MASTITIS VACCINE ANTIBODIES (MASTIVAC I) PASIVELY PROTECT MICE AGAINST STAPHYLOCOCCUS AUREUS ISOLATES FROM USA, GERMANY AND ITALY

Similar documents
Development of a Staphylococcus aureus vaccine against mastitis in dairy cows. II. Field trial

Long influence of Escherichia coli intramammary infections on milk quality

Vaccination as a Tool to Reduce Mastitis and Improve Milk Quality in Dairy Goats. F.M. Kautz, S. C. Nickerson, and L. O. Ely.

ENVIRACOR J-5 aids in the control of clinical signs associated with Escherichia coli (E. coli) mastitis

Use of a novel adjuvant to enhance the antibody response to vaccination against Staphylococcus aureus mastitis in dairy heifers.

Use of a staphylococcal vaccine to reduce prevalence of mastitis and lower somatic cell counts in a registered Saanen dairy goat herd

Subclinical mastitis in small ruminants: prevalence, comparative aspects and prevention

Options for Handling Mastitis during Lactation in Modern Dairy Farms

Udder infection and milk somatic cell count, NAGase activity and milk composition fat, protein and lactose in Israeli-Assaf and Awassi sheep

Evaluation of a computerized antimicrobial susceptibility system with bacteria isolated from animals

April Boll Iowa State University. Leo L. Timms Iowa State University. Recommended Citation

Mastitis cows and immunization

Finnzymes Oy. PathoProof Mastitis PCR Assay. Real time PCR based mastitis testing in milk monitoring programs

Mastitis: Background, Management and Control

Mastitis MANAGING SOMATIC CELLS COUNTS IN. Somatic Cell Count Are Affected by. Somatic Cells are NOT Affected by:

Milk quality & mastitis - troubleshooting, control program

Diagnosis of Heartworm (Dirofilaria immitis) Infection in Dogs and Cats by Using Western Blot Technique

Mastitis Management and SCC Control in Once a Day Herds. Don Crowley- Teagasc

Changes in milk constituents of Awassi ewes after experimental infection with Staphylococcus aureus

Tel: Fax:

Milk Quality Management Protocol: Fresh Cows

Presented at Central Veterinary Conference, Kansas City, MO, August 2013; Copyright 2013, P.L Ruegg, all rights reserved

Int.J.Curr.Microbiol.App.Sci (2018) 7(8):

Strep. ag.-infected Dairy Cows

Update on Staphylococcus aureus Mastitis. John R. Middleton College of Veterinary Medicine, University of Missouri, Columbia

The organism Infection process Tissue reaction SCC response Prevention Treatment

Gina M Pighetti & Raul Almeida. University of Tennessee

Bovine Mastitis Products for Microbiological Analysis

SUMMARY OF PRODUCT CHARACTERISTICS. Lincomycin (as Lincomycin hydrochloride) Neomycin (as Neomycin sulphate) Excipients Disodium edetate

MILK COMPOSITIONAL CHANGES DURING MASTITIS

Effect of omitting post-milking teat disinfection on the mastitis infection rate of dairy cows over a full lactation

Prevalence of sub clinical mastitis in small holder dairy farms in Selale, North Shewa Zone, Central Ethiopia

POSSIBLE PROTECTIVE EFFECT OF AN AUTOVACCINE AGAINST HIGH VIRULENCE STAPHYLOCOCCUS AUREUS IN A RABBIT SKIN INFECTION MODEL

Mastitis Prevention and Cure Rates in Heifers Treated with Spectramast Dry Cow Therapy and/or Orbeseal Dry Cow Teat Sealant

Controlling Contagious Mastitis

Using SCC to Evaluate Subclinical Mastitis Cows

TEAT DIP- POST DIP- PRE DIP- STRIPING

Walter M. Guterbock, DVM, MS Veterinary Medicine Teaching and Research Center University of California, Davis

1. INTRODUCTION. Sayed, M.L. 1, Shell, W.S. 1, Hanan, A. Ahmed 1, Hanan, M. Ibrahim 1, Nasr, E.A. 2 and Ali, A.M. 2

Using DHIA and bacteriology to investigate herd milk quality problems.

Mastitis in ewes: towards development of a prevention and treatment plan

Evaluation of Different Antigens in Western Blotting Technique for the Diagnosis of Sheep Haemonchosis

Quad Plate User s Manual

Mastitis vaccines in dairy cows: Recent developments and recommendations of application

Quality Milk on Pasture Based Dairy Farms. Scott E. Poock, DVM University of Missouri Clinical Assistant Professor DABVP Beef and Dairy Cattle

Methicillin-Resistant Staphylococcus aureus

Minna Koivula & Esa Mäntysaari, MTT Agrifood Research Finland, Animal Production Research, Jokioinen, Finland

STUDY ON CLINICAL MASTITIS IN BUFFALOES CAUSED STAPHYLOCOCCAL SPECIES

New York State Cattle Health Assurance Program Fact Sheet Udder Health Herd Goals

Decision tree analysis of treatment strategies for mild and moderate cases of clinical mastitis occurring in early lactation

Interpretation of Bulk Tank Milk Results

On-farm milk culture training workshop. Christina Petersson-Wolfe Department of Dairy Science Virginia Tech

Dairy/Milk Testing Report Detecting Elevated Levels of Bacteria in Milk-On-Site Direct- From-The-Cow Within Minutes as Indicator of Mastitis

Milk Quality Evaluation Tools for Dairy Farmers

Field Efficacy of J-VAC Vaccines in the Prevention of Clinical Coliform Mastitis in Dairy Cattle

CERTIFICATION. Certificate No. The AOAC Research Institute hereby certifies that the performance of the test kit known as: Compact Dry X SA

MATERIALS AND METHODS

On- farm milk culture training workshop

Lab Exercise: Antibiotics- Evaluation using Kirby Bauer method.

