Advances in Animal and Veterinary Sciences

Download PDF Download ePUB
AAVS_9_8_1211-1222

 

 

Research Article

 

In-vitro Antimicrobial Activity and In-vivo Prophylactic Influence of Tulathromycin against Respiratory Diseases in Dairy Heifers Exposed to Cold Stress

 

Essam S. Soliman1*, Ahmed E. Mahmoud2

1Department of Animal Hygiene, Zoonosis, and Animal Behavior, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt; 2Department of Animal Medicine, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.

 

Abstract | Calf health was prioritized as one of the most important issues facing the dairy industry. The in-vitro antimicrobial and the in-vivo prophylactic actions of tulathromycin in dairy heifers exposed to harsh macroclimate concerning hematological, biochemical, and bacteriological profiles were evaluated. Tulathromycin 15, 20, and 25* mg were tested against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida (1.8 × 108 CFU/ml), and E. coli O6 (3.5 × 109 CFU/ml) at 0.25, 0.5, 1.0, and 2.0 h using minimal inhibitory concentration procedures. Experimental 30 dairy heifers with an average body weight of 30 kg were divided into two groups, the first was injected with 2.5 ml tulathromycin 25* mg, and the second was kept as control. A total number of 690 samples including 240 in-vitro samples (bacterial- tulathromycin mixes) and 450 in-vivo samples (150 whole blood samples, 150 sera, and 150 nasal swabs) were collected.The in-vitro study revealed that tulathromycin 25* mg achieved highly significant (P<0.01) efficiency with 100% killing against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6 at 1.0, 1.0, 0.5, 0.5, and 0.5 h, respectively. In-vivo measured hematological and biochemical parameters revealed no significant differences between the injected and control heifers with significant (P≤0.05) declines in alanine aminotransferase and triglycerides, as well as, significant (P≤0.05) increases in total cholesterol at the 4th-week post-injection. The bacteriological assessments revealed highly significant (P<0.01) declines of total bacterial, Enterobacteriaceae, Streptococcus, Haemophilus, and Pasteurella counts in injected dairy heifers compared to the control. Temperature and humidity revealed non-significant weak correlations with hematological and biochemical parameters. The study concluded an efficient in-vitro antimicrobial, as well as, protective and preventive in-vivo activities of tulathromycin without any modifications in the hematological and biochemical parameters of dairy heifers exposed to cold macroclimatic conditions.

 

Keywords | Antimicrobial, Cold Stress, Dairy Heifers, Macroclimate, Tulathromycin, Preventive.

 

Received | March 31, 2021; Accepted | May 06, 2021; Published | July 01, 2021

*Correspondence | Essam S Soliman, Department of Animal Hygiene, Zoonosis, and Animal Behavior, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt; Email: soliman.essam@vet.suez.edu.eg

Citation | Soliman ES, Mahmoud AE (2021). In-vitro antimicrobial activity and in-vivo prophylactic influence of tulathromycin against respiratory diseases in dairy heifers exposed to cold stress. Adv. Anim. Vet. Sci. 9(8): 1211-1222.

DOI | http://dx.doi.org/10.17582/journal.aavs/2021/9.8.1211.1222

ISSN (Online) | 2307-8316; ISSN (Print) | 2309-3331

Copyright © 2021 Soliman and Mahmoud. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

 

INTRODUCTION

 

The growth, performance, and survival of newly born dairy heifers depend on their ability to withstand and endure the impact of the harsh surrounding macroclimatic conditions concerning wet and cold weather during winter and early spring (Van De Stroet et al., 2016). These harsh macroclimatic circumstances might present negative impacts on heifer’s welfare, growth, and survivability (Chester-Jones et al., 2017), as well as, contribute to hypothermia in heifers. The modern biosecurity programs aim to reduce the influence of cold stress on heifers through efficient hygienic practices and strict preventive programs (Pineda et al., 2016). The preventive measures taken included a proper housing system with a suitable and sanitary parlor, good bedding with high insulation degrees, a proper heating system, sufficient food supply, proper disinfection procedures, rodent control, fly proof, efficient management strategies, proper handling of waste and manure, and hygienic carcasses disposal (Ghasemi et al., 2017).

 

Good management practice encourages heifers to resist the cold harsh macroclimatic conditions through several actions including shivering and increase the basal metabolic rates to increase the rate of thermogenesis and reduce the rate of thermolysis (Cannon and Nedergaard 2011). Once a dairy heifer is overwhelmed by the macroclimatic cold stress, she will be more susceptible to disease concerning the respiratory system (Borderas et al., 2009; Drackley, 2008; Lago et al., 2006). Bovine respiratory diseases (BRDs) considered the most common diseases affecting back-grounding and feedlot cattle as they caused higher losses as a result of poor performance and deaths (Hulbert and Moisá, 2016; Butler et al., 2006).

 

Tulathromycin is a macrolide antibiotic that is relatively safe and highly effective in the field of prophylaxis and control of bovine diseases caused by Mannheimia haemolytica, Pasteurella multocida, Histophilus somni, and Mycoplasma bovis in cattle, as well as, against infectious diseases in high-risk calves (Murray et al., 2016). Tulathromycin has been used for treating bovine footrot caused by Fusobacterium necrophorum and Porphyromonas levii (Papich 2016). Tulathromycin once administered and absorbed, can concentrate in the cytoplasm of the white blood cells and enhance its effectiveness against intracellular micro-organisms (Nowakowski et al., 2004). Tulathromycin 25* mg/ml (Recommended by the manufacturer) at a single dose of 2.5 ml is rapidly absorbed, widely distributed, and achieved higher concentrations in the lung for long periods (Nutsch et al., 2005; Skogerboe et al., 2005; Rooney et al., 2005).

 

The current study investigated the in-vitro antimicrobial activity of different concentrations of tulathromycin (15, 20, and 25* mg/ml) against Streptococcus pneumonie (1.8 × 108 CFU/ml), Streptococcus pyogenes (1.8 × 108 CFU/ml), Haemophilus influenzae (1.8 × 108 CFU/ml), Pasteurella multocida (1.8 × 108 CFU/ml), and E. coli O6 (3.5 × 109 CFU/ml), as well as, the preventive and prophylactic influences of tulathromycin 25* mg in dairy heifers exposed to adverse cold weather concerning its impact on some hematological parameters, biochemical parameters, and nasal microbiota.

 

MATERIALS AND METHODS

 

The materials, methodology, and study design were approved by the Scientific Research Ethics Committee on animal and poultry researches, Faculty of Veterinary Medicine, Suez Canal University, Egypt with approval number (2021001).

 

In-vitro evaluation of tulathromycin

Tulathromycin: Tulathromycin 25 mg (Zoetis®) injectable solution was purchased from a veterinary clinic, Ismailia Governorate, Egypt. The tulathromycin solution was subjected to serial dilutions using distilled water to produce final concentrations of 15, 20, and 25* mg (recommended by the manufacturer) into 50 ml capacity Falcon tubes and held at 4°C until testing.


