to mastitis therapeutics.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Continue progress on projects outlined above. Also, conducting field study of the long term impact (efficacy, production, milk quality) of antimicrobial therapy on mild clinical mastitis.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Evaluate the expression of Galectin variants in blood from cattle sheep and goats</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Mammary blood vessel structure will be examined in different regions of calf mammary glands that had been stimulated to experience different degrees of mammary growth and development in order to determine how blood vessel structure is altered during mammary growth. This vasculature examination has relevance and can be translated to understanding how blood vessels adapt during cases of mastitis and recruiting immune cells to localized sites of inflammation.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>We will examine the stimuli for the SrrAB and SaeSR, which are primary transcriptional regulators that control toxin, exoprotein, and biofilm formation in S. aureus. Determining the stimuli will ultimately be use to design small molecules to modulate the behavior of the organism.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>A. Crooker, S. M. Godden, J. D. Lippolis and J. B. Cole. Reducing mastitis in the dairy cow by increasing the prevalence of beneficial polymorphisms in genes associated with mastitis resistance. We have repeated the intramammary <em>E. coli</em> challenge with another 5 UH and 7 CH cows and will evaluate milk somatic cells from both studies to determine differences in expression of genes associated with immune response. We anticipate this will identify differential expression of genes that play important beneficial and detrimental roles in the magnitude and effectiveness of the immune response to mastitis. Data analysis will be completed and reported in 2020. Subsequent sequencing and identification of polymorphic differences in the genes and / or their regulatory components (transcription factors, etc.) between the Holstein genotypes will occur in 2020.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Continue evaluating the impact of heat stress on the microbiomes of dairy cattle.&nbsp; We are evaluating fecal, rumen, skin, and milk microbial populations of dairy cows pre, during, and post heat stress. Additionally, we plan to evaluate the fecal microbiome of cows that have experienced heat abatement strategies compared to those that have not.&nbsp; This component of our work is in collaboration with NE-1748 members from Mississippi (Stone).</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>In the coming year, we will describe incidence and elimination rates of non-aureus staphylococci intramammary infections during the dry and lactating periods and investigate host-related risk factors associated with these.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Zhao Lab at VT will study the mechanism underlying the repression of milk protein expression by LPS.</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Evaluate the effect of recombinant galectins and their antagonists on inflammatory gene expression in blood and isolated neutrophils from cows, sheep and goats</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>We are examining novel antimicrobials containing metal ions (tin, copper, zinc) in combination with organic antimicrobials. We hope to show that combining the compound with the metal ion will have a synergistic effect and increased antimicrobial activity.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Illinois</span></p><br /> <p>Characterize the clinical utility of quarter lactose concentration in diagnosing quarters that have inflammation or microbial growth present. Apply the new pharmacokinetic model for intramammary administration to new data sets to evaluate the models performance in characterizing the determinants of withdrawal time.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Noyes, L. Caixeta, B. Heins, V. Machado, P. Pinedo, A. Formiga, D. Van Nydam, J. Velez, H. Karremann. Catalyzing an open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case.</p><br /> <p>The long-term goal of this proposal is to ignite robust, systematic and application-driven microbiome research activity within the livestock community, thus producing a panoply of new, evidence-based tools to combat livestock disease and production challenges. Project objectives described above.&nbsp; Field to collect samples, plus laboratory analysis will continue into 2020.</p><br /> <p>Godden, S., P. Raynor, R. Singer, E. Royster, J. Hadrich, K. Janni, B. Crooker, J. Bender, B. Alexander. Investigation of the Relationship between Method of Processing Recycled Manure Solids Bedding, Udder Health, Milk Quality, Air Quality, Antimicrobial Resistance and Economics on Midwest Dairy Farms.&nbsp; Project objectives described above. We plan to resample all herds again in winter 2020 specifically for the udder health and milk quality objectives. Results will be forthcoming in 2020.&nbsp;&nbsp;</p><br /> <p>S.M. Rowe,* S.M. Godden, E. Royster, J. Timmerman, B.A. Crooker, M. Boyle. Cross-sectional study of the relationship among bedding materials, bedding bacteria counts and intramammary infection in late lactation dairy cows. Project details described above. Prepare and submit a second manuscript in 2020.</p><br /> <p>Nusrat Annie Jahan<sup>*</sup>, S. Godden, T. Schoenfus, C. Gebhart, E. Royster, S. Wells, J. Timmerman, and R. Fink. Evaluation of the Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS) system in the detection of Mastitis pathogens from Bovine milk samples. Project details described above. Submit manuscript in 2020.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Develop imaging techniques to evaluate the association between teat morphology and intramammary infection and mastitis in lactating cows, peripartum heifers, and dry cows. Additionally, we plan to finish our characterization of antimicrobial susceptibility among Gram-positive bacteria isolated from the feces of dairy heifers pre and post exposure to intramammary pirlimycin.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>The Barlow lab will continue studies of <em>S. aureus</em> strain variation and transmission dynamics on small dairy farms. The Barlow lab will continue the observational field studies comparing milk quality and mastitis prevalence on organic dairy herds utilizing different bedding management practices. NE-1748 members from Minnesota (Godden) and Missouri (Adkins) will collaborate in this research</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>OBJECTIVE 3</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Continue evaluation of botanical formulations from plants for antimicrobial activity against mastitis pathogens.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Continue progress on projects outlined above. Also, conducting field study of the long term impact (efficacy, production, milk quality) of antimicrobial therapy on mild clinical mastitis.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Determine the effect of dietary galectin antagonists on immune gene expression in small ruminants</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>We are going to evaluate the use of a teat sealant in non-lactating dairy heifers in their efficacy of preventing new intramammary infections at different stages of gestation. This will be done to determine if there is a better time to administer teat sealant to prevent more infections during late gestation than others.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>We have extracted the RNA from the liver (-10 and 7 day relative to parturition) and macrophages isolated from milk (10 day post-partum) from the Jersey cows from the above experiment. We plan to have the RNAseq performed in those samples (see Project # ORE00174).</p><br /> <p>We will perform an experiment on sheep where we will test the hypothesis that Se biofortified chicory improve the response to intramammary infection (see Project # ORE00170).</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>We examining the mechanisms by which copper ions kills <em>S. aureus</em>. We are describing and determining the mode of action of how a novel antimicrobial with potent activity against <em>S. aureus</em>.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">University of Illinois</span></p><br /> <p>Apply segmented regression to existing data sets to identify the SCC cut point that differentiates healthy from inflamed quarters at dry-off and at freshening in dairy cattle.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>S.M. Rowe,<sup>1</sup> S.M. Godden, D.V. Nydam, P.J. Gorden, A. Lago, A.K. Vasquez, E. Royster, J. Timmerman, M.J. Thomas. Randomized controlled non-inferiority trial investigating the effect of two selective dry cow therapy protocols on antibiotic use and udder health: quarter-level outcomes. Project details described above. Submit third manuscript in 2020.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>We will investigate the impact of recycled manure bedding on subclinical and clinical mastitis incidence and on distribution of the pathogens involved (Dufour). We will also evaluate the precision and economic impact of different selection strategies at drying off (Dufour). We will also evaluate diagnostic precision of different sampling approaches for diagnosing <em>Salmonella </em>Dublin in dairy herd (Dufour). We are also investigating the reduction or shift in use of antimicrobials on dairies following the implementation of a provincial regulation limiting the use of antimicrobials that are deemed to be of critical importance for humans (Dufour). We will described antimicrobial resistance in isolates obtained from dairy herds (Archambault).</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>In collaboration with Tom McFadden&rsquo;s group at the University of Missouri, Zhao Lab at VT is investigating oxidative damage and the role of Nrf2 in antioxidation in the mammary gland during mastitis. The aim of this work is to find natural means to activate Nrf2 signaling to control mastitis&rsquo;s damage to the udders.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>One publication near submission, others in preparation regarding casein hydrolysate as a dry treatment.&nbsp; This includes histological measurements to quantify mammary involution in different regions of the gland compared among treatments.</p>

