Next Article in Journal
Bats, Pathogens, and Species Richness
Next Article in Special Issue
Dry Period or Early Lactation—Time of Onset and Associated Risk Factors for Intramammary Infections in Dairy Cows
Previous Article in Journal
Towards New Horizons: Climate Trends in Europe Increase the Environmental Suitability for Permanent Populations of Hyalomma marginatum (Ixodidae)
Previous Article in Special Issue
A Paradox in Bacterial Pathogenesis: Activation of the Local Macrophage Inflammasome Is Required for Virulence of Streptococcus uberis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Characterization of Enterococci- and ESBL-Producing Escherichia coli Isolated from Milk of Bovides with Mastitis in Egypt

1
Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Naumburger Str. 96a, 07743 Jena, Germany
2
Department of Microbiology, Faculty of Veterinary Medicine, Benha University, Moshtohor Toukh P.O. Box 13736, Benha 13511, Egypt
3
Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Str. 9, 07745 Jena, Germany
4
InfectoGnostics Research Campus Jena e. V., Philosophenweg 7, 07743 Jena, Germany
*
Author to whom correspondence should be addressed.
Pathogens 2021, 10(2), 97; https://doi.org/10.3390/pathogens10020097
Submission received: 27 December 2020 / Revised: 13 January 2021 / Accepted: 19 January 2021 / Published: 21 January 2021
(This article belongs to the Collection Mastitis in Dairy Ruminants)

Abstract

:
This study aimed to investigate the prevalence and antimicrobial resistance of enterococci- and ESBL-producing E. coli isolated from milk of bovine mastitis cases in Egypt. Fifty milk samples of dairy animals were collected from localities in the Nile Delta region of Egypt. Isolates were identified using MALDI-TOF MS, and antibiotic susceptibility testing was performed by the broth microdilution method. PCR amplifications were carried out, targeting resistance-associated genes. Seventeen Enterococcus isolates and eight coliform isolates could be cultivated. Vancomycin resistance rate was high in Ent. faecalis. The VITEK 2 system confirmed all E. coli isolates as ESBL-producing. All Ent. faecalis isolates harbored erm(B), tetL and aac-aphD genes. The vanA gene was detected in Ent. faecalis isolate, vanB was found in other Enterococcus, while one isolate of E. casseliflavus exhibited the vanA gene. E. coli isolates exhibited high prevalence of erm(B) and tetL. E. coli isolates were analyzed by DNA microarray analysis. Four isolates were determined by O-serotyping as O8 (n = 1), O86 (n = 2) and O157 (n = 1). H-serotyping resulted in H11, H12, H21 (two isolates each) and one was of H16 type. Different virulence-associated genes were detected in E. coli isolates including lpfA, astA, celB, cma hemL, intI1 and intI2, and the iroN gene was identified by DNA microarray analysis.

1. Introduction

Bovine mastitis is the most common and costly disease affecting dairy cattle throughout the world [1]. It leads to a reduction of the amount and quality of milk produced, increasing of veterinary costs and culling of severely infected animals [2].
Coliform bacteria are the most common pathogens that have been isolated from bovine mastitis [3]. The most important coliform pathogen is Escherichia (E.) coli that causes mastitis with high incidence in comparison to other coliforms. Especially, strain type O157:H7 is of great importance because of its zoonotic character [4].
Beside mastitis, E. coli is the most common pathogen responsible for various severe gastrointestinal or urinary tract infections, and even bacteremia in humans, causing thousands of deaths worldwide every year. Emergence of extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae, especially E. coli, has been increased over the last decades. Possibly, a cause is the usage of cephalosporins as preferred agents to avoid mastitis-associated economic losses in dairy cattle [5]. High prevalence of ESBL-producing E. coli in animals has been demonstrated in many studies and varies between countries and animal species [6].
Enterococci are one of the environmental causative agents of mastitis. These opportunistic bacteria are part of normal physiological gut flora in humans and animals, but over the last years, they have become one of the main pathogens causing numerous infections in humans, mainly those hospital-acquired, such as bacteremia and infections of the urinary tract, skin, soft tissue, abdomen and pelvis and central nervous system. These infections are caused mainly by Enterococcus (Ent.) faecalis (about 80.0%) and Ent. faecium (10.0–15.0%). The high tolerance of enterococci to disadvantageous conditions allows their long survival in the environment, including in abattoirs [7]. In addition to their importance in causing diseases, enterococci can evolve resistance to antibiotics, and additionally to their intrinsic resistance properties. Enterococci may harbor multidrug resistance determinants for antimicrobial agents such as cephalosporins and aminoglycosides [8]. Generally, bacteria can produce antibiotic resistance with remarkably new mechanisms [9]. Resistance genes are often easily transferred to other species through conjugative transposons and plasmids, showing a broad host profile [10].
Due to the little information known about enterococci and E. coli from mastitis in bovides in Egypt, the aim of this study was to investigate the prevalence and antimicrobial resistance of enterococci and ESBL-producing E. coli isolated from milk of bovine mastitis cases in Egypt. The study completes the first part regarding staphylococci and streptococci from milk [11].