Microbial cultures and propagation

Streptococcus pneumonie (Thermo ScientificTM Culti-LoopsTM Streptococcus pneumoniae ATCCTM 49619TM), Streptococcus pyogenes (Thermo ScientificTM Culti-LoopsTM Streptococcus pyogenes ATCCTM 12384TM), Haemophilus influenzae (Thermo ScientificTM Culti-LoopsTM Haemophilus influenzae ATCCTM 35540TM), Pasteurella multocida (MicrobiologiscTM Pasteurella multocida ATCCTM 12945TM), and E. coli O6 suspension (2.5 × 105 CFU/ml) were purchased from Animal Health Research Institute (AHRI), Cairo, Egypt.

 

Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, and Pasteurella multocida culti-loops were propagated as recommended by Herigstad et al. (2001) into tryptone soy broth (Thermo Scientific™ Oxoid™ Tryptone Soya Broth, CM0129, 500 g) at 37°C for 24 hours. Ten microliters were transferred aseptically from Streptococcus pneumonie and Streptococcus pyogenes tubes onto K-F Streptococcus agar (Thermo Scientific™ Oxoid™ K-F Streptococcus Agar, CM0701B, 500 g), and from Haemophilus influenza and Pasteurella multocida tubes onto Columbia blood agar with 5% sheep blood (Thermo Scientific™ Oxoid™ Columbia Blood Agar Base, CM0331, 500 g) at 37°C for 24 hours. The typical colonies were counted, picked up, and resuscitated in buffered peptone water (Thermo Scientific™ Oxoid™ Buffered Peptone Water, CM0509B, 500 g) to obtain a suspension of 1.8 × 108 CFU/ml for each microorganism.

 

E. coli O6 suspension was propagated as recommended by Soliman et al. (2018) into Mac-Conkey broth (Thermo Scientific™ Oxoid™ MacConkey Broth, CM0505, 500 g) at 42°C for 24 hours. Ten microliters were transferred aseptically onto eosine methylene blue agar (EMB, Modified Levine Eosine Methylene Blue Thermo ScientificTM OxoidTM, CM0069B, 500 g) at 37°C for 24 hours. The typical colonies were counted, picked up, and resuscitated in buffered peptone water (Thermo Scientific™ Oxoid™ Buffered Peptone Water, CM0509B, 500 g) to obtain a suspension of 3.5 × 109 CFU/ml.

 

In-vitro antimicrobial activity of tulathromycin

The in-vitro evaluation of tulathromycin antimicrobial activity was carried out using minimal inhibitory concentration (MIC) procedures according to Soliman et al. (2016). One ml from Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6 suspensions were added to four replicates 9 ml of each of the tulathromycin different concentrations (15, 20, 25* mg/ml), and mixed thoroughly using vortex (Vortex Mixer XH-D, 2800r/m, 30 W, Bowel and disk shapes). After 0.25, 0.5, 1.0, and 2.0 h contact times, one ml was transferred from each mix, added to 9 ml physiological saline resuscitation tubes held previously at 4°C, and mixed thoroughly using the vortex. The tubes were transferred for the bacteriological assessment.

 

In-vivo evaluation of tulathromycin

Study area and time: The study was carried out in a private sector dairy farm located in El-Sharkia Governorate, Egypt. The farm was located at coordinates 30°44′28″N 32°00′23″E. The experimental study was carried out through the 2nd week of January to the end of the 3rd week of February 2020.

 

The dairy farm was designed as a milking housing system that was composed of a loose housing system associated with non-shaded yards and a herringbone milking parlor. The farm was supported with isolation pens for the suspected and diseased animals in the southern part of the farm. The drainage system was based on a dirty floor system with weekly scratching and removal of the top 5 cm monthly to discourage the anaerobic conditions.

 

The claves were reared in an artificial outdoor rearing system in which the calves are separated from their dams after six hours of birth to allow them to suck colostrum. Each calf was housed in a separate pen for two weeks then removed to a collective alternative hutch system to facilitate the cleaning and scratching of the dirty floor. Calves were feed on milk substitutes twice daily in buckets at a distance from the floor to encourage the passage of the milk to the abomasum directly via the esophageal groove.

 

The floors were disinfected from time to time using slaked lime in the presence and/or absence of animals without stimulating the dustiness to minimize the development of respiratory diseases.

 

Experimental animals and treatment

Thirty (30) dairy heifers with an average body weight of 30 kg were selected at the time of birth. Heifers were divided into two groups (15 heifers each, 3 replicates of five heifers). The heifers of the first group (G1) were injected with 2.5 ml tulathromycin 25mg*/ml subcutaneous (SC) at birth and the second group (G2) was used as control. Dairy heifers were monitored for four weeks post-injection for the general health status and the development of any respiratory manifestations.

 

The macroclimatic minimum and maximum temperature and relative humidity were recorded during each sampling time using thermometers (ThermoPro® TP50 Digital LCD Thermometer Hygrometer Temperature Humidity Meter) and Thermohygrometer (Digital Thermometer Hygrometer Indoor Outdoor Temperature Meter Humidity Monitor with LCD Alarm Clock, 3M Probe Cord).

 

Sampling

A total number of 690 samples including 240 in-vitro bacterial- tulathromycin mixes (3 concentrations × 4 replicates × 5 microbial cultures × 4 contact times) and 450 in-vivo samples (150 whole blood samples, 150 sera, and 150 nasal swabs) were collected during the study. The whole blood samples, sera, and nasal swabs samples were collected at the injection time (zero time), one-week post-injection (P1), two weeks post-injection (P2), three weeks post-injection (P3), and four weeks post-injection (P4). All samples were preserved in a dry ice-box and transferred to the laboratory for analysis.

 

Whole blood samples were collected on sterile ready to use vacutainer tubes (VACUETTE® TUBE 5 ml K3E K3EDTA 13x100 lavender cap-black ring, PREMIUM), mixed thoroughly, and transferred to the laboratory for immediate hematological examination. Sera samples were collected on serum vacutainers (BD Vacutainer® Serum tubes, 10.0mL, 16 x 100mm, Plastic, Additive: Clot Activator, Silicone Coated, Red Conventional Closure, and Paper Label), centrifuged (Fisher®Thermo Scientific CL10 Centrifuge w/ F-G3 Rotor with a max RPM of 4000) at 3000 rpm for 10-15 min, clear non-hemolyzed sera were pipetted using an automatic pipette (Thermo Scientific™ Finnpipette™ Adjustable Volume Single-Channel Micro Pipettor, 100 to 1000 μL microliter Volume) into Eppendorf tubes, and stored at -20°C until biochemical examination. Nasal swabs were collected on 9 ml buffered peptone water (Thermo Scientific™ Oxoid™ Buffered Peptone Water, CM0509B, 500g), transferred to the laboratory, and preserved at 4°C until bacteriological assessment (Soliman et al., 2017).

 

Hematological and biochemical profile

The whole blood samples (150 samples, 5 samples per animal, one per each sampling time) were examined for red blood cells count (RBCs, ×106/µl), the white blood cells count (WBCs, ×103/µl), hemoglobin concentrations (Hb expressed as g/dl), platelet counts (×103/µl), mean corpuscular hemoglobin concentrations (MCHC expressed as g/dl) using Sysmex XP-300 Automated Hematology Analyzer. Sera (150 samples, 5 samples per animal, one per each sampling time) were examined for total protein (TP expressed as g/dl), alanine aminotransferase (ALT expressed as IU/L), creatinine (CREAT expressed as mg/dl), glucose (GLUCO expressed as mg/dl), triglycerides (TG expressed as mg/dl), and total cholesterol (TC expressed as mg/dl) using Chemical Analyzer Semi-auto Photometer 5010 (Germany).