HardyCHROM MRSA, Contact Plate

Detection of Mastitis

MASTITIS CASE MANAGEMENT

Caused by microorganisms (usually bacteria) that invade the udder, multiply, and produce toxins that are harmful to the mammary gland

Veterinaria.com.pt 2009; Vol. 1 Nº 1: e13 (publicação inicial em Julho de 2008) Disponível em

MASTITIS. Therefore, mastitis is an inflammation of the mammary gland.

Development of a Breeding Value for Mastitis Based on SCS-Results

Exploring simvastatin, an antihyperlipidemic drug, as a potential topical antibacterial agent

Mastitis control in bred heifers: Use of dry cow therapy and teat sealant for curing existing intramammary infections and preventing new ones

Estimate of milk and curd yield loss of sheep and goats with intrammamary infection and its relation to somatic cell count

Effect of subclinical intramammary infection on somatic cell counts, NAGase activity and gross composition of goats milk

How to Decrease the Use of Antibiotics in Udder Health Management

The mastitis situation in Canada where do you stand?

LOOKING FOR PROFITS IN MILK QUALITY

THIS ARTICLE IS SPONSORED BY THE MINNESOTA DAIRY HEALTH CONFERENCE.

Clostridial Vaccination Efficacy on Stimulating and Maintaining an Immune Response in Beef Cows and Calves 1,2

Management Practices and Intramammary Infections: New Ideas for an Old Problem

Association between teat skin colonization and intramammary infections with Staphylococcus aureus and Streptococcus agalactiae

Isolation and identification of major causing bacteria from bovinemastitis R. Lakshmi 1 and K.K. Jayavardhanan 2

S. P. Oliver, R. A. Almeida, B. E. Gillespie, S. J. Ivey, H. Moorehead, P. Lunn, H. H. Dowlen, D. L. Johnson, and K. C. Lamar

Subclinical mastitis assessed by deviations in milk yield and electrical resistance

Somatic Cell Count as an Indicator of Subclinical Mastitis. Genetic Parameters and Correlations with Clinical Mastitis

CLINICAL MASTITIS PERCEPTIONS OF KANSAS DAIRY PRODUCERS. J.R. Roberson 1

Proper Dry-Off Procedures to Prevent New Infections and Cure Existing Cases of Mastitis. Stephen C. Nickerson University of Georgia

Selective toxicity. Antimicrobial Drugs. Alexander Fleming 10/17/2016

Validation of the PathoProof TM Mastitis PCR Assay for Bacterial Identification from Milk Recording Samples

Prevalence and Drug Resistance Patterns of Staphylococcus Aureus in Lactating Dairy Cow s Milk in Wolayta Sodo, Ethiopia

Guideline on the conduct of efficacy studies for intramammary products for use in cattle

Herd Navigator and mastitis management

Check that milk is suitable to go in the vat

PCR detection of Leptospira in. stray cat and

Mastitis in non-bovine dairy species, companion animals and breastfeeding mothers. Chris Knight

Northern NY Agricultural Development Program 2016 Project Report

SUMMARY OF PRODUCT CHARACTERISTICS. NUFLOR 300 mg/ml solution for injection for cattle and sheep

SCC. Staphylococcus aureus Streptococcus uberis Staphylococcus epidermidis. Antibiotic. coli

STATISTICAL REPORT. Preliminary Analysis of the Second Collaborative Study of the Hard Surface Carrier Test

International Journal of Advances in Pharmacy and Biotechnology Vol.3, Issue-2, 2017, 1-7 Research Article Open Access.

Diurnal variation in microfilaremia in cats experimentally infected with larvae of

Mastitis Reminders and Resources LAURA SIEGLE EXTENSION AGENT VIRGINIA COOPERATIVE EXTENSION AMELIA COUNTY

BIOLACTAM. Product Description. An innovative in vitro diagnostic for the rapid quantitative determination of ß-lactamase activity

Last 2-3 months of lactation

Transcription:

MASTITIS VACCINE ANTIBODIES (MASTIVAC I) PASIVELY PROTECT MICE AGAINST STAPHYLOCOCCUS AUREUS ISOLATES FROM USA, GERMANY AND ITALY Leitner G.,'* Krifucks O.', Weisblit L.', Arnon E. 1, Yagana K. 2 and Trainin Z. 3 'National Mastitis Reference Center, Kimron Veterinary Institute, Ministry of Agriculture and Rural Development, RO. Box 12, Bet Dagan 50250, Israel 3 2 MASTIVAC, P.O. Box 15578, Rishon Le Zion 75054, Israel Israel Dairy Board, P.O. Box 15578, Rishon Le Zion 75054, Israel. * Corresponding author: Gabriel Leitner Phone: 972-3-9681745 Fax: 972-3-9681692 E-mail: leitnerg@moag.gov.il (G. Leitner) ABSTRACT Previously we described the development of a new Staphylococcus aureus vaccine denoted MASTIVAC I. The present study tested the ability of antibodies elicited by MASTIVAC I to protect mice against S. aureus isolates derived from infected bovine udders in Germany, the USA and Italy,. Variations were found among isolates but not between countries iusing the hemolytic slide-latex agglutination response on Baird Parker agar. No differences were found in the susceptibility tests and virulence determinations in mice. Cluster analysis performed by the unweighted pair-group method, using arithmetic averages (UPGMA), and calculation of the immunoblot relatedness with serum from MASTIVAC I- immune mice, revealed three groups with similarity coefficients of protection of 82-84%. The first 2 groups contained all S. aureus isolates without segregation regarding the origin of the strain. Staphylococcus chromogenes 36173/1 was separate in group 2 and group 3 included three Staphylococcus haemoliticus isolates with a coefficient of 76% from groups 1 and 2. Overall mortality rates of the groups vaccinated with MASTIVAC I and sham controls were 7.3 and 28.6%, respectively, and the morbidity rates were 21.4% and 46.4%, respectively. Between-group differences (vaccinated and control) were significant for mortality (P < 0.001), morbidity (P < 0.05), and mortality + morbidity (P < 0.0001) with variations among isolates. S. aureus strains from the USA, Germany and Italy did not differ from the Israeli isolates in their antibody recognition and protection patterns. Moreover, the isolates possess crucial immunogenic determinants, which reacted with the antibodies elicited by the MASTIVAC I vaccine and probably will protect cows against these bacteria Key Words: Staphylococcus aureus, mastitis, vaccine, dairy cow INTRODUCTION Staphylococcus aureus is still one of the major pathogens causing mastitis worldwide whose treatment necessitates the extensive use of antibiotics in dairy herds. The increasing public concern over food safety, expressed as the desire to minimize antibiotic residues in milk on the one hand, and the need to reduce somatic cell counts (SCC) on the other hand, vindicates our effort to combat S. aureus mastitis by vaccination. Nevertheless, previous attempts to develop a vaccine against S. aureus mastitis have failed to provide a substantial solution [3,5,13,16,19]. MASTIVAC I is a newly introduced vaccine (Patent no. IL122829, PTC/IL 98/00627, AU 746285, USA09/582692) designed to protect cows from S. aureus mastitis, which has been used commercially in Israel since 2004. The vaccine is composed of three field strains of S. aureus, which exhibit a broad spectrum of antigenic and immunogenic properties [9]. In controlled experiments, the vaccine was found effective in protecting cows challenged with a virulent field strain of S. aureus [10]. A largescale field trial, lasting over 2 consecutive years and involved a total of452 (vaccinated and control) Israeli-Holstein heifers in 7 dairy farms, resulted in 40% lower incidence of SCC in vaccinated cows in the first and second lactations than in unvaccinated control cows. Moreover, the vaccinated cows yielded 0.5 kg/ d more milk than the unvaccinated ones [11]. For registering MASTIVAC I, a highly infected herd of267 multiparous Israeli- Holstein cows, of which 22.1 % were chronically infected with S. aureus mastitis, was vaccinated and monitored for a year under research regulations [12]. The field study again demonstrated the ability of MASTIVAC I to significantly reduce S. aureus new infection in vaccinated animals, all of which were uninfected at time of vaccination, and to cure vaccinated compared with placebo-treated S. aureus infected-animals. Although antibodies generated by MASTIVAC I recognized a wide range of Israeli 77 WEBSITE: www.isrvma.org VOLUME 63 (3) 2008