2. Materials and Methods

2.1. Sample Collection and Cultivation

The present study was carried out in 2018 and 2019 on 50 milk samples of dairy animals from 50 different localities in Qalyubia and Monufia governorates in the Nile Delta region of Egypt. All dairy cattle and buffalo were local Egyptian breed and kept by smallholders (1–5 animals). They were hand-milked twice daily.
All animals were subjected to clinical examination. Animals with clinical mastitis were defined when one or more of the following signs were observed: cardinal signs of inflammation in one or more of the udder quarters, signs of systemic reaction such as fever, depression and disturbed appetite, and abnormal physical character of milk such as clot formation, discoloration, alterations in viscosity, aberrant smell or presence of blood. Due to the absence of observable clinical signs in animals, the presumptive diagnosis of subclinical mastitis was done based on laboratory diagnostic tests of milk samples, including the California Mastitis Test (CMT).
Milk samples were taken and stored as previously described [11].
Isolation of bacteria from milk samples was carried out as described by the National Mastitis Council [12]. A loopful of milk sample was streaked on blood agar (Oxoid Deutschland GmbH, Wesel, Germany) supplemented with 5% sheep red blood cells and then sub-cultured on selective media: Mannitol Salt Agar, Edwards Medium and Brilliance ESBL Agar (Oxoid Deutschland GmbH) for identification of ESBL-producing microorganisms. All plates were incubated aerobically at 37 °C for 24 h. The plates were examined for colony morphology, pigmentation and hemolytic characteristics after 24 and 48 h.

2.2. Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry (MALDI-TOF MS)

Isolates were identified using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) [13]. Briefly, bacteria from overnight cultures were suspended in 300 µL of bi-distilled water and mixed with 900 µL of ethanol (96% vol/vol; Carl Roth GmbH, Karlsruhe, Germany) for precipitation. After centrifugation for 5 min at 10,000× g, the supernatant was removed, and the pellet was re-suspended in 50 µL of 70% (vol/vol) formic acid (Sigma-Aldrich Chemie GmbH, Steinheim, Germany). Fifty microliters of acetonitrile (Carl Roth GmbH) were added, mixed and centrifuged for 5 min at 10,000× g. One and a half microliters of the supernatant were transferred onto a MTP 384 Target Plate Polished Steel TF (Bruker Daltonik GmbH, Bremen, Germany). After air-drying, the material was overlaid with 2 µL of a saturated solution of α-cyano-4-hydroxycinnamic acid (Sigma-Aldrich Chemie GmbH) in a mix of 50% acetonitrile and 2.5% trifluoroacetic acid (Sigma-Aldrich Chemie GmbH). After air-drying, spectra were acquired with an Ultraflex instrument (Bruker Daltonik GmbH). The instrument was calibrated with the IVD Bacterial Test Standard (Bruker Daltonik GmbH). Analysis was carried out with the Biotyper 3.1 software (Bruker Daltonik GmbH). Interpretation of results was performed according to the manufacturer’s recommendation: score of ≥ 2.3 represented reliable species level identification, score 2.0–2.29, probable species level identification, score 1.7–1.9, probable genus level identification, and score ≤ 1.7 was considered an unreliable identification.