 

Bacteriological examination

Nasal swabs on buffered peptone water (150 samples, 5 samples per animal, one per each sampling time) and bacterial- tulathromycin mixes (240 samples, 3 concentrations × 4 replicates × 5 microbial cultures × 4 contact times) were prepared by ten-fold serial dilutions up to 107 to cover all the chances of the microbial growth from the samples as recommended by American Public Health Association; APHA, (2017).

 

Total bacterial counts onto Standard Plate count agar (SPA, Thermo Scientific™ Oxoid™ Plate Count Agar, CM0325, 500 g), Enterobacteriaceae counts onto eosine methylene blue agar (EMB, Modified Levine Eosine Methylene Blue Thermo ScientificTM OxoidTM, CM0069B, 500 g), Streptococcus counts onto K-F Streptococcus agar (Thermo Scientific™ Oxoid™ K-F Streptococcus Agar, CM0701B, 500 g), and Haemophilus and Pasteurella counts onto Columbia blood agar with 5% sheep blood (Thermo Scientific™ Oxoid™ Columbia Blood Agar Base, CM0331, 500 g) were carried out at 37°C for 24-36 hours. The microbial counts were conducted using the drop plate technique as recommended by Kim and Lee, (2016). The plates were counted as recommended by Murray et al. (2015) using dark filed colony counter (R164109 Reichert-Jung Quebec Darkfield 3325 Colony Counter).

 

Statistical analysis

The statistical analysis was carried out using the statistical package for social sciences (SPSS version 21, IBM, SPSS Inc., USA; SPSS, 2016). The data were analyzed using multifactorial analysis of variance (two-tailed ANOVA) to investigate the in-vitro antimicrobial activity of tulathromycin different concentrations (15, 20, and 25* mg/ml) against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenza, Pasteurella multocida, and E. coli O6 concerning different exposure times (0.25, 0.5, 1.0, and 2.0 h), as well as, the prophylactic influence of tulathromycin on hematological, biochemical, and bacteriological parameters concerning different sampling times (zero-time, P1, P2, P3, and P4). The statistical model was summarized as follow:

Yijk= µ + αi + βj + (αβ)ij + Ɛijk

Where Yijk was the measurement of dependent variables; µ was overall mean; αj was the fixed effect of the tulathromycin, βj was the fixed effect of the exposure times or sampling times, (αβ)ij was the interactions, and Ɛijk was the random error. Pearson’s correlation (r) was calculated to determine the correlation coefficient between macroclimatic conditions like temperature and relative humidity with the bacterial counts, measured bacterial, and hematological parameters. The bacterial counts were transformed and expressed as logarithmic counts (Log10) using Microsoft Excel 2016. The significance was expressed as highly significant at (P<0.01), significant at (P≤0.05), and non-significant at (P>0.05).

 

RESULTS

 

In-vitro antimicrobial activity of tulathromycin

The overall means in Table-1 revealed highly significant (P<0.01) efficiency of tulathromycin 25* mg (recommended by the manufacturer) against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6 compared with the other tested concentrations (15 and 20 mg/ml). The overall means concerning the exposure times (Table-1) revealed highly significant (P<0.01) increases in the killing percentages with the increased exposure times in all tested concentrations against all microorganisms.

 

Tulathromycin 15 mg/ml achieved highly significant (P<0.01, Table-1) efficiency with 34.3, 36.0, 28.0, 27.4, and 72.8% killing against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6, respectively at 2.0 h. Tulathromycin 20 mg/ml in Table-1 achieved highly significant (P<0.01) efficiency with 62.4, 64.2, 53.3, 48.0, and 100.0% killing against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6, respectively at 2.0 h. Meanwhile, tulathromycin 25* mg/ml achieved highly significant (P<0.01, Table-1) efficiency with a 100% killing percentage against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6 at 1.0, 1.0, 0.5, 0.5, and 0.5 h, respectively.

Clinical examinations

The dairy heifers injected with tulathromycin revealed neither abnormalities nor development of any clinical manifestations during the study period. On the other hand, 40% of the control animals under study exhibited pneumonia

 

Table 1: In-vitro antimicrobial activity (killing % ±SE) of tulathromycin concentrations against different microorganisms via minimal inhibitory concentration test.

 

Conc.

Contact

times

Strep pneumonie Strep pyogenes Haemoph influenzae Past multocida

E. coli O6

Overall means concerning tulathromycin concentrations
15 mg

26.3c±0.046

24.2c±0.062

16.7c±0.084

18.7c±0.066

47.4c±0.075

20 mg

43.8b±0.059

50.2b±0.039

39.8b±0.078

36.3b±0.020

73.9b±0.057

25 mg*

82.2a±0.079

88.3a±0.038

96.3a±0.018

96.3a±0.005

88.3a±0.077

P-value 0.000 0.001 0.000 0.000 0.000
Overall means concerning exposure times
0.25 h

34.2d±0.086

37.6d±0.024

41.2d±0.036

39.1d±0.061

36.8d±0.001

0.5 h

45.1c±0.026

49.9c±0.064

49.6c±0.071

51.0c±0.054

69.1c±0.002

1.0 h

58.2b±0.002

62.3b±0.024

52.6b±0.038

53.2b±0.092

82.8b±0.094

2.0 h

65.5a±0.052

66.7a±0.025

60.4a±0.054

58.5a±0.092

90.9a±0.053

P-value 0.000 0.000 0.002 0.001 0.00
Tulathromycin concentrations by exposure times interactions
15 mg 0.25 h

12.7d±0.011

14.0d±0.037

10.1d±0.011

12.8d±0.066

12.9d±0.012

  0.5 h

27.8c±0.033

17.2c±0.027

12.3c±0.017

15.5c±0.048

43.9c±0.060

  1.0 h

30.2b±0.032

29.5b±0.020

16.3b±0.038

19.1b±0.039

60.0b±0.079

  2.0 h

34.3a±0.071

36.0a±0.016

28.0a±0.020

27.4a±0.018

72.8a±0.067

20 mg 0.25 h

30.2d±0.032

29.5d±0.020

28.0d±0.020

19.1d±0.039

44.0d±0.030

  0.5 h

38.1c±0.012

48.6c±0.045

36.5c±0.045

37.4c±0.058

63.3c±0.020

  1.0 h

44.3b±0.078

57.6b±0.039

41.5b±0.027

40.4b±0.004

88.4b±0.052

  2.0 h

62.4a±0.021

64.2a±0.033

53.3a±0.053

48.0a±0.023

100.0a±0.000

25 mg* 0.25 h

59.5c±0.054

69.3c±0.092

85.4b±0.021

85.4b±0.021

53.4b±0.028

  0.5 h

69.3b±0.092

83.8b±0.042

100.0a±0.000

100.0a±0.000

100.0a±0.000

  1.0 h

100.0a±0.000

100.0a±0.000

100.0a±0.000

100.0a±0.000

100.0a±0.000

  2.0 h

100.0a±0.000

100.0a±0.000

100.0a±0.000

100.0a±0.000

100.0a±0.000

P-value 0.000 0.000 0.000 0.001 0.001


Means carrying different superscripts in the same column are significantly different at (P ≤ 0.05) or highly significantly different at (P < 0.01). Means carrying the same superscripts in the same column are non-significantly different at (P < 0.05).