field strains of S. aureus, it was crucial to examine the capacity of the vaccine to recognize S. aureus isolates found in other dairy industries. This study therefore aimed to test the ability of antibodies elicited by MASTIVAC I to recognize (identify) S. aureus strains isolated from infected cattle in Germany, USA and Italy, and determine the protection provided by MASTIVAC I against challenge with these S. aureus strains in a mouse model. MATERIALS AND METHODS 2.1. Mice Female Swiss line of 6-8 weeks old mice were maintained in the animal facility of the Kimron Veterinary Institute. Three mice were housed in each cage under standard conditions of light (12/12 h light/dark) and temperature (22 C) and were fed standard laboratory chow and water ad lib. At the end of the experiments, the mice were euthanized with C0 2. Animal ethics approval was granted for all animal experiments by the Israeli ethics committee (Kimron Veterinary Institute Animal Ethics Committee). 2.2. Phenotypes of field isolates and susceptibility test. 2.2.1. Bacterial Identification Twenty-eight staphylococci field isolates, comprising 10 from Germany and 9 from the USA, were provided by Boehringer (Ingelheim Vetmdica GmbH, Germany), and 8 isolates were provided by FATRO (Pharmaceutical Veterinary Industry, Ozzano, Italy). An aliquot of 10 ul from each sample was spread over blood agar plates (Hy-labs, Park Tamar, Rehovot, Israel) containing 5% washed sheep erythrocytes and incubated at 37 C for 24 h. The following tests were performed: Coagulase (tube test) (Anilab, Tal-Shachar, Israel) [6] slide latex agglutination test (BACTI Staph, Remel, Santa Fe Drive, Lenexa, KS) and selective media: Baird Parker (Difco, Laboratories, Detroit, Ml). In the following step, isolates were identified by the ID- 32-API STAPH test (BioMerieux S.A., France). Isolates were considered as being S. aureus if the isolate identification was > 98% with T > 65%. Phage typing was performed according to Blair and Williams [4], with phages issued by the International Reference Laboratory, Colindale, UK, as modified by Samra and Gadba[17]. 2.2.2. Susceptibility Test Antimicrobial susceptibility test (ATB) was performed in accordance with NCCLS guidelines [15] using commercial test disks (Beckton Dickinson, Le Pont de Claix, France) and the MIC test. Commercially available disks (Dispens-O-Disc, Susceptibility Test System, Difco) or BBL Sensi-Disc Antimicrobial Susceptibility Test Discs (Becton Dickinson, MD) were used as recommended, and the plates were incubated at 30 C for methicillin (5 ug/disk) and 37 C for other antibiotics, penicillin G (10 units/disk), erythromycin (15 ug/disk), cephalothin (30 ug/disk), neomycin (30 ug/disk), trimethoprimsulfamethoxazol (SXT)( 1.25-23.75 ug/disk). Susceptibility or resistance was interpreted according to the manufacturers' recommendations. 2.2.3. Electrophoresis and immunohlotting Onc-dimcnsional sodium dodccyl sulfate-polyacrylamide gel electrophoresis was performed [7]. The bacterial cells were disrupted with glass beads in a homogenizer (Braun Melsungen AG, Germany) for 10-15 min. The glass beads and the remaining bacteria were removed by centrifugation at 1,000 x g for 15 min, and the supernatant was filtered through 0.2-um filters. Protein concentrations of the disrupted bacteria were determined with the Bio-Rad protein assay. The samples (antigens) were adjusted to a final protein concentration of 1 mg/ml, and 33 ul of each sample was mixed with 25 ul of 4 * NuPAGE sample buffer, 32 ul of ultrapure water and 10 ul of reducing agent. The sample mixtures were heated to 70 C for 10 min and loaded onto the gels at 30 ul per lane. The gel was 10% (Bis-Tris Gel with w/mops) (NOVEX, San Diego, CA), stained with colloidal blue. Molecular-weight markers for these gels were: See Blue Pre-Stained, 191-14 kda for the 10% gel. For the immunoblot assay, a 0.2-um nitrocellulose membrane was blocked with 3% casein and incubated with 1:100 dilutions of serum from mice that had been immunized with the MASTIVAC I (maximum dilution that gives a positive result in ELISA is 1:10.000). The blot was developed with goat anti-mouse IgG (H+L) alkaline peroxidase conjugate (1:1000), with a substrate of DAB (3,3- diaminobenzidine tetrahydrochloride) (ICN Pharmaceuticals, Inc., Costa Mesa, CA). Molecular-weight markers for these gels were: NOVEX Marker 12 Standard Bands (2.5-200 kda). Cluster analysis of the immunoblot was performed by the unweighted pair-group method, using arithmetic averages (UPGMA), and the calculation of their relatedness was based on the Dice coefficient [ 1 ]. 2.3. Virulence vaccination and protection Virulence and resistance to challenge after MASTIVAC I vaccination were determined in mice, with the German and US isolates in two sets of experiments. 2.3.1. Virulence in a mouse model [9]. Each of nineteen groups of nine mice was divided into three subgroups of three mice. The mice in each subgroup were inoculated intramuscularly in the left hind limb with one of the S. aureus strains with one of 3 doses of live bacteria (1 x 10 6 to 1 x 10 9 CFU). The mice were examined individually; the injected limb was visually inspected daily for visible erythrema and gangrene. Macroscopic inspections during 20 days of observation revealed either: (0) normal appearance; (1) morbidity - erythrema gangrene; or (2) mortality. Virulence was calculated as the CFU of bacteria that resulted in death (mortality) or erythrematous gangrene (morbidity) in 50% of the inoculated mice. Isolates whose virulence was not determined by these series of tests were retested as mentioned above, but the dosage of bacteria was increased to > 1 x 10 9 CFU/mouse or decreased to 1 x 10 5 CFU/mouse. VOLUME 63 (3) 2008 WEBSITE: www.isrvma.org 78