2.3. Antibiotic Susceptibility Testing using Broth Microdilution

The antibiotic susceptibility testing of all Enterococcus isolates was performed with the MICRONAUT system for Gram-positive bacteria (MICRONAUT-S MRSA/GP; Merlin, Bornheim, Germany) according to the manufacturer’s recommendations. It allowed the determination of minimum inhibitory concentrations (MICs) of 22 antimicrobial agents, including ampicillin (β-lactam), cefoxitin (β-lactam; cephamycin), ceftaroline (cephalosporin 5th generation), clindamycin (lincosamide), daptomycin (cyclic lipopeptide), erythromycin (macrolide), erythromycin/clindamycin, fosfomycin (epoxide antibiotic), fusidic acid (steroide antibiotic), gentamicin (aminoglycoside), linezolid (oxazolidinone), moxifloxacin (fluorchinolone 4th generation), mupirocin, oxacillin (β-lactam), penicillin G (β-lactam), rifampicin (ansamycine), synercid (streptogramine), teicoplanin (glycopeptide), tigecycline (glycylcycline), trimethoprim/sulphamethoxazole (trimethoxybenzyl pyrimidine/sulfonamide) and vancomycin (glycopeptide).
The MICRONAUT-S FLI MHK plates allowed the determination of MICs for E. coli against 14 antimicrobial agents including AMK (amikacin), AMC (amoxicillin/clavulanic acid), CAZ (ceftazidim), CMP (chloramphenicol), CIP (ciprofloxacin), ERY (erythromycin), GEN (gentamicin), IMP (imipenem), LEV (levofloxacin), PEN (penicillin G), RAM (rifampicin), STR (streptomycin), TET (tetracycline) and T/S (trimethoprim/sulfamethoxazole) in serial dilutions of the antibiotics.
Overnight grown bacteria were suspended in NaCl solution (0.9%) to obtain a turbidity corresponding to a McFarland standard of 0.5 (Dr. Lange, CADAS photometer 30, Berlin, Germany). Three hundred microliters of the suspension were diluted with 11 mL of Mueller–Hinton broth (Oxoid Deutschland GmbH), resulting in a concentration of approximately 106–107 colony forming units (cfu)/mL. In total, 100 µL of the inoculum were given in each well of the plate. After sealing the plates, they were incubated for 18 to 24 h at 37 °C. Reading of plates was done optically. Interpretation was carried out as recommended by the Clinical and Laboratory Standards Institute [14].

2.4. Antibiotic Susceptibility Testing using the VITEK 2 System

All E. coli isolates suspected as ESBL producers were subsequently confirmed using an automated microdilution system (VITEK 2, bioMérieux Deutschland GmbH, Nürtingen, Germany) according to the instructions of the manufacturer. For this study, the test card AST-N289 (bioMérieux Deutschland GmbH) was used, which included the following antibiotics: piperacillin (PIP), piperacillin/tazobactam (TZP), cefotaxime (CTX), ceftazidime (CAZ), cefepime (FEB), aztreonam (ATM), imipenem (IMP), meropenem (MEM), amikacin (AMK), gentamicin (GEN), tobramycin (TOP), ciprofloxacin (CIP), moxifloxacin (MXF), tigecycline (TGC), fosfomycin (FOS), colistin (CT) and trimethoprim/sulfamethoxazole (T/S).