Strep=Streptococcus, Haemoph= Haemophilus, Past=Pasteurella, E. coli=Escherichia coli, SE=Standard error.

 

Table 2: Hematological parameters (Mean ±SE) in dairy heifers exposed to cold stress conditions.

 

Groups Time Hb (g / dl)

RBCs (×106 / µl)

MCHC (g / dl)

WBCs (×103 / µl)

Platelets (×103 / µl)

Overall means among groups
G1

7.6a±0.012

6.00a±0.017

38.0a±0.002

6.61a±0.631

249a±0.076

Gc

7.5a±0.011

6.01a±0.015

32.3b±0.001

6.64a±0.023

264a±0.055

P-value 0.641 0.930 0.000 0.921 0.010
Overall means among sampling times
Zero

8.0a±0.022

6.14a±0.001

33.0c±0.021

6.98a±0.033

261a±0.044

1st wk

8.0a±0.032

6.12a±0.009

34.0bc±0.001

7.03a±0.061

259a±0.024

2nd wk

7.8a±0.021

6.27a±0.011

34.0bc±0.002

6.82a±0.045

246a±0.011

3rd wk

7.0b±0.011

6.02a±0.002

36.8ab±0.002

6.45ab±0.014

262a±0.001

4th wk

6.8b±0.001

5.50b±0.002

37.9a±0.002

5.82b±0.055

254a±0.040

P-value 0.000 0.001 0.025 0.062 0.378
Treatment by sampling times interactions
G1 Zero

8.0a±0.003

6.10a±0.008

32.9b±0.026

6.80a±0.074

245a±0.013

 

1st wk

8.2a±0.002

6.14a±0.002

35.4b±0.017

7.11a±0.049

266a±0.016

 

2nd wk

7.7a±0.001

6.12a±0.001

35.7b±0.022

6.68a±0.021

229b±0.021

 

3rd wk

6.9b±0.031

6.28a±0.002

41.8a±0.022

6.90a±0.023

249a±0.004

 

4th wk

7.0a±0.014

5.38b±0.006

44.1a±0.001

5.54b±0.068

255a±0.003

G2 Zero

8.0a±0.001

6.18a±0.006

33.2a±0.026

7.16a±0.068

277a±0.001

 

1st wk

7.9a±0.002

6.09a±0.001

32.6a±0.026

6.96a±0.071

251a±0.006

 

2nd wk

7.8a±0.001

6.41a±0.004

32.4a±0.021

6.96a±0.044

262a±0.033

 

3rd wk

7.2a±0.006

5.76b±0.006

31.8a±0.022

6.13a±0.042

275a±0.002

 

4th wk

6.5b±0.004

5.63b±0.004

31.6a±0.017

6.10a±0.086

253a±0.001

P-value 0.726 0.115 0.001 0.561 0.029


Means carrying different superscripts in the same column are significantly different at (P ≤ 0.05) or highly significantly different at (P < 0.01). Means carrying the same superscripts in the same column are non-significantly different at (P < 0.05).

G1=Tulathromycin injected group, Gc=Control group, Hb=Hemoglobin, RBCs=Red blood cells, MCHC=Mean corpuscular hemoglobin concentration, WBCs=White blood cells, SE=Standard error.

 

Table 3: Biochemical parameters (Mean ±SE) in dairy heifers exposed to cold stress conditions.

 

Gs Time

TP

g/dl

ALT

IU/L

CREAT

mg/dl

GLUCO

mg/dl

TG

mg/dl

TC

mg/dl

Overall means among groups
G1

5.7a±0.014

3.0a±0.022

0.9a±0.027

89a±0.032

110a±0.052

57a±0.098

Gc

5.8a±0.016

2.6a±0.013

0.9a±0.029

78a±0.080

100a±0.087

57a±0.015

P-value 0.672 0.098 0.757 0.072 0.015 0.985
Overall means among sampling times
Zero

5.4a±0.033

3.1a±0.029

1.0a±0.061

93a±0.074

118a±0.078

48b±0.025

1st w

5.9a±0.029

3.4a±0.030

1.0a±0.049

87a±0.075

105ab±0.073

48b±0.051

2nd w

5.9a±0.022

2.5ab±0.016

0.9a±0.034

86a±0.052

99b±0.099

47b±0.077

3rd w

5.8a±0.018

3.0a±0.043

0.9a±0.039

73a±0.096

104ab±0.023

69a±0.079

4th w

5.8a±0.013

2.1b±0.011

0.9a±0.032

81a±0.061

99b±0.059

71a±0.089

P-value 0.470 0.013 0.161 0.334 0.055 0.000
Treatment by sampling times interactions
G1 Zero

5.5a±0.042

3.2a±0.042

1.1a±0.089

104a±0.073

124a±0.015

54c±0.036

 

1st w

5.9a±0.046

3.4a±0.043

0.9a±0.059

90a±0.083

121a±0.047

48c±0.087

 

2nd w

6.0a±0.026

2.7ab±0.025

1.0a±0.038

99a±0.086

98b±0.021

39d±0.070

 

3rd w

5.8a±0.026

3.7a±0.082

0.9a±0.048

72a±0.097

104b±0.018

63b±0.040

 

4th w

5.4a±0.013

2.1b±0.019

0.9a±0.042

83a±0.076

105b±0.005

81a±0.054

G2 Zero

5.2a±0.052

2.9a±0.040

0.9a±0.077

82a±0.049

111a±0.071

43b±0.054

 

1st w

6.0a±0.036

3.4a±0.043

1.1a±0.074

84a±0.059

89b±0.05

49b±0.051

 

2nd w

5.8a±0.037

2.4ab±0.021

0.9a±0.053

73a±0.065

100a±0.097

55ab±0.082

 

3rd w

5.8a±0.027

2.3b±0.011

0.9a±0.063

74a±0.022

105a±0.086

76a±0.090

 

4th w

6.2a±0.020

2.1b±0.011

0.9a±0.049

78a±0.012

93ab±0.081

62a±0.091

P-value 0.567 0.363 0.024 0.545 0.099 0.023


Means carrying different superscripts in the same column are significantly different at (P ≤ 0.05) or highly significantly different at (P < 0.01). Means carrying the same superscripts in the same column are non-significantly different at (P < 0.05).

TP=Total protein, ALT=Alanine aminotransferase, CREAT=Creatinine, GLUCO=Glucose, TG=Triglycerides, TC=Total cholesterol, Gs=Groups, SE=Standard error.

 

Table 4: Temperature and relative humidity correlation coefficient with hematological parameters in dairy heifers exposed to cold stress conditions.

 

r Temp Hb RBCs MCHC WBCs Plat
RH 1 -0.056 -0.108 -0.039 0.131 -0.088
HB -0.107 1 0.647** -0.101 0.089 0.066
RBCs -0.080 0.647** 1 -0.131 0.059 0.049
MCHC -0.059 -0.101 -0.131 1 -0.133 -0.014
WBCs 0.132 0.089 0.059 -0.133 1 -0.031
Platelets -0.142 0.066 0.049 -0.014 -0.031 1


**. Correlation is highly significant (P < 0.01). *. Correlation is significant (P < 0.05). NS. Correlation is non-significant (P < 0.05).

r= Person’s correlation coefficient, Temp=Temperature, RH=Relative humidity, Hb=Hemoglobin, RBCs, RBCs=Red blood cells, MCHC=Mean corpuscular hemoglobin concentration, WBCs=White blood cells.