Publications

<p><strong>Peer-Reviewed Literature</strong></p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Moore-Foster, R., B. Norby,&nbsp;R. L. Schewe, R. Thomson, P. C. Bartlett&nbsp;and R. J. Erskin. Short communication: Herd-level variables associated with overmilking in Michigan dairy herds. J.Dairy Sci.&nbsp;2019 102:8400-8404.</p><br /> <p>Erskine, R.J., B. Norby, L. M. Neuder and R. S. Thomson. 2019. Decreased milk yield is associated with delayed milk ejection. J. Dairy Sci. 102:6477-6484.</p><br /> <p>Moore-Foster R, B. Norby, R. L. Schewe, R. Thomson, P. C. Bartlett, and R. J. Erskine. 2019. Herd-level variables associated with pre-milking stimulation time in Michigan dairy herds.&nbsp;J. Dairy Sci. 102:2544-2550.&nbsp;</p><br /> <p>Moore-Foster R, B. Norby, R. L. Schewe, R. Thomson, P. C. Bartlett, and R. J. Erskine. 2019. Herd-level variables associated with delayed milk ejection in Michigan dairy herds.&nbsp;J. Dairy Sci. 102:696-705.</p><br /> <p>Contreras G.A., J. De Koster, J. de Souza, J. Laguna, V. Mavangira, R. K. Nelli, J. Gandy, A. L. Lock, and L. M. Sordillo. 2019. Lipolysis modulates the biosynthesis of inflammatory lipid mediators derived from linoleic acid in adipose tissue of periparturient dairy cows. J. Dairy Sci.&nbsp;2019 Nov 20. pii: S0022-0302(19)31041-0. doi: 10.3168/jds.2019-17256. [Epub ahead of print]</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Emmanuel K. Asiamah, Mario Vailati-Riboni, Zheng Zhou, Tianle Xu, Juan J. Loor, Keith Schimmel, Mulumebet Worku Rumen-protected methionine supplementation during the peripartal period alters the expression of galectin genes associated with inflammation in peripheral neutrophils and secretion in plasma of Holstein cow 2019 Journal of Dairy Research</p><br /> <p>Eluka-Okoludoh, E.,&nbsp;Ekwemalor, K.,&nbsp;Adjei-Fremah, S.,&nbsp;Mulakala, B.,&nbsp;&amp; Worku, M. (2019).&nbsp;Galectin-8 Modulates Innate and Adaptive Immune Response Genes in Bovine Neutrophils.&nbsp;Journal of Molecular Biology Research,&nbsp;9(1),&nbsp;24-32. <em>2019 Molecular Biology Research<strong>.</strong></em></p><br /> <p>S Adjei-Fremah, M Worku, MO De Erive, F He, T Wang, G Chen . Effect of microfluidization on microstructure, protein profile and physicochemical properties of whole cowpea flours. Innovative Food Science &amp; Emerging Technologies. 57, 102207<em>2019, Elsevier</em></p><br /> <p>EK Asiamah, K Ekwemalor, S Adjei-Fremah, B Osei, R Newman, M Worku . Natural and synthetic pathogen associated molecular patterns modulate galectin expression in cow blood. Journal of animal science and technology 61 (5), 245.<em>2019, J Anim Sci Technol.</em></p><br /> <p>S Adjei-Fremah, M Worku . Cowpea polyphenol extract regulates galectin gene expression in bovine blood. Animal Biotechnology, 1-12.<em>2019, Animal Biotechnology.</em></p><br /> <p>JE Koltes, JB Cole, R Clemmens, RN Dilger, LM Kramer, JK Lunney. A vision for development and utilization of high-throughput phenotyping and big data analytics in livestock. Frontiers in Genetics 10, 1197.<em>2019, Frontiers in Genetics</em></p><br /> <p>&nbsp;Mulakala, B., Adjei-Fremah, S., Eluka-Okoludoh, E.,&nbsp;Ekwemalor, K.,&nbsp;Ibrahim, S. and Worku, M. (2019). Cellular Glycans and their Binding Proteins.&nbsp;EC Microbiology&nbsp;15.2 (2019): 108-111.<em>2019, EC Microbiology&nbsp;</em></p><br /> <p>Ekwemalor, K.,&nbsp;Asiamah, E., Eluka-Okoludoh, E., Mulakala, B., Adjei-Fremah, S., &amp; Worku, M. (2018). Shifts in Gut Microbe Population in Periparturient Goats. CPQ Microbiology (2018) 1: 5 Research Article.&nbsp;Microbiology,&nbsp;1(5), 01-06.<em>2018, Microbiology</em></p><br /> <p>Osei, B., Worku, M.,&nbsp;Eluka-Okoludoh,&nbsp;Adjei-Fremah, S.,&nbsp;Asiamah, E.,&nbsp;E.,&nbsp;Ekwemalor, K.,&nbsp;&amp;&nbsp;Mulakala, B.&nbsp;(2018).Galectin secretion and modulation in sheep blood.&nbsp;Journal&nbsp;of Molecular Biology Research,&nbsp;8(1),&nbsp;183.&nbsp;<em>2018, Molecular Biology Research</em></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Enger, B.D. 2019. Invited Review: Reevaluating how mastitis reduces milk yield: Discussion of competitive substrate utilization. Appl. Anim. Sci. 35:408&ndash;415.</p><br /> <p>Enger, B.D., H.L.M. Tucker, S.C. Nickerson, C.L.M. Parsons, and R.M. Akers. 2019. Effects of Staphylococcus aureus intramammary infection on the expression of estrogen receptor &alpha; (ESR1) and progesterone receptor (PGR) in mammary glands of nonlactating cows administered estradiol and progesterone to stimulate mammary growth. J. Dairy Sci. 102:2807-2617.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Shana Jaaf, Angela Krueger, Brandon Batty, Charles T. Estill$, and Massimo Bionaz. One% BW of selenium biofortified alfalfa improves selenium status and glutathione peroxidase activity in peripartum dairy cows and their calves. Submitted to Journal of Dairy Research</p><br /> <p>Shana Jaaf. PhD thesis. Effect of agronomically Se biofortified hay on oxidative status, metabolic and inflammation markers, and immune response of transition primiparous dairy cows and their calves. Defended the 3 December 2019.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Dubovoy V., Ganti A., Zhang T., Al-Tameemi H.M., Cerezo J., Boyd J.M.*, Asefa T.* One-pot hydrothermal synthesis of benzalkonium-templated mesoporous silica antimicrobial agents. <em>Journal of the American Chemical Society. </em>2018 Oct. PMID: 30260224</p><br /> <p>Rosario-Cruz Z., Eletsky A., Daigham N.S., Al-Tameemi H.M., Swapna G.V.T., Szyperski T., Montelione G.T., and Boyd J.M.* The <em>copBL</em> operon protects <em>Staphylococcus aureus</em> from copper toxicity: Cbl is an extracellular membrane-associated copper-binding protein. <em>Journal of Biological Chemistry.</em> 2019 Jan. PMID: 30655293</p><br /> <p>Bezar I.F., Mashruwala A.A., Boyd J.M., Stock A.M., Drug-like Fragments Inhibit <em>agr-</em>Mediated Virulence Expression in <em>Staphylococcus aureus</em>. <em>Nature Scientific Reports. </em>2019 May PMID: 31043623</p><br /> <p>Austin C.M., Garabaglu S., Krute C.N., Ridder M.J., Seawell N.A., Markiewicz M.A., Boyd J.M., Bose J.L. Contribution of YjbIH to virulence factor expression and host colonization in <em>Staphylococcus aureus</em>. <em>Infection and Immunity.</em> 2019 March. PMID: 30885928</p><br /> <p>Mashruwala A.A., Eilers B.J., Fuchs A., Earle C.A., Van De Guchte A., Copi&eacute; V., Boyd J.M. The ClpCP complex modulates respiratory metabolism in <em>Staphylococcus aureus</em> and is regulated in a SrrAB-dependent manner. <em>Journal of Bacteriology.</em> 2019 July. PMID: 31109995</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Illinois</span></p><br /> <p>Kandeel, S.A., Megahed, A.A., Ebeid, M.H. and Constable, P.D., 2019. Ability of milk pH to predict subclinical mastitis and intramammary infection in quarters from lactating dairy cattle. J. Dairy Sci. 102(2):1417-1427.</p><br /> <p>Kandeel, S.A., Megahed, A.A. and Constable, P.D., 2019. Evaluation of hand‐held sodium, potassium, calcium, and electrical conductivity meters for diagnosing subclinical mastitis and intramammary infection in dairy cattle. J. Vet. Intern. Med. 1-11. https://doi.org.10.1111/jvim.15550.</p><br /> <p>Kandeel, S.A., Megahed, A.A., Ebeid, M.H. and Constable, P.D., 2019. Evaluation of 3 esterase tests for the diagnosis of subclinical mastitis at dry-off and freshening in dairy cattle. J. Dairy Sci. 102(2):1402-1416.</p><br /> <p>Megahed, A.A., Hiew, M., Gr&uuml;nberg, W., Trefz, F.M. and Constable, P.D., 2019. Evaluation of the analytical performance of a portable ion-selective electrode meter for measuring whole-blood, plasma, milk, abomasal-fluid, and urine sodium concentrations in cattle. J. Dairy Sci. 102(8):7435-7444.</p><br /> <p>El Badawy, S.A., Amer, A.M., Kamel, G.M., Eldeib, K.M. and Constable, P.D., 2019. Pharmacokinetics and pharmacodynamics of intramammary cefquinome in lactating goats with and without experimentally induced <em>Staphylococcus aureus</em> mastitis. J. Vet. Pharmacol. Therap. 42:452-460.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>M.A. Crookenden, K.M. Moyes<strong>,</strong> B. Kuhn-Sherlock, K. Lehnert, C.G. Walker, J.J. Loor, M.D. Mitchell, A. Murray, V.S.R. Dukkipatti, M. Vailati-Riboni, A. Heiser and J.R. Roche.&nbsp;&nbsp; 2019. Transcriptomic analysis of circulating neutrophils in metabolically stressed peripartal grazing dairy cows. J. Dairy Sci. 102:7408-7420.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Patel, K.,* S.M. Godden, E. Royster, B.A. Crooker, J. Timmerman and L. Fox. 2019. Relationships between Bedding Materials, Bedding Bacteria Counts, Udder Hygiene, Milk Quality and Udder Health on United States Dairy Herds. J. Dairy Sci. 102:10213&ndash;10234</p><br /> <p>Murphy, S., D. Kent, N. Martin, R. Evanowski, K. Patel, S. Godden and M. Wiedmann.&nbsp;2019. Bedding and bedding management practices are associated with mesophilic and thermophilic spore levels in bulk tank raw milk. J. Dairy Sci. 102: 102:6885&ndash;6900</p><br /> <p>Jenkins,S., E. Okello, P.V. Rossito, T.W. Lehenbauer, J.Champagne, M.C.T. Penedo, A.G. Arruda, S. Godden, P. Rapnicki, P.J. Gorden, L.L. Timms, and S.S. Aly. 2019. Molecular epidemiology of coagulase-negative Staphylococcus species isolated at different lactation stages from dairy cattle in the United States.&nbsp; PeerJ&nbsp;7:e6749&nbsp;<a href="https://doi.org/10.7717/peerj.6749">https://doi.org/10.7717/peerj.6749</a></p><br /> <p>Afifi, M., F. Kabera, H. Stryhn, J. Roy, L. Heider, S. Godden, W. Montelpare, J. Sanchez and S. Dufour. 2018. Antimicrobial-based dry cow therapy approaches for cure and prevention of intramammary infections: a protocol for a systematic review and meta-analysis.&nbsp; Animal Health Research Reviews. 19:74-48. https://doi.org/10.1017/S1466252318000051</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Adkins PRF, Middleton JR.&nbsp; 2018.&nbsp; Methods for diagnosing mastitis.&nbsp; <em>Vet Clin North Am Food Anim Pract.</em>&nbsp; 34(3):479-491.&nbsp; Invited article.&nbsp;</p><br /> <p>Wilson D, Middleton J, Adkins P, Goodell G.&nbsp; 2019.&nbsp; Test agreement among biochemical methods, MALDI-TOF and 16S rRNA sequencing for the identification of microorganisms isolated from bovine milk.&nbsp; <em>J Clin Microbiol</em>.&nbsp; 57(3):1-8&nbsp; pii: JCM.01381-18. doi: 10.1128/JCM.01381-18. [Epub 19 Mar 19]</p><br /> <p>Bernier Gosselin V, Dufour S, Adkins PRF, Poock S, Pithua P, Middleton JR.&nbsp; 2019.&nbsp; Longitudinal microbiological evaluation of subclinical non-aureus staphylococcal intramammary infections in a dairy goat herd.&nbsp; <em>Vet Microbiol</em>.&nbsp; 230:156-163.&nbsp;</p><br /> <p>Bernier Gosselin V, Dufour S, Calcutt MJ, Adkins PRF, Middleton JR.&nbsp; 2019.&nbsp; Staphylococcal intramammary infection dynamics and their relationship with milk quality parameters in dairy goats over the dry period.&nbsp; <em>J Dairy Sci</em>.&nbsp; 102(5):4332-4340.</p><br /> <p>Bernier Gosselin V, Dufour S, Adkins PRF, Middleton JR.&nbsp; 2019.&nbsp; Persistence of coagulase negative staphylococcal intramammary infection in dairy goats.&nbsp; <em>J Dairy Res</em>.&nbsp; 86(2):211-216.&nbsp;</p><br /> <p>Bernier Gosselin V, Dufour S, Middleton JR.&nbsp; 2019.&nbsp; Association between species-specific staphylococcal intramammary infection and milk somatic cell score over time in dairy goats.&nbsp; <em>Prev Vet Med. </em>174:104815 <em>&nbsp;</em>doi: 10.1016/j.prevetmed.2019.104815.&nbsp; [Epub 2 Nov 19].</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Mass&eacute;, J., S. Dufour, and M. Archambault, Characterization of Klebsiella and Raoultella isolates obtained from dairy cattle clinical mastitis cases. Journal of&nbsp; Dairy Science, Submitted.</p><br /> <p>Lard&eacute;, H., et al., Assignment of Canadian Defined Daily Doses and Canadian Defined Course Doses for quantification of antimicrobial usage in cattle. Frontiers Veterinary Science, Submitted.</p><br /> <p>Kabera, F., et al., Evaluation of a quarter-based selective dry cow therapy using Petrifilm on-farm milk culture: A randomized controlled trial. Journal of Dairy Science, Submitted.</p><br /> <p>Lasprilla-Mantilla, M., et al., Impact of Recycled Manure Solids Bedding on Spread of Gastro-intestinal Parasites in Environment of Dairies and Milk. Journal of Dairy Science, In press.</p><br /> <p>Belage, E., et al., A qualitative study of Ontario dairy farmer attitudes and perceptions towards implementing recommended milking practices. Journal of&nbsp; Dairy Science, Accepted. 102(10): p. 9548-9557.</p><br /> <p>Naaushad, S., et al., Staphylococcus debuckii sp. nov., a coagulase-negative species from bovine milk. Journal of Systematic and Evolutionary Microbiology, 2019. 69(8): p. 2239-2249.</p><br /> <p>Gagnon, M., et al., Effect of recycled manure solids as bedding on bulk tank milk and cheese microbiological quality. Journal of&nbsp; Dairy Science, 2019. 103.</p><br /> <p>Dufour, S., et al., Systematic review of non-antimicrobial approaches at drying-off for treating and preventing intramammary infections in dairy cows. Part 1. Meta-analyses on efficacy of using an internal teat sealant without a concomitant antimicrobial treatment. Animal Health Research Reviews, 2019.</p><br /> <p>Dufour, S., J. Labrie, and M. Jacques, The Mastitis Pathogens Culture Collection. Microbiology - Resource Announcements, 2019. 8(15): p. 1-2.</p><br /> <p>Corbeil, A., et al., Short communication: Search for superantigen genes in coagulase-negative staphylococci isolated from bovine milk in Canada Journal of&nbsp; Dairy Science, 2019. 102: p. 2008-2010.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Alexander Jonathan Spitzer, Qing Tian, Ratan Choudhary, and Feng-Qi Zhao. (2020). Bacterial endotoxin induces oxidative stress and reduces milk protein expression and hypoxia in the mammary gland. Oxidative Medicine and Cellular Longevity, minor revision</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Kurz JP, Yang Z, Weiss RB, Wilson DJ, Rood KA, Liu GE, Wang Z: A genome-wide association study for mastitis resistance in phenotypically well-characterized Holstein dairy cattle using a selective genotyping approach. Immunogenetics Sept 30, doi: 10.1007/s00251-018-1088-9. (Epub ahead of print), 2018. 71:1:35-47, 2019.</p><br /> <p>Wilson DJ, Middleton JR, Adkins PRF, Goodell GM: Test agreement among biochemical methods, MALDI-TOF and 16S rRNA sequencing for the identification of microorganisms isolated from bovine milk.&nbsp; J Clin Microbiol 57:3: doi: 10.1128/JCM.01381-18., 2019.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Abstracts</strong></p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>E Eluka-Okoludoh, E Asiamah, K Ekwemalor, B Mulakala, S Adjei-Fremah. PSVIII-37 Modulation of innate and adaptive immune response genes in bovine neutrophil by Galectin-8. Journal of Animal Science 96 (suppl_3), 31-32<em>21018, Journal of Animal Science</em></p><br /> <p>B Osei, E Asiamah, K Ekwemalor, S Adjei-Fremah, M Worku. PSVI-21 Coproantibodies as indicators of the periparturient rise in fecal egg counts in St Croix Sheep. Journal of Animal Science 96 (suppl_3), 461-461<em>2018, Journal of Animal Science</em></p><br /> <p>E Asiamah, K Ekwemalor, S Adjei Fremah, M Worku, B Osei. PSVIII-41 Unique Signatures of Galectin Expression in Cow Blood Exposed to Microbial Cell Wall Antigens. Journal of Animal Science 96 (suppl_3), 33-33<em>Journal of Animal Science, 2018</em></p><br /> <p>B Mulakala, E Eluka-Okoludoh, S Adjei-Fremah, E Asiamah, K Ekwemalor. PSVIII-29 Galectin 9 secretion in cow milk a marker for homostasis and health. Journal of Animal Science 96 (suppl_3), 30-31<em>2018, Journal of Animal Science</em></p><br /> <p>K Ekwemalor, E Asiamah, S Adjei-Fremah, B Osei, E Eluka-Okoludoh. PSVI-32 Pathogen-associated molecular patterns induce differential galectin-3 activation in Goat blood. Journal of Animal Science 96 (suppl_3), 464-464<em>2018, Journal of Animal Science</em></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Jaaf S., M. Mezzetti, A. Krueger, B. Batty, M. Premi, F. Perrotta, J. Foster, M. Hanlon, E. Trevisi, and M. Bionaz. Selenium biofortified alfalfa hay supplemented during transition period improves oxidative stress and some metabolic and inflammation parameters in dairy cows and their calves. 37th ADSA Discovery Conference, October 28-31, Chicago, IL</p><br /> <p>Jaaf S., B. Batty, A. Krueger, G. Bobe, C. Estill, and M. Bionaz. Effect of agronomic selenium biofortified alfalfa hay on selenium status and glutathione peroxidase activity in transition dairy cows and their calves. 2019 PNW Animal Nutrition Conference. Boise (ID)</p><br /> <p>Jaaf S., Mezzetti M., Krueger A., Batty B., Belveal J., Premi M., Foster J., Trevisi E., Bobe G., Estill C., and M. Bionaz. 2018. Effect of selenium-enriched hay on Se concentration in blood and milk, immune function, and performance in dairy cows during the transition period. 2018 ADSA Annual Meeting Abstract#268</p><br /> <p>Mezzetti M., Jaaf S., Busato S., Premi M., Trevisi E., Bobe G., and M. Bionaz. 2018. Calves born from cows fed with alfalfa enriched with selenium have higher Se in blood and higher phagocytosis. 2018 ADSA Annual Meeting Abstract#154</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>Hanna M. Mader, C.M. Scholte, D. Biswas and K.M. Moyes.&nbsp; 2019. Immune response to <em>Escherichia coli </em>induced bovine mastitis in lactating dairy cows.&nbsp; American Dairy Science Association (ADSA) Annual Meetings, Cincinnnati, OH.</p><br /> <p>S.S. Potts, K.M. Brady, C.M. Scholte, K.M. Moyes and R.A. Erdman.&nbsp; 2019. Rumen- protected choline or methionine affects the choline metabolite profile of plasma&nbsp; and milk and alters expression of genes associated with choline and lipid metabolism in periparturient cows. American Dairy Science Association (ADSA)&nbsp; Annual Meetings, Cincinnnati, OH.</p><br /> <p>C.M. Scholte, A.M. Fisher, H.M. Mader, D. Biswas and K.M. Moyes*.&nbsp; 2019. The effects of citral as a therapeutic treatment for <em>Escherichia coli</em> mastitis in lactating dairy cattle. American Dairy Science Association (ADSA) Annual Meetings, Cincinnnati, OH.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Lippolis, J. D., E. J. Putz, T. A. Reinhardt, E. Casas, W. J. Weber and B. A. Crooker. 2019. What 50 years of breeding has done to the ability of Holsteins to fight mastitis. Conference of Research Workers in Animal Diseases. November 3. Chicago, IL.</p><br /> <p>Dean, C., Slizovskiy, I., Crone, K., Heinz, B., Caixeta, L., Noyes, N. Sampling considerations for conducting large-scale microbial ecology studies of cow udder epithelium. Proceedings of the 99<sup>th</sup>&nbsp;Annual Conference of Research Workers in Animal Diseases, Chicago, IL, 2018. Abstract 198.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Wattenburger K, Schmidt R, Placheta L, Middleton J, Adkins P. 2019.&nbsp; Evaluation of aspectic techniques used to collect bovine milk samples. MU Life Sciences Week.&nbsp;University of Missouri, Columbia, MO.&nbsp; April 15-20, 2019. Poster.</p><br /> <p>Witzke MC, Rodrigues RO, Shangraw EM, Ericsson AC, McFadden TB, Adkins PRF. 2019.