2.5. DNA Extraction and Detection of Resistance-Associated Genes

Genomic DNA was extracted from bacterial cultures using the High Pure PCR Template Purification Kit (Roche Diagnostics, Mannheim, Germany) according to the instructions of the manufacturer.
For E. coli, PCR amplifications were carried out for colistin resistance genes (mcr-1, mcr-2, mcr-3, mcr-4 and mcr-5), erythromycin resistance genes (erm(A) and erm(B)), macrolide resistance genes (msrC), aminoglycoside resistance genes (aac6-aph2) and tetracycline resistance genes (tetK, tetL and tetM). Primer sequences and target genes are given in Table 1.
Enterococci PCR amplifications (primers, see Table 1) were done for vancomycin resistance genes (vanA, vanB and vanC1), erythromycin resistance genes (erm(B), erm(A) and erm(C)), penicillin resistance gene (blaZ), linezolide resistance genes (optrA and cfr), macrolide resistance gene (msrC), aminoglycoside resistance genes (aac-aphD), tetracycline resistance genes (tetK, tetM, tetL and tetO) and lincosamide resistance genes (lnuA and lnuD).
PCR products were analyzed by electrophoresis on 2% agarose gel following staining with ethidium bromide and visualizing under UV.

2.6. GenoSerotyping, Detection of Antibiotic Resistance and Virulence-Associated Genes of E. coli Isolates by Microarray Analysis

Serotypes of E. coli isolates were determined using the E. coli SeroGenoTyping AS-1 Kit (Alere Technologies GmbH, Jena, Germany). Five microliters of extracted RNA-free DNA (with a concentration of at least 100 ng/μL) were biotin-labeled by a primer extension amplification using the E. coli SeroGenoTyping AS-1 Kit according to the manufacturer’s instructions. The procedures for multiplex labeling, hybridization and data analysis were carried out as described in a previous study [29].
Antimicrobial resistance (AMR) genotypes and resistance-associated genes were ascertained using the CarbDetect AS-2 Kit and E. coli PanType AS-2 Kit, respectively (Alere Technologies GmbH). The data were automatically summarized by the “result collector”, a software tool provided by Alere Technologies GmbH. The detection of virulence-associated genes was performed using the E. coli PanType AS-2 Kit. Twenty-eight different gene loci connected with resistance to antibiotics and virulence factors associated with adhesion, fimbriae production, secretion systems, SPATE (serine protease auto-transporters), toxins and miscellaneous genes were detected. Analysis was done as described above.

3. Results

3.1. Bacterial Isolation and Identification by MALDI-TOF MS

In this study, 17 Enterococcus isolates (34.0%) were obtained from 50 milk samples of cattle and buffaloes. Identification by MALDI-TOF MS resulted in Ent. faecalis (n = 13; 26.0%), Ent. casseliflavus (n = 2; 4.0%) and Ent. hirae (n = 2; 4.0%). Additionally, 8 coliform isolates (7 E. coli and one Enterobacter cloacae) could be cultivated. Distribution of isolates from cattle and buffalo is given in Table 2.

3.2. Antimicrobial Susceptibility Profiles of Enterococcus Isolates

All Enterococcus isolates were examined for their susceptibility to 22 antimicrobial agents. Table 3 shows that all Ent. faecalis isolates were resistant to clindamycin, erythromycin, gentamicin, rifampicin, synercid, trimethoprim/sulfamethoxazole and daptomycin. The resistance rate for linezolide, moxifloxacin, erythromycin and others reached 92.3%. Vancomycin resistance rate was high, too (76.9%). Resistance rates regarding other antibiotics ranged between 38.4% for ampicillin and 84.6% for oxacillin.
Ent. casseliflavus and Ent. hirae isolates were resistant to clindamycin, erythromycin, fosfomycin, fusidic acid, daptomycin, rifampicin, trimethoprim/sulphomethoxazole, oxacillin, moxifloxacin and gentamicin. Resistance rate for vancomycin was 25.0%.

3.3. Antimicrobial Susceptibility Profiles of Escherichia coli Isolates

All E. coli isolates were resistant to penicillin, streptomycin, erythromycin, chloramphenicol, rifampicin and trimethoprim/sulfamethoxazole (Table 4). Ceftazidim, ciprofloxacin, gentamicin, levofloxacin and tetracycline followed with a resistance rate of 85.7%. Other antibiotics showed resistance rates of 42.9% for amoxicillin/clavulanic acid and imipenem and of 71.4% for amikacin, respectively.
The VITEK 2 system confirmed all 7 E. coli isolates as ESBL-producing. They showed resistance to piperacillin, cefotaxime, ceftazidime, astreonam, cefepime, gentamicin and trimethoprim/sulfamethoxazole, as shown in Table 5. All isolates were susceptible to imipenem and meropenem.