 

Table 5: Temperature and relative humidity correlation coefficient with biochemical parameters in dairy heifers exposed to cold stress conditions.

 

r Temp TP ALT CREAT GLUCO TG TC
RH 1 0.059 0.004 -0.061 0.013 -0.026 0.039
TP 0.105 1 -0.017 0.051 -0.126 -0.041 0.158
ALT -0.075 -0.017 1 -0.078 0.237** 0.338** -0.266**
CREAT -0.044 0.051

-0.078

1 0.100 -0.115 -0.009
GLUCO -0.008 -0.126 0.237** 0.100 1 -0.003 -0.203*
TG -0.083 -0.041 0.338** -0.115 -0.003 1 -0.021
TC -0.008 0.158 -0.226** -0.009 -0.203* -0.021 1

**. Correlation is highly significant (P < 0.01). *. Correlation is significant (P < 0.05). NS. Correlation is non-significant (P < 0.05).

r= Person’s correlation coefficient, Temp=Temperature, RH=Relative humidity, TP=Total protein, ALT=Alanine aminotransferase, CREAT=Creatinine, GLUCO=Glucose, TG=Triglycerides, TC=Total cholesterol.

 

like manifestations including sneezing, coughing, serous nasal discharges, hyperthermia, reduced feed intake, and the dull sound of the lung on auscultation.

 

Hematological examinations

The overall means in Table-2 revealed no significant differences in all the measured hematological parameters between the injected and control heifers.

 

On a time scale, red blood cells and platelet counts revealed no significant differences in Table-2 between the injected and control heifers. While, hemoglobin and white blood cells (Table-2) revealed highly significant (P<0.01) increases at the zero, 1st, and 2nd weeks post-injection compared with the rest of sampling times during the study period with no significant differences between the three weeks. Mean corpuscular hemoglobin concentrations revealed in Table-2 highly significant (P<0.01) increases at the 4th week post-injection compared with all other monitoring times.

 

Hemoglobin, red blood cells, white blood cells, and platelets (Table-2) revealed highly significant (P<0.01) declines only at the 3rd, 4th, 4th, and 2nd-week post-injection in the injected animals with no significant differences between the values at the other sampling times. Mean corpuscular hemoglobin concentrations in Table-2 revealed highly significant (P<0.01) increases at the 3rd and 4th weeks post-injection.

 

Biochemical examinations

The overall means revealed no significant differences (Table-3) in all the measured parameters between the injected and control calves.

 

Total protein, creatinine, and glucose revealed in Table-3 no significant differences in all animals under study. Alanine aminotransferase and triglycerides revealed significant (P≤0.05) declines as time passes on injected animals compared to the control. Total cholesterol, on the other hand, revealed significant (P≤0.05) increases at the 4th-week post-injection in the injected animals compared to the control.

 

Macroclimatic conditions

Temperature and relative humidity (Table-4) revealed weak negative non-significant correlations with hemoglobin, red blood cells, mean corpuscular hemoglobin concentrations, and platelet counts, as well as, weak positive non-significant correlations with white blood cells.

 

The temperature in Table-5 revealed weak positive non-significant correlations with total protein, alanine aminotransferase, glucose, and total cholesterol, as well as, weak negative non-significant correlations with creatinine and triglycerides. Relative humidity (Table-5) revealed weak positive non-significant correlations with total protein and weak negative non-significant correlations with alanine aminotransferase, creatinine, glucose, triglycerides, and total cholesterol.

 

Bacteriological examinations

The overall means revealed in Figure-1A highly significant (P<0.01) declines of total bacterial, Enterobacteriaceae, Streptococcus, Haemophilus, and Pasteurella counts in injected dairy calves compared to the control animals. The measured bacterial counts revealed (Figure-1B) highly significant (P<0.01) declines as the time of the study proceed.

 

The animal groups by the sampling times interactions in Figures 2A and 2B revealed highly significant (P<0.01) declines of total bacterial, Enterobacteriaceae, Streptococcus, Haemophilus, and Pasteurella counts in animals injected with tulathromycin compared to the control dairy heifers.

 

 

 

DISCUSSION

 

Newly born calves should respond properly to the surrounding environment with a certain degree of adaptation that might be difficult under certain circumstances depending on the biosecurity and management system in the dairy farm (Sjaastad et al., 2010). The dairy industry has been directed with greater extent toward the intensification, reproduction at the 2nd instead of the 3rd year of life, the early separation of the newly born calves from their dams, foster rearing, synchronization of estrus to focus calving in the cold seasons (winter and early spring), these circumstances contributed to the existence of stressful conditions on calves starting from day one (Ellingsen et al., 2015).

 

At the time of birth, calves exhibited higher degrees of thermolysis that are aggravated by extreme macroclimatic conditions and the evaporation action of the fetal fluids surrounding the newly born (Kirovski, 2015). Supplementing calves at the early hours of life with colostrum plays an important role in raising immunity and adjusting the physical conditions of calves (McGrath et al., 2015; Savino et al., 2011).

 

An emphasis on the preventive measures in dairy farms is critical, limiting the need for subsequent intervention. These preventive measures should include a complete accurate physical examination of the heifers’ bodies to confirm the physiological status of the heifers according to Silva et al. (2016) and Windeyer et al. (2014). Also, the preventive measures included the administration of some prophylactic drugs like tulathromycin that might protect the calves from adverse circumstances that might contribute to the development of some respiratory diseases as recommended by Holman et al. (2018) and Fontes Novo et al., (2015).

 

The current study recorded an extreme and significant in-vitro antimicrobial activity of tulathromycin against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6 with a significant 100% killing percentage. The results were consistent with those recorded by Gorden and Plummer (2010) who recorded the significant influence of tulathromycin on Pasteurella multocida, Pasteurella hemolytica, and Mycoplasma dispar that have been recorded as the most common infectious causes of respiratory diseases in calves. Keith and McGuirk, (2009), and McGuirk (2008) reported that respiratory diseases might be caused by non-infectious causes like insufficient or lack of passive immunity and/or improper calve housing macroclimate.

 

Smith et al. (2017) revealed a significant influence of tulathromycin and enrofloxacin in 8-months-old calves against Salmonella and Campylobacter for 28 days post-treatment. Pereira et al. (2016) explained the broad-spectrum action of tulathromycin macrolides on the enteric microorganisms that might attack and contribute to diarrhea in newly born calves. Foditsch et al. (2019) and Tempini et al. (2018) recorded significant influence of tulathromycin and enrofloxacin with no significant relative gene-linked resistance in calves up to 14 days. Timsit et al. (2017) also recorded the high efficient antimicrobial activities of tulathromycin against some bacterial pathogens using the traditional cultural means and sensitivity test.

 

The current study recorded clinically ill animals up to 41% in the control group compared to zero% illness in the injected animals. These results were synchronized with those reported by Stanton et al. (2010) who recorded a significant reduction in the incidence of respiratory diseases up to 4.9% compared to the control animals that recorded about 46.8% during the adverse environmental conditions. They also recorded a significant decrease in morbidity up to 9.3% compared to 34.5% in the control groups with the increase in the performance and livability of the injected animals. Abell et al. (2017) recorded the metaphylactic and therapeutic actions of tulathromycin as a single or combined form and in turn, reduced the incidence of respiratory diseases in dairy animals at the time of birth.