&nbsp; Experimental heat stress alters the fecal microbiome of dairy cattle. MU Life Sciences Week.&nbsp;University of Missouri, Columbia, MO.&nbsp; April 15-20, 2019. Poster.</p><br /> <p>Wattenburger K, Schmidt R, Placheta L, Middleton JR, Adkins PRF.&nbsp; 2019.&nbsp; Evaluation of aseptic techniques used to collect bovine milk samples.&nbsp; Proceedings the 42<sup>nd</sup> Annual CVM Research Day (Phi Zeta).&nbsp; 10 May 2019.&nbsp; Abstract #26.&nbsp; Poster.</p><br /> <p>Schmidt R, Ierardi R, Placheta L, Adkins PRF, Middleton JR.&nbsp; 2019.&nbsp; Epidemiology of mastitis pathogens on Amish Dairy Farms in Missouri.&nbsp; Proceedings the 42<sup>nd</sup> Annual CVM Research Day (Phi Zeta). 10 May 2019.&nbsp; Abstract #52.&nbsp; Oral presentation.</p><br /> <p>Rivero L, Witzke M, Adkins PRF, Middleton JR.&nbsp; 2019.&nbsp; Understanding bovine mammary gland niches for staphylococci.&nbsp; Proceedings the 42<sup>nd</sup> Annual CVM Research Day (Phi Zeta). 10 May 2019.&nbsp; Abstract #62.&nbsp; Oral presentation.</p><br /> <p>Witzke MC, Rodrigues RO, Shangraw EM, Ericsson AC, McFadden TB, Adkins PRF. 2019.&nbsp; Bovine fecal microbiome is altered by experimental heat stress and feed restriction. Proceedings of the 42<sup>th</sup> Annual CVM Research Day (Phi Zeta).&nbsp; University of Missouri, May 10, 2019. Oral presentation.</p><br /> <p>Bernier Gosselin V, Middleton JR.&nbsp; 2019.&nbsp; Advances in understanding of epidemiology of subclinical staphylococcal mastitis in dairy goats.&nbsp; Ontario Small Ruminant Veterinary Conference.&nbsp; June 17-19. Oral presentation.</p><br /> <p>Ankney S, Rivero L, Witzke M, Naclerio A, Adkins PR, Middleton JR.&nbsp; 2019.&nbsp; Defining causes of intramammary infection in dairy cattle using a novel sampling technique.&nbsp; National Veterinary Scholars Symposium. Worcester, Massachusetts.&nbsp; July 26<sup>th</sup>.&nbsp; Poster.</p><br /> <p>Naclerio A, Witzke M, Adkins P. Antimicrobial resistance in bacteria of dairy cattle exposed to intramammary pirlymcin hydrochloride. National Veterinary Scholars Symposium.&nbsp; Cummings School of Veterinary Medicine hosted at Worcester State University. Worcester, MA, July 25-27, 2019.&nbsp; Poster</p><br /> <p>Rivero L, Witzke M, Adkins PRF, Middleton JR.&nbsp; 2019.&nbsp; Understanding bovine mammary gland niches for staphylococci using different milk sampling techniques.&nbsp; American Association of Bovine Practitioners.&nbsp; St. Louis, Missouri.&nbsp; September 12-14.&nbsp; Oral presentation.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Kabera, F., et al. Quarter-based Selective Dry Cow Therapy using on-farm diagnostics: results of a Randomized Controlled Trial. in National Mastitis Council annual meeting. 2019. Savannah, Georgia.</p><br /> <p>Beauchemin, J., et al. Comparison of microbiological contamination of recycled manure solid bedding with straw bedding in dairies farms. in 5th International Congress on Pathogens at the Human-Animal Interface. 2019. Qu&eacute;bec, QC, Canada.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>F.-Q. Zhao, T.B. McFadden, R.K. Choudhary, E.M. Shangraw, R.O. Rodrigues, A.J. Spitzer. Intramammary endotoxin challenge elicits time-dependent local and systemic effects on lactating bovine mammary glands. CRWAD 2019, Chicago. Oral</p><br /> <p>Choudhary RK, Spitzer A, McFadden TB, Shangraw EM, Rodrigues RO, Linder HF, Zhao F-Q (2019) Quantitative histological changes in lactating bovine mammary gland after endotoxin challenge. ADSA Annual Meeting 2019, Cincinnati, Ohio. Oral</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Conference Proceedings </strong></p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Erskine, R. J. How should we milk a cow? Let the cows decide! Seminar presented to the staff of the U.S. Congress on behalf of the National Coalition for Food and Agricultural Research, Washington. D.C., October, 9, 2018.</p><br /> <p>Erskine, R. J. Association of delayed milk yield with delayed (bimodal) milk ejection. Seminar presented at the Conference of Research Workers in Animal Disease, Chicago, IL, December 3, 2018.</p><br /> <p>Erskine, R.J. and J. R. Middleton. Mastitis Therapy: Why, What, When, and How? Short course presented at the 59<sup>th</sup> Annual Meeting of the National Mastitis Council, Savannah, GA, January 30, 2019.</p><br /> <p>Erskine, R.J. Effect of bimodal milk letdown on milk production. Research Summary presented at the 52<sup>nd</sup> Annual Conference of the Am. Assoc. Bovine Pract. St. Louis, MO, September 13, 2019.</p><br /> <p>Erskine, R. J. Animal Agriculture and Antimicrobial Resistance in Michigan. Seminar presented at the Michigan One Health Antimicrobial Resistance Summit. Dearborn, MI, September 18, 2019.</p><br /> <p>Erskine, RJ, Martinez, RO, and Kayitsinga, J. Labor Needs on the Dairy Farm-From the Workers Perspective. Seminar and Panel Discussion presented the 30<sup>th</sup> Anniversary of the Julian Samora Research Institute, East Lansing, MI, November 1, 2019.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Enger, B.D. 2019. Contemplating the energetic consequences of bovine mastitis. Tri-State Dairy Nutrition Conference Annual Meeting Proceedings. Pages 89-100.</p><br /> <p>Enger, B.D. 2019. Mastitis in dairy heifers: The damage we cannot see. National Mastitis Council 58th Annual Meeting Proceedings. Pages 54-61.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Boyd J.M. Dissecting <em>Staphylococcus aureus</em> physiology to decrease disease burden. University of Wisconsin-Madison, Dept. of Bacteriology, Madison, WI 11/2018</p><br /> <p>Boyd J.M. Dissecting <em>Staphylococcus aureus</em> physiology to decrease disease burden. Biology department. Georgetown University, Washington, DC, 11/2018.</p><br /> <p>Boyd J.M. Dissecting <em>Staphylococcus aureus</em> physiology to decrease disease burden. Theobald smith society-New Jersey Branch of ASM. 11/2018.</p><br /> <p>Boyd J.M. Metal ion homeostasis in <em>Staphylococcus aureus</em>. Staphylococcal Diseases Gordon Conference, Castelldefels, Spain, 8/2019</p><br /> <p>Boyd J.M. The role of Bacillithiol in metal ion homeostasis. International meeting on: Thiol-based switches and redox regulation - from microbes to men Sant Feliu de Guixols Spain, 9/2019</p><br /> <p>Boyd J.M. Metal ion homeostasis in <em>Staphylococcus aureus</em>. Dept. of Cell Biology, Microbiology, and Molecular Biology, University of South Florida 11/2019.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Rowe, S.M.*, Godden, S.M., Nydam, D.V., Lago, A., Royster, E., Vasquez, A., Randomized controlled trial evaluating the efficacy of two commercial internal teat sealants in dairy cows. <em>American</em> <em>Association of Bovine Practitioners </em>Proc. 52nd Annual Conference. Sept. 12-14, 2019. St Louis, MO</p><br /> <p>Rowe, S.M.*, Godden, S.M., Nydam, D.V., Gorden, P., Lago, A., Royster, E., Vasquez, A., Thomas, M. Selective dry cow therapy on U.S. dairy farms: Impact on udder health. <em>American Association of Bovine Practitioners </em>Proc. 52nd Annual Conference. Sept. 12-14, 2019. St Louis, MO</p><br /> <p>Godden, S.M.*, Royster, E., Rowe, S.M., Patel, K., Timmerman, J., Crooker, B., Fox, L., Chipping Away at the Tough Questions about Bedding Management and Mastitis. <em>American Association of Bovine Practitioners </em>Proc. 52nd Annual Conference. Sept. 12-14, 2019. St Louis, MO</p><br /> <p>Rowe, S.M*. Godden, S.M., Nydam, D.V., Gorden, P., Lago, A., Royster, E., Vasquez, A. Late Breaking - Selective dry cow therapy on U.S. dairy farms: Impact on udder health. <em>American Dairy Science Association </em>annual meeting. June 21-23, 2019. Cincinnati, OH: page 1<br /> Rowe, S.M.*, Godden, S.M., Royster, E., Timmerman, J., Boyle, M. Investigation Of The Relationship Between Bedding Bacteria Count And Intramammary Infection In Late Lactation Dairy Cows: Interim Findings. Proc. 58th Annual meeting of the <em>National Mastitis Council. Jan 29-Feb. 1, 2019. Savannah, GA. Pg.</em> 153-154.</p><br /> <p>Rowe, S.M.*, Godden, S.M., Royster, E., Timmerman, J., Boyle, M. Investigation of the Relationship Between Udder Towel Hygiene, Udder Towel Management and Intramammary Infection in Late-lactation Dairy Cows. Proc. 58th Annual meeting of the <em>National Mastitis Council. Jan 29-Feb. 1, 2019. Savannah, GA.</em>: 153-154.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Wattenburger K, Schmidt R, Middleton JR, Adkins PRF.&nbsp; 2019.&nbsp; Evaluation of aseptic techniques used to collect bovine milk samples.&nbsp; Proceedings of the 58<sup>th</sup> Annual Meeting of the National Mastitis Council, Savannah, GA.&nbsp; Jan 30-Feb 1.&nbsp; Poster.</p><br /> <p>Bernier Gosselin V, Dufour S, Adkins PRF, Middleton JR<sup>b</sup>.&nbsp; 2019.&nbsp; Infection dynamics of staphylococcal intramammary infections over the dry period in dairy goats.&nbsp; Proceedings of the 58<sup>th</sup> Annual Meeting of the National Mastitis Council, Savannah, GA.&nbsp; Jan 30-Feb 1.&nbsp; Poster.</p><br /> <p>Middleton JR, Adkins PRF, Bernier Gosselin V.&nbsp; 2019.&nbsp; Advances in our understanding of staphylococcal mastitis in dairy ruminants.&nbsp; Proceedings of the 2019 ACVIM Forum.&nbsp; Phoenix, AZ.&nbsp; June 7<sup>th</sup>.</p><br /> <p>Witzke M, Rodrigues R, Shangraw E, Ericsson A, McFadden T, Adkins P.&nbsp; 2019.&nbsp; Experimental heat stress alters the fecal microbiome of lactating dairy cattle. American Dairy Science Association Annual Meeting.&nbsp; Cincinnati, OH, June 23-26, 2019.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Lard&eacute;, H., et al. Quantification of antimicrobial usage in 101 Qu&eacute;bec dairy herds. in Annual Conference of the Canadian Association of Veterinary Epidemiology and Preventive Medicine. 2019. Saint-Hyacinthe, QC, Canada.</p><br /> <p>Lard&eacute;, H. Antibiotiques de cat&eacute;gorie 1 : Qui sont-ils et pourquoi devons-nous diminuer leur utilisation?&nbsp; in Rencontre annuelle des producteurs de la clinique ambulatoire de la facult&eacute; de m&eacute;decine v&eacute;t&eacute;rinaire. 2019. Saint-Hyacinthe, QC, Canada.</p><br /> <p>Lard&eacute;, H. &Eacute;valuation des pratiques d&rsquo;utilisation des antibiotiques par les producteurs de bovins laitiers et les m&eacute;decins v&eacute;t&eacute;rinaires du Qu&eacute;bec. in Sommet Laitier Zoetis. 2019.</p><br /> <p>Dufour, S., H. Lard&eacute;, and D. Francoz. Utilisation des antibiotiques sur les fermes laiti&egrave;res au Qu&eacute;bec. in Congr&egrave;s v&eacute;t&eacute;rinaire qu&eacute;b&eacute;cois. 2019. Saint-Hyacinthe, QC, Canada.</p><br /> <p>Dufour, S. Les scientifiques qui parlent &agrave; l'oreille des animaux. in Pint of science festival. 2019. Saint-Hyacinthe, QC, Canada.</p><br /> <p>Dufour, S. Nouvelles m&eacute;thodes de diagnostic mol&eacute;culaire pour le suivi des infections intra-mammaires et &eacute;pid&eacute;miologie des agents pathog&egrave;nes causant la mammite. in Congr&egrave;s AMVPQ 2019 2019. Est&eacute;rel, QC, Canada.</p><br /> <p>Dufour, S. Mastitis research in Canada. 2019. Massey University, New Zealand.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Wilson DJ, Middleton J, Adkins P, Goodell GM: Conventional culture, MALDI-TOF and 16S rRNA compared for agreement in diagnosis of bovine mastitis pathogens. Proc 51st Ann Conv Am Assoc Bov Pract: 294, 2018.</p><br /> <p>Wilson DJ, Middleton JR, Adkins PRF, Goodell GM: Microbial identification of bovine milk isolates compared between conventional culture, MALDI-TOF and 16S rRNA. Proc 12<sup>th</sup> Euroglobal Summit on Veterinary and Animal Sciences: 19, 2018.</p><br /> <p>Wilson DJ, Middleton JR, Adkins PRF, Goodell GM: Culture, MALDI-TOF and 16S rRNA bovine milk microbial diagnostic methods - blind comparison for test agreement. Proc 61st Ann Conf Am Assoc Vet Lab Diag: 10, 2018.</p><br /> <p>Wilson DJ: Dairy goat milking and milk quality. Proc Urban and Small Farms Conf: 190, 2019.</p><br /> <p>Wilson DJ, Britten JE, Rood KA: Intramammary infusion of casein hydrolysate for involution of a single mastitic quarter for the remainder of lactation in dairy cows. Proc 23<sup>rd</sup> Cong An Sci and Prod Assoc: 35, 2019.</p><br /> <p>Wilson DJ, Middleton J, Adkins P, Goodell GM: Conventional culture, MALDI-TOF and 16S rRNA compared for test agreement in diagnosis of bacteria in bovine milk samples. Proc 23<sup>rd</sup> Cong An Sci and Prod Assoc: 42, 2019.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Poster Presentations</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>M Worku, S Adjei-Fremah, K Ekwemalor, EK Asiamah. A Look at the Galectin Gene Expression and Modulation in Ruminants. Plant and Animal Genome XXVII Conference (January 12-16, 2019Emmanuel Asiamah, Kingsley Ekwemalor, Sarah Adjei-Fremah, and Mulumebet Worku, Rumen-protected Methionine supplementation during the peripartal period reduces the expression of LGALS-1, -3 and -4 in polymorphonuclear leukocytes (PMNL) and secretion of Gal-2 and Gal-12 in&nbsp;plasma of&nbsp;Holstein cows, ASAS 2019 <em>Poster, 2019, ASAS.</em></p><br /> <p>Kingsley Ekwemalor, Sarah Adjei-Fremah, Emmanuel Asiamah, Bharath Kumar Mulakala, Eboghoye ElukaOkoludoh and Mulumebet Worku. The effect of oligodeoxynucleotides on Toll-like receptor pathway genes in goat blood, ASAS 2019 <em>Poster, 2019, ASAS</em></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Jaaf S., M. Mezzetti, A. Krueger, B. Batty, M. Premi, F. Perrotta, J. Foster, M. Hanlon, E. Trevisi, and M. Bionaz. Selenium biofortified alfalfa hay supplemented during transition period improves oxidative stress and some metabolic and inflammation parameters in dairy cows and their calves. 37<sup>th</sup> ADSA Discovery Conference, October 28-31, Chicago, IL</p><br /> <p>Jaaf S., B. Batty, A. Krueger, G. Bobe, C. Estill, and M. Bionaz. Effect of agronomic selenium biofortified alfalfa hay on selenium status and glutathione peroxidase activity in transition dairy cows and their calves. 2019 PNW Animal Nutrition Conference. Boise (ID)</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Al-Tamemmi H., Beavers, W.N., Norambuena-Morales, J., Skaar E., Boyd J.M. Copper Stress in <em>Staphylococcus aureus</em> Involves Perturbed Metal Ion Homeostasis. <em>Rutgers Microbiology Symposium</em>, Rutgers University, New Brunswick, NJ, 2/2019.</p><br /> <p>Esquil&iacute;n-Lebr&oacute;n K.J., Foley M., Carabetta V.J., Boyd J.M. Iron-sulfur protein assembly in <em>Staphylococcus aureus. American Society of Microbiology national meeting.</em> San Francisco CA. 6/2019.</p><br /> <p>Al-Tamemmi H., Beavers, W.N., Norambuena-Morales, J., Skaar E., Boyd J.M. <em>Staphylococcus aureus</em> lacking a functional MntABC manganese import system have increased resistance to copper. <em>Gordon Research Conference on Staphylococcal Biology.</em> Catelldefels Spain 8/2019.</p><br /> <p>Boyd J.M. Esquilin-Lebron K., Foley M., Carabetta V., Beavers W., Skaar E. Iron-sulfur protein assembly in Gram positive bacteria. Annual Conference for NSF CAREER awardees. Alexandria VA. 10/2019.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Melson, S., W. J. Weber, E. Powell, D. Zimmerman, J. Lippolis, and B. A. Crooker. 2019. Is the contemporary Holstein more susceptible to E. coli mastitis than her ancestors? Minnesota Dairy Health Conference. April 17, 2019. St. Paul, MN.</p><br /> <p>Dean C.*, Ray, T., Heins, B., Machado, V., Pinedo, P., Formiga, A., Caixeta, L., Noyes, N. (2019, April) An open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case. Poster session presented at the Minnesota Dairy Health Conference, St. Paul, MN. April 17, 2019.<br /> Rowe, S.M.*, Godden, S.M., Nydam, D.V., Lago, A., Royster, E., Vasquez, A. Selective Dry Cow Therapy in U.S. Dairy Herds: Impacts on Health. <em>University of Minnesota Points of Pride day, </em>St Paul, MN, USA. Oct. 2, 2019.</p><br /> <p>Rowe, S.M.*, Godden, S.M., Royster, E., Timmerman, J., Crooker, B., Boyle, M. Investigation of the Relationship Between Bedding Bacteria Count and Intramammary Infection in Late-lactation Dairy Cows: Interim Findings. <em>National Mastitis Council annual meeting, </em>Savannah, GA, USA. <em>Jan 29-Feb. 1, 2019</em></p><br /> <p><em>&nbsp;</em></p><br /> <p><span style="text-decoration: underline;">University of Montreal</span></p><br /> <p>Schwartz, D., et al. Differential somatic cell count as a new and inexpensive supplementary tool for mastitis screening. in International Dairy Federation mastitis conference 2019. 2019. Copenhagen, Denmark.</p><br /> <p>Mass&eacute;, J., et al. La r&eacute;sistance aux antibiotiques: un regard dans nos fermes. in Symposium bovins laitiers du Qu&eacute;bec. 2019. Drumondville.</p><br /> <p>Lard&eacute;, H., et al. Comparison of 3 methods to estimate antimicrobial use for surveillance purposes in Qu&eacute;bec dairy farms. in Annual Conference of the Canadian Association of Veterinary Epidemiology and Preventive Medicine. 2019. Saint-Hyacinthe, QC, Canada.</p><br /> <p>Beauchemin, J., et al. Determination of microbiological contamination of recycled manure solid bedding in dairy farms. in Rencontre annuelle du CRIPA 2019. 2019. St-Hyacinthe, QC, Canada.</p><br /> <p>Beauchemin, J., S. Dufour, and P. Fravalo. Determination of microbiological contamination of recycled manure solid bedding in dairies farms. in 12i&egrave;me symposium du CRIPA. 2019. Montr&eacute;al, QC.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Shangraw E, Rodrigues R, Witzke M, Linder HF, Choudhary R, Spitzer A, Zhao, F-Q, McFadden TB (2019) Intramammary lipopolysaccharide infusion elicits local or systemic effects depending on milk component. ADSA Annual Meeting 2019, Cincinnati, Ohio. Poster</p><br /> <p>&nbsp;</p><br /> <p><strong>Other Publications</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Ruegg PL and RJ Erskine. 2019. Mammary Gland Health and Disorders. In Large Animal Internal Medicine, 6<sup>th</sup> edition, BP Smith, editor, pp. 1118-1050.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">North Carolina A&amp;T State University</span></p><br /> <p>Worku M, Asiamah EK, Adjei-Fremah S, Ekwemalor K. 2019. Global gene expression analysis of neutrophils from periparturient Holstein cows.&nbsp;&nbsp;<a href="https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE129639">https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE129639</a></p><br /> <p>Eboghoye ElukaOkoludoh &ndash; Studies on the expression and the Effect of Galectin-8 on bovine neutrophils- Dissertation</p><br /> <p>Bharath Kumar Mulakala - Comparative studies of galectin gene expression and secretion in cow milk and blood<strong> -</strong>Thesis</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Maryland</span></p><br /> <p>K.M. Moyes.&nbsp; 2018.&nbsp; Maximizing Labor Efficiency In The Milking Parlor- Adapted from Rhyannon Moore, Michigan State University.&nbsp; Maryland Dairy Reporter.</p><br /> <p>K.M. Moyes.&nbsp; 2018.&nbsp; Are you implementing the 10-Point mastitis control plan? Maryland Dairy Reporter.</p><br /> <p>K.M. Moyes.&nbsp; 2019. Judicious use of antimicrobials on dairy farms. Maryland Dairy Reporter.</p><br /> <p>&nbsp;</p>