2.3.2. Vaccination and protection Mice were vaccinated subcutancously with MASTIVAC I at 0.2 mg/mousc in the right limb. Groups of 4 or 5 mice were vaccinated with MASTIVAC I and after 25 days they were challenged with one of the S. aureus isolates. Each vaccinated group was matched by a control group comprising the same number of mice. The level of challenge used for each of the isolates was that which resulted in virulence in 50% of the inoculated mice in the first series of tests. 2.4. Statistical Analysis Data were analyzed with the JMP statistical software [18]. The analyzed parameters were: percentage mortality, percentage morbidity and percentage (mortality + morbidity). The group effect (vaccinated vs. control) was determined by applying a one-way ANOVA design in blocks (bacteria) with the linear model: Y = u + a, + Bj+ c in which u is the grand mean; a, is the effect of the /th group; Bj is the variance between bacteria (random effect); and e,, represent the random error. RESULTS 3.1. Phenotyping the field isolates Of the 28 staphylococci, one (USA 18) was found to be contaminated and was not identified. Isolate 36173/1 was identified as S. chromogenes, and isolates 937/6, VMTRC- AB019 and 931/1 were identified as S. haemoliticus by the coagulase and ID32 STAPH tests (Table I). All the remaining 23 identified S. aureus isolates were coagulase positive, 6/23 (26%) were not slide-latex agglutinated and 14/23 (61%) did not show a positive response on Baird Parker. Of the hemolysis reactions, 2 (8.7%) were non-hemolytic and 6 (26.1%) were a-hemolytic, all of which were from Germany; 6, of which 5 were from the USA, (26.1%) were P-hemolytic; 6, mostly from Italy, (26.1%) were (a + P) hemolytic; and 3 (13%) were (P + y) hemolytic (Tab. I). All six German isolates,, were insensitive to any of the phages used, while the others showed a variety of patterns. All S. aureus isolates were sensitive to methicillin and cephalothin. All were sensitive to neomycin except for two isolates from Italy, All the Italian isolates were resistant to penicillin and all the German and American isolates were penicillin-sensitive. About half of the isolates from the three sources showed intermediate sensitivity to erythromycin and SXT (data not shown). Antigenic patterns of the 27 staphylococcal isolates (from USA, Germany, Italy) and one Israeli S. aureus isolate (Z03984), i.e., one of the three S. aureus strains included in the vaccine, were analyzed by electrophoresis and were further analyzed by immunoblotting with 1:100 dilutions of serum from mice immunized with MASTIVAC I. Fig. 1 shows the 1- dimension SDS-PAGE results and Fig. 2 shows the immunoblot results obtained with the same isolate. Cluster analysis by the unweighted pair-group method, using arithmetic averages (UPGMA), and calculation of the immunoblot relatcdncss (US and German isolates) revealed 3 groups (Fig. 3) with similarity coefficients of 82-84%. The first 2 groups contained S. aureus isolates from both countries, as well as isolates Z03984 (Israel) and ATCC 29740, with no segregations to the source. In group 2, but separately, was found 5. chromogenes 36173/1. Group 3 included the three S. haemoliticus isolates: 931/1, 937/6 and VMTRC-AB019 with a similarity coefficient of 76% with respect to both groups 1 and 2. The Italian isolates had similar immunoblotting patterns to those of groups 1 and 2. 3.2. Virulence in mice The virulence of the S. aureus isolates varied among the German and US isolates (Tab. II). Virulence of ATCC 29740 was 5 x 10 7 (ii and that of most of the German isolates was similar, whereas about half of the US isolates were about half as virulent. The S. chromogenes and the three S. haemoliticus isolates did not cause clinical symptoms even when the highest dose (5 x 10'' CFU) of bacteria was injected into the mice (data not shown). Protection results of mice vaccinated with MASTIVAC I vaccine and then challenged with one of the 11 German and US S. aureus isolates are presented in Tab. III. The vaccine protected the mice as compared with the relevant control, against all isolates except 36220/3. Overall mortality rates of the vaccinated and control mice were 7.3 and 28.6%, respectively, and morbidity rates were 21.4 and 46.4%, respectively. The significance levels P[F] of the ANOVA effects (group and bacteria), the R : and the between-bacteria percentage of variance from the overall variance, for percentage mortality, percentage morbidity and percentage (mortality + morbidity) arc presented in Tab. IV. The (vaccinated and control) differences between groups were significant for mortality (P < 0.001), morbidity (P < 0.05), and (mortality + morbidity) (P< 0.0001) with high variation among the isolates. DISCUSSION Under laboratory and field conditions, MASTIVAC I was found to have a wide range of recognition of Israeli S. aureus strains (9-11]. This was attributed to the unique features of the aureus strains in the vaccine as well as its realistic production method. In order to manufacture and use this vaccine to combat S. aureus mastitis in other countries, it was necessary to verify its ability to recognize S. aureus isolates from different target locations. Bacterial phenotypic and genotypic characterization is important for management and treatment. However, this provides only a rough estimate of the ability of antibodies to protect animals against infection. MASTIVAC I, which includes bacterial fragments of two S. aureus strains and the secretion of a third strain, is designed to recognize and protect against a wide range of S. aureus strains and also to partially CNS strains. IgGi and IgGi antibody,isotypes, are the principal immunoglobulins of the mammary gland immune system, and are responsible for promoting polymorphonuclear phagocytosis [2,14]. The level of IgG in milk is low compared with that in the blood. However, IgGi is transferred to the mammary gland 79 WEBSITE: www.isrvma.org VOLUME 63 (3) 2008