 

The study revealed non-significant changes in hematological and biochemical parameters in the injected animals compared to control. The results also showed significant declines in the levels of alanine aminotransferase and triglycerides of the injected animals confirming the absence of any stresses on the animals from injection or the surrounding macroclimatic conditions. The current results were supported by those recorded by Amir et al. (2013) who revealed maintaining of the biochemical and hematological conditions in calves injected with tulathromycin. Also, Ignǎtescu et al. (2018); Soliman et al. (2020) and Soliman et al. (2021) reported that the usage of preventive and biosecurity measures like the injection of some prophylactic drugs (antibiotics as tulathromycin, prebiotics, probiotics, synbiotics, and herbal additives), proper housing design, proper building direction, maximum interior arrangement, outdoor or mixed calving system, good parlor hygienic practices, sufficient disinfection program, fly and rodent-proof, control of pet animal access to the farm, hygienic disposal of carcasses, and waste management, as well as, artificial colostrum might present solutions for the high incidence of many diseases in newly born calves with more reference to the respiratory diseases. Also, O’Connor et al. (2016) recorded significant maintaining of the sera parameters in calves with no significant differences between tulathromycin and enrofloxacin treatment. Crosby et al. (2018) revealed significant superior efficiency of tulathromycin (33.7%) over enrofloxacin (18.3%) in calves during the first 45 days of life and ensured the metaphylactic effect of tulathromycin in newborn calves.

 

The current study also revealed significant reductions in total bacterial, Enterobacteriaceae, Streptococcus, Haemophilus, and Pasteurella counts in the nasal swabs collected from the newly born calves. The results were consistent with these reported by Toutain et al. (2016) who found that subcutaneous injection of newborn calves with tulathromycin 2.5 mg/kg was able to significantly reduce Mannheimia haemolytica up to 66% and Pasteurella multocida up to 87%. Baptiste and Kyvsgaard, (2017) explained that tulathromycin therapy in newborn calves produced a good metaphylaxis with a significant reduction of respiratory disease incidence. Collingnon et al. (2016) recorded a significant influence of tulathromycin against Salmonella, Campylobacter, and multi-drug resistant Shigella.

 

Dennehy (2019) and Ferguson et al. (2018) recorded significant declines in the Escherichia coli population in the samples collected from calves treated with tulathromycin and enrofloxacin. Pereira et al. (2020) recorded that tulathromycin and enrofloxacin treatment in calves revealed a higher rate of metaphylaxis with the development of some E. coli resistant bacteria. Lin et al. (2019) revealed that extra-labeled withdrawal intervals of tulathromycin should be considered in calves when administered as a prophylactic and treatment against pneumonia. Bartram et al. (2016) also recorded high efficiency of tulathromycin against Mycoplasma bovis in calves with lower lung lesions that might be caused by Mycoplasma bovis compared with other prophylactic and treating antibiotics. Doster et al. (2018) recorded a greater prophylactic influence of tulathromycin in dairy calves against resistance fecal resistome and microbiome over the changes in geography, diet, macroclimatic exposure, and transition during the early feeding periods in the feedlot.

 

CONCLUSION

 

Tulathromycin was able to produce efficient and significant in-vitro antimicrobial activity with 100% killing efficacy against Streptococcus pneumonie, Streptococcus pyogenes, Haemophilus influenzae, Pasteurella multocida, and E. coli O6 at 1.0, 1.0, 0.5, 0.5, and 0.5 h, respectively.

 

Tulathromycin as a single dose was able to exhibit sufficient protective and prophylactic activities in the dairy heifers exposed to cold stress via significant reduction of the total bacterial, Enterobacteriaceae, Streptococcus, Haemophilus, and Pasteurella counts that might contribute to a higher incidence of respiratory diseases in dairy heifers under such circumstances (cold weather), as well as, maintaining the hematological and biochemical parameters in injected calves at control levels.

 

ACKNOWLEDGMENTS

 

Sincere thanking should be provided to the private sector dairy farm in El-Sharkia Governorate, Egypt for their patience and allowance to run the current experiment. Also, we sincerely thank Prof. TS Nafie and Prof. MA Sobieh for their tremendous directions during the experiment. Also, we would like to express our sincere thanking to Dr. OF Mohamed for her help in the in-vitro experiment. The current article received no specific grant from any funding agency in the public and commercial fields, and not for profit sectors.

 

conflict of interest

 

The authors declare that they have no financial or personal conflicts which may have inappropriately influenced them in writing this manuscript.

 

AUTHORS’ CONTRIBUTION

 

AEM designed the experimental design, participated in the execution of the in-vivo experiment, and writing of the manuscript. ESS conducted the in-vitro evaluation, participated in the execution of the in-vivo experiment, and writing of the manuscript.

 

REFERENCES

 