Impact Statements

Back to top

Date of Annual Report: 01/22/2021

Report Information

Annual Meeting Dates: 12/01/2020 - 12/03/2020
Period the Report Covers: 12/03/2019 - 12/03/2020

Participants

Pamela Adkins, University of Missouri
John Middleton, University of Missouri
Feng-Qi Zhao, University of Vermont
John Barlow, University of Vermont
Pamela Ruegg, Michigan State University
Bill Owens, Louisiana State University
Sandra Godden, University of Minnesota
Benjamin Enger, Ohio State University
Massimo Bionaz, Oregon State University
Paolo Moroni, Cornell University
Amanda Stone, Mississippi State University
Ernest Hovingh, Pennsylvania State University

Brief Summary of Minutes

1.  The 2020 (FY20) annual business meeting of the NE-1748 Multistate research project was called to order at approximately 1pm.


2.  A total of 12 members were present at the meeting, representing 10 stations. Approximately 30 people attended the conference.


3.  Open discussion of delinquent experiment stations. Need to discuss with the project administrative advisor, Kumar Venkitanarayanan (not in attendance).


4.  Discussed future hosting sites of meeting. Several options that were proposed were to: 1) Return to University of Missouri; 2) Host at Mississippi State; 3) Host at OARDC; 4) Return to Chicago, in relation with CRWAD; 5) Return to Chicago without participating in CRWAD. It was decided that participating in CRWAD presented too many logistic’s, especially with the CRWAD meeting changing meeting dates. The discussion was tabled to see how the SARS-CoV-2 pandemic resolved and would affect future meetings sites.


5.  Open discussion of possibly having future meetings utilize a hybrid in person and distance meeting presentation format.


6.  Sam Rowe was nominated to serve as the NE-1748 secretary.


7.  Discussed the annual report (needs to be completed within 60days after the annual meeting). The report will be appended to these minutes and filed in NIMSS.


8.  Meeting was adjourned.


 

Accomplishments

<p><strong>OBJECTIVE 1: Characterize host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance to improve economic outcomes and animal welfare.</strong></p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Member: William E. Owens</p><br /> <p>Identify and evaluate bacteria causing bovine mastitis in Louisiana. Test bacteria for antimicrobial resistance to mastitis therapeutics.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Members: Ronald Erskine, Lorraine Sordillo, Pamela Ruegg</p><br /> <p>The Sordillo lab has continued to study the impact of negative energy balance and fat mobilization on vascular endothelial and adipose inflammatory responses and are defining the role of n-3 fatty acid content in altering the profile of vasoactive eicosanoids and the role of poly-unsaturated fatty acids on attenuating endothelial cell inflammatory responses.&nbsp;</p><br /> <p>The Ruegg lab is working on defining differences among mastitis pathogens in pathogenesis and transmission, with focus on Klebsiella pneumoniae and the group of organisms broadly characterized as Streptococcal like organisms.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Member: Benjamin D. Enger</p><br /> <p>An evaluation of how mammary blood vessels grow and develop in the bovine mammary gland was completed to understand how these blood vessels achieve successful delivery of nutrients for milk synthesis and secretion and also immune cells to the mammary gland that are instrumental for resolving cases of mastitis. Calf mammary glands experiencing different degrees of growth were microscopically examined to determine how these blood vessels form for the first time. It was observed that blood vessels branch outward with the growing mammary tissues to support these new tissue structures. This indicates that the needs of the mammary tissues direct the development of blood vessels which enhances our understanding of how blood vessels grow and adapt during cases of mastitis. Such knowledge will also be important for understanding how the mammary gland heals after a case of mastitis and resumes its productive capacity.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Member: Jeffrey M. Boyd, Paulmi Rudra, Javiera Norembuena, Erin Price</p><br /> <p>Characterize host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance to improve economic outcomes and animal welfare. We have examined and characterized the SrrAB two component regulatory system which sences changes in cellular respiration. When respiration is inhibited or during fermentative growth, SrrAB responds by increasing expression of a number of virulence factors.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: P. Gorden (Iowa State), D. Nydam (Cornell), S. Sreevatsan (Michigan State), R. Fink (St. Cloud State)</p><br /> <p>B. A. Crooker, S. M. Godden, J. D. Lippolis and J. B. Cole. Reducing mastitis in the dairy cow by increasing the prevalence of beneficial polymorphisms in genes associated with mastitis resistance. Our premise is that previous selection practices have successfully increased the presence of genetic polymorphisms associated with increased milk yield but have decreased the presences of polymorphisms associated with disease resistance. We are using contemporary Holsteins (CH) and unique unselected Holsteins (UH) that have not been subjected to selection since 1964 to determine differences in immune and inflammatory responses and mammary gene expression. We will link differential phenotypic responses with whole genome sequencing to identify polymorphisms that impact mastitis resistance. These polymorphisms could enhance gene-assisted selection efforts to increase mastitis resistance. Intramammary challenge studies and data analysis are underway. Our first intramammary challenge study will be submitted in 2020.</p><br /> <p>N. Noyes, L.S. Caixeta, B.J. Heins, V.S. Machado, P. Pinedo. Catalyzing an open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case. The two objectives of this study are (1) to describe the teat skin microbiome of dairy heifers in certified organic herds in the US and (2) to investigate the association between teat skin microbiome throughout the transition period and the occurrence of intramammary infections. Approximately 500 nulliparous heifers from 5 organic dairy herds in MN, CO, TX, and NM were enrolled in the summer and fall of 2019. Milk culture for the first 5 weeks post-partum for all the cows enrolled in the study was carried out during fall 2019 and spring 2020. DNA extraction and sequencing started in fall 2020 and it is expected to be completed in spring 2021. Data analysis and reported will be completed in 2022-2023.</p><br /> <p>K. Patel, S. Godden, E. Royster, B. Crooker, T. Johnson, E. Smith, S. Sreevatsan. Prevalence, antibiotic resistance, virulence and genetic diversity of Staphylococcus aureus isolated from bulk tank milk samples of U.S. dairy herds. The objective of this study was to investigate the prevalence, antibiotic resistance gene content, virulence determinants and genomic diversity of S. aureus in bulk tank milk samples (BTM) from U.S. dairy herds. Of 365 pooled BTM samples collected from 189 herds across the United States, S. aureus was cultured from 170 samples, indicating a sample prevalence of 46.6% in the BTM and a herd prevalence of 62.4% (118 out of 189 herds). Among a subset of 138 S. aureus isolates that were stored for further analysis, 124 were genome sequenced after being confirmed as S. aureus using phenotypic tests. Our findings indicated a high prevalence of S. aureus in BTM, with little evidence of antibiotic resistance but often carrying genes for the various enterotoxins. This study identified predominant genetic clones and provides continued surveillance of S. aureus strains found in U.S. dairy herds. Manuscript submitted in 2020 (in review).</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Smantha Haw, Monica Witzke, Thomas McFadden, Scott Poock, Aaron Ericsson</p><br /> <p>We evaluated the impact of increased environmental heat and humidity on the microbiomes of lactating dairy cattle by conducting challenge trials including control, heat stress, and feed restriction groups. When evaluating the microbiomes of skin and bedding samples, we determined that <em>Enterococcus</em> appeared to be heat-responsive as evidenced by an increased abundance in bedding and on the inguinal and teat skin samples throughout the challenge period in heat stress cows. Although the milk microbiome exhibited some responses to heat and feed restriction challenges, it was overall highly variable over time.&nbsp;&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>In collaboration with Tom McFadden&rsquo;s group at the University of Missouri, Zhao Lab at VT investigated the timing and magnitude of acute local and systemic mammary responses to intramammary LPS challenge in lactating cows and revealed distinct local and systemic effects of LPS on milk yield and composition of milk and blood. In addition, Zhao&rsquo;s lab found differential effects of LPS on milk protein expression in mouse mammary gland and mammary epithelial cells.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>We have analyzed the whole transcriptome of the liver and macrophages isolated from the milk from Jersey cows fed with 1 kg of Alfalfa biofortified with Se/100 kg of BW. The results were partly presented to the 2020 ADSA Annual meeting. We detected very little effect of the treatment on the liver and we discovered that few macrophage samples were more close to mammary epithelial than macrophages (due to very high expression of casein genes and lactalbumin). Thus, the analysis of macrophages would need to be repeated.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Cornell University </span></p><br /> <p>Members: Paolo Moroni</p><br /> <p>Characterization <em>of Staphylococcus aureus</em> host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance in clinical and subclinical mastitis. Genome wide association study and the correlation between gene carriage, lineage and clinical outcome of IMI in a collection of S. aureus isolates from cattle from different country.We investigated the distribution of pathogenic non-agalactiae gram-positive, catalase-negative cocci (GPCN) in a convenience sample of New York State dairy farms. Our primary objective with the clinical mastitis (CM) GPCN samples was to evaluate somatic cell count (SCC) resolution and bacteriological cure of Streptococcus dysgalactiae or Streptococcus uberis versus Lactococcus lactis or Lactococcus garvieae in cows that received an approved intramammary treatment.A large-scale comparative analysis of <em>M. bovis</em> genomes by integrating 103 publicly available genomes and our dataset (250 total genomes). Whole genome single nucleotide polymorphism (SNP) based phylogeny using <em>M.agalactiae</em> as an outgroup revealed that <em>M. bovis</em> population structure is composed of five different clades.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Idaho</span></p><br /> <p>Member: Pedram Rezamand</p><br /> <p>The Rezamand lab continues to work on identifying relationships between nutrient metabolism during the periparturient period and health issues (mastitis) in Pacific Northwest dairy herds. An objective of the present study was to determine the relationship between the serum lipid-soluble vitamins of dairy cows and calves with mastitis status of the cows during the periparturient period. Blood samples of cows were obtained from early Spring to Winter of 2018 for a total of 645 periparturient cows and April to Jun of 2019 of total 599 periparturient cows on d-21, d-7, d+1, d+7 and d+14 relative to calving. In addition, we select a total of 230 cows focusing on mastitic and non-mastitic cows to evaluate IgG1 to IgG2 ratios during the periparturition period via a novel nanoparticle probe. Results showed that cows with mastitis had greater serum retinol compared with that of healthy cows postpartum in 2018. There was evidence of a &alpha;-tocopherol &times; mastitis interaction at various timepoints in 2018. for &alpha;-tocopherol and &beta;-carotene in cows with mastitis tended to be lower than those of healthy cows in 2019. The IgG results show that the test detected an immune status change associated with IgG1/IgG2 relative quantity around the time of parturition.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Members: Gina M Pighetti, Oudessa Kerro Dego</p><br /> <p>Our prior research identified areas of the bovine genome and genes that potentially influence the strength and duration of the dairy cows&rsquo; immune response to intramammary infection. The genes identified suggest how Streptococcus uberis is transported and degraded within the mammary epithelial cell and could explain why some cows are better able to resist an infection compared to others. To investigate these as potential therapeutic targets, we initiated studies examining expression of these genes in mammary epithelial cells and how that changes with Strep. uberis infection. We also initiated how to reduce expression of these genes for future studies that will investigate if it improves or reduces the ability of Strep. uberis to survive within mammary epithelial cells.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2: Assess and apply new technologies that advance mastitis control, milk quality and/or dairy food safety.</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Member: Jeffrey M. Boyd, Paulmi Rudra, Javiera Norembuena, Erin Price</p><br /> <p>Assess and apply new technologies that advance mastitis control, milk quality and/or dairy food safety. We have been examining the mechanisms of metal toxicity to Staphylococcus aureus. Specifically, we have determined that Cu enters S. aureus cells to poison Fe-S cluster prosthetic groups of enzymes. We have also examined some novel molecules that are bactericidal to S. aureus.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: P. Gorden (Iowa State), D. Nydam (Cornell), S. Sreevatsan (Michigan State), R. Fink (St. Cloud State)</p><br /> <p>&nbsp;</p><br /> <p>L.S. Caixeta. Applying precision dairy farming and diagnostic technologies to detect mastitis at the time of and following dry off in dairy cows in a field study. The main objective of this study was evaluate accuracy of precision dairy monitoring and diagnostic technologies (i.e. differential somatic cell counter) in collecting data that effectively identifies appropriate animals for dry cow therapy compared to SCC and bacteriology. In this study, milk samples were collected 1d prior to dry-off and 3 and 5 days after calving. Milk samples were used for the measurement of differential SCC, SCC, and milk culture to identify intramammary infection. Cow enrollment and health and performance was completed between fall 2019 and fall 2020. Data analysis and reported will be completed in 2021.</p><br /> <p>&nbsp;</p><br /> <p>Nusrat Annie Jahan, S. Godden, T. Schoenfus, C. Gebhart, E. Royster, S. Wells, J. Timmerman, and R. Fink. Evaluation of the Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS) system in the detection of Mastitis pathogens from Bovine milk samples. In the present study, we prospectively compared MALDI-TOF MS (Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry or MALDI-ToF) to the conventional 16S rDNA sequencing method for the identification of&nbsp; mastitis isolates (481). Among the 481 milk isolates, 445 (93%) were putatively identified to the genus level by MALDI-TOF MS and 355 (74%) were identified to the species level, but no reliable identification was obtained for 16 (3.3%), and 20 (4.2%) discordant results were identified. In conclusion, our results show that MALDI-TOF MS is a fast and reliable technique which has the potential to replace conventional identification methods for most dairy pathogens, routinely isolated from the milk and dairy products. Manuscript submitted in 2020 (in review).</p><br /> <p>&nbsp;</p><br /> <p>Godden, S., F. Pe&ntilde;a Mosca, E. Royster, B. Crooker, P. Raynor, and K. Janni. Management of Recycled Manure Solids Bedding on Midwest Dairy Farms. Our objective was to investigate if recycled manure solids (RMS) processing method was associated with udder health, milk production, and with bedding bacteria counts (BBC) in ready-to-use RMS bedding samples. Twenty-nine premises in MN and WI were enrolled. Processing systems represented included green (GRN; n=7), drum composted (COM; n=4), digested (DIG; n=6) or mechanically dried (DRY; n=12). Farms were visited twice (summer 2019/winter 2020) to collect samples and data. Ready-to-use bedding was cultured to describe BBC. Results showed that herds using DRY or COM RMS processing systems generally had improved udder health and, for DRY, improved milk production, as compared to herds using DIG or GRN solids. Also, BBC were most consistently lowest in ready-to-use DRY or COM samples.&nbsp; Manuscript is in preparation to submit in 2021.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>A study was initiated to determine if a commercially available teat sealant was effective in preventing new cases of mastitis from occurring in dairy heifers before they calve and begin producing milk. The study is currently ongoing. Results are expected to identify and refine management practices for farmers to better control this disease and prevent dairy heifers from being immediately impinged by a preventable disease immediately after starting to produce milk and recoup the investment imparted by the producer.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Smantha Haw, Monica Witzke, Thomas McFadden, Scott Poock, Aaron Ericsson</p><br /> <p>Associations between staphylococcal species intramammary infection, milk somatic cell count, and persistence of infection during lactation and over the dry period were found in dairy goats. We showed an association between the use of intramammary pirlimycin and short term changes in the fecal micrbiome of dairy cattle being treated for staphylococcal intramammary infections. We demonstrated an association between teat end preparation techniques and contamination of milk samples collected for milk culture with more contaminants being present when teats were not scrubbed with alcohol prior to sample collection. We also evaluated 16S rRNA gene amplicon sequencing of milk samples and our results supported the use of higher PCR cycle numbers to evaluate these low microbial biomass samples.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab has completed a study of the mammary microbiome of lactating organic dairy cattle and we have submitted this work for publication (in revision). The Barlow lab has completed a study applying machine learning to identify key predictors of pathogen strain type from multilocus sequence typing databases for Staphylococcus aureus, Streptococcus agalactiae, and Streptococcus uberis. The Barlow lab has initiated a study comparing milk quality and mastitis prevalence on organic dairy herds utilizing different bedding management practices. NE-1748 members from Minnesota (Godden), Missouri (Adkins) will collaborate in this research.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Members: Oudessa Kerro Dego, Gina M Pighetti</p><br /> <p>A study was conducted to tracking and understand the sources of antimicrobial resistance on regional dairy farms. Most antimicrobial resistant bacteria were Escherichia coli isolated from fecal samples and not bulk tank milk. Of these genes, an extended spectrum beta lactamase (ESBL) gene was expressed by E. coli. In a separate study, this gene was noted to be present in E.coli isolated from bulk tank milk, farm floor, feeding, and watering troughs. A total of 122 suspected ESBLs-E. coli (106 from dairy cattle, and 16 from farm environments) were stocked for further confirmation by molecular analysis. Bacteria with potential to cause human illness (E. coli, Klebsiella spp and Pseudomona aeruginosa) were present and some demonstrated resistance to tetracycline, cefotaxime and nalidixic acid. This suggests dairy cattle and farm environments can harbor antimicrobial resistant bacteria with potential to cause human disease.</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 3: Identify and apply new strategies associated with the control of mastitis that can reduce the use of antibiotics in dairy herds.</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Member: William E. Owens</p><br /> <p>Continue evaluation of botanical formulations from plants for antimicrobial activity against mastitis pathogens.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Members: Ronald Erskine, Lorraine Sordillo, Pamela Ruegg</p><br /> <p>The Ruegg lab conducted a randomized clinical trial that includes evaluation of differences among selected strep like organism causing mastitis in response to differing durations of antimicrobial therapy.&nbsp; We also characterized usage of antimicrobials on 40 large dairy farms and have identified the proportion of antimicrobial usage that is associated with prevention and treatment of mastitis.</p><br /> <p>We are developing novel applications of vacuum analysis to determine milking efficiency in dairy herds.&nbsp; This will provide an on-farm education platform for producers and/or employees regarding milking protocols.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Member: Jeffrey M. Boyd</p><br /> <p>Examine the specific Staphylococcus aureus enzymes that are toxified by metals. Determine internal and external stimuli that trigger the SaeSR regulatory system to increase expression of virulence factors. Determine if we can utilize small molecules to modulates S. aureus behavior by stimulating the Sae and Srr regulatory systems.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: P. Gorden (Iowa State), D. Nydam (Cornell), S. Sreevatsan (Michigan State), R. Fink (St. Cloud State)</p><br /> <p>&nbsp;</p><br /> <p>S.M. Rowe, S.M. Godden, D.V. Nydam, P.J. Gorden, A. Lago, A.K. Vasquez, E. Royster, J. Timmerman, M.J. Thomas. Randomized controlled non-inferiority trial investigating the effect of two selective dry cow therapy protocols on antibiotic use and udder health: quarter-level outcomes. The objective of this study was to compare culture- and algorithm-guided SDCT programs to blanket dry cow therapy (BDCT) in a multi-site, randomized, natural exposure, non-inferiority trial on quarter-level infection dynamics during the dry period and on cow health and performance during the first 120 DIM. Results showed that the two SDCT approaches (culture- and algorithm-guided) each reduced antibiotic use at dry-off by 55%, without causing any negative impacts on intramammary infection dynamics during the dry period. An economic analysis showed an estimated positive economic return, on average, with either SDCT program. We conclude that SDCT can be used in appropriate U.S. dairy herds to reduce antibiotic use while maintaining udder health. Three manuscripts have been published in 2020, with a fourth in press.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab has completed collecting data from a field study exploring Staphylococcus aureus strain variation among dairy cattle and farm workers on 21 dairy farms producing farmstead of artisan cheeses. Strain typing and characterization of antimicrobial susceptibility phenotypes and genotypes has been completed for approximately 160 isolates and cross species (zoonotic) transmission dynamics of S. aureus will be explored using these data.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Members: David Wilson</p><br /> <p>Contributors: Kerry Rood, Justine Britten, and Chad Clancy</p><br /> <p>Udder halves were compared between one of 4 dry treatment groups including casein hydrolysate (CH) intramammary infusion alone or in combinations, and control (dry cow antibiotic plus teat sealant) in a split udder design.&nbsp; Microscopic morphometry measured changes in alveolar epithelial cell height, alveolar luminal diameter, and interstitial stromal thickness during the first 7 days dry.&nbsp; CH alone or combined with antibiotic and/or teat sealant was associated with some histological indications of increased mammary involution compared to controls at d 2 and d 7 dry. Casein hydrolysate may be a useful adjunct or replacement for dry cow antibiotic treatment.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>We received funding to carry out an experiment with the objective to determine if the combination of feeding chicory and supplementing selenium would improve the response to intramammary infection with <em>Strep. Uberis</em>. The experiment was planned to be carried out in April 2020; however, due to the lockdown as response to Covid19, we had to post-pone our experiment to 2021.</p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Members: Paolo Moroni</p><br /> <p>The group conducted a randomized clinical trial that includes evaluation of differences antibiotics. We sampled healthy cows under DCT and implemented a within-subject experimental design based on udder quarters: each quarter received a different treatment: cephalonium dihydrate (first-generation cephalosporin), benzathine cloxacillin, and bismuth subnitrate (internal teat sealant); the last quarter was left untreated and served as experimental control. We hypothesize that antibiotic and non-antibiotic treatments for SDCT do not alter significantly the milk microbiome of healthy dairy cows: this would further support the replacement of antibiotics with teat-sealant for SDCT. We investigated the distribution of pathogenic non-agalactiae gram-positive, catalase-negative cocci (GPCN) in a convenience sample of New York State dairy farms. Our primary objective with the clinical mastitis (CM) GPCN samples was to evaluate somatic cell count (SCC) resolution and bacteriological cure of Streptococcus dysgalactiae or Streptococcus uberis versus Lactococcus lactis or Lactococcus garvieae in cows that received an approved intramammary treatment.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Members: Oudessa Kerro Dego, Gina M Pighetti.</p><br /> <p>A year-long field-study was conducted to evaluate the efficacy of using Staphylococcus aureus surface proteins (SASP) and Staphylococcus chromogenes surface proteins (SCSP) as a vaccine against staphylococci pathogens. An alternative vaccine strategy was initiated for E. coli, using enterobactin conjugated to keyhole limpet hemocyanin and will be assessing antibody titers in this pilot study. Dr. Pighetti also is in the process of evaluating field data from an 18 month study assessing the impact of nutritional supplementation on the number of mastitis cases occurring during high risk time periods, when selective antibiotic therapy of dry cows is employed.</p><br /> <p>&nbsp;</p><br /> <p><strong>WORK PLANNED FOR THE COMING YEAR, LISTED BY OBJECTIVE:</strong></p><br /> <p><strong>OBJECTIVE 1</strong></p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Identify and evaluate bacteria causing bovine mastitis in Louisiana. Test bacteria for antimicrobial resistance to mastitis therapeutics.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Mammary blood vessel growth will be examined in different regions of calf mammary glands that had been stimulated to experience different degrees of mammary growth and VEGFR2 will be localized and described in calf mammary glands.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>B. A. Crooker, S. M. Godden, J. D. Lippolis and J. B. Cole. Reducing mastitis in the dairy cow by increasing the prevalence of beneficial polymorphisms in genes associated with mastitis resistance. To increase the power of the genotype scan, we have repeated the intramammary E. coli challenge with another 5 UH and 7 CH cows and have a 3rd study scheduled for early 2021. We will evaluate milk somatic cells from these studies to determine differences in expression of genes associated with responses to mastitis. We anticipate this will identify differential expression of genes that play important beneficial and detrimental roles in the magnitude and effectiveness of the immune response to mastitis. Data analysis will be completed and reported in 2021. Subsequent sequencing and identification of polymorphic differences in the genes and / or their regulatory components (transcription factors, etc.) between the Holstein genotypes will occur in late 2021.</p><br /> <p>C. Dean, L.S. Caixeta, N. Noyes, S. Godden, B. Crooker, F. Pena Mosca. Impact of the cow udder microbiome on the biodiversity of milk. Exploratory project to investigate the microbiome in different parts of the mammary gland. Sample collection scheduled for December 2020. DNA extraction, DNA sequencing and data analysis will be completed in spring 2021 with reporting in summer 2021.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Longitudinal evaluation of intramammary infection and somatic cell count in Jersey cattle.&nbsp; Evaluate associations between MALDI-TOF identified bacteria and milk somatic cell count to better define mastitis pathogens versus sample contaminants.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Zhao Lab at VT will study the mechanism underlying the repression of milk protein expression by LPS.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>We would extract RNA from additional macrophage samples and, after testing their expression of CSN3 and LALBA using RTqPCR, we will perform the RNAseq analysis. After this we plan to publish the results of liver and macrophages.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Members: Paolo Moroni</p><br /> <p>Epidemiology study on Mycoplasma and longitudinal evaluation of intramammary infection and somatic cell count in cow positive to Mycoplasma bovis.&nbsp; Evaluate associations between MALDI-TOF identified bacteria and milk somatic cell count to better define mastitis pathogens versus sample contaminants.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Idaho</span></p><br /> <p>We will continue to examine the relationship between nutrient metabolism and mastitis.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Investigate the movement and degradation <em>of Streptococcus uberis</em> strains within mammary epithelial cells and macrophages relative to host proteins. Investigate the prevalence of Shiga toxin-producing E. coli in dairy farms in East Tennessee.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>L.S. Caixeta, S. Godden, W. Knauer, J. Hadrich. G. Cramer. Randomized clinical trial evaluating the effect of administering Bovikalc&reg; dry at dry off on udder health, early lactation health and performance, and economics in dairy cows. Randomized clinical trial investigating the effects of pre dry-off administration of acidogenic bolus on udder health during the dry period and in early lactation. Enrollment of cows is schedule for the spring 2021 and follow up period in fall 2021. Lactation health and performance for the first 100 DIM will continue into late fall 2021. Data analysis and reporting will be completed in 2022.</p><br /> <p>Godden, S., P. Raynor, R. Singer, E. Royster, J. Hadrich, K. Janni, B. Crooker, J. Bender, B. Alexander. Investigation of the Relationship between Method of Processing Recycled Manure Solids Bedding, Udder Health, Milk Quality, Air Quality, Antimicrobial Resistance and Economics on Midwest Dairy Farms.&nbsp; Project objectives described above. Final results will be analyzed and manuscripts submitted in 2021.&nbsp;</p><br /> <p>N. Noyes, L. Caixeta, B. Heins, V. Machado, P. Pinedo, A. Formiga, D. Van Nydam, J. Velez, H. Karremann. Catalyzing an open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case. The long-term goal of this proposal is to ignite robust, systematic and application-driven microbiome research activity within the livestock community, thus producing a panoply of new, evidence-based tools to combat livestock disease and production challenges. Project objectives described above.&nbsp; Field to collect samples, plus laboratory analysis will continue into 2020.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>The Barlow lab will continue studies of <em>S. aureus</em> strain variation and transmission dynamics on small dairy farms. The Barlow lab will continue the observational field studies comparing milk quality and mastitis prevalence on organic dairy herds utilizing different bedding management practices. NE-1748 members from Minnesota (Godden) and Missouri (Adkins) will collaborate in this research.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Screen a large panel of blood and serum clinical pathology tests on dairy calves and develop the most practical reduced panel of tests for prognosis and treatment of sick dairy calves during the first 60 days of life.&nbsp; This will be compared to a clinical scale of calf disease signs as well as the treatment and disease or health outcomes of the calves.&nbsp; The goal is to develop an affordable, rapid blood test panel that can be used on calves showing early signs of disease for better prognostic and treatment purposes, especially in large scale calf rearing.</p><br /> <p>Study different concentrations of a preservative for milk, colostrum and milk replacer fed to calves.&nbsp; First phase will examine association with bacteria counts over time under the conditions which exist when those products are fed to dairy calves, in comparison with untreated milk.&nbsp; The goal is to determine a safe but effective concentration of preservative that can be added to colostrum and milk fed to dairy calves.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Members: Paolo Moroni</p><br /> <p>Characterization <em>of Streptococcus uberis </em>host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance in clinical mastitis.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Investigate the molecular epidemiology of extended-spectrum &beta;-lactamase producing <em>E. coli</em> in dairy farms. Develop a review article on extended-spectrum beta-lactamase-producing Enterobacteriaceae in the US dairy cattle production system and their implication on public health.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>OBJECTIVE 3</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">Louisiana State University</span></p><br /> <p>Continue evaluation of botanical formulations from plants for antimicrobial activity against mastitis pathogens.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Continue progress on projects outlined above. Also, conducting field study of the long term impact (efficacy, production, milk quality) of antimicrobial therapy on mild clinical mastitis. We are also developing an online Continuing Education course for veterinarians to build their capacity to evaluate milk quality for client dairy herds.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>We are going to evaluate the use of a teat sealant in non-lactating dairy heifers in their efficacy of preventing new intramammary infections at different stages of gestation. This will be done to determine if there is a better time to administer teat sealant to prevent more infections during late gestation than others.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>S. Rowe, L.S. Caixeta, E. Royster, S. Godden. Clinical trial evaluating the health effects of reducing antibiotic use at dry-off in dairy cows. Randomized clinical trial comparing blanket dry cow therapy and no dry cow therapy in a low input commercial dairy farm. Enrollment of cows is schedule for the summer 2021 and follow up period in fall 2021. Lactation health and performance for the first 100 DIM will continue until winter 2021. Data analysis and reporting will be completed in 2022.</p><br /> <p>&nbsp;</p><br /> <p>Godden, S. and E. Royster. Pilot Study: Refining a Culture-Guided Selective Dry Cow Therapy Program (SDCT) to Reduce Antibiotic Use on Dairy Farms. We hypothesize that by identifying and selectively treating only IMI caused by SSLO, we can dramatically reduce antibiotic use at dry-off by as much as 90%, while maintaining udder health, cow health and performance in the subsequent lactation (versus blanket DCT).&nbsp; Our major objective will be to conduct a pilot study in 2 commercial herds to evaluate the effect of implementing a SDCT program that identifies and treats only IMI caused by SSLO on measures of quarter health, cow health and performance and antibiotic use (vs. BDCT). Covid permitting, field work will commence in spring, 2021.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>In collaboration with Tom McFadden&rsquo;s group at the University of Missouri, Zhao Lab at VT is investigating oxidative damage and the role of Nrf2 in antioxidation in the mammary gland during mastitis. The aim of this work is to find natural means to activate Nrf2 signaling to control mastitis&rsquo;s damage to the udders.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>We are planning to start the experiment with the objective to determine if the combination of feeding chicory and supplementing selenium would improve the response to intramammary infection with <em>Strep. uberis</em> this coming April 2021.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Evaluate efficacies of <em>Staphylococcus aureus</em> surface proteins (SASP), <em>Staphylococcus chromogenes</em> surface proteins (SCSP) vaccines against Staphylococcal and other mastitis pathogens in dairy cows. Develop nanoparticle-based delivery of SASP and SCSP vaccines. Evaluate the immunogenicity of enterobactin-KLH vaccine against <em>E. coli</em> mastitis in dairy cows. Analyze, summarize, and share the results regarding the potential use of a nutritional supplement to enhance the effectiveness of selective antibiotic therapy of dry cows.</p>