continuously, regardless of infection with S. aureus [8], which renders the vaccinated cows better able to confront a new infection. Therefore, high affinity IgG 1 antibodies that recognize a wide range of 5. aureus strains and varieties are a crucial factor in protecting cows against S. aureus mastitis. In fact, the cluster analysis of the immunoblot relatedness revealed that the antibody response of MASTIVAC I recognized all 5. aureus with a similarity coefficient of 82-84%. This assay also revealed that MASTIVAC I antibodies were able to recognize coagulate negative staphylococci (CNS), though to a lesser extent. This is important because, although the vaccine was directed to protect cows mainly from 5. aureus, CNS comprise the group of Staphylococci that represent the most commonly found bacteria in dairy mastitis, mainly in its subclinical form. The most important finding of this study is the significantly enhanced protection of mice vaccinated with MASTIVAC I vaccine. MASTIVAC I provided full protection from mortality to all but one (36220/3) of the vaccinated mice that were challenged with all of the US and German 5. aureus strains tested. Moreover, it also provided significant protection against erythrematous gangrene, as indicated by comparison with the unvaccinated control mice. Similar results were obtained when mice were challenged with various Israeli S. aureus isolates [9]. In conclusion, S. aureus strains derived from USA, Germany and Italy did not differ in their antibody recognition and protection properties from the Israeli S. aureus isolates. Moreover, the isolates possess, with high probability, the crucial antigenic determinants that will react with MASTIVAC I vaccinal antibodies, and probably will protect cows against these bacteria. ACKNOWLEDGMENTS The authors thank Dr. Francesca Berti of Fatro S.p.A. and Dr. Fausto Toni of Kriton S.r.l. for supplying the S. aureus field isolates from Italy. REFERENCES 1. Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic- Acids Res. 25:3389-3402, 1997. 2. Avery VM, Gordon LD. Antibacterial properties of breast milk: requirements for surface phagocytosis and chemiluminescence. Europ. J. Clinical Microb. and Inf. Dis. 10:1034-1039, 1991. 3. Calzolari A, Giraudo EJ, Rampone H, Odierno L, Giraudo TA, Frigerio C, Bettera S, Raspanti C, Hernandez J, Wehbe M, Mattea M, Forrari M, Larriestra A, Nagel R. Field trials of vaccine against bovine mastitis. 2. Evaluation in two commercial dairy herds. J. Dairy Sci. 80:854-858, 1997. 4. Blair JE, Williams REO. Phage type of Staphylococci. Bulletin of the World Health Organization 24:771-784, 1961. 5. Giraudo JE, Calzolari A, Rampone H, Rampone A, Giraudo TA, Bogni C, Larriestra A, Nagel R. Field trials of vaccine against bovine mastitis. 1. Evaluation in heifers../. Dairy Sci. 80:45-853, 1997. 6. Koneman W, Allen SD, Bowell VR, Janda WM, Sommers HM, Winn WC. Color and Textbook of Diagnostic Microbiology. In: Color Atlas and Textbook of Diagnostic Microbiology, 3 rj Edn, Vol. 3, pp. 311-364. 1988. J.B Lippincott Co., Philadelphia. 7. Laemmli UK.. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227:680 685, 1970. 8. Leitner G, Yadlin B, Glickman A, Chaffer M, Saran A. Systemic and local immune response of cows to intramammary infection with Staphylococcus aureus. Res. Vet. Sci. 69:181-184, 2000. 9. Leitner G, Lubashevsky E, Trainin Z. Staphylococcus aureus vaccine against mastitis in dairy cows, composition and evaluation in mouse model. Vet. Immunol. Immunopath. 93:159-167, 2003a. 10. Leitner G, Lubashevsky E, Glickman A, Winkler M, Saran A, Trainin Z. Development of a Staphylococcus aureus vaccine against mastitis in dairy cows. I. Challenge trials. Vet. Immunol. Immunopath. 93:31-38, 2003b. 11. Leitner G, Yadlin N, Lubashevsky E, Ezra E, Glickman A, Chaffer M, Winkler M, Saran A, Trainin Z. Development of a Staphylococcus aureus vaccine against mastitis in dairy cows. II. Field trials. Vet. Immunol. Immunopath. 93:153 158, 2003c. 12. Leitner G, Krifucks O, Glickman A, Vaadia Y, Friedman S, Ezra E, Saran A, Trainin Z. MASTIVAC I: Staphylococcus aureus vaccine, prevention of new infection and therapeutic effect on cows chronically infected with S. aureus under field conditions. Israel J. Vet. Med. 59:68-72, 2004. 13. Middlcton JR, Rinchart LC, Taylor NV, Luby DC, Steevens JB. Efficacy of different Lysigin formulations in the prevention of Staphylococcus aureus intramammary infection in dairy heifers. J. Dairy Res. 73:9-10, 2006. 14. Miller RH, Guidry J A, Paape JM, Dulin MA, Fulton AL. Relationship between immunoglobulin concentrations in milk and phagocytosis by bovine neutrophils. Am. J. Vet. Res. 49:42^15, 1988. 15. NCCLS, Performance standards for antimicrobial disk and dilution susceptibility tests of bacteria isolates from animals. Approved Standard M31-A, Wayne, PA. 1999. 16. Nordhoug ML, Nesse LL, Norcross LN, Gudding R. A field trial with an experimental vaccine against Staphylococcus aureus mastitis in cattle. I. Clinical parameters. J.Dairy Sci. 77:1267-1275,1994. 17. Samra Z, Gadba R. Antibiotic susceptibility and phage typing of Methicillin resistant Staphylococcus aureus clinical isolation from blood cultures of 962 patients in 15 Israeli hospitals. Europ. J. Epidem. 9:559-562, 1993. 18. SAS Institute, JMP Statistics and Graphics Guide, Version 5. SAS Inst., Cary, NC, USA. 2002. 19. Watson DL, McColl LM, Davies HI. Field trial of staphylococcal mastitis vaccine in dairy heifers: clinical, subclinical and microbiological assessments. Australian Vet. J. 74:447-450, 1996. VOLUME 63 (3) 2008 WEBSITE: www.isrvma.org 80