  • Abell KM, Theurer ME, Larson RL, White BJ, Apley M. (2017). A mixed treatment comparison meta-analysis of metaphylaxis treatments for bovine respiratory disease in beef cattle. J. Anim. Sci., 95(2):626-635. https://doi.org/10.2527/jas2016.1062
  • American Public Health Association, American Water Works Association, Water Environment Federation. (2017). Standard methods for the examination of water and wastewater. By E.W. Rice, R.B. Baird, A.D. Eaton, American Water Work Association Publications 23rd Ed, Washington D.C.
  • Amir PRS, Babak FH, Asghar A, Hamed M, Amir RY, Behnam AA, Amir D, Javad K. (2013). Control of clinical pneumonia in calves by antibiotic therapy. Europ. J. Exper. Biol., 3(4): 61-65.
  • Baptiste KE, Kyvsgaard NC (2017). Do antimicrobial mass medications work? A systematic review and meta-analysis of randomised clinical trials investigating antimicrobial prophylaxis or metaphylaxis against naturally occurring bovine respiratory disease. Pathog. Dis., 75 (2017), Article ftx083. https://doi.org/10.1093/femspd/ftx083
  • Bartram DJ, Moyaert H, Vanimisetti BH, Ramage CP, Reddick D, Stegemann MR (2016). Comparative efficiency of tulathromycin and tildipirosin for the treatment of experimental Mycoplasam bovis infection in calves. Vet. Medi. Sci., 2(3): 170-178. https://doi.org/10.1002/vms3.31
  • Borderas FT, de Passillé AMB, Rushen J (2009). Temperature preferences and feed level of the newborn dairy calf. Appl. Anim. Behav. Sci., 120(1-2): 56–61. https://doi.org/10.1016/j.applanim.2009.04.010
  • Butler L, Daly R, Wright C (2006). Cold stress and newborn calves. Extension Extra, South Dakota State University Extension, Paper 73:1–3.
  • Cannon B, Nedergaard J (2011). Nonshivering thermogenesis and its adequate measurement in metabolic studies. J. Exper. Biol., 214(2):242–253. https://doi.org/10.1242/jeb.050989
  • Chester-Jones H, Heins BJ, Ziegler D, Schime KD, Schuling S, Ziegler B, de Ondarza MB, Sniffen CJ, Broadwater N. (2017). Relationships between early-life growth, intake, and birth season with first-lactation performance of Holstein dairy cows. J. Dairy Sci., 100(5):3697–3704. https://doi.org/10.3168/jds.2016-12229
  • Collignon PJ, Conly JM, Andremont A, McEwen SA, Aidara-Kan A. (2016). World Health Organization Advisory Group, Bogotá Meeting on Integrated Surveillance of Antimicrobial Resistance (WHO-AGISAR) World Health Organization ranking of antimicrobials according to their importance in human medicine: A critical step for developing risk management strategies to control antimicrobial resistance from food animal production. Clin. Infec. Dis., 63 (8): 1087-1093. https://doi.org/10.1093/cid/ciw475
  • Crosby S, Credille B, Giguere S, Berghaus R. (2018). Comparative efficacy of enrofloxacin to that of tulathromycin for the control of bovine respiratory disease and prevalence of antimicrobial resistance in Mannheimia haemolytica in calves at high risk of developing bovine respiratory disease. J. Anim. Sci., 96(4): 1259-1267. https://doi.org/10.1093/jas/sky054
  • Dennehy PH. (2019). Infectious gastroenteritis. Introduction to Clinical Infectious Disease: A Problem-Based Approach. (1st ed.), Springer, Syracuse, NY. https://doi.org/10.1007/978-3-319-91080-2_15
  • Doster E, Rovira P, Noyes RN, Burgess BA, Yang X, Weinroth MD, Lakin SM, Dean CJ, Linke L, Magnuson R, Jones KI, Boucher C, Ruiz J, Belk K, Morley PS. (2018). Investigating effects of tulathromycin metaphylaxis on the fecal resistome and microbiome of commercial feedlot cattle early in the feeding period. Front. Microbiol., 9: 1715. https://doi.org/10.3389/fmicb.2018.01715
  • Drackley JK. (2008). Calf nutrition from birth to breeding. Vet. Clin. North Am. Food Anim., 24(1):55–86. https://doi.org/10.1016/j.cvfa.2008.01.001
  • Ellingsen K, Mejdell Cecilie M, Ottesenb Nina, Larsen S, Grøndahl Ann Margaret. (2015). The effect of large milk meals on digestive physiology and behaviour in dairy calves. Physiol. Behavr., 154(February): 169–174. https://doi.org/10.1016/j.physbeh.2015.11.025
  • Ferguson KM, Jacob ME, Theriot CM, Callahan BJ, Prange T, Papich MG, Foster DM. (2018). Dosing regimen of enrofloxacin impacts intestinal pharmacokinetics and the fecal microbiota in steers. Front. Microbiol., 9 (2018), Article 2190. https://doi.org/10.3389/fmicb.2018.02190
  • Foditsch C, Pereira RVV, Siler JD, Altier C, Warnick LD. (2019). Effects of treatment with enrofloxacin or tulathromycin on fecal microbiota composition and genetic function of dairy calves. PLoS One, 14 (2019), Article e0219635. https://doi.org/10.1371/journal.pone.0219635
  • Fontes Novo MS, Freitas RL, da Costa e Silva CP, Baldacim VAP, Baccili CC, dos Reis JF, Hagiwara MK, Gomes V. (2015). Hematological adaptation in Holstein calves during the neonatal period. Brazillian J. Vet. Res. Anim. Sci., São Paulo, 52(3), 212-216. https://doi.org/10.11606/issn.1678-4456.v52i3p212-216
  • Ghasemi E, Azad-Shahraki M, Khorvash M. (2017). Effect of different fat supplements on performance of dairy calves during cold season. J. Dairy Sci., 100(7):5319–5328. https://doi.org/10.3168/jds.2016-11827
  • Gorden PJ, Plummer P. (2010). Control, management, and prevention of bovine respiratory disease in dairy calves and cows. Vet. Clin. North Am. Food Anim., 26(2):243-259. https://doi.org/10.1016/j.cvfa.2010.03.004
  • Herigstad B, Hamilton M, Heersink J. (2001). How to optimize the drop plate method for enumerating bacteria. J. Microbiol. Meth., 44(2):121-129. https://doi.org/10.1016/S0167-7012(00)00241-4
  • Holman DB, Timsit E, Booker CW, Alexander TW. (2018). Injectable antimicrobials in commercial feedlot cattle and their effect on the nasopharyngeal microbiota and antimicrobial resistance. Vet. Microbiol., 214 (February):140-147. https://doi.org/10.1016/j.vetmic.2017.12.015
  • Hulbert LE, Moisá SJ. (2016). Stress, immunity, and the management of calves. J. Dairy Sci., 99(4):3199–3216. https://doi.org/10.3168/jds.2015-10198
  • Ignǎtescu RM, Goanţǎ AM, Mihal A, Ioniţǎ L. (2018). A review of the adaptation of the newborn calf to its environment. Scientific Papers. Series D. Anim. Sci., LXI(1): 52-57.
  • Keith PP, McGuirk SM. (2009). Respiratory disease of the bovine neonate. Vet. Clin. North Am. Food Anim., 25(1): 121–137. https://doi.org/10.1016/j.cvfa.2008.10.007
  • Kim SK, Lee JH. (2016). Biofilm modeling systems. Korean J. Microbiol., 52(2): 125-139. https://doi.org/10.7845/kjm.2016.6027
  • Kirovski D. (2015). Endocrine and metabolic adaptations of calves to extra-uterine life. Acta Vet., Beograd, 65(3), 297-318. https://doi.org/10.1515/acve-2015-0025
  • Lago A, McGuirk SM, Bennett TB, Cook NB, Nordlund KV. (2006). Calf respiratory disease and pen microenvironments in naturally ventilated calf barns in winter. J. Dairy Sci., 89(10):4014–4025. https://doi.org/10.3168/jds.S0022-0302(06)72445-6
  • Lin Z, He C, Magstadt DR, Cooper VL, Kleinhenz MD, Smith JS, Gorden PJ, Wulf LW, Coetzee JF. (2019). Tissue residue depletion and estimation of extralabel meat withdrawal intervals for tulathromycin in calves after dart administration. J. Anim. Sci., 97(9): 3714-3726. https://doi.org/10.1093/jas/skz231