3.4. Detection of Resistance-Associated Genes in Enterococcus Isolates

All Ent. faecalis isolates harbored the erm(B) gene, associated with erythromycin resistance, the tetL gene, connected with tetracycline resistance, and aac-aphD, responsible for aminoglycoside resistance (Table 6). Other frequently detected resistance determinants were blaZ, associated with penicillin resistance, and tetM, associated with tetracycline resistance (84.6%). The vanA gene was detected in 53.8% of Ent. faecalis isolates, while vanB and vanC1 genes were not found. Other detected resistance-associated genes were msrC (n = 3), optrA (n = 2), lnuA (n = 2), lnuD (n = 1) and erm(A) (n = 1).
Other Enterococcus (n = 4) isolates exhibited high prevalence of resistance genes erm(B), tetL, aac-aph2 and vanB, with 100%, 100%, 75.0% and 75.0%, respectively. There were two Ent. casseliflavus isolates, one exhibited the vanA gene and the other one carried the vanB gene.

3.5. Detection of Resistance-Associated Genes in Coliform Bacteria

E. coli isolates exhibited high prevalence of the erm(B) gene (71.4%), followed by tetL (57.1%), while the tetK and msrC genes, responsible for macrolide resistance, were found only in 2 isolates (28.6%). Aminoglycoside resistance-associated genes aac6-aph2 were detected only in one isolate (14.3%). No colistin resistance genes have been detected.
In this study, one Enterobacter cloacae isolate was examined for the presence of antibiotic resistance genes. It harbored blaZ, erm(B), tetL, tetO and aac6-aph2 genes.

3.6. GenoSerotyping and Analysis of Escherichia coli Isolates by Microarray Investigation

Table 7 shows the data detected by microarray analysis. All of the 7 isolates were detected to be E. coli. Four isolates were determined by O-serotyping as O8 (n = 1), two were O86 and one isolate was O157. Analysis of three isolates failed. H-serotyping resulted in H11, H12, H21 (two isolates each) and one was of H16 type.
Different virulence-associated genes were detected in E. coli isolates, including the fimbrea-associated gene (lpfA). Toxin genes astA, cba, celB and cma were detected in 6 isolates, whereas only isolates 19CS0069 and 19CS0092-1 carried all four determinants (Table 7). Others were carriers of one or two of these genes. Miscellaneous virulence-associated genes like hemL and intI1 were detected in all isolates, intI2 was found in one isolate. Two isolates carried iroN and iss genes, respectively. No Shiga toxin gene or those responsible for adhesion or secretion systems were detected.
Various antibiotic resistance-associated genes were detected (Table 7). Associated with aminoglycoside resistance, the aadA1 gene was identified in 6 isolates, aadA4 in 2, aphA in 4, strA in 5 and strB in 3 isolates. The most frequently detected chloramphenicol resistance genes were cmlA1 and floR, found in 6 isolates. Two isolates carried the catA1 gene. The mphA gene responsible for macrolide resistance was detected in 3 isolates. Quinolone resistance-associated genes qnrA1 and qnrS were found in two isolates. The tetA gene connected with tetracycline resistance was found in 3 isolates. Three sulphonamide resistance determinants including sul1, sul2 and sul3 were detected in two, two and 6 isolates respectively, while four different genes associated with trimethoprim resistance were identified, including dfrA12 in 3 isolates and dfrA17, dfrA1 and dfrA14 in 2 isolates each.
Genes characteristic for ESBL-producing E. coli were detected in all isolates to different degrees: blaCTX-M9 in 5 isolates, blaCTX-M1, M15 in 2 isolates and blaTEM in 4 isolates.