Publications

<p><strong>Peer-Reviewed Literature</strong></p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Fuenzalida, M.J. and P.L. Ruegg.&nbsp; 2020.&nbsp; Molecular epidemiology of non-severe clinical mastitis caused by Klebsiella pneumoniae occurring in cows on 2 Wisconsin dairy farms.&nbsp; J. Dairy Sci.&nbsp; 103:3779-3792.&nbsp; https://doi.org/10.3168/jds.2019-17464</p><br /> <p>Leite de Campos, J., A. Kates, A. Steinberger, A. Sethi, G. Suen, J. Shutske, N. Safdar, T. Goldberg, and P.L. Ruegg.&nbsp; 2021.&nbsp; Quantification of antimicrobial usage in adult cows and preweaned calves on 40 large Wisconsin dairy farms using dose-based and mass-based metrics.&nbsp; Accepted J Dairy Sci, Nov 2020</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Enger, B.D., S.N. Lehner, C.L.M. Parsons, R.M. Akers, and N.R. Hardy. 2020. Short Communication: Effect of intramammary infection and parenchymal region on collagen abundance in non-lactating bovine mammary glands. Appl. Anim. Sci. 36:688-693. doi:10.15232/aas.2020-02003.</p><br /> <p>Enger, B.D., S.C. Nickerson, R.M. Akers, L.E. Moraes, and C.E. Crutchfield. 2020. Use of commercial somatic cell counters to quantify somatic cells in non-lactating bovine mammary gland secretions. Vet. Prev. Med. 174:104775. doi:10.1016/j.prevetmed.2019.104775.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Ferrer-Gonzalez E., Fujita J., Yoshizawa T., Nelson J., Pilch A., Hillman E., Ozawa M., Kuroda N., Al-Tameemi H., Boyd J.M., LaVoie E., Matsumura H., and Pilch D. Structure-Guided Design of a Fluorescent Probe for the Visualization of FtsZ in Clinically Important Gram-Positive and Gram-Negative Bacterial Pathogens. Nature Scientific Reports, 2019 PMID: 31882782</p><br /> <p>Norambuena J., Miller M., Boyd J.M.*, Barkay T.*, Expression and regulation of the mer operon in Thermus thermophilus. Environmental Microbiology, 2020 PMID: 32090420</p><br /> <p>Rudra, P., Boyd J.M. Metabolic control of virulence factor production in Staphylococcus aureus. Current Opinion in Microbiology, 2020 PMID: 32388086.</p><br /> <p>Tiwari N., L&oacute;pez-Redondo M., Miguel-Romero L., Kulhankova K., Cahill M.P., Al-Tameemi H., Herfst C.A., Kirby J.R., Boyd J.M., McCormick J.K., Salgado-Pab&oacute;n W., Marina A., Schlievert P.M., Fuentes E.J., The SrrAB two-component system regulates Staphylococcus aureus pathogenicity through redox sensitive cysteines. Proceedings of the National Academy of Sciences, 2020 PMID: 32354997.</p><br /> <p>Price E.E., Boyd J.M., Genetic control of metal ion homeostasis in Staphylococcus aureus. Trends in Microbiology, 2020 PMID: 32381454.</p><br /> <p>Dubovoy V., Nawrocki S., Verma G., Wojtas L., Desi P., Al-Tameemi H., Brinzari T.V., Stranick M., Chen D., Xu S., Ma S., Boyd J.M., Asefa T., Pan L., Synthesis, characterization, and investigation of the antimicrobial activity of cetylpyridinium tetrachlorozincate. ACS Omega, 2020 PMID: 32426592.</p><br /> <p>Dubovoy V., Desai P., Hao Z., Cheng C., Verma G., Wojtas L., Brinzari T.V., Boyd J.M., Ma S., Asefa T., Pan L., Synthesis, Characterization, and Antimicrobial Investigation of a Novel Chlorhexidine Cyclamate Complex. ACS Crystal Design and Growth, 2020 doi.10.1021/acs.cgd.0c00107</p><br /> <p>Carabetta, V.J., Esquilin-Lebron K., Zelzion E., Boyd J.M., Genetic approaches to uncover gene products involved in iron-sulfur protein maturation: High throughput genomic screening using transposon-sequencing. Methods in Molecular Biology, 2020 accepted.</p><br /> <p>Al-Tameemi H., Beavers W.N., Norambuena-Morales J., Skaar E., Boyd J.M. Staphylococcus aureus lacking a functional MntABC manganese import system have increased resistance to copper. Molecular Microbiology. 2020 PMID: 33034093.</p><br /> <p>Juttukonda L.J., Beavers W.N., Horning K.J., Unsihuay D., Horvath D.J., Kim K., Weiss A., Pishchany G., Al-Tameemi H., Boyd J.M., Sulikowski G., Bowman E.B., and Skaar E.P. A small molecule modulator of metal homeostasis is toxic to Gram-positive pathogens. mBio. 2020 PMID: 33109764.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Rowe, S. , S. Godden, D. Nydam, P. Gorden, A. Lago, A. Vasquez, E. Royster, J. Timmerman, M. Thomas. 2020. Randomized controlled non-inferiority trial investigating the effect of two selective dry cow therapy protocols on antibiotic use at dry-off and dry period intramammary infection dynamics. J. Dairy Sci. Vol. 103:6473-6492</p><br /> <p>Rowe, S. , S. Godden, D. Nydam, P. Gorden, A. Lago, A. Vasquez, E. Royster, J. Timmerman, M. Thomas.&nbsp; 2020. Randomized controlled trial investigating the effect of two selective dry cow therapy protocols on udder health and performance in the subsequent lactation. J. Dairy Sci. Vol. 103:6493-6503</p><br /> <p>Rowe, S. , S. Godden, D. Nydam, A. Lago, A. Vasquez, E. Royster, J. Timmerman. 2020. Randomized Equivalence Study Comparing the Efficacy of Two Commercial Internal Teat Sealants in Dairy Cows. J. Dairy Sci. 103:5398-5413</p><br /> <p>Rowe, S. , S. Godden, D. Nydam, P. Gorden, A. Lago, A. Vasquez, E. Royster, J. Timmerman, M. Thomas.&nbsp; 2020. Evaluation of rapid culture, a predictive algorithm, esterase somatic cell count and lactate dehydrogenase to detect intramammary infection in quarters of dairy cows at dry-off. Prev. Vet. Med. 179:1-11&nbsp;&nbsp;</p><br /> <p>Rowe, S., S. Godden, E. Royster, J. Timmerman, B. Crooker and M. Boyle. 2019. Cross-sectional study of the relationship between bedding materials, bedding bacteria counts and intramammary infection in late lactation dairy cows. J. Dairy Sci. 102:11384&ndash;11400</p><br /> <p>Rowe, S., S. Godden, E. Royster, J. Timmerman, B. Crooker and M. Boyle.&nbsp; 2019. Cross-sectional study of the relationship between cloth udder towel management, towel bacteria counts and intramammary infection in late lactation dairy cows. J. Dairy Sci. 102:11401&ndash;11413</p><br /> <p>Dean, C.J., Slizovskiy, I.B., Crone, K.K., Pfennig, A.X., Heins, B.J., Caixeta, L.S., and Noyes, N.R. 2020. Investigating the skin and teat canal microbiomes of the bovine udder using differing sampling and sequencing approaches. J Dairy Sci. 2020 Oct 29;S0022-0302(20)30860-2. doi: 10.3168/jds.2020-18277.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Bernier Gosselin V, Dufour S, Middleton JR. 2020. Association between species-specific staphylococcal intramammary infection and milk somatic cell score over time in dairy goats. Prev Vet Med. Jan 174:104815</p><br /> <p>Adkins PRF, Ericsson A, Middleton JR, Witzke M. 2020. The effect of intramammary pirlimycin hydrochloride on the fecal microbiome of early lactation heifers. J Dairy Sci. 103(4):3459-3469</p><br /> <p>Wattenburger K, Schmidt R, Placheta L, Middleton JR, Adkins PRF. 2020. Evaluation of four different teat disinfection methods prior to collection of milk samples for bacterial culture in dairy cattle. J Dairy Sci. 103(5):4579-4587</p><br /> <p>Shwani A, Adkins PRF, Ekesi NS, Alrubaye A, Calcutt MJ, Middleton JR, Rhoads DD. 2020. Whole genome comparisons of Staphylococcus agnetis isolates from cattle and chickens. Appl Environ Microbiol. Jun 2;86(12):e00484-20</p><br /> <p>Witzke MC, Gullic A, Yang P, Bivens NJ, Adkins PRF, Ericsson AC.&nbsp; Influence of PCR cycle number on 16S rRNA gene amplicon sequencing of low biomass samples.&nbsp; J Microbiol Methods.&nbsp; Sep 176: 106033.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Alexander Jonathan Spitzer, Qing Tian, Ratan Choudhary, and Feng-Qi Zhao. (2020). Bacterial endotoxin induces oxidative stress and reduces milk protein expression and hypoxia in the mammary gland. Oxidative Medicine and Cellular Longevity, Volume 2020, Article ID 3894309, 16 pages. DOI: 10.1155/2020/3894309</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Tsai, C., Rosa, F., Bionaz, M., &amp; Rezamand, P. (2020). Effects of 2,4-thiazolidinedione (TZD) on milk fatty acid profile and serum vitamins in dairy goats challenged with intramammary infusion of Streptococcus uberis. Journal of Dairy Research, 1-8</p><br /> <p>Jaaf, S.; Batty, B., Krueger A., Estill, C.T.; Bionaz, M. (2020). Selenium biofortified alfalfa hay fed in low quantities improves selenium status and glutathione peroxidase activity in transition dairy cows and their calves. Journal of Dairy Research, May;87(2):184-190</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Cornell University</span></p><br /> <p>Monistero V, Barberio A, Biscarini F, Cremonesi P, Castiglioni B, Graber HU, Bottini E, Ceballos-Marquez A, Kroemker V, Petzer IM, Pollera C, Santisteban C, Veiga Dos Santos M, Bronzo V, Piccinini R, Re G, Cocchi M, Moroni P. Different distribution of antimicrobial resistance genes and virulence profiles of Staphylococcus aureus strains isolated from clinical mastitis in six countries. J Dairy Sci. 2020 Apr;103(4):3431-3446. doi: 10.3168/jds.2019-17141. Epub 2020 Jan 31. PMID: 32008788.</p><br /> <p>Bronzo V, Lopreiato V, Riva F, Amadori M, Curone G, Addis MF, Cremonesi P, Moroni P, Trevisi E, Castiglioni B. The Role of Innate Immune Response and Microbiome in Resilience of Dairy Cattle to Disease: The Mastitis Model. Animals (Basel). 2020 Aug 11;10(8):1397. doi: 10.3390/ani10081397. PMID: 32796642; PMCID: PMC7459693.</p><br /> <p>Hoekstra J, Zomer AL, Rutten VPMG, Benedictus L, Stegeman A, Spaninks MP, Bennedsgaard TW, Biggs A, De Vliegher S, Mateo DH, Huber-Schlenstedt R, Katholm J, Kov&aacute;cs P, Kr&ouml;mker V, Lequeux G, Moroni P, Pinho L, Smulski S, Supr&eacute; K, Swinkels JM, Holmes MA, Lam TJGM, Koop G. Genomic analysis of European bovine Staphylococcus aureus from clinical versus subclinical mastitis. Sci Rep. 2020 Oct 23;10(1):18172. doi: 10.1038/s41598-020-75179-2. PMID: 33097797; PMCID: PMC7584570.</p><br /> <p>Wemette M, Safi AG, Beauvais W, Ceres K, Shapiro M, Moroni P, Welcome FL, Ivanek R. New York State dairy farmers' perceptions of antibiotic use and resistance: A qualitative interview study. PLoS One. 2020 May 27;15(5):e0232937. doi: 10.1371/journal.pone.0232937. PMID: 32459799; PMCID: PMC7252592.</p><br /> <p>Kumar R, Register K, Christopher-Hennings J, Moroni P, Gioia G, Garcia- Fernandez N, Nelson J, Jelinski MD, Lysnyansky I, Bayles D, Alt D, Scaria J. Population Genomic Analysis of Mycoplasma bovis Elucidates Geographical Variations and Genes associated with Host-Types. Microorganisms. 2020 Oct</p><br /> <p>10;8(10):1561. doi: 10.3390/microorganisms8101561. PMID: 33050495; PMCID: PMC7650767.</p><br /> <p>Calonzi D, Romano A, Monistero V, Moroni P, Luini MV, Biscarini F, Castiglioni B, Cremonesi P. Technical note: Development of multiplex PCR assays for the molecular characterization of Streptococcus uberis strains isolated from bovine mastitis. J Dairy Sci. 2020 Jan;103(1):915-921. doi: 10.3168/jds.2019-16823. Epub 2019 Nov 6. PMID: 31704014.</p><br /> <p>Scillieri Smith JC, Moroni P, Santisteban CG, Rauch BJ, Ospina PA, Nydam DV. Distribution of Lactococcus spp. in New York State dairy farms and the association of somatic cell count resolution and bacteriological cure in clinical mastitis samples. J Dairy Sci. 2020 Feb;103(2):1785-1794. doi: 10.3168/jds.2018-16199. Epub 2019 Dec 19. PMID: 31864733.</p><br /> <p>Biscarini F, Cremonesi P, Castiglioni B, Stella A, Bronzo V, Locatelli C, Moroni P. A Randomized Controlled Trial of Teat-Sealant and Antibiotic Dry-Cow Treatments for Mastitis Prevention Shows Similar Effect on the Healthy Milk Microbiome. Front Vet Sci. 2020 Sep 2;7:581. doi: 10.3389/fvets.2020.00581. PMID: 32984415; PMCID: PMC7492605.</p><br /> <p>Mann S, Curone G, Chandler TL, Moroni P, Cha J, Bhawal R, Zhang S. Heat treatment of bovine colostrum: I. Effects on bacterial and somatic cell counts, immunoglobulin, insulin, and IGF-I concentrations, as well as the colostrum proteome. J Dairy Sci. 2020 Oct;103(10):9368-9383. doi: 10.3168/jds.2020-18618. Epub 2020 Aug 20. PMID: 32828510.</p><br /> <p>Wemette M, Greiner Safi A, Wolverton AK, Beauvais W, Shapiro M, Moroni P, Welcome FL, Ivanek R. Public perceptions of antibiotic use on dairy farms in the United States. J Dairy Sci. 2021 Jan 14:S0022-0302(21)00003-5. doi: 10.3168/jds.2019-17673. Epub ahead of print. PMID: 33455793.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Idaho</span></p><br /> <p>Tsai, C. Y. R. Hassan, H. C. Hung, T. Weber, W. J. Price, P. Rezamand*, and Q. Huo. 2020. A Rapid Blood Test to Monitor the Immune Status Change of Dairy Cows and to Evaluate their Disease Risk during the Periparturient Period.&nbsp; Sensors International. <a href="https://doi.org/10.1016/j.sintl.2020.100078">https://doi.org/10.1016/j.sintl.2020.100078</a></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Amenu K, Agga GE, Kumbe A, Shibiru A, Desta H, Tiki W, Kerro Dego O, Wieland B, Grace D, S. A. 2020. Community-tailored training to improve the knowledge, attitudes, and practices of women regarding hygienic milk production and handling in the Borana pastoral area of southern Ethiopia. Journal of Dairy Science 103:9748 &ndash; 9757. DOI:<a href="https://doi.org/10.3168/jds.2020-18292">https://doi.org/10.3168/jds.2020-18292</a><span style="text-decoration: underline;">.</span></p><br /> <p>&nbsp;</p><br /> <p>Kerro Dego O. Bovine mastitis part I, IntechOpen, DOI: 10.5772/intechopen.93483, Published online (September 2, 2020), Available from, <a href="https://www.intechopen.com/online-first/bovine-mastitis-part-i">https://www.intechopen.com/online-first/bovine-mastitis-part-i</a><span style="text-decoration: underline;">. (chapter)</span></p><br /> <p>Kerro Dego O. Control and Prevention of Mastitis: Part Two. IntechOpen, DOI: 10.5772/intechopen.93484, Published online (August 27, 2020). Available from, <a href="https://www.intechopen.com/online-first/control-and-prevention-of-mastitis-part-two">https://www.intechopen.com/online-first/control-and-prevention-of-mastitis-part-two</a><span style="text-decoration: underline;">. (chapter)</span></p><br /> <p>Kerro Dego O. Current Status of Antimicrobial Resistance and Prospect for New Vaccines against Major Bacterial Bovine Mastitis Pathogens.IntechOpen, <em>DOI: http://dx.doi.org/10.5772/intechopen.94227</em>, Published online (October 19, 2020), Available from: <a href="https://www.intechopen.com/online-first/current-status-of-antimicrobial-resistance-and-prospect-for-new-vaccines-against-major-bacterial-bov">https://www.intechopen.com/online-first/current-status-of-antimicrobial-resistance-and-prospect-for-new-vaccines-against-major-bacterial-bov</a><span style="text-decoration: underline;"> (chapter)</span></p><br /> <p>Kerro Dego O, Pacha PA, Gillespie BE, Pighetti GM. Experimental <em>Staphylococcus aureus</em> Mastitis Infection Model by Teat Dipping in Bacterial Culture Suspension in Dairy Cows. MDPI Animals <strong>2020</strong>, 10(5), 751. &nbsp;<a href="https://doi.org/10.3390/ani10050751">https://doi.org/10.3390/ani10050751</a>.</p><br /> <p>Lee, A, S Schexnayder, L Schneider, S Oliver, G Pighetti, C Petersson-Wolfe, J Bewley, S Ward, P Krawczel. 2019. Dairy producers in the Southeast United States are concerned with cow care and welfare. J Dairy Res. 1-4. DOI: <a href="https://doi.org/10.1017/S0022029919000943">https://doi.org/10.1017/S0022029919000943</a>.</p><br /> <p>Vaughn JM, Abdi RD, Gillespie BE, Kerro Dego O. 2020. Genetic diversity and virulence characteristics of <em>Staphylococcus aureus</em> isolates from cases of bovine mastitis. Microb Pathog 144:104171. <a href="https://doi.org/10.1016/j.micpath.2020.104171">https://doi.org/10.1016/j.micpath.2020.104171</a>.</p><br /> <p>&nbsp;</p><br /> <p><strong>Abstracts</strong></p><br /> <p><span style="text-decoration: underline;">Oregon State University</span></p><br /> <p>Busato S. and M Bionaz. 2020. Selenium-biofortified alfalfa hay supplementation modulates liver and macrophage gene expression in periparturient dairy cows. J. Dairy Sci. 103 (Suppl. 1):124</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Enger, K.M., N.R. Hardy, and B.D. Enger. 2020. Effect of intramammary infection on colostrum antibody concentrations. Pages 209-210 in Natl. Mastitis Counc. Annu. Mtg. Proc., Orlando, FL, Natl. Mastitis Council Inc., New Prague, MN.</p><br /> <p>Hardy, N.R., K.M. Enger, and B.D. Enger. 2020. Quantifying mammary growth and proliferative effects of estradiol in Holstein heifer calves. J. Dairy Sci. 102 (Suppl. 2):XXX. Accepted.</p><br /> <p>Hardy, N.R., K.M. Enger, and B.D. Enger. 2020. Mammary blood vessel development in response to estradiol administration in heifer calves. J. Dairy Sci. 103. (Suppl. 1):168.</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Lippolis, J. D., E. J. Putz, T. A. Reinhardt, E. Casas, W. J. Weber, B. A. Crooker. 2020. The effect of 50 years of breeding on the ability of Holsteins to fight mastitis. International Milk Genomics Consortium. 17th International Symposium (Virtual). October 13-16. Hosted by UC Davis.</p><br /> <p>Godden, S., F. Pe&ntilde;a Mosca, E. Royster, B. Crooker, J. Hadrich, P. Raynor, R. Singer, and K. Janni. Investigation of the Relationship between Method of Processing Recycled Manure Solids Bedding and Udder Health and Production on Midwest Dairy Farms. Proc. Annu. Mtg. Am Assoc. Bovine Pract. Sept. 24-26, 2020. (virtual).&nbsp;</p><br /> <p>Godden, S., F. Pe&ntilde;a Mosca, E. Royster, B. Crooker, J. Hadrich, P. Raynor, R. Singer, and K. Janni. Investigation of the Relationship between Method of Processing and Bacteria Counts in Ready-to-Use Recycled Manure Solids Bedding on Midwest Dairy Farms Proc. Annu. Mtg. Am Assoc. Bovine Pract. Sept. 24-26, 2020. (virtual).&nbsp;</p><br /> <p>Godden, S., E. Royster, J. Timmerman and N. O&rsquo;Sell. Efficacy of a Stabilized Liquid Potassium Sorbate Preservative to Reduce Bacterial Proliferation in Colostrum or Tank Milk Stored by Refrigeration or at Room Temperature. Proc. Annu. Mtg. Am Assoc. Bovine Pract. Sept. 24-26, 2020. (virtual).&nbsp;</p><br /> <p>Rowe, S.M*., Nydam, D.V., Godden, S.M., Gorden, P., Lago, A., Royster, E., Vasquez, A., Thomas, M. 2020. Partial budget analysis of selective dry cow therapy strategies. American Dairy Science Assoc. Annual Meeting, USA. June 22-24, 2020 (virtual)</p><br /> <p>Rowe, S.M.*, Godden, S.M., Nydam, D.V., Lago, A., Thomas, M., Royster, E., Vasquez, A., Timmerman. J., Thomas. M.. 2020. Randomized controlled trial investigating the effect of two selective dry cow therapy protocols on antibiotic use and udder health, Proc. 59th Annual meeting of the National Mastitis Council. Jan 28-30, 2020. Orlando, FL.</p><br /> <p>Rowe, S.M.*, Godden, S.M., Nydam, D.V., Lago, A., Royster, E., Vasquez, A., 2020. Randomized controlled trial evaluating the efficacy of two commercial internal teat sealants in dairy cows. Proc. 59th Annual meeting of the National Mastitis Council. Jan. 28-30, 2020. Orlando, FL</p><br /> <p>Rowe, S.*, S. Godden, E. Royster, J. Timmerman, D.V. Nydam, A.K. Vasquez, P. Gorden, A. Lago, and M. Thomas. Selective Dry Cow Therapy on US Dairy Farms: Impact on Udder Health and Productivity. 2019 Conference of Research Workers in Animal Diseases. Nov. 2-5, 2019. Chicago, IL.</p><br /> <p>Rowe, S.*, S. Godden, E. Royster, J. Timmerman, D.V. Nydam, A.K. Vasquez, A. Lago. 2019. Randomized controlled trial evaluating the efficacy of two commercial internal teat sealants in dairy cows. 2019 Conference of Research Workers in Animal Diseases. Nov. 2-5, 2019. Chicago, IL</p><br /> <p>Dean, C., Fernandes, L., Sharpe, K., Junior, A., Ray, T., Baumann, C., Wehri, T., Heins, B., Pinedo, P., Machado, V., Caixeta, L.S., Noyes, N. &ldquo;Community research and education program to use the microbiome for the advancement of organic livestock production&rdquo;. Proceedings of the 100th Annual Conference of Research Workers in Animals Diseases, Chicago, IL, 2019.</p><br /> <p>Baumann, C., Dean, C., Fernandes, L., Sharpe, K., Antunes Junior, A. M., Ray, T., Wehri, T., Heins, B., Pinedo, P., Formiga, A., Machado, V., Caixeta, L.S., Noyes, N. &ldquo;Incidence rates of mastitis in heifers on organic dairy farms&rsquo;. Proceedings of the 100th Annual Conference of Research Workers in Animals Diseases, Abstract 041. Poster presentation. Chicago, IL, 2019.</p><br /> <p>Fernandes, L., Guimaraes, I., Noyes, N.R., Caixeta, L.S., Machado, V.S. &ldquo;Impact of subclinical mastitis during the first month of lactation on milk yield, fertility, and culling of dairy cows on USDA-certified organic herds&rdquo;. ADSA Conference. Virtual Conference, 2020.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Kurban D, Bolduc E, Roy JP, DeVries T, Gervais R, Adkins P, Middleton JR, Keefe G, France A, Dufour S.&nbsp; 2020.&nbsp; An observational transversal study on the prevalence of staphylococcal intramammary infections after calving in primirparous vs multiparous cows milked using an automatic milking system.&nbsp; Journ&eacute;e de la recherche FMV - Research day at FVM. University of Montreal, Quebec, Canada.&nbsp; 19 March 2020.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>F.-Q. Zhao, T.B. McFadden, R.K. Choudhary, E.M. Shangraw, R.O. Rodrigues, A.J. Spitzer. Intramammary endotoxin challenge elicits time-dependent local and systemic effects on lactating bovine mammary glands. CRWAD 2019, Chicago. Oral</p><br /> <p>Choudhary RK, Spitzer A, McFadden TB, Shangraw EM, Rodrigues RO, Linder HF, Zhao F-Q (2019) Quantitative histological changes in lactating bovine mammary gland after endotoxin challenge. ADSA Annual Meeting 2019, Cincinnati, Ohio. Oral</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Conference Proceedings </strong></p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Erskine, R.J. and A. J. Swan. The Veterinarian&rsquo;s/Consultant&rsquo;s Role in Milk Quality. Short course presented at the 60th Annual Meeting of the National Mastitis Council, Orlando, FL, January 30th, 2020.</p><br /> <p>Erskine, R.J. Three ways to lose money on a farm: A view from the udder. Seminar presented at the Western Canadian Dairy Seminar, Red Deer, Alberta, March 11th, 2020.</p><br /> <p>Erskine R.J. Dry cow management for udder health. Webinar presented for University of Wisconsin webinar series-Dry cow management for a successful lactation. June 11th, 2020.&nbsp;</p><br /> <p>Erskine R.J. and R.S. Thomson. Podcast presented for the A.A.B.P. Why udder prep matters. November 19th, 2020.</p><br /> <p>Ruegg, P.L.&nbsp; Understanding the economic impact of mastitis.&nbsp; The role of duration and drug selection.&nbsp; 2020. Pp 84-91 in Proc. 3rd Am. Assoc. Bov. Pract. Recent Graduate Conf. Columbus OH, Feb 21-22, 2020.&nbsp; Available online:&nbsp; www.aabp.org&nbsp;&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Bedding management and udder health &ndash; Getting better results from recycled manure solids and sand bedding. Godden, S., F. Pena Mosca, E. Royster, J. Timmeran, B. Crooker. 2020. Proc. Annu. Mtg. MN Dairy Health Conference. Aug. 6, 2020. (virtual).</p><br /> <p>Using Rapid Culture Systems to Guide Selective Treatment of Clinical Mastitis and at Dry-off. S. Godden, E. Royster and J. Timmerman. 3 hrs Short Course #1 at 59th Annu Meeting of the National Mastitis Council. Jan 28-30, 2020. Orlando, FL.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Missouri</span></p><br /> <p>Kurban D, Bolduc E, Roy JP, DeVries T, Gervais R, Adkins P, Middleton JR, Keefe G, France A, Dufour S.&nbsp; 2020.&nbsp; An observational transversal study on the prevalence of staphylococcal intramammary infections after calving in primirparous vs multiparous cows milked using an automatic milking system.&nbsp; Proceedings of the 59th Annual Meeting of the National Mastitis Council, Orlando, FL.&nbsp;</p><br /> <p>Ankney SR, Rivero LA, Witzke MC, Naclerio A5, Adkins PRF, Middleton JR. 2020.&nbsp; Comparison of two techniques for collecting milk samples to diagnose intramammary infection in dairy cattle.&nbsp; Proceedings of the 59th Annual Meeting of the National Mastitis Council, Orlando, FL</p><br /> <p>Witzke MC, Rodrigues RO, Shangraw EM, Ericsson AC, McFadden TB, Adkins PRF. 2020 Feed restriction and heat stress alters the fecal microbiome.&nbsp; Proceedings of the 59th Annual Meeting of the National Mastitis Council, Orlando, FL</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>F.-Q. Zhao, T.B. McFadden, R.K. Choudhary, E.M. Shangraw, R.O. Rodrigues, A.J. Spitzer. Intramammary endotoxin challenge elicits time-dependent local and systemic effects on lactating bovine mammary glands. CRWAD 2019, Chicago. Oral</p><br /> <p>Choudhary RK, Spitzer A, McFadden TB, Shangraw EM, Rodrigues RO, Linder HF, Zhao F-Q (2019) Quantitative histological changes in lactating bovine mammary gland after endotoxin challenge. ADSA Annual Meeting 2019, Cincinnati, Ohio. Oral</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Utah State University</span></p><br /> <p>Wilson DJ, Britten JE, Clancy CS, Rood KA: Bovine mammary gland involution histology and morphometry following casein hydrolysate and other dry treatments. Proc 63rd Ann Conf Am Assoc Vet Lab Diag: 2020.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Poster Presentations</strong></p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Michigan State University</span></p><br /> <p>Leite de Campos, J., A. Steinberger, T. Goldberg, N. Safdar, A. Kates, J. Sutske, A. Sethi, G. Suen and P. Ruegg.&nbsp; 2020.&nbsp; Distribution of antimicrobial usage for mastitis using two metrics.&nbsp; Pp 223-224 in Proceedings 59th Ann. Meeting NMC.&nbsp; Jan 28-31, 2020.&nbsp; Available www.NMConline.org</p><br /> <p>Kolar, Q. K., S. M. Godden, and P.L. Ruegg.&nbsp; 2020.&nbsp; Duration of subclincial mastitis prior to occurrence of non-severe clinical mastitis &ndash; results of a preliminary analysis. Proceedings 59th Ann. Meeting NMC.&nbsp; Jan 28-31, 2020.&nbsp; Available www.NMConline.org</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><span style="text-decoration: underline;">Rutgers University</span></p><br /> <p>Schaeffer L.M., Begum-Gafur R., Zaidel L., Crane S., Norabuena-Morales J., Boyd J.M., The effect of Zinc and tin of the growth and oxidative stress response of key oral bacteria. International Association for Dental Research Meeting. Washington D.C. 3/2020.</p><br /> <p>Norambuena J.,&nbsp; Boyd J.M., Aeration Influences Copper Toxicity in Staphylococcus aureus. Rutgers Microbiology Symposium. New Brunswick, NJ. 2/2020.</p><br /> <p>Esquilin-Lebron K., Foley M., Carabetta V., Beavers W., Skaar E., Boyd J.M., Iron-Sulfur Protein Assembly in Gram Positive Bacteria. Rutgers Microbiology Symposium. New Brunswick, NJ. 2/2020.</p><br /> <p>Almeda-Ahmadi A., McGinley C.M., Boyd J.M., The Effects of Varying Concentrations of Cetylpyridinium Tetrachloride with Tin on the Growth of Common Oral Cavity Bacteria. Rutgers Microbiology Symposium. New Brunswick, NJ. 2/2020.</p><br /> <p>Norambuena J., Al-Tameemi H., Boyd J.M. Staphylococcus aureus lacking a functional MntABC manganese import system have increased resistance to copper. American Society National Meeting.&nbsp; Virtual 7/2020.</p><br /> <p>Price, E.E. Mashruwala A.A., Boyd J.M. Examining Activators of SaeRS in S. aureus Fermentative Biofilm Formation American Society National Meeting.&nbsp; Virtual 7/2020.</p><br /> <p>Norambuena J., Al-Tameemi H., Boyd J.M. Staphylococcus aureus lacking a functional MntABC manganese import system have increased resistance to copper. Boston Bacteriology Meeting.&nbsp; Virtual 7/2020.</p><br /> <p>Esquil&iacute;n-Lebr&oacute;n K., Foley M., Carabetta V., Beavers W., Skaar E.P., Boyd J.M. Investigating the role of YlaN in iron homeostasis in Staphylococcus aureus. Boston Bacteriology Meeting.&nbsp; Virtual 7/2020.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Minnesota</span></p><br /> <p>Rowe, S.M.*, Godden, S.M., Nydam, D.V., Lago, A., Royster, E., Vasquez, A. 2020. Randomized controlled trial evaluating the efficacy of two commercial internal teat sealants in dairy cows. Poster presentation. National Mastitis Council Annual Meeting. Jan. 28-30, 2020. Orlando, FL, USA</p><br /> <p>Rowe, S.M.*, S.M. Godden, E. Royster, J. Timmerman and M. Boyle. Cross-sectional study of the relationship between cloth udder towel management, towel bacteria counts and intramammary infection in late lactation dairy cows. Poster Presentation. 2019 Conference of Research Workers in Animal Diseases. Nov. 2-5, 2019. Chicago, IL.</p><br /> <p>Rowe, S.M.*, S.M. Godden, E. Royster, J. Timmerman and M. Boyle. Cross-sectional study of bedding bacteria counts and intramammary infection in late lactation dairy cows. Poster presentation. 2019 Conference of Research Workers in Animal Diseases. Nov. 2-5, 2019. Chicago, IL.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Vermont</span></p><br /> <p>Shangraw E, Rodrigues R, Witzke M, Linder HF, Choudhary R, Spitzer A, Zhao, F-Q, McFadden TB (2019) Intramammary lipopolysaccharide infusion elicits local or systemic effects depending on milk component. ADSA Annual Meeting 2019, Cincinnati, Ohio. Poster</p><br /> <p>&nbsp;</p><br /> <p><strong>Other Publications</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">Ohio State University</span></p><br /> <p>Enger, B.D. 2020. Blanket, selective, or no dry cow therapy: Which should I choose? OARDC Extension. Dairy in brief (DIB) # 22-20.</p><br /> <p>Enger, B.D. 2020. Therapy of Clinical Mastitis in Tough Economic Times. OARDC Extension. Dairy in brief (DIB) # 41-20.</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">University of Tennessee</span></p><br /> <p>Eckelkamp, E., K. Hash, and G. Pighetti. 2020. On-Farm Culture Setup. UT Extension Publication, W 946.</p><br /> <p>Hash, K., E. Eckelkamp, and G. Pighetti. 2020. Aseptic Milk Sampling Guide: Taking Clean Samples. UT Extension Publication, W 945.</p><br /> <p>Cox, M., E. Eckelkamp, G. Pighetti, P. Krawczel, and R. Trout Fryxell. 2019. Dairy parlor management: Fly control. UT Extension Publication, W 817.</p><br /> <p>Couture, V. G. Pighetti, P. Krawczel, and E. Eckelkamp. 2019. Managing mastitis in heifers and dry cows. UT Extension Publication, W 813.</p>

Impact Statements

Back to top

Date of Annual Report: 01/24/2022

Report Information

Annual Meeting Dates: 11/03/2021 - 11/05/2021
Period the Report Covers: 11/01/2020 - 11/04/2021

Participants

Pamela Adkins University of Missouri
John Middleton University of Missouri
John Barlow University of Vermont
Sandra Godden University of Minnesota
Benjamin Enger Ohio State University
Paolo Moroni Cornell University
Sam Rowe University of Sydney
David Wilson Utah State University
Oudessa Kerro Dego University of Tennessee