  • McGrath BA, Fox PF, McSweeney PLH, Kelly AL. (2015). Composition and properties of bovine colostrum: a review. Dairy Sci. Technol., 96(2): 133–158. https://doi.org/10.1007/s13594-015-0258-x
  • McGuirk SM. (2008). Disease management of dairy calves and heifers. Vet. Clin. North Am. Food Anim., 24(1): 139–153. https://doi.org/10.1016/j.cvfa.2007.10.003
  • Murray GM, Cassidy JP, Clegg TA, Tratalos JA, McClure J, O’Neill RG, Sammin DJ, Casey MJ, McElroy M, Earley B, Bourke N, More SJ. (2016). A retrospective epidemiological analysis of risk factors for a primary necropsy diagnosis of bovine respiratory disease. Prev. Vet. Med., 132 (September): 49-56. https://doi.org/10.1016/j.prevetmed.2016.08.009
  • Murray PR, Rosenthal KS, Pfaller MA. (2015). Medical Microbiology, 8th Edition, Elsevier Health Sciences, Philadelphia, PA, USA.
  • Nowakowski MA, Inskeep P, Risk J, Skogerboe TL, Benchaoui HA, Meinert TR, Sherington J, Sunderland SJ. (2004). Pharmacokinetics and lung tissue concentrations of tulathromycin, a new triamilide antibiotic, in cattle. Vet. Ther., 5(1):60-74.
  • Nutsch RG, Skogerboe TL, Rooney KA, Weigel DJ, Gajewski K, Lechtenberg KF. (2005). Comparative efficacy of tulathromycin, tilmicosin and fl orfenicol in the treatment of bovine respiratory disease in stocker cattle. Vet. Ther., 6(2):167-179.
  • O’Connor AM, Yuan C, Cullen JN, Coetzee JF, da Silva N, Wang C. (2016). A mixed treatment meta-analysis of antibiotic treatment options for bovine respiratory disease—An update. Prev. Vet. Med., 132(September): 130-139. https://doi.org/10.1016/j.prevetmed.2016.07.003
  • Papich MG. (2016). Saunders handbook of veterinary drugs (Fourth edition). Small and Large Animal. Pages 824-825. https://doi.org/10.1016/B978-0-323-24485-5.00578-7
  • Pereira RV, Altier C, Siler JD, Mann S, Jordan D, Warnick LD. (2020). Longitudinal effects of enrofloxacin or tulathromycin use in preweaned calves at high risk of bovine respiratory disease on the shedding of antimicrobial-resistant fecal Escherichia coli. J. Dairy Sci., 103(11): 10547-10599. https://doi.org/10.3168/jds.2019-17989
  • Pereira RV, Lima S, Siler JD, Foditsch C, Warnick LD, and Bicalho RC. (2016). Ingestion of milk containing very low concentration of antimicrobials: Longitudinal effect on fecal microbiota composition in preweaned calves. PLoS One, 11, Article e0147525. https://doi.org/10.1371/journal.pone.0147525
  • Pineda A, Ballou MA, Campbell JM, Cardoso FC, Drackley JK. (2016). Evaluation of serum protein-based arrival formula and serum protein supplement (Gammulin) on growth, morbidity, and mortality of stressed (transport and cold) male dairy calves. J. Dairy Sci., 99(11):9027–9039. https://doi.org/10.3168/jds.2016-11237
  • Rooney KA, Nutsch RG, Skogerboe TL, Weigel DJ, Kimberly K, Kilgore WR. (2005). Efficacy of tulathromycin compared with tilmicosin and florfenicol for the control of respiratory disease in cattle at high risk of developing bovine respiratory disease. Vet. Ther., 6(2):154-166.
  • Savino F, Liguori SA, Sorrenti M, Fissore MF, Oggero R. (2011). Breast milk hormones and regulation of glucose homeostasis. Int. J. Pediat., 803985. https://doi.org/10.1155/2011/803985
  • Silva BT, Henklein A, Marques R, De S, de Oliveira PL, Leite SBP, Novo SMF, Baccili CC, Reis JF, Gomes V. (2016). Vital parameters of Holstein calves from birth to weaning. Rev. Bras. de Med. Vet., 38(3), 299- 304.
  • Sjaastad V, Hove K, Sand O. (2010). Physiology of domestic animals. 2nd ed. Scandinavian Veterinary Press, Oslo.
  • Skogerboe TL, Rooney KA, Nutsch RG, Weigel DJ, Gajewski K, Kilgore WR. (2005). Comparative efficacy of tulathromycin versus florfenicol and tilmicosin against undifferentiated bovine respiratory disease in feedlot cattle. Vet. Ther., 6(2):180-196.
  • Smith AB, Renter DG, Cernicchiaro N, Shi X, Nickell JS, Keil DJ, Nagaraja TG. (2017). A randomized trial to assess the effect of fluoroquinolone metaphylaxis on the fecal prevalence and quinolone susceptibilities of Salmonella and Campylobacter in feedlot cattle. Foodborne Pathog. Dis., 14 (10): 600-607. https://doi.org/10.1089/fpd.2017.2282
  • Soliman ES, Abdallah MS (2020). Assessment of biosecurity measures in broiler’s farms in the Suez Canal area – Egypt using a seasonal prevalence of Salmonellosis. Veterinary World, 13(4): 622-632. https://doi.org/10.14202/vetworld.2020.622-632
  • Soliman ES, Hamad RT, Abdallah MS (2021). Preventive antimicrobial action and tissue architecture ameliorations of Bacillus subtilis in challenged broilers. Veterinary World, 14(2): 523-536. https://doi.org/10.14202/vetworld.2021.523-536
  • Soliman ES, Hamad RT, Ahmed A (2017). Prophylactic and immune modulatory influences of Nigella sativa Linn. in broilers exposed to biological challenge. Veterinary World, 10(12):1447-1455. https://doi.org/10.14202/vetworld.2017.1447-1455
  • Soliman ES, Moawed SA, Ziaan AMG. (2016). Assessing cleaning and disinfection regime in a slaughterhouse against carcasses contamination. Adv. Anim. Vet. Sci., 4(9): 449-457. https://doi.org/10.14737/journal.aavs/2016/4.9.449.457
  • Soliman ES, Sallam NH, Abouelhassan EM. (2018). Effectiveness of poultry litter amendments on bacterial survival and Eimeria oocyst sporulation. Vet. World, 11(8): 1064-1073. https://doi.org/10.14202/vetworld.2018.1064-1073
  • SPSS. (2016). Statistical Packages of Social Sciences. Version 21 for windows. SPSS. Inc. USA.
  • Stanton AL, Kelton DF, Leblanc SJ, Millman ST, Wormuth J, Dingwell RT, Leslie KS. (2010). The effect of treatment with long-acting antibiotic at postweaning movement on respiratory disease and on growth in commercial dairy calves. J. Dairy Sci., 93(2):574-581. https://doi.org/10.3168/jds.2009-2414
  • Tempini PN, Aly SS, Karle BM, and Pereira RV. (2018). Multidrug residues and antimicrobial resistance patterns in waste milk from dairy farms in Central California. J. Dairy Sci., 101 (9): 8110-8122. https://doi.org/10.3168/jds.2018-14398
  • Timsit E, Hallewell J, Booker C, Tison N, Amat S, Alexander TW. (2017). Prevalence and antimicrobial susceptibility of Mannheimia haemolytica, Pasteurella multocida, and Histophilus somni isolated from the lower respiratory tract of healthy feedlot cattle and those diagnosed with bovine respiratory disease. Vet. Microbiol., 208(September):118-125. https://doi.org/10.1016/j.vetmic.2017.07.013
  • Toutain PL, Potter T, Pelligand L, Lacroix M, Illambas J, Lees P. (2016). Standard PK/PD concepts can be applied to determine a dosage regimen for a macrolide: the case of tulathromycin in the calf. J. Vet. Pharmacol. Ther., 40(1): 16-27. https://doi.org/10.1111/jvp.12333
  • Van De Stroet DL, Calderón Díaz JA, Stalder KJ, Heinrichs AJ, Dechow CD. (2016). Association of calf growth traits with production characteristics in dairy cattle. J. Dairy Sci., 99(10):8347–8355. https://doi.org/10.3168/jds.2015-10738
  • Windeyer MC, Leslie KE, Godden SM, Hodgins DC, Lissemore KD, Leblanc SJ. (2014). Factors associated with morbidity, mortality, and growth of dairy heifer calves up to 3 months of age. Prev. Vet. Med., 113(2): 231-240. https://doi.org/10.1016/j.prevetmed.2013.10.019
  •