4. Discussion

Raw milk consumption could present a potential risk for public health due to the presence of foodborne pathogens and spoilage bacteria from raw milk samples.
Enterococcus species were encountered in studies regarding milk connected with mastitis. In this study, prevalence of Enterococcus species in milk of cattle and buffalo was 34.0%, which is similar to a report of the authors of Reference [30], who found that Enterococcus species were present in 31.0% of cow milk samples in Iraq.
One of the most important environmental pathogens causing mastitis is Ent. faecalis. In this study, the most frequently isolated Enterococcus species was Ent. faecalis, which is in agreement with other authors’ previous work [30,31,32].
Ent. faecalis was found in 26.0% of isolates, similar to other studies that reported the prevalence of Ent. faecalis in bovine mastitis cases to be 19.5% in Egypt [33] and 20.9% in Czech Republic [34]. Other enterococci, Ent. casseliflavus (4.0%) and Ent. hirae (4.0%), were found in similar percentages, as reported in Reference [35].
Enterococci have developed various intrinsic and acquired resistance mechanisms to antibiotics. They have an intrinsic resistance to β-lactams, cephalosporins, clindamycin and low concentrations of aminoglycosides, while acquired resistance to erythromycin, linezolid, daptomycin, tetracyclines, ciprofloxacin and vancomycin was recognized [36].
Vancomycin-resistant enterococci (VRE) were first reported in 1998 and their incidence has increased rapidly through the world after that. Vancomycin-resistance determinants are van genes which can be transferred to other Gram-positive bacteria [8]. In this study, phenotypic determined vancomycin resistance was found in 76.9% of E. faecalis isolates: 53.8% of them carried the vanA gene, which is in contrast to results of the authors of Reference [37], who found E. faecalis originated from mastitis cases in Turkey resistant to vancomycin at low levels (1.06%).
All Enterococcus isolates were found to be phenotypically resistant to erythromycin. This result can be explained by the presence of the erm(B) gene in all of them, which is in agreement to reports given by other authors [31,38]. They also found that the erm(B) gene was the most prevalent erythromycin resistance gene found in enterococci from both human and animals. Tetracycline resistance in enterococci is often connected with the presence of tet genes. Here, tetL and tetM genes were found frequently, which is nearly in agreement with data reported in Reference [39]. The high percentage of erythromycin and tetracycline resistance found in this and in other studies [38,39,40,41] is due to common and prolonged usage of these antibiotics in the dairy industry for prophylaxis and treatment of mastitis-diseased cattle.
One of the most significant bacterium causing mastitis is E. coli. It is isolated in high incidence and it is more dangerous for public health, as a serotype like O157:H7 is enteropathogenic and can cause gastroenteritis, food intoxication, hemorrhagic colitis and hemolytic uremic syndrome. Furthermore, E. coli is one of the pathogens that is related to severe clinical mastitis, it is considered as a fatal mastitis pathogen, in some cases it has led to animal death [42]. In this study, in 14.0% of milk samples, E. coli was isolated, which is comparable to other Egyptian studies on mastitis-diseased cattle in Egypt with 13.3% and 18.7%, respectively [43,44]. Microarray analysis of E. coli isolates revealed that one was serotyped as O157. O157 serotype was reported in some studies in Egypt as a mastitis-causing pathogen [42,43,45] as well as one of the most harmful STEC that can cause severe human infections. Other isolates belonged to serotype O86 within the EPEC group. Isolates of this serogroup were also described as connected with bovine mastitis [46].