Brief Summary of Minutes

Accomplishments

<p><strong>OBJECTIVE 1: Characterize host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance to improve economic outcomes and animal welfare.</strong></p><br /> <p><strong><span style="text-decoration: underline;">Louisiana State University</span></strong></p><br /> <p>Member: William E. Owens</p><br /> <p>Identify and evaluate bacteria causing bovine mastitis in Louisiana. Test bacteria for antimicrobial resistance to mastitis therapeutics.</p><br /> <p><strong><span style="text-decoration: underline;">Ohio State University</span></strong></p><br /> <p>Member: Benjamin D. Enger, Kellie M. Enger</p><br /> <p>An animal trial was completed that investigated how intramammary infections affect mammary gland growth and development in dairy heifers.&nbsp; First results indicate that IMI reduce the amount of epithelial tissues present in infected mammary glands and increase the abundance of stromal tissues.</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: Sam Rowe, Pat Gordon, Daryl Nydam</p><br /> <p>Patel, S. Godden, E. Royster, B. Crooker, T. Johnson, E. Smith, S. Sreevatsan. Prevalence, antibiotic resistance, virulence and genetic diversity of Staphylococcus aureus isolated from bulk tank milk samples of U.S. dairy herds. The objective of this study was to investigate the prevalence, antibiotic resistance gene content, virulence determinants and genomic diversity of S. aureus in bulk tank milk samples (BTM) from U.S. dairy herds. Of 365 pooled BTM samples collected from 189 herds across the United States, S. aureus was cultured from 170 samples, indicating a sample prevalence of 46.6% in the BTM and a herd prevalence of 62.4% (118 out of 189 herds). Among a subset of 138 S. aureus isolates that were stored for further analysis, 124 were genome sequenced after being confirmed as S. aureus using phenotypic tests. Our findings indicated a high prevalence of S. aureus in BTM, with little evidence of antibiotic resistance but often carrying genes for the various enterotoxins. This study identified predominant genetic clones and provides continued surveillance of S. aureus strains found in U.S. dairy herds. Manuscript published in 2021.</p><br /> <p>Noyes, L.S. Caixeta, B.J. Heins, V.S. Machado, P. Pinedo. Catalyzing an open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case. The two objectives of this study are (1) to describe the teat skin microbiome of dairy heifers in certified organic herds in the US and (2) to investigate the association between teat skin microbiome throughout the transition period and the occurrence of intramammary infections. Approximately 500 nulliparous heifers from 5 organic dairy herds in MN, CO, TX, and NM were enrolled in the summer and fall of 2019. Milk culture for the first 5 weeks post-partum for all the cows enrolled in the study was carried out during fall 2019 and spring 2020. Total DNA was extracted and 16S V4 amplicons were sequenced for ~8,000 gauze samples of the external teat apex epithelium and ~2,000 samples of the internal teat canal epithelium. Sequencing of all samples is nearly complete and full data analysis will occur in late 2021 through 2022.</p><br /> <p>Dean, L.S. Caixeta, N. Noyes, S. Godden, B. Crooker, F. Pena Mosca. Impact of the cow udder microbiome on the biodiversity of milk. Exploratory project to investigate the microbiome in different parts of the mammary gland, and to better understand how microbiome-host interactions influence response to mastitis challenge. Sample collection and sequencing will be completed in early 2022, and data analysis will be completed by end of 2022.</p><br /> <p>A. Crooker, S. M. Godden, L. Caixeta, A. Seykora, M. Schutz, J. D. Lippolis, J. B. Cole and B. Rosen. Reducing mastitis in the dairy cow by increasing the prevalence of beneficial polymorphisms in genes associated with mastitis resistance. Our premise is that previous selection practices have successfully increased the presence of genetic polymorphisms associated with increased milk yield but have decreased the presences of polymorphisms associated with disease resistance. We are using contemporary Holsteins (CH) and unique unselected Holsteins (UH) that have not been subjected to selection since 1964 to determine differences in immune and inflammatory responses and mammary gene expression. We will link differential phenotypic responses with whole genome sequencing to identify polymorphisms that impact mastitis resistance. These polymorphisms could enhance gene-assisted selection efforts to increase mastitis resistance. Intramammary challenge studies and data analysis are underway. Intramammary challenge studies with <em>Escherichia coli</em> strain P4 and <em>Streptococcus uberis</em> strain 0140J have been conducted and analyses of collected milk, blood and DNA samples are underway. Impact of Holstein genotype on immune response to lipopolysaccharide (LPS) and lipoteichoic acid (LTA) was assessed in an ex-vivo whole blood study demonstrated greater cytokine responses in the UH cows.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Missouri</span></strong></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Samantha Haw</p><br /> <p>Ongoing collaborations with University of Montreal and University of Calgary to better define the role of non-aureus staphylococci in bovine mastitis. A collaboration with the University of Maryland has been established to evaluate big-data genomics to improve dairy cattle health, including investigating mastitis resistance.</p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>In collaborations with Drs. Jana Kraft and Thomas McFadden, Zhao lab has investigated that the effects of intramammary challenge of lipopolysaccharide (LPS) on fatty acid profiles in blood and milk. It was shown that LPS challenge significantly changed the fatty acid profiles in triglyceride and phospholipid fractions in blood, but not in milk. (VT MO)</p><br /> <p><strong><span style="text-decoration: underline;">Oregon State University</span></strong></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>Contributors: Serkan Ates, Hunter Ford</p><br /> <p>Characterize host mechanisms and pathogenic virulence factors associated with mastitis susceptibility and resistance to improve economic outcomes and animal welfare.</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Member: Paolo Moroni</p><br /> <p>Contributors: Gloria Gioia, Anja Sipka, Paul Virkler, Daryl Nydam</p><br /> <p>We will continue the research program investigating the opportunity to understand the epidemiology of Mycoplasma spp and prevalence in US farms. In collaborations with different group, we investigated genotyping and antimicrobial susceptibility profiling of <em>Streptococcus uberis</em> isolated from a clinical bovine mastitis outbreak in a dairy farm. Molecular characterization contributed to understanding their modes of dissemination, and revealed that <em>Strep. uberis</em> infections could be related to a restricted number of predominant cow-adapted strains.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Member: Oudessa Kerro Dego</p><br /> <p>Contributors: Benti Deresa Gelalcha, Aga Edema Gelgie, Jessica Vidlund, Barbara Gillespie, Ximin Zeng, Jun Lin</p><br /> <p>Bovine mycoplasmoses, which is mostly caused by <em>Mycoplasma bovis</em>, is a significant problem in the dairy and beef industry. Mycoplasma is an important etiologic agent of bovine mastitis leading to significant economic losses and affecting animal welfare. It is highly contagious and resistant to antibiotics and lacks cell wall rendering certain classes of antibiotics ineffective. Intramammary <em>Mycoplasma bovis </em>infections cause elevation in milk somatic cell count which consequently result in poor milk quality. Current mastitis control measures are not effective against mycoplasma mastitis. In addition, the current lack of information on virulence factors hampered the development of immunological or other alternatives to control <em>M. bovis</em> mastitis. Thus, the lack of preventive measures or treatment options renders infected animal culling as the recommended practice for controlling this disease. The objectives of this study are 1) determine <em>M. bovis </em>IMI status in East TN dairy farms, 2) identify genes responsible for development of <em>M. bovis</em> mastitis using transposon-based mutagenesis and <em>M. bovis</em> transcriptome analysis during early host-<em>M. bovis</em> interactions and 3) Evaluate detailed transcriptomic, histopathological and immunological changes during pathogenesis of experimentally induced <em>M. bovis</em> mastitis.</p><br /> <p><strong>Summary preliminary results</strong>:</p><br /> <ul><br /> <li>Transposon (Tn4001) mutant library of <em> bovis</em> PG45 were created (Collaborative BARD project with Kimron Veterinary Institute, Israel)</li><br /> <li>Mutant clones&rsquo; library were characterized for loss of important virulence by in vitro adhesion and invasion assays</li><br /> <li>Clones that showed defect to cause in vitro infection of mammary epithelial cells were selected and we will be tested in vivo by experimental challenge infection</li><br /> </ul><br /> <p><strong><span style="text-decoration: underline;">University of Idaho</span></strong></p><br /> <p>Members: Pedram Rezamand</p><br /> <p>The Rezamand lab continues to work on identifying relationships between nutrient metabolism during the periparturient period and health issues (mastitis) in Pacific Northwest dairy herds.&nbsp;An objective of the study is to assess the relationship between the milk fatty acid profile of dairy cows with mastitis status of the cows during the periparturient period. 115 healthy and mastitic and lameness cows were randomly selected for milk fatty acid composition (gas chromatography). Health records were categorized based on the occurrence of postpartum diseases. Data were analyzed using linear mixed models in SAS with significance declared at <em>P </em>&le; 0.05.&nbsp; results showed that under specific conditions of the experiment, no significant difference in milk fatty acid composition between diseased and healthy cows was observed</p><br /> <p>&nbsp;</p><br /> <p><strong>OBJECTIVE 2: Assess and apply new technologies that advance mastitis control, milk quality and/or dairy food safety.</strong></p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: Sam Rowe, Pat Gordon, Daryl Nydam</p><br /> <p>&nbsp;</p><br /> <p>Nusrat Annie Jahan, S. Godden, T. Schoenfus, C. Gebhart, E. Royster, S. Wells, J. Timmerman, and R. Fink. Evaluation of the Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS) system in the detection of Mastitis pathogens from Bovine milk samples. In the present study, we prospectively compared MALDI-TOF MS (Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry or MALDI-ToF) to the conventional 16S rDNA sequencing method for the identification of&nbsp; mastitis isolates (481). Among the 481 milk isolates, 445 (93%) were putatively identified to the genus level by MALDI-TOF MS and 355 (74%) were identified to the species level, but no reliable identification was obtained for 16 (3.3%), and 20 (4.2%) discordant results were identified. In conclusion, our results show that MALDI-TOF MS is a fast and reliable technique which has the potential to replace conventional identification methods for most dairy pathogens, routinely isolated from the milk and dairy products. Manuscript published in 2021.</p><br /> <p>L.S. Caixeta. Applying precision dairy farming and diagnostic technologies to detect mastitis at the time of and following dry off in dairy cows in a field study. The main objective of this study was evaluate accuracy of precision dairy monitoring and diagnostic technologies (i.e. differential somatic cell counter) in collecting data that effectively identifies appropriate animals for dry cow therapy compared to SCC and bacteriology. In this study, milk samples were collected 1d prior to dry-off and 3 and 5 days after calving. Milk samples were used for the measurement of differential SCC, SCC, and milk culture to identify intramammary infection. Cow enrollment and health and performance was completed between fall 2019 and fall 2020. Data analysis and reported will be completed in 2021.</p><br /> <p><strong><span style="text-decoration: underline;">University of Missouri</span></strong></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Samantha Haw</p><br /> <p>Collaborated with University of Montreal and other NE-1748 stations in the U.S. on work led by Simon Dufour and Daryna Kurban (University of Montreal) to define intramammary infection using MALDI-TOF identification of bacteria isolated from bovine milk samples.&nbsp; Studied the prevalence of mastitis pathogens in Jersey cattle and their association with subclinical mastitis.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab collaborated with Dr. De Vliegher and Dr. De Visscher from Ghent University and Dr. Fox from Washington State University in the development of a multilocus sequence strain typing scheme for <em>Staphylococcus chromogenes</em>. (VT, WA, Ghent)</p><br /> <p>The Barlow lab completed a survey of organic dairy herds in Vermont that identified the frequency of different housing and bedding management systems used for winter housing of lactating cows. Tie-stall barns using wood shavings&nbsp; bedding was the most common system used on this population of farms.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Utah State University</span></strong></p><br /> <p>Members: David Wilson</p><br /> <p>Contributors: Kerry Rood, Justine Britten&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;</p><br /> <p>Screen a large panel of blood and serum clinical pathology tests on dairy calves and develop the most practical reduced panel of tests for prognosis and treatment of sick dairy calves during the first 60 days of life.&nbsp; This will be compared to a clinical scale of calf disease signs as well as the treatment and disease or health outcomes of the calves.&nbsp; The goal is to develop an affordable, rapid blood test panel that can be used on calves showing early signs of disease for better prognostic and treatment purposes, especially in large scale calf rearing.</p><br /> <p>Study different concentrations of a preservative for milk, colostrum and milk replacer fed to calves.&nbsp; First phase will examine association with bacteria counts over time under the conditions which exist when those products are fed to dairy calves, in comparison with untreated milk.&nbsp; The goal is to determine a safe but effective concentration of preservative that can be added to colostrum and milk fed to dairy calves.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Member: Paolo Moroni</p><br /> <p>Contributors: Gloria Gioia, Anja Sipka, Paul Virkler, Daryl Nydam</p><br /> <p>We will continue to investigate new technology for early identification of Mycoplasma from milk and compare different type of on farm culture. Survey and interviews on the use of antibiotics in New York State farm could be also one of our topics.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Member: Oudessa Kerro Dego</p><br /> <p>Contributors: Benti Deresa Gelalcha, Aga Edema Gelgie, Jessica Vidlund, Barbara Gillespie, Ximin Zeng, Jun Lin</p><br /> <p><strong>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; Prevalence of Antimicrobial Resistant and Extended Spectrum Beta-Lactamase producing</strong><strong>&nbsp;<em>Escherichia coli </em>in Dairy Cattle Farms in East Tennessee</strong></p><br /> <p>Antimicrobials have been widely used in dairy farms to prevent and control dairy cattle diseases since the1960s. This led to the emergence of antimicrobial resistant bacteria (ARB) that, along with their antimicrobial resistance genes (ARGs), can spread from dairy farms to humans. Therefore, regular antimicrobial resistance (AMR) monitoring is important to implement proper mitigation measures. The objective of this study was to determine prevalence of extended-spectrum beta-lactamases producing <em>E. coli</em> (ESBL-<em>E. coli</em>) in in East Tennessee dairy&nbsp; farms. A total of eighty samples consisting of twenty samples each of bulk tank milk, feces, dairy cattle manure amended soil, and prairie soil adjacent to the farms were collected and cultured for the isolation of <em>E. coli</em>. &nbsp;Tetracycline (TET<sup>r</sup>)-, third-generation cephalosporin (TGC<sup>r</sup>)- and nalidixic acid (NAL<sup>r</sup>)-resistant <em>E. coli</em> (n=88) were isolated and identified on agar media supplemented with tetracycline, cefotaxime, and nalidixic acid, respectively. TGC<sup>r</sup> <em>E. coli</em> were tested for ESBLs and other co-selected ARGs. &nbsp;</p><br /> <p><strong>Results:</strong> TET<sup>r</sup> (74%, n=88) was the most common, followed by TGC<sup>r</sup> (20%) and NAL<sup>r</sup> (8%). Farms had significant (<em>P</em>&lt;0.001) differences: the highest prevalence of TGC<sup>r</sup> (55%) and TET<sup>r</sup> (100%) were observed in farm D, while all NAL<sup>r</sup> isolates were from farm C. Over 83% of TGC<sup>r</sup> isolates (n=18) harbored ESBL gene<em> bla</em><sub>CTX-M</sub>. Majority (78%) of the <em>E. coli</em> isolates were multidrug-resistant (MDR) being positive for beta-lactams (<em>bla</em><sub>CTX-M</sub>), tetracyclines <em>tet</em>(<em>A</em>)<em>, tet</em>(<em>B</em>)<em>, tet</em>(<em>M</em>)), sulphonamides (<em>sul</em>2), aminoglycosides (<em>strA</em>)<em>, </em>and phenicols (<em>floR</em>).</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Molecular Epidemiology of Extended-Spectrum &beta;-lactamase Producing&nbsp;<em>E. coli</em>&nbsp;in East Tennessee Dairy Farms</strong></p><br /> <p>Most dairy farms in the USA- use third-generation cephalosporins (TGC), a medically important class of antibiotics, for the treatment and prevention of infectious diseases of dairy cattle (1). Recent reports are indicating that resistance to a third-generation cephalosporin (TGC) is rising among <em>Enterobacteriaceae</em> such as <em>Escherichia coli</em> (<em>E. coli</em>) in dairy operations and human health settings. We hypothesized that ESBLs-<em>E. coli</em> and -<em>Klebsiella</em> spp.&nbsp; are widespread in dairy farms and their environments, and they co-carry other resistance genes. The objectives of this study are 1) determine the prevalence of ESBLs-<em>E. coli</em> and -<em>Klebsiella</em> spp.&nbsp; in dairy cattle and manure, 2) determine the genomic relationship among all ESBLs -E. coli and -Klebsiella spp.</p><br /> <p><strong>Preliminary results</strong></p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; A total of 512 rectal fecal samples were collected from 512 cattle from 14 dairy&nbsp; farms</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; A total of 22 bulk tank milk, 25 manure (stored manure, pooled manure from farm floor, lagoon, etc.), 13 &nbsp;&nbsp;&nbsp;&nbsp;&nbsp;</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; feed samples, and 17 water samples were collected and screened for ESBL-<em>E. coli</em> and -<em>Klebsiell</em>a spp.</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; From rectal fecal samples, a total of 278 presumptive ESBLs-<em>E. coli </em>and 208 ESBLs-<em>Klebsiell</em>a spp. were &nbsp;&nbsp;</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; isolated and stocked for further confirmation and molecular analysis</p><br /> <p>&nbsp; &nbsp;&nbsp;&nbsp;&nbsp;&nbsp;From other farm samples (Bulk Tank Milk, manure, feed and water) 27 presumptive ESBL-<em>E. coli, and </em>26 &nbsp;&nbsp;</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; ESBLs<em>-Klebsiell</em>a spp. were isolated and stocked for further confirmation and molecular analysis</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; Suspected ESBL-<em>E. coli</em> and -<em>Klebsiell</em>a spp. were isolated from all sample types (rectal fecal sample, bulk &nbsp;&nbsp;</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; tank milk, manure, feed, and water).</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>OBJECTIVE 3: Identify and apply new strategies associated with the control of mastitis that can reduce the use of antibiotics in dairy herds.</strong></p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Louisiana State University</span></strong></p><br /> <p>Member: William E. Owens</p><br /> <p>Continue evaluation of botanical formulations from plants for antimicrobial activity against mastitis</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: Sam Rowe, Pat Gordon, Daryl Nydam</p><br /> <p>&nbsp;</p><br /> <p>S.M. Rowe, S.M. Godden, D.V. Nydam, P.J. Gorden, A. Lago, A.K. Vasquez, E. Royster, J. Timmerman, M.J. Thomas. Randomized controlled non-inferiority trial investigating the effect of two selective dry cow therapy protocols on antibiotic use and udder health: quarter-level outcomes. The objective of this study was to compare culture- and algorithm-guided SDCT programs to blanket dry cow therapy (BDCT) in a multi-site, randomized, natural exposure, non-inferiority trial on quarter-level infection dynamics during the dry period and on cow health and performance during the first 120 DIM. Results showed that the two SDCT approaches (culture- and algorithm-guided) each reduced antibiotic use at dry-off by 55%, without causing any negative impacts on intramammary infection dynamics during the dry period. An economic analysis showed an estimated positive economic return, on average, with either SDCT program. We conclude that SDCT can be used in appropriate U.S. dairy herds to reduce antibiotic use while maintaining udder health.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p>Godden, S., S. Wells, E. Royster, B. Crooker. Management of Recycled Manure Solids Bedding on Midwest Dairy Farms. Part 1 - Relationship between Processing Method, Udder Health and Production, and Bedding Bacteria Counts. Godden. Our objective was to investigate if recycled manure solids (RMS) processing method was associated with udder health, milk production, and with bedding bacteria counts (BBC) in ready-to-use RMS bedding samples. Twenty-nine premises in MN and WI were enrolled. Processing systems represented included green (GRN; n=7), drum composted (COM; n=4), digested (DIG; n=6) or mechanically dried (DRY; n=12). Farms were visited twice (summer 2019/winter 2020) to collect samples and data. Ready-to-use bedding was cultured to describe BBC. Results showed that herds using DRY or COM RMS processing systems generally had improved udder health and, for DRY, improved milk production, as compared to herds using DIG or GRN solids. Also, BBC were most consistently lowest in ready-to-use DRY or COM samples. &nbsp;</p><br /> <p>Godden, S., S. Wells, E. Royster, B. Crooker. Investigation of the Relationship between Method for Processing Recycled Manure Solids and Pathogen Control on Midwest Dairy Farms. Our objectives were to Investigate if method of recycled manure solids (RMS) processing reduces or eliminates viable mastitis pathogens as well as zoonotic pathogens in RMS. In summer of 2021 slurry and pre- and post-processed RMS were sampled from 27 herds in MN and WI.