Table I. Phage type, coagulase, Baird Parker, agglutination and hemolysis patterns of 28 isolates from German, US and Italian mastitic cows. Isolate Origin Phage Type Coagulase B.P. Agglutination Hemolysis Indentification (API) 1 M165 Ahlemer Inst 0[0] Pos Neg Pos a S. aureus 2 VMTRC-AB01 USA 29 [80 90 95] Pos Neg Neg P S. aureus 3 VMTRC-AB03 USA 80 3/A 3/C 42E 92 94 95 96 D117HK2[29] Pos Neg Pos a + p S. aureus 4 36220/3 Ahlemer Inst 0[0] Pos Neg Pos a S. aureus 5 Lufa Munster 0[0] Pos Neg Pos a S. aureus 6 VMTRC-AB06 USA 29 80 71 42E 75 90j92[52 95] Pos Pos Neg P S. aureus 7 VMTRC-AB02 USA 79 80 3/A(3/C 90 92 95 Pos Neg Neg P+y S. aureus 8 36173/1 Ahlemer Inst 0[0] Neg Neg Neg Neg S. chromogenes 9 VMTRC-AB010 USA 52 80 42E 75 90 92 Pos Neg Neg P S. aureus 10 Ml 674 Ahlemer Inst 29 79 80 71 90 92 95 D117HK2[52] Pos Neg Neg P+y S. aureus 11 ATCC 29740 USA 42E Pos Neg Pos P S. aureus 12 VMTRC-AB08 USA 6 29 52 80 3/A 3/C 55(71 42E 54 75 88 89 90 92 95 [47 85] Pos Pos Neg P+y S. aureus 13 VMTRC-AB01I USA 42E 75 [6 47 54] Pos Pos Pos P S. aureus 14 36173/31 Ahlemer Inst 0[0] Pos Neg Pos a S. aureus 15 Lufa Munster 92 Pos Neg Pos a S. aureus 16 937/6 Ahlemer Inst 0[0] Neg Neg Neg Neg S. haemoliticus 17 Lufa Munster 0[0] Pos Neg Pos a S. aureus 19 VMTRC-AB019 USA 0[0] Neg Neg Neg Neg S. haemoliticus 20 931/1 Ahlemer Inst 0[0] Neg Neg Neg Neg S. haemoliticus 21 1645 Italy Pos Neg Pos a + P S. aureus 22 954 Italy Pos Pos Pos P S. aureus 23 845 Italy Pos Pos Pos a + p S. aureus 24 598 Italy Pos Pos Pos a + p S. aureus 25 1449 Italy Pos Pos Pos Neg S. aureus 26 622 Italy Pos Pos Pos a + p S. aureus 27 1425 Italy Pos Neg Pos Neg S. aureus 28 1640 Italy Pos Pos Pos a + P S. aureus 81 WEBSITE: www.isrvma.org VOLUME 63 (3) 2008

Table II. Virulence (CFU of bacteria result in mortality + morbidity of 50% of mice) derived from inoculation with several concentrations of various strains of S. aureus isolates from USA and Germany. Bacterium CFU/mouse Mortality a Morbidity a Virulence 1 x io 7 0/3 M165 1 x io 8 3/3 5 x 10 7 1 x io 9 1/3 2/3 1 x io 6 0/3 VMTRC-AB01 1 x io 7 1/3 5 x 10 7 1 x io 8 3/3 3 x 10 6 0/3 VMTRC-AB03 3 x 10 7 0/3 5 x 10 8 3 x 10 8 2/3 1 x io 6 0/3 36220/3 1 x io 7 2/3 5 x 10 7 1 x io 8 2/3 1/3 3 x io 5 0/3 Lufa Munster (5) 3 x 10 6 0/3 5 x 10 7 3 x io 7 2/3 1 x io 7 0/3 VMTRC-AB06 1 x io 8 0/3 >1 x io 9 1 x io 9 0/3 5 x 10 5 0/3 VMTRC-AB02 5 x 10 6 1/3 1 x io 7 5 x 10 7 2/3 1/3 1 x io 7 0/3 VMTRC-AB010 1 x io 8 0/3 >1 x io 9 1 x io 9 1/3 2 x io 6 0/3 Ml 674 2 x io 7 1/3 1 x io 7 2 x io 8 2/3 5 xlo 5 0/3 ATCC 29740 5 x 10 6 0/3 5 x 10 7 5 x 10 7 1/3 1/3 1 x io 7 0/3 VMTRC-AB08 1 x io 8 0/3 1 x io 9 1 x io 9 1/3 1/3 5 x 10 6 0/3 VMTRC-AB011 5 x 10 7 1/3 1 x io 8 5 x 10 8 3/3 5 x 10 6 0/3 36173/31 5 x io 7 1/3 1 x io 8 5 x 10 8 1/3 1/3 5 x 10 5 0/3 Lufa Munster (15) 5 x 10* 1/3 5 x 10 7 5 x IO 7 1/3 1/3 5x 10* 0/3 Lufa Munster (17) 5 x 10 6 1/3 1 x io 7 5 x 10 7 273 1/3 * Mice mortality during 20 days post inoculation. VOLUME 63 (3) 2008 WEBSITE: www.isrvma.org 82