ESBL-encoding genes have been categorized into three main types: blaCTX-M, blaSHV and blaTEM. The blaCTX-M isolates have been further categorized into five sub-groups (blaCTX-M-1, blaCTX-M-2, blaCTX-M-8, blaCTX-M-9, blaCTX-M-25) and more than 150 variants have been documented (http://www.lahey.org/studies). In our study, all E. coli isolates were ESBL-producing, as confirmed by the VITEK 2 system, and this finding was explained by the presence of one or two types of ESBL genes. It agrees with results obtained by several authors for E. coli isolates from bovine mastitis cases [5,47,48]. Previous studies suggest that the blaCTX-M type, predominantly blaCTX-M-15, was the most prevalent ESBL type worldwide [49]. This result was alarming because E. coli which produce ESBLs of blaCTX-M-15 type are important causal agents of healthcare-oriented as well as community-based infections in humans [50] and have recently also been increasingly reported from food-producing animals [51]. The cause for frequent occurrence of ESBL-producing bacteria could be the use of β-lactams and even fourth generation cephalosporins in veterinary medicine [52]. Another reason is possibly co-selection by multiple resistance mechanisms through the usage of various antibiotics due to the fact that resistance genes for aminoglycosides, tetracyclines and trimethoprim-sulfamethoxazole are frequently placed on single conjugative plasmids, as is often also the case with blaESBL genes [53]. Generally, ESBL genes are located on plasmids that could spread easily among commensal and pathogenic bacteria within herds and the environment. All ESBL-producing E. coli were multidrug-resistant and showed resistance to cephalosporins, β-lactam and non-β-lactam antibiotics, such as ampicillin, aminoglycosides, macrolides, tetracyclines and fluoroquinolones. Many studies confirmed that ESBL-producing E. coli were multidrug-resistant independent from the source, like cattle [54,55], poultry [56], pigs [51] and humans [57].
Erythromycin resistance can be connected with the presence of different genes like erm(B), msrC or mphA, which were found in E. coli isolates of the milk samples. Similar data have been reported [58]. Other resistance determinants regarding erythromycin resistance played no role.
Phenotypic chloramphenicol resistance was detected in all E. coli isolates and DNA microarray analysis resulted in the detection of genes cmlA1, floR and catA1 responsible for it. Previously, the floR gene was detected in Egyptian E. coli isolates from neonatal calves [59] as well as floR and cmlA in E. coli from bovine mastitis cases [60].
Resistance to trimethoprim/sulphamethoxazole was detected in all 7 E. coli isolates by both the microdilution method and the VITEK 2 system, which resulted from the presence of sulphonamide resistance genes sul1, sul2 and sul3. The isolates carried at least one or two of these genes, which was already reported previously [61], while carriage of genes dfrA1, dfrA12, dfrA14 and dfrA17 responsible for resistance to trimethoprim were described for Egyptian isolates from calves and mastitis cases [59,60].
All E. coli isolates carried at least one aminoglycoside resistance determinant like aadA1, aadA4, aac6-aph2, aphA, strA or strB genes. Similar results have been found previously in E. coli isolates from bovine mastitis cases in Egypt [61] and in Iran [62]. Plasmid-mediated quinolone resistance genes qnrA1 and qnrS were present in only a few isolates, a fact which was reported in the past, too [59,60]. Nevertheless, the detection of these genes is very important due to the possibility of spread of these resistance determinants between bacteria through plasmid mobility [62].
High detection rates of tet genes described here were also found in other investigations previously [63,64], in which mainly tetA was present in E. coli isolates from bovine mastitis cases. Here, the cause of the high presence rate of tetracycline resistance genes is the widespread and uncontrolled usage of this antibiotic for treatment and prevention of infections in livestock in Egypt.
In general, increasing Enterococcus and E. coli resistance to antibiotics results in excessive use of antimicrobials in different genetic resistance mechanisms, vertically by inheriting genes to new generations or horizontally by exchanging genetic materials among bacteria.