&nbsp; Laboratory analysis of samples is ongoing.</p><br /> <p>Godden, S. and E. Royster. Pilot Study: Refining a Culture-Guided Selective Dry Cow Therapy Program (SDCT) to Reduce Antibiotic Use on Dairy Farms. We hypothesize that by identifying and selectively treating only IMI caused by SSLO, we can dramatically reduce antibiotic use at dry-off by as much as 90%, while maintaining udder health, cow health and performance in the subsequent lactation (versus blanket DCT).&nbsp; In summer, 2020, we conducted a pilot study in 2 commercial herds to evaluate the effect of implementing a SDCT program that identifies and treats only IMI caused by SSLO on measures of quarter health, cow health and performance and antibiotic use (vs. BDCT). Lab culture of samples is ongoing.</p><br /> <p>Noyes, L. Caixeta, B. Heins, V. Machado, P. Pinedo, A. Formiga, D. Van Nydam, J. Velez, H. Karremann. Catalyzing an open-community research and education program to leverage the microbiome for the advancement of organic livestock production, using mastitis as a test case. The long-term goal of this proposal is to foster application-driven microbiome research activity within the dairy scientific community, thus supporting the discovery and development of novel, evidence-based tools to combat dairy disease and production challenges, with a focus on mastitis. Project objectives described above.&nbsp;</p><br /> <p>Rowe, L.S. Caixeta, E. Royster, S. Godden. Clinical trial evaluating the health effects of reducing antibiotic use at dry-off in dairy cows. Randomized clinical trial comparing blanket dry cow therapy and no dry cow therapy in a low input commercial dairy farm. Enrollment of cows started in the summer 2021 and will continue in the spring of 2022. Lactation health and performance for the first 100 DIM will continue until winter 2022. Data analysis and reporting will be completed in 2023.</p><br /> <p>L.S. Caixeta, S. Godden, W. Knauer, J. Hadrich. G. Cramer. Randomized clinical trial evaluating the effect of administering Bovikalc&reg; dry at dry off on udder health, early lactation health and performance, and economics in dairy cows. Randomized clinical trial investigating the effects of pre dry-off administration of acidogenic bolus on udder health during the dry period and in early lactation. Enrollment of cows was completed in the summer and fall of 2021. Cows will be follow until the spring of 2022. Data analysis and reporting will be completed in 2022.</p><br /> <p><strong><span style="text-decoration: underline;">Oregon State University</span></strong></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>Contributors: Serkan Ates, Hunter Ford</p><br /> <p>The objective was to assess the potential of using chicory silage in combination with organic selenium in improving the repose to intramammary infection. For the purpose we used 30 Polypay lactating sheep in a 2x2 experimental design where animals were fed for 40 days during pregnancy and 45 days during lactation with isocaloric and isonitrogenous rations containing either chicory silage or grass silage and receiving 0 or 3.6 mg of Se/day as Se-yeast into the concentrate. At 35 days in lactation 2&acute;10<sup>7</sup> CFU of <em>Strep uberis</em> were infused into both mammary glands. Animals were left untreated for 10 days after which intramammary antibiotics were infused and the sheep dried off. Daily feed and water intake were recorded, milk yield and rectal temperature were measured twice daily, blood was collected prior to and for 10 days after IMI for a complete blood count, leukocyte migration and phagocytosis, and large metabolic, oxidative, liver function, and immune profiling, and milk was collected for milk component analysis, including fatty acid profiling. Preliminary data indicated a minor effect of Se-yeast on the response to mastitis and a more prominent effect of feeding chicory silage.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong><span style="text-decoration: underline;">Ohio State University</span></strong></p><br /> <p>Member: Benjamin D. Enger, Kellie M. Enger</p><br /> <p>An animal trial was completed that investigated the ability of teat sealants to prevent intramammary infections is non lactating heifers. Results indicate that properly administered teat sealants can provide a significant protective effect and are likely to improve first lactation performance given fewer animals begin lactation with infections. Animals with fewer infections are likely to require less antibiotic treatments.</p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Missouri</span></strong></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Samantha Haw</p><br /> <p>Co-authorship on invited reviews led by Herman Barkema and his team at University of Calgary on selective dry cow therapy and selective treatment of clinical mastitis along with authors from other NE-1748 stations in the U.S.A, Canada, and Europe.</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Member: Paolo Moroni</p><br /> <p>Contributors: Gloria Gioia, Anja Sipka, Paul Virkler, Daryl Nydam</p><br /> <p>Co-authorship on invited reviews led by Sandra Godden&nbsp; and his team at University of Minnesota on selective dry cow therapy and selective treatment of clinical mastitis. Validation of commercial PCR for identification of <em>Mycoplasma spp</em> and <em>Staphylococcus aureus</em></p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Member: Oudessa Kerro Dego</p><br /> <p>Contributors: Benti Deresa Gelalcha, Aga Edema Gelgie, Jessica Vidlund, Barbara Gillespie, Ximin Zeng, Jun Lin</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Member: Oudessa Kerro Dego</p><br /> <p>Contributors: Benti Deresa Gelalcha, Aga Edema Gelgie, Jessica Vidlund, Barbara Gillespie, Ximin Zeng, Jun Lin</p><br /> <p><strong>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; Evaluation of Efficacy of Staphylococcal Surface Proteins Vaccine to Control Mastitis in&nbsp;</strong><strong>Dairy Cows</strong></p><br /> <p>Bovine staphylococcal mastitis is a major cause of economic losses in the dairy industry throughout the world. Coagulase-positive <em>Staphylococcus aureus</em> is the most common pathogen that causes mastitis in dairy cows. <em>Staphylococcus chromogenes</em> is one of coagulase-negative staphylococci frequently isolated from intramammary infection (IMI) characterized by high somatic cell count (SCC). Sustainable alternatives to antimicrobials such as improved effective vaccines are critically required to control mastitis.</p><br /> <p>We developed <em>Staphylococcus aureus </em>surface proteins (SASP) and <em>Staphylococcus chromogenes</em> surface proteins (SCSP) based vaccines. Previously, we completed two evaluations of the protective efficacy of these vaccines using controlled experimental vaccination and challenge studies. Our results showed that both SASP and SCSP vaccines are immunogenic and the induced immunity partially protected vaccinated cows from <em>S. aureus</em> mastitis. &nbsp;In the current study, we evaluated the immunological responses and protection from mastitis in dairy cows vaccinated with&nbsp; SCSP and&nbsp; SASP vaccines over one full lactation of 300 days. A total of 45 dairy cows in their 1<sup>st</sup>&nbsp;or 2<sup>nd</sup>&nbsp;lactation from the East Tennessee Research and Education Center dairy herd were randomly enrolled in vaccination with SCSP (n=16) or SASP (n=15) with Emulsigin-D&reg; adjuvant&nbsp;(Phibro Animal Health)&nbsp;&nbsp;at the drying off (D0), 21, and 42 days after drying off (D21 and D42). Control cows (n=14) were injected with PBS (pH 7.4) mixed with Emulsigin-D&reg; at similar time points.&nbsp;Cows in the SCSP and SASP groups were vaccinated with 1.2 mg of SCSP and SASP vaccine, respectively. All cows were vaccinated subcutaneously (SC) in the neck area.&nbsp;The 1st dose was administered on the left side of the neck, the 2nd dose on the right side of the neck, and the 3rd dose on the lower left side of the neck.Serum and milk antibody titers (IgG, IgG1, IgG2, and IgA) induced by SCSP or SASP were measured by ELISA.&nbsp;We monitored them for antibody titers in milk and serum, mastitis status (clinical, subclinical, no mastitis), somatic cell count (SCC), milk yield, bacterial count in CFU/mL of milk over one full lactation cycle.</p><br /> <p><strong>Results</strong>: Of the total 45 cows, 9 were removed from the study due to non-vaccine-related complications. All vaccinated cows had no allergic reactions or vaccine-related complications, so these vaccines are safe. Similar to the previous two studies, SASP and SCSP vaccines induced significantly increased antibody titers. The subsequent experimental challenge results showed that both vaccines confer partial protection, but SCSP conferred better protection than SASP. The duration of induced immunity was about 4 months. A total of 4 &nbsp;vaccine injections are required throughout lactation periods at drying off (D0), at days 21 and 42 after drying off, and at 120 days in milk or 120 days after calving.</p><br /> <p>&nbsp;</p><br /> <p><strong>Evaluation of Immunogenicity of Enterobactin Conjugate Vaccine for the Control of <em>E. coli</em> Mastitis in Dairy Cows</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p>Coliform bacteria, such as <em>Escherichia coli</em>, <em>Klebsiella,</em> and <em>Enterobacter</em>, widely exist in the environment and are common causes of clinical mastitis in dairy cows. <em>Escherichia coli</em> is the most common species isolated in more than 80% of cases of coliform mastitis. Despite decades of vaccine trials to develop an effective vaccine against <em>E. coli</em> mastitis in dairy cows, the efficacy of the current J5 based vaccine is limited only to reducing clinical disease and production losses. The limited effectiveness is probably due to antibody inaccessibility and antigenic diversity/variation issues. Therefore, a novel vaccine, with a new target of less antigenic changes, is highly warranted. Recently, a novel essential nutrient deprivation-based immunization approach, which targets Enterobactin (Ent), has been under investigation.</p><br /> <p>The objective of this study was to determine Ent-specific immune response to Ent-KLH vaccine in dairy cows during early lactation. A total of 12 healthy Holstein dairy cows from East Tennessee Research and Education Center Little River Animal and Environmental Unit Herd (ETREC-LAEU) were divided into two groups of 6 cows each. Cows in group 1 were vaccinated subcutaneously with 200 &mu;g Ent-KLH vaccine with Emulsigen-D (first vaccination) and Freund&rsquo;s incomplete adjuvant (2<sup>nd</sup> and 3<sup>rd</sup> vaccination) in the total volume of 5 mL. Cows in group 2 were injected with PBS (pH7.4) with Emulsigen-D (first injection) and Freund&rsquo;s incomplete adjuvant (2<sup>nd</sup> and 3<sup>rd</sup> injections). The serum and milk samples were analyzed for the Ent-specific IgG1, IgG2, and IgA immune responses by indirect ELISA.</p><br /> <p><strong>Results:</strong> KLH-Ent vaccine induced significantly higher antibody in serum and milk of vaccinates compared with control cows. In particular, in vaccinated cows, the IgG and IgG2 response against KLH-Ent conjugate and Ent reached the peak at calving (C), and continued increasing until 30 days after calving (C30). Ent specific IgG1 and IgA peaked at D42 and remained high at C and C14, and then quickly declined. The KLH-Ent successfully triggered strong Ent-specific immune response in dairy cows without significantly affecting the microbiota diversity and gut health. Thus, Ent conjugate vaccine may serve as effective vaccine against <em>E. coli</em> mastitis in dairy cows.</p><br /> <p>&nbsp;</p><br /> <p><strong>Developing a universal nanoparticle-based vaccine delivery system against infectious diseases </strong>(collaborative project between Dr. Paul Dalhaimer from Department of Chemical and Biomolecular Engineering, Tickle College of Engineering, UTK, Knoxville and Dr. Oudessa Kerro Dego, Department of Animal Science, UTIA, Knoxville). The objective of this study was to develop nanoparticles that have antigens of interest on the same nanoparticle. we propose to use the spherical nanoparticles (SNPs), diblock copolymers: poly(ethylene-oxide)-<em>b</em>-poly(butadiene) (PEO-b-PBD). PEO is FDA-approved for use in the body. Current nanoparticles tend to have just one functionality group &ndash; usually the antigen. Our nanoparticles have many attributes that make them ideal for use as vaccines. First, they are made from benign polymers that have been extensively administered to rodents and shown low toxicity. Second, they are stable even at temperatures ~80&ordm;C. This is a very important requirement for vaccines to be used in refrigeration-poor countries. Third, they have ideal physical dimensions. They are ~50 nm in diameter, which is ideal for reaching the lymph system from site of administration.&nbsp; Our nanoparticle allows for the attachment of multiple combinations of antigens and targeting peptides which makes it highly advantageous because you can attach multiple antigens and targeting peptides on one nanoparticle.</p><br /> <p><strong>Summary of preliminary results:</strong></p><br /> <p>We constructed nanoparticle-Staphylococcus aureus surface protein peptide (SASP-peptide) Vaccinated two groups of mice with Nanopartilce alone (Group 1) and nanoparticle-attached to SASP- peptide at days 0, 14 and 28. Blood samples were collected at days 0, 14, 28 before each vaccination and at day 42&nbsp; two weeks after last vaccination.We measured induced immune responses (IgG, IgM and IgA) against nanoparticle and nanoparticle-SASP-peptide conjugate by ELISA. Serum&nbsp; IgG antibody specific to&nbsp; nanoparticle-SASP peptide increased&nbsp; significantly from base line at D28 and D42. Both Nanoparticle alone and Nanoparticle-SASP peptide induced increased IgM antibody in serum at D28. We are currently running second vaccination and immune response evaluation in mice.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>WORK PLANNED FOR THE COMING YEAR, LISTED BY OBJECTIVE:</strong></p><br /> <p><strong>OBJECTIVE 1</strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Missouri</span></strong></p><br /> <p>Members: John Middleton, Pamela Adkins</p><br /> <p>Contributors: Samantha Haw</p><br /> <p>We will be investigating the antibacterial activity of <em>Staphylococcus chromogenes </em>isolates originating from dairy cattle, specifically looking at their <em>in vitro</em> ability to inhibit growth of Gram-positive mastitis pathogens. We will also begin a collaborative project with the University of Vermont, working to do whole genome sequencing and strain typing of <em>S. chromogenes</em> isolates, looking to determine genetic determinants of phenotypic behavior.&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Louisiana State University</span></strong></p><br /> <p>Member: William E. Owens</p><br /> <p>Identify and evaluate bacteria causing bovine mastitis in Louisiana. Test bacteria for antimicrobial resistance to mastitis therapeutics.</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: Sam Rowe, Pat Gordon, Daryl NydamA. Crooker, S. M. Godden, L. Caixeta, A. Seykora, M. Schutz, J. D. Lippolis and B. Rosen. Reducing mastitis in the dairy cow by increasing the prevalence of beneficial polymorphisms in genes associated with mastitis resistance. Analysis of collected milk, blood and DNA samples from unselected and contemporary Holsteins subjected to intramammary challenges of <em>Escherichia coli</em> strain P4 or <em>Streptococcus uberis</em> strain 0140J will continue. Impact of Holstein genotype on immune response to gram negative and gram positive bacteria that commonly cause mastitis will be assessed using ex-vivo incubation models.</p><br /> <p>Caixeta, S. Dow, N. Noyes, B. Crooker, S. Godden, D. Nydam, B. Walcheck. Effect of mucosal immune stimulation on mammary gland immune responses during the dry period and early lactation period in dairy cows. The two objectives of this project are to determine the effect of MucosImmune on (1) the innate immune response in the mammary gland during the period immediately before and after parturition, and (2) the resistance to bacterial infection of the mammary gland. Initial dose titration experiment will be carried out in the spring of 2022. The three main experiments proposed in this project will be completed between the summer of 2022 and winter 2023.</p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>Zhao lab will study the effects of LPS and different cytokines on milk synthesis in mammary epithelia cells.</p><br /> <p><strong><span style="text-decoration: underline;">Ohio State University</span></strong></p><br /> <p>Member: Benjamin D. Enger, Kellie M. Enger</p><br /> <p>Continue to investigate how mammary growth is altered by mastitis by assessing proliferation and apoptosis of epithelial cells in heifer mammary gland tissues and also assess how myoepithelial cells and their migration into the fat pad are affected by inflammatory processes.</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Member: Paolo Moroni</p><br /> <p>Contributors: Gloria Gioia, Anja Sipka, Paul Virkler, Daryl Nydam</p><br /> <p>We will be investigating the antibacterial activity of <em>Lactococcus garvieae &nbsp;</em>isolates originating from milk bedding and water.</p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Idaho</span></strong></p><br /> <p>Members: Pedram Rezamand</p><br /> <p>We will continue to examine the relationship between nutrient metabolism and periparturient diseases (i.e., mastitis).&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong>OBJECTIVE 2</strong></p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: Sam Rowe, Pat Gordon, Daryl Nydam</p><br /> <p>&nbsp;</p><br /> <p>Godden, S., S. Wells, E. Royster, B. Crooker. Management of Recycled Manure Solids Bedding on Midwest Dairy Farms. Part 1 - Relationship between Processing Method, Udder Health and Production, and Bedding Bacteria Counts. &nbsp;Final results will be analyzed and manuscripts submitted in 2022.&nbsp;&nbsp;</p><br /> <p>Godden, S., S. Wells, E. Royster, B. Crooker. Investigation of the Relationship between Method for Processing Recycled Manure Solids and Pathogen Control on Midwest Dairy Farms. Our objectives were to Investigate if method of recycled manure solids (RMS) processing reduces or eliminates viable mastitis pathogens as well as zoonotic pathogens in RMS. In summer of 2021 slurry and pre- and post-processed RMS were sampled from 27 herds in MN and WI.&nbsp; Laboratory analysis of samples is ongoing. Data analysis and reporting will occur in 2022.</p><br /> <p><strong><span style="text-decoration: underline;">Oregon State University</span></strong></p><br /> <p>Member: Massimo Bionaz</p><br /> <p>Contributors: Serkan Ates, Hunter Ford</p><br /> <p>Complete the analyses including blood profiling and milk fatty acid profiling and submit two manuscripts to peer-reviewed journals</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Member: Oudessa Kerro Dego</p><br /> <p>Contributors: Benti Deresa Gelalcha, Aga Edema Gelgie, Jessica Vidlund, Barbara Gillespie, Ximin Zeng, Jun Lin</p><br /> <p>Molecular Epidemiology of Extended-Spectrum &beta;-lactamase Producing&nbsp;<em>E. coli</em>&nbsp;in East Tennessee Dairy Farms</p><br /> <p>Identification of Virulence Factors &amp; Pathogenesis of Mycoplasma bovis Mastitis (MBM) in Dairy</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab will collaborate with Dr. Pamela Adkins at the University of Missouri to explore the epidemiology and pathogenesis of <em>Staphylococcus chromogenes</em> intramammary infections. The Barlow lab will complete whole genome sequencing of <em>Staphylococcus</em> species isolates to identify potential virulence factors and antimicrobial resistance genes, and explore the phylogeny and ecology of <em>Staphylococcus </em>species and mobile genetic elements among these species. The Barlow lab will continue to explore the epidemiology of <em>Staphylococcus aureus</em> and non-aureus staphylococci in small to medium sized dairy farms.</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Member: Paolo Moroni</p><br /> <p>Contributors: Gloria Gioia, Anja Sipka, Paul Virkler, Daryl Nydam</p><br /> <p>Investigation of the Relationship between different type of bedding and control of mastitis.</p><br /> <p><strong>OBJECTIVE 3</strong></p><br /> <p><strong><span style="text-decoration: underline;">&nbsp;</span></strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Members: John Barlow, Feng-Qi Zhao</p><br /> <p>The Barlow lab will continue to explore the relationship between housing and bedding management practices and mastitis risk using culture-based and culture independent (metagenomic) methods. Barlow lab will continue to explore the potential role of endogenous inhibitor bacteria and bacteriocins in the epidemiology of Staphylococcus mastitis. We will use in vitro co-culture systems and metagenomic methods to describe bacterial factors influencing colonization and infection of mammary glands.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Member: Oudessa Kerro Dego</p><br /> <p>Contributors: Benti Deresa Gelalcha, Aga Edema Gelgie, Jessica Vidlund, Barbara Gillespie, Ximin Zeng, Jun Lin</p><br /> <p>Optimization of vaccination regimen for staphylocococcal, streptococcal and <em>E. coli</em> vaccines:</p><br /> <p>Evaluation of Efficacy of Staphylococcal and Streptococcal Surface Proteins Vaccine to Control Mastitis in Dairy Cows</p><br /> <p>Evaluation of Immunogenicity of Enterobactin Conjugate Vaccine for the Control of <em>E. coli</em> Mastitis in Dairy Cows</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Members: S. Godden, E. Royster, L. Caixeta, N. Noyes, and B. Crooker</p><br /> <p>Contributors: Sam Rowe, Pat Gordon, Daryl Nydam</p><br /> <p>Godden, S. and E. Royster. Pilot Study: Refining a Culture-Guided Selective Dry Cow Therapy Program (SDCT) to Reduce Antibiotic Use on Dairy Farms. Data analysis and reporting will occur in 2022.</p><br /> <p><strong><span style="text-decoration: underline;">Louisiana State University</span></strong></p><br /> <p>Member: William E. Owens</p><br /> <p>Continue evaluation of botanical formulations from plants for antimicrobial activity against mastitis pathogens.</p>