Table III. Mortality and morbidity rates of MASTIVAC I vaccinated and control mice inoculated with S. aureus isolates from the USA and Germany. Bacterium CFU/mouse Vaccinated (%) Control (%) # mortality morbidity # mortality morbidity M165 1 x io 8 5 0 60 5 40 60 VMTRC-AB01 3 x 10 7 4 0 0 4 25 25 36220/3 5 x 10 7 5 80 0 5 100 0 Lufa Munster (5) 1 x io 8 4 0 100 4 50 50 VMTRC-AB06 1 x 10" 4 0 0 4 25 25 VMTRC-AB02 5 x 10 7 4 0 0 4 25 25 Ml 674 5 x 10 8 4 0 0 4 0 100 VMTRC-AB08 1 x 10 9 4 0 0 4 25 25 VMTRC-AB011 1 x io 8 4 0 0 4 0 50 36173/31 5 x 10 7 4 0 50 4 25 75 Lufa Munster (17) 5 x 10 7 4 0 25 4 0 75 Mean 7.3 21.4 28.6 46.4 Mice mortality during 20 days post inoculation. Table IV. The significance level P[F] of the ANOVA effects (group and bacteria), the R square (R 2 ) and the percentage of variance between bacteria from the overall variance, for percentage mortality, percentage morbidity and percentage (mortality + morbidity) of inoculated mice with US or German S. aureus isolates post vaccinated with MASTIVAC I. Df P [ F ] % mortality % morbidity % (mortality + morbidity) Group 1 0.0014 0.0493 <.0001 Bacteria 10 0.0006 0.1503.0047 n 22 22 22 R 2 0.921 0.712 0.909 Variance between bacteria 81.43% 32.63% 71.24% 83 WEBSITE: www.isrvma.org VOLUME 63 (3) 2008

Figure 1. One-dimension SDS-polyacrylamide gel electrophoresis (10%) of disrupted Staphylococcus strains. 30 ug protein of disrupted Staphylococcus strains supernatant was applied to each lane. Lane 1: MW markers; Lane 2: S. haemoliticus 931/1; Lane 3: S. haemoliticus VMTR-AB19; Lane 4: S.aureus Z03984 (Positive control, Israel); Lane 5: S. aureus Lufa Munster (17); Lane 6: S. haemoliticus 937/6; Lane 7: S. aureus Lufa Munster (15); Lane 8: S. aureus 36173/31; Lane 9: S. aureus VMTR-AB11; Lane 10: S. aureus VMTR-AB08; Lane 11:5. aureus ATCC29740. VOLUME 63 (3) 2008 WEBSITE: www.isrvma.org 84

Figure 2. Immunoblot patterns of disrupted Staphylococcus strains with 1:100 dilutions of serum from mice immunized with MASTIVAC I. Lane 1: MW markers; Lane 2: S. haemoliticus 931/1; Lane 3: S. haemoliticus VMTR-AB19; Lane 4: S. aureus Z03984 (Positive control, Israel); Lane 5: S. aureus Lufa Munster (17); Lane 6: S. haemoliticus 937/6; Lane 7: S. aureus Lufa Munster (15); Lane 8: S. aureus 36173/31; Lane 9: S. aureus VMTR-AB11; Lane 10:5. aureus VMTR-AB08; Lane 11: S. aureus ATCC29740. 191 O - U o 03 ea ««co a t y TT 3 a a u o ri r a = T i S u ON.!*5 L_ LTJ DC t" H 1 MW $J S j S J H 1 < < > > h 64 51 < 19 1 4»» 85 WEBSITE: www.isrvma.org VOLUME 63 (3) 2008

Figure 3. Cluster analysis performed by the unweighted pair-group method using arithmetic averages (UPGMA), and calculation of the immunoblot relatedness (US and German isolates).?.,.1 S s! * f f t t ' c ATCC 29740 VMTRC-A B011 LUFA MUNSTER 17 36173/31 LUFA MUNSTER 18 VMTRC-A BOS Z03984 VMTRC-A B03 M-1674 VMTRC-A 802 LUFA MUNSTER i VMTRC-A B01 VMTRC-A BOS M16S VMTRC-A B10 3622013 3617311 931/1 93716 VMTRC-A B019 VOLUME 63 (3) 2008 WEBSITE: www.isrvma.org 86