Publications

<p><strong>PUBLICATION LIST:</strong>&nbsp;</p><br /> <p><strong>Peer-Reviewed Literature</strong></p><br /> <p><strong><span style="text-decoration: underline;">Ohio State University</span></strong></p><br /> <p>L.R. Larsen, P.H. Baker, K.M. Enger, L.E. Moraes, P.R.F. Adkins, J.A. Pempek, C.A. Zimmerly, S.M. Gauta, R.L. Bond and B.D. Enger. Administration of internal teat sealant in primigravid dairy heifers at different times of gestation to prevent intramammary infections at calving. J. Dairy Sci. 104: 12773-12784.</p><br /> <p>Enger, B.D. and J. R. Middleton. 2021. Letter to the Editor: Comments on &ldquo;Mammary microbial dysbiosis leads to the zoonosis of bovine mastitis: a One-Health perspective&rdquo; by Maity and Ambatipudi. FEMS Microbiol. Ecol. 97:fiab076.</p><br /> <p>Enger, K.M., N.R. Hardy, E.M. Hist, and B.D. Enger. 2021. Relationship between intramammary infection and antibody concentrations in Jersey and Holstein colostrum. J. Dairy Sci. 104:6124-6133.</p><br /> <p>Hardy, N.R., K.M. Enger, J. Hanson, M.L. Eastridge, L.E. Moraes, and B.D. Enger. 2021. Organization of mammary blood vessels as affected by mammary parenchymal region and estradiol administration in Holstein heifer calves. J. Dairy Sci. 104:6200-6211.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Kerro Dego O, Almeida R, Ivey S, Agga GE. 2021. Evaluation of Streptococcus uberis Surface Proteins as Vaccine Antigens to Control S. uberis Mastitis in Dairy Cows. MDPI Vaccines 9:868<strong>.</strong><strong>&nbsp; </strong><a href="https://doi.org/10.3390/vaccines9080868">https://doi.org/10.3390/vaccines9080868</a></p><br /> <p>Abdi RD, Gillespie BE, Ivey S, Pighetti GM, Almeida RA, Kerro Dego O. 2021. Antimicrobial Resistance of Major Bacterial Pathogens from Dairy Cows with High Somatic Cell Count and Clinical Mastitis. Animals (Basel) 11. DOI:&nbsp;<a href="https://doi.org/10.3390/ani11010131">10.3390/ani11010131</a></p><br /> <p>Balemi A, Gumi B, Amenu K, Girma S, Gebru M, Tekle M, Rius AA, D'Souza DH, Agga GE, Kerro Dego O. 2021. Prevalence of Mastitis and Antibiotic Resistance of Bacterial Isolates from CMT Positive Milk Samples Obtained from Dairy Cows, Camels, and Goats in Two Pastoral Districts in Southern Ethiopia. Animals (Basel) 11.DOI:&nbsp;<a href="https://doi.org/10.3390/ani11061530">10.3390/ani11061530</a></p><br /> <p>Gelalcha, B. D., D. B. Ensermu, G. E. Agga, M. Vancuren, B. E. Gillespie, D. H. D&rsquo;Souza, C. Okafor, and O. Kerro Dego. 2021. Prevalence of Antimicrobial Resistant and Extended Spectrum Beta-Lactamase producing <em>Escherichia coli </em>in Dairy Cattle Farms in East Tennessee, Foodborne pathogens and disease, In press.</p><br /> <p>Gelalcha, B. D., S. Brown, H. Crocker, G. E. Agga, and O. Kerro Dego. 2021. Regulation Mechanisms of Virulence Genes in Enterohemorrhagic <em>Escherichia coli</em> (EHEC): A review, Foodborne pathogens and disease, In press.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Fernandes LM, Guimaraes I, Noyes NR, Caixeta L, Machado, V. Impact of subclinical mastitis detected in the first month of lactation on somatic cell count linear scores, milk yield, fertility, and culling of dairy cows in certified organic herds. Journal of Dairy Science. February 2021. https://doi.org/10.3168/jds.2020-19153</p><br /> <p>Rowe, S. , S. Godden, E. Royster, J. Timmerman, M. Boyle. 2021. Post-calving udder health and productivity in cows approaching dry-off with intramammary infections caused by non-aureus Staphylococcus, Aerococcus, Enterococcus, Lactococcus and Streptococcus species. <em>J. Dairy Sci. 104(5) 6061-6079</em></p><br /> <p>Rowe, S. , A. Vasquez, S. Godden, D. Nydam, E. Royster, J. Timmerman, M. Boyle. 2021. Evaluation of four predictive algorithms for intramammary infection status in late lactation cows. <em>J. Dairy Sci. 104(10): 11035-11046. </em></p><br /> <p>Rowe, S. , D. Nydam, S. Godden, P. Gorden, A. Lago, A. Vasquez, E. Royster, J. Timmerman, M. Thomas, R. Lynch. 2021. Partial budget analysis of culture and algorithm guided selective dry cow therapy. <em>J. Dairy Sci. 104:5652&ndash;5664&nbsp; </em><a href="https://doi.org/10.3168/jds.2020-19366"><em>https://doi.org/10.3168/jds.2020-19366</em></a></p><br /> <p>Patel, K., S. Godden, E. Royster, B. Crooker, T. Johnson, E. Smith, S. Sreevatsan. 2021. Prevalence, antibiotic resistance, virulence and genetic diversity of <em>Staphylococcus aureus</em> isolated from bulk tank milk samples of U.S. dairy herds. BMC Genomics 22:367<a href="https://doi.org/10.1186/s12864-021-07603-4"><em>https://doi.org/10.1186/s12864-021-07603-4</em></a></p><br /> <p>Jahan, N. , S. Godden, E. Royster, T. Schoenfuss, C. Gebhart, J. Timmerman and R. Fink. 2021. Evaluation of the Matrix-Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS) System in the Detection of Mastitis Pathogens from Bovine Milk Samples. J. Microbiol Methods. 182: 106168. <a href="https://doi.org/10.1016/j.mimet.2021.106168"><em>https://doi.org/10.1016/j.mimet.2021.106168</em></a></p><br /> <p>Lippolis, J. D., E. J. Putz, T. A. Reinhardt, E. Casas, W. J. Weber and B. A. Crooker. <em><span style="text-decoration: underline;">Submitted</span></em>. Effect of Holstein genotype on immune response to an intramammary Escherichia. coli challenge. <em><span style="text-decoration: underline;">Submitted. J. Dairy Sci. JDS.2021-21166</span>. Under revision.</em></p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Missouri</span></strong></p><br /> <p>DeBuck J, Ha V, Naushad S, Nobrega DB, Luby C, Middleton JR, De Vliegher S, Barkema HW.&nbsp; 2021.&nbsp; Non-aureus staphylococci and udder health.&nbsp; Front Vet Sci.&nbsp; April 15;8:658031. Invited Review.&nbsp; <a href="https://doi.org/10.3389/fvets.2021.658031">https://doi.org/10.3389/fvets.2021.658031</a>&nbsp;&nbsp;</p><br /> <p>Enger BD, Middleton JR.&nbsp; 2021.&nbsp; <em>Letter to the Editor &ndash; </em>Reply to the article: &ldquo;Mammary microbial dysbiosis leads to the zoonosis of bovine mastitis: a One-Health perspective&rdquo; <em>FEMS Microbiology Ecology</em>.&nbsp; 2021 Jun 9: <a href="https://doi.org/10.1093/femsec/fiab076">https://doi.org/10.1093/femsec/fiab076</a></p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Miyuki Takashima, Christian Lalonde, Laura Olszanski, and Feng-Qi Zhao. (2021). Localized and systemic inflammation mediators in a murine acute mastitis model. <em>Journal of Inflammation Research</em> 14:4053&ndash;4067. doi: 10.2147/JIR.S313799</p><br /> <p>Andrews T, Jeffrey Cc, Gilker R, Neher D, Barlow JWa. 2021. Survey design and implementation quantifies winter housing and bedding types used on Vermont organic dairy farms. J. Dairy Science Apr 14;S0022-0302(21)00517-8 DOI: 10.3168/jds.2020-19832</p><br /> <p>Huebner R, Mugabi Rc, Hetesy Gd, Fox L, De Vliegher S, De Visscher A, Barlow JWa, Sensabaugh G. 2021. Characterization of genetic diversity and population structure within Staphylococcus chromogenes by multilocus sequence typing. PLoS One. Mar 15; 6(3): e0243688. <a href="https://www.biorxiv.org/content/10.1101/2020.11.30.403683v1">https://www.biorxiv.org/content/10.1101/2020.11.30.403683v1</a> DOI: 10.1371/journal.pone.0243688</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Hoekstra J, Zomer AL, Rutten VPMG, Benedictus L, Stegeman A, Spaninks MP, Bennedsgaard TW, Biggs A, De Vliegher S, Mateo DH, Huber-Schlenstedt R, Katholm J, Kov&aacute;cs P, Kr&ouml;mker V, Lequeux G, Moroni P, Pinho L, Smulski S, Supr&eacute; K, Swinkels JM, Holmes MA, Lam TJGM, Koop G. Genomic analysis of European bovine Staphylococcus aureus from clinical versus subclinical mastitis. Sci Rep. 2020 Oct 23;10(1):18172. doi: 10.1038/s41598-020-75179-2. PMID: 33097797; PMCID: PMC7584570.</p><br /> <p>Kumar R, Register K, Christopher-Hennings J, Moroni P, Gioia G, Garcia- Fernandez N, Nelson J, Jelinski MD, Lysnyansky I, Bayles D, Alt D, Scaria J. Population Genomic Analysis of Mycoplasma bovis Elucidates Geographical Variations and Genes associated with Host-Types. Microorganisms. 2020 Oct 10;8(10):1561. doi: 10.3390/microorganisms8101561. PMID: 33050495; PMCID: PMC7650767.</p><br /> <p>Gioia G, Addis MF, Santisteban C, Gross B, Nydam DV, Sipka AS, Virkler PD, Watters RD, Wieland M, Zurakowski MJ, Moroni P. Mycoplasma species isolated from bovine milk collected from US dairy herds between 2016 and 2019. J Dairy Sci. 2021 Apr;104(4):4813-4821. doi: 10.3168/jds.2020-19171. Epub 2021 Feb 19. PMID: 33612245.</p><br /> <p>Wieland M, Geary CM, Gioia G, Case KL, Moroni P, Sipka A. Vacuum Dynamics as an Alternative Method for Detection of Bimodal Milk Ejection in Dairy Cows. Animals (Basel). 2021 Jun 23;11(7):1860. doi: 10.3390/ani11071860. PMID: 34201426; PMCID: PMC8300128.</p><br /> <p>Gioia G, Addis MF, Goodman LB, Mitchell PK, Thompson B, Goodrich E, Moroni P. Draft Genome Sequence of Acholeplasma laidlawii Isolated from the Conjunctiva of a Heifer with Infectious Bovine Keratoconjunctivitis. Microbiol Resour Announc. 2021 Jan 28;10(4):e01345-20. doi: 10.1128/MRA.01345-20. PMID: 33509992; PMCID: PMC7844077.</p><br /> <p>Bulut E, Stout A, Wemette M, Llanos-Soto S, Schell RC, Greiner Safi A, Shapiro MA, Moroni P, Ivanek R. How does public perception of antibiotic use on dairy farms contribute to self-reported purchasing of organic? J Food Sci. 2021 May;86(5):2045-2060. doi: 10.1111/1750-3841.15720. Epub 2021 May 6. PMID: 33955540; PMCID: PMC8251749.</p><br /> <p>Monistero V, Barberio A, Cremonesi P, Castiglioni B, Morandi S, Lassen DCK, Astrup LB, Locatelli C, Piccinini R, Addis MF, Bronzo V, Moroni P. Genotyping and Antimicrobial Susceptibility Profiling of &lt;i&gt;Streptococcus uberis&lt;/i&gt; Isolated from a Clinical Bovine Mastitis Outbreak in a Dairy Farm. Antibiotics (Basel). 2021 May 28;10(6):644. doi: 10.3390/antibiotics10060644. PMID: 34071296; PMCID: PMC8229259.</p><br /> <p>Mann S, Curone G, Chandler TL, Moroni P, Cha J, Bhawal R, Zhang S. Heat treatment of bovine colostrum: I. Effects on bacterial and somatic cell counts, immunoglobulin, insulin, and IGF-I concentrations, as well as the colostrum proteome. J Dairy Sci. 2020 Oct;103(10):9368-9383. doi: 10.3168/jds.2020-18618. Epub 2020 Aug 20. PMID: 32828510.</p><br /> <p>Llanos-Soto SG, Vezeau N, Wemette M, Bulut E, Greiner Safi A, Moroni P, Shapiro MA, Ivanek R. Survey of perceptions and attitudes of an international group of veterinarians regarding antibiotic use and resistance on dairy cattle farms. Prev Vet Med. 2021 Mar;188:105253. doi: 10.1016/j.prevetmed.2020.105253. Epub 2021 Jan 15. PMID: 33524793.</p><br /> <p>Penati M, Sala G, Biscarini F, Boccardo A, Bronzo V, Castiglioni B, Cremonesi P, Moroni P, Pravettoni D, Addis MF. Feeding Pre-weaned Calves With Waste Milk Containing Antibiotic Residues Is Related to a Higher Incidence of Diarrhea and Alterations in the Fecal Microbiota. Front Vet Sci. 2021 Jul 8;8:650150. doi: 10.3389/fvets.2021.650150. PMID: 34307516; PMCID: PMC8298036.</p><br /> <p>Capoferri R, Cremonesi P, Castiglioni B, Pisoni G, Roccabianca P, Riva F, Filipe J, Del Corvo M, Stella A, Williams JL, Rupp R, Moroni P. Comparison of the response of mammary gland tissue from two divergent lines of goat with high and low milk somatic cell scores to an experimental Staphylococcus aureus</p><br /> <p>infection. Vet Immunol Immunopathol. 2021 Apr;234:110208. doi: 10.1016/j.vetimm.2021.110208. Epub 2021 Feb 19. PMID: 33640660.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Utah State University</span></strong></p><br /> <p>Britten JE, Rood KA, Wilson DJ: Comparison of bovine mammary involution and intramammary infections following intramammary treatment with casein hydrolysate and other conventional treatments at dry-off. Animals 11:8:1-12, 2021</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Abstracts</strong></p><br /> <p><strong><span style="text-decoration: underline;">Oregon State University</span></strong></p><br /> <p>Hunter Ford, Massimo Bionaz, Serkan Ates, Joe Klopfenstein, Jorge Vanegas, Gracia Puerto Hernandez, Callan Stowell, Paige Tafoya, Michele McCann, Mustafa Bozkus, 64 Feeding Chicory-plantain Silage and Se-yeast on Lactating Ewe Subjected to Intramammary Infection: Effect on Performance and Milk Quality, Journal of Animal Science, Volume 99, Issue Supplement_3, November 2021, Pages 34&ndash;35, <a href="https://doi.org/10.1093/jas/skab235.060">https://doi.org/10.1093/jas/skab235.060</a></p><br /> <p>Hunter Ford, Massimo Bionaz, Serkan Ates, Joe Klopfenstein, Jorge Vanegas, Sebastiano Busato, Daniella Hasan, Shelby Park, Carolyn Pearce, Tyler Quinones, Kate Ness, Mariangel Aleman Torres, 62 Feeding Chicory-plantain Silage and Se-yeast to Lactating Ewes Subjected to Intramammary Infection: Effect on the Immune System, Journal of Animal Science, Volume 99, Issue Supplement_3, November 2021, Pages 32&ndash;33, https://doi.org/10.1093/jas/skab235.056</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Ohio State University</span></strong></p><br /> <p>Enger, B.D., P.H. Baker, M.A. McGuire, R.M. Akers, S.K. Jacobi, and K.M. Enger. 2021. Effect of intramammary infections on heifer mammary gland growth and development. Conference of Research Workers in Animal Disease Proceedings:244</p><br /> <p>Larsen, L.R., P.H. Baker, K.M. Enger, L.E. Moraes, and B.D. Enger. 2021. Intramammary infection prevalence and secretion characteristics of primigravid dairy heifers. J. Dairy Sci. 104 (Suppl. 1):256.</p><br /> <p>Larsen, L.R., P.H. Baker, K.M. Enger, L.E. Moraes, and B.D. Enger. 2021. Earlier administration of an internal teat sealant in primigravid dairy heifers to prevent intramammary infections at calving. J. Dairy Sci. 104 (Suppl. 1):15.</p><br /> <p>Rodriguez, M., W.P. Weiss, B.D. Enger, K. Lee, and C. Lee. 2021. Effects of different levels of vitamin A supply on production and blood parameters of transition cows. J. Dairy Sci. 104 (Suppl. 1):310.</p><br /> <p>Larsen, L.R., P.H. Baker, K.M. Enger, L.E. Moraes, and B.D. Enger. 2021. Earlier administration of internal teat sealant in dairy heifers to prevent intramammary infections at calving and lessen potential impacts on performance. Proc. 2021 Tri-State Nutrition Conference:169.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Rowe, S.M., M. Dziuba, B. Boyum, S. Godden, E. Royster, L. Caixeta. 2021. Negatively Controlled Trial Investigating the Effect of Dry Cow Therapy on Clinical Mastitis and Culling. ADSA /Annu Mtg. (Virtual) July 11-14, 2021.</p><br /> <p>Rowe, S.M., S.M. Godden, D.V. Nydam, A. Vasquez. 2021. Selective dry-cow therapy protocols to enhance udder health from dry-off to calving. ADSA Annu Mtg. July 11-14, 2021.</p><br /> <p>Brink, A.A., W.J. Weber, J.D. Lippolis, J.B. Cole, S.M. Godden, P.A. Crooker. Effect of Holstein milk yield genotype on ex-vivo innate immune response to lipopolysaccharide (LPS) and lipoteichoic acid (LTA) during the periparturient period. ADSA Annu Mtg (virtual). July 11-14, 2021.</p><br /> <p>Rowe, S.M., A. Vasquez, S.M. Godden, D.V. Nydam, E. Royster, J. Timmerman, M. Boyle. Evaluation of four predictivealgorithms for intramammmary infection status in late lactation cows. Proc. 60<sup>th</sup> Annu. Conf National Mastitis Council. Jan. 25-28, 2021 (virtual).&nbsp;</p><br /> <p>Rowe, S.M., D.V. Nydam, S.M. Godden, P. Gorden, A. Lago., E. Royster, A. Vasquez, M. Ghomas, R. Lynch. Partial budget analysis of selective dry cow strategies. Proc. 60<sup>th</sup> Annu. Conf National Mastitis Council. Jan. 25-28, 2021 (virtual).</p><br /> <p>Vasquez, D.V. Nydam, S.M. Rowe, S.M. Godden, P. Gorden, A. Lago, E. Royster, M. Thomas, R. Lynch. Impact of dry cow antimicrobial use on the milk microbiome: Caracterization of dry-off and fresh-cow samples from 6 U.S. dairy farms. Proc. 60<sup>th</sup> Annu. Conf National Mastitis Council. Jan. 25-28, 2021 (virtual).</p><br /> <p>Godden, S., F. Pe&ntilde;a Mosca, E. Royster, B. Crooker, P. Raynor, and K. Janni. 2021. Management of Recycled Manure Solids Bedding on Midwest Dairy Farms. Part 1 - Relationship between Processing Method, Udder Health and Production, and Bedding Bacteria Counts. Proc. 60<sup>th</sup> Annu. Conf National Mastitis Council. Jan. 25-28, 2021 (virtual).</p><br /> <p>Pe&ntilde;a Mosca, F., S. Godden, E. Royster, B. Crooker, P. Raynor, and K. Janni. 2021. Management of Recycled Manure Solids Bedding on Midwest Dairy Farms. Part 2 - Relationship between Processing Method, Bedding Characteristics and Bedding Bacterial Count. Proc 60<sup>th</sup> Annu. Conf National Mastitis Council. Jan. 25-28, 2021 (virtual).</p><br /> <p>Brink, A. A., W. J. Weber, J. D. Lippolis, J. B. Cole, S. M. Godden and B. A. Crooker. 2021. Effect of Holstein milk yield genotype on ex-vivo innate immune response to lipopolysaccharide (LPS) and lipoteichoic acid (LTA) during the periparturient period. ADSA Annual Meeting. J. Dairy Sci. July 11-14. (virtual).</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Missouri</span></strong></p><br /> <p>Kurban D, Roy JP, Kabera F, Frechette A, Um MM, Albaaj A, Rowe S, Godden S, Adkins PRF, Middleton JR, Gauthier ML, Keefe G, DeVries TJ, Kelton DF, Moroni P, Veiga dos Santos M, Barkema HW, Dufour S.&nbsp; 2020.&nbsp; Diagnosing intramammary infection: a scoping review and meta-analysis on frequency and udder health relevance of microorganism species retrieved in bovine milk samples.&nbsp; Journ&eacute;e de recherch&eacute;, Faculty of Veterinary Medicine, University of Montreal, Quebec, Canada.&nbsp; 15-18 March 2021.</p><br /> <p>Resendiz-Biermaier K5, Ringen DR, Middleton JR.&nbsp; 2021.&nbsp; Epidemiology of staphylococcal mastitis in primiparous heifers on a pasture-based dairy.&nbsp; Proceedings of the 43rd Annual CVM Research Day. May 7, 2021.&nbsp; Abstract #48.Adkins PRF. Heat stress and the microbiome of dairy cows<strong>. </strong>American College of Veterinary Internal Medicine Annual Forum, June, 2021</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Gelalcha, B. D., B. Gillespie and O. Kerro Dego. Extended-Spectrum Beta-Lactamases Producing <em>E. coli </em>in Dairy Cattle Farms, MRW 2021&nbsp; annual meeting Nov. 3 &ndash; 4, Virtual.</p><br /> <p>Gelgie, A. E., R. Almeida and O. Kerro Dego. 2021. Identification of Virulence Factors and Pathogenesis of Mycoplasma bovis Mastitis in Dairy Cattle, A Proposal, MRW 2021&nbsp; Annual meeting Nov. 3 &ndash; 4, Virtual.</p><br /> <p>Vidlund, J. B. Gillespie and o. Kerro Dego. 2021. Efficacy of Staphylococcal surface associated protein Vaccines against Mastitis. MRW 2021&nbsp; Annual meeting Nov. 3 &ndash; 4, Virtual.</p><br /> <p>Kerro Dego, O., X. Zeng, J. Vidlund, B. Gillespie, L. Cao, H. Wang and J. Lin. 2021. Evaluation of Immunogenicity of Enterobactin Conjugate Vaccine for Controlling <em>E. coli</em> Mastitis in Dairy Cows, MRW 2021&nbsp; Annual meeting Nov. 3 &ndash; 4, Virtual.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Addis F., Pisanu S., Penati M., Monistero V., Gazzola A., Castiglioni B., Cremonesi P., Moroni P., Pagnozzi D., and R. Piccinini. Comparative Secretome Analysis of <em>Staphylococcus aureus </em>Strains Belonging to Sequence Types with Different Within-herd Mastitis Prevalence Proceeding of the 60<sup>nd</sup> National Mastitis Council Annual Meeting pp.90-91. January 26-28, 2021 Virtual.</p><br /> <p>Monistero V., Cremonesi P., Morandi S., Barberio A., Castiglioni B., Locatelli C., Piccinini R., and P. Moroni.&nbsp; Molecular Characterization of <em>Streptococcus uberis </em>Strains Isolated from a Clinical Bovine Mastitis Outbreak in an Italian Dairy Farm. Proceeding of the 60<sup>nd</sup> National Mastitis Council Annual Meeting pp.122-123. January 26-28, 2021 Virtual.</p><br /> <p>Penati M., Sala G., Biscarini F., Boccardo A., Bronzo V., Castiglioni B., Cremonesi P., Pravettoni D., Locatelli C., Moroni P., and M. F. Addis. Waste Milk Containing Antimicrobials Affects Gut Health and Microbiota Diversity in Calves. Proceeding of the 60<sup>nd</sup> National Mastitis Council Annual Meeting pp.124-125. January 26-28, 2021 Virtual.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Idaho</span></strong></p><br /> <p>Tsai C. Y., H. H. Hung, T. Weber, Q. Huo, and P. Rezamand. 2021. Relationship between serum</p><br /> <p>metabolites and milk fatty acid with periparturient diseases in Pacific Northwest dairy farms. <em>J.</em></p><br /> <p><em>Dairy Sci. Vol. 104, Suppl. 1): 37 (abstr.).</em></p><br /> <p>&nbsp;</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Conference Proceedings </strong></p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Fernandes, L., Celestino, M. L., Menta, P. R., Silva, T. H., Paiva, D., Ribeiro, T. L., Caixeta, L. S., Noyes, N. R., Machado, V. S. (2021). Risk factors associated with intramammary infections in primiparous dairy cows in organic herds. (J. Dairy Sci, vol. 104, Suppl. 1., pp. 215--216).</p><br /> <p>Mosca FP, Dean CJ, Heins BJ, Machado VS, Pinedo PJ, Noyes NR, Caixeta LR. Description of Staphylococcus sp. intramammary infections in early lactation heifers on organic dairy farms. American Dairy Science Association. Virtual, July 2021. Abstract 399.</p><br /> <p>Mosca FP, Dean CJ, Ray T, Doster E, Fernandes L, Antunes A, Sharpe K, Feijoo V, Baumann C, Wehri T, Heins BJ, Pinedo PJ, Machado VS, Caixeta LR, Noyes NR. Associations Between Intramammary Infections Caused by Non-aureus Staphylococci, Streptococcus spp. and Streptococcus-like organisms and Staphylococcus aureus in Early Lactation in Organic Dairy Cows. National Mastitis Council Annual Meeting. Virtual, January 2021.</p><br /> <ol start="2020"><br /> <li>Fernandes, I. Guimaraes, N. Noyes, L. Caixeta, V.S. Machado. Impact of subclinical mastitis detected in the first month of lactation on performance of organic dairy cows. Proceedings of the 101st Conference of Research Workers in Animal Diseases, Virtual, December 2020. Abstract 410.</li><br /> </ol><br /> <p>Dean, C, Felipe Pena Mosca, Tui Ray, Bradley Heins, Pablo Pinedo, Vinicius Machado, Luciano Caixeta, Noelle Noyes. What Is the Microbiome, and Why Is It Important for Organic Livestock Production? 2020 ASA-CSSA-SSSA International Annual Meeting. Virtual, November 2020.</p><br /> <p>&nbsp;</p><br /> <p>Pena Mosca, F., Dean, C.J., Caixeta, L.S., Ray, T., Heins, B., Machado, V.S., Pinedo, P.J., Noyes, N.R. &ldquo;Description of the intramammary infections dynamics in early lactation heifer on organic dairy farms.&rdquo; 54th Annual Conference of the American Association of Bovine Practitioners. Salt Lake City, UT, 2021</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Tennessee</span></strong></p><br /> <p>Zeng, X., J. Vidlund, B. Gillespie, L. Cao, H. Wang,&nbsp; J. Lin&nbsp; and O. Kerro Dego. 2021. Immunogenicity of the Novel Enterobactin Conjugate Vaccine in Dairy Cows, Conference of Research Workers On Animal Diseases (CRWAD), December 3 &ndash; 7, Chicago, IL. USA.</p><br /> <p>Gelalcha, B. D.,&nbsp; D. B. Ensermu, S. Brown, B. E. Gillespie, G. E. Agga, D. H. D&rsquo;Souza, C. Okafor, and O. Kerro Dego. Detection of Multidrug-Resistant Extended-Spectrum &beta;-lactamase Producing <em>E. coli</em> from Bulk tank Milk Obtained from Dairy Cattle Farms, Conference of Research Workers On Animal Diseases (CRWAD), December 3 &ndash; 7, Chicago, IL. USA.</p><br /> <p>Vidlund, J., B. E. Gillespie, G. E. Agga and O. Kerro Dego. Efficacy of Novel Staphylococcal Surface Protein Vaccines Against Mastitis in Dairy Cattle, Conference of Research Workers On Animal Diseases (CRWAD), December 3 &ndash; 7, Chicago, IL. USA.</p><br /> <p><span style="text-decoration: underline;">&nbsp;</span></p><br /> <p><strong><span style="text-decoration: underline;">University of Vermont</span></strong></p><br /> <p>Lalonde C, Kraft J, Choudhary RK, Bourne DE, Shangraw EM, McFadden TB, Zhao F-Q (2021) Intramammary lipopolysaccharide infusion alters the fatty acid profile in blood triacylglycerides and phospholipids of lactating dairy cows. ADSA Annul Meeting 2021.</p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">Cornell University</span></strong></p><br /> <p>Virtual&nbsp;MEX&nbsp;ConneXion Event &ldquo;Selective Treatment of Mastitis Science into practice from a US perspective&nbsp; 22<sup>th</sup> June 2021 Ghent University. Virtual</p><br /> <h3>Workshop &rdquo;Prevention and control of environmental mastitis&rdquo;. April 21<sup>th </sup>2021. <sup>&nbsp;</sup>Meeting con liberi professionisti scandinavi. Virtual.</h3><br /> <h3>1<sup>st</sup> Master Class in Udder Health. Onfarm Academy &ldquo;Manejo de ambiente para controle da mastite&rdquo; February 2<sup>th</sup> 2021. Virtual.</h3><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>Poster Presentations</strong></p><br /> <p>&nbsp;</p><br /> <p><strong><span style="text-decoration: underline;">University of Minnesota</span></strong></p><br /> <p>Using Rapid Culture Systems to Guide Selective Treatment of Clinical Mastitis and at Dry-off. E. Royster, S. Godden, E. Royster and S. Rowe. 3 hrs Short Course #1 at 60<sup>th</sup> Annu Meeting of the National Mastitis Council. Jan 20, 2021. (virtual)</p><br /> <p>Decision support tools for selective dry cow therapy. Rowe, S.M., A. Vasquez, S.M. Godden, D.V. Nydam. Proc. 60<sup>th</sup> Annu. Conf National Mastitis Council. Jan. 25-28, 2021 (virtual).&nbsp; &nbsp;</p><br /> <p>3 Bs of Udder Health: Bedding, Bacteria Load and Better Management. S. Godden, F. Pena Mosca, E. Royster, S. Rowe, J. Timmeran, B. Crooker. National Mastitis Council Webinar. Nov. 19, 2020 (virtual).</p><br /> <p>3 B&rsquo;s of Udder Health: Bedding, Bacterial Load and Better Management. IFRAMIX CCPA Czech Producer Webinar. 1.5 hr. Mar. 18, 2021 (virtual)</p><br /> <p>Is selective dry cow therapy an opportunity for your herd?&nbsp; Carver County Dairy Expo. Feb. 15, 2021 (virtual)</p><br /> <p>&nbsp;</p><br /> <p>Relationship between recycled manure solids processing methods, bedding bacteria counts and udder health. Carver County Dairy Expo. Feb. 15, 2021. (virtual)</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p>

Impact Statements

Back to top
Log Out ?

Are you sure you want to log out?

Press No if you want to continue work. Press Yes to logout current user.

Report a Bug
Report a Bug

Describe your bug clearly, including the steps you used to create it.