5. Conclusions

This study clearly showed that milk from small farmers in Egypt, which is unpasteurized and used as food, is traded or will be processed further, was contaminated with bacteria which are potentially hazardous for human health. To make matters worse, many of these bacteria became multidrug-resistant, whcih makes therapy of resulting diseases hard.
As a consequence, training of farmers is desirable regarding a better hygiene system and pasteurization of produced milk without exception. A surveillance system by governmental institutions should be introduced.
Concerning antibiotic resistance, a reduction of uncontrolled administering of antibiotics should be a primary aim. Only in cases of diseases after serious diagnosis should antimicrobials be used, not as prophylaxis or even as growth promoters. The usage of last-line antibiotics like vancomycin is not allowed in veterinary medicine.

Author Contributions

W.A., H.H., H.T. and A.A.A.E.-T. participated in the conception and design of the study; W.A., H.H., F.I.E.H. and S.M. performed farm and laboratory work; W.A., H.H., H.N., H.T. and S.M. analyzed the data, wrote the manuscript and contributed to manuscript discussion. All authors read and approved the final manuscript.

Funding

There was no external funding for this study.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the Egyptian Cultural and Educational Office of the Arab Republic of Egypt in Germany and the Egyptian Ministry of Higher Education for financial support. The authors would like to thank Byrgit Hofmann, Peggy Methner, Elke Müller and Annett Reissig for their excellent technical assistance.

Conflicts of Interest

The authors declare no conflict of interests.

References

  1. Yang, F.; Liu, L.H.; Li, X.P.; Luo, J.Y.; Zhang, Z.; Yan, Z.T.; Zhang, S.D.; Li, H.S. Short communication: N-Acetylcysteine-mediated modulation of antibiotic susceptibility of bovine mastitis pathogens. J. Dairy Sci. 2016, 99, 4300–4302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Seegers, H.; Fourichon, C.; Beaudeau, F. Production effects related to mastitis and mastitis economics in dairy cattle herds. Vet. Res. 2003, 34, 475–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. El-Khodery, S.A.; Osman, S.A. Acute coliform mastitis in buffaloes (Bubalus bubalis): Clinical findings and treatment outcomes. Trop. Anim. Health Prod. 2008, 40, 93–99. [Google Scholar] [CrossRef] [PubMed]
  4. Cursons, R.T.; Williamson, J.; Bean, A. Shiga-toxin genes from Escherichia coli strains isolated from mastitis milk. In Mastitis in dairy production: Current Knowledge and Future Solutions, Proceedings of the 4th IDF International Mastitis Conference, Maastricht, The Netherlands, 12–15 June 2005; Hogeveen, H., Ed.; Wageningen Academic Publishers: Wageningen, The Netherlands, 2005; pp. 671–676. [Google Scholar]
  5. Ali, T.; Rahman, S.U.; Zhang, L.; Shahid, M.; Han, D.; Gao, J.; Zhang, S.; Ruegg, P.L.; Saddique, U.; Han, B. Characteristics and genetic diversity of multi-drug resistant extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli isolated from bovine mastitis. Oncotarget 2017, 8, 90144–90163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Valentin, L.; Sharp, H.; Hille, K.; Seibt, U.; Fischer, J.; Pfeifer, Y.; Michael, G.B.; Nickel, S.; Schmiedel, J.; Falgenhauer, L.; et al. Subgrouping of ESBL-producing Escherichia coli from animal and human sources: An approach to quantify the distribution of ESBL types between different reservoirs. Int. J. Med. Microbiol. 2014, 304, 805–816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Różańska, H.; Lewtak-Piłat, A.; Kubajka, M.; Weiner, M. Occurrence of enterococci in mastitic cow’s milk and their antimicrobial resistance. J. Vet. Res. 2019, 63, 93–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Kang, H.M.; Jung, B.Y.; Moon, J.S.; Kang, M.S.; Kim, J.M.; Chung, C.I. Distribution of Vancomycin-Resistant Enterococci (VRE) and van Gene Types in Domestic Animals. Available online: http://210.101.116.28/W_files/kiss3/0b700851_pv.pdf (accessed on 12 February 2015).
  9. Klare, I.; Werner, G.; Witte, W. Enterococci. Habitats, infections, virulence factors, resistances to antibiotics, transfer of resistance determinants. Contrib. Microbiol. 2001, 8, 108–122. [Google Scholar] [PubMed]
  10. Cöleri, A.; Cökmüs, C. Molecular mechanisms of resistance to glycopeptide antibiotics in Enterococcus species and modes of gene transfer. Turk. Hij. Den. Biyol. Derg. 2008, 65, 87–96. [Google Scholar]
  11. Ahmed, W.; Neubauer, H.; Tomaso, H.; El Hofy, F.I.; Monecke, S.; Abdeltawab, A.A.; Hotzel, H. Characterization of staphylococci and streptococci from milk of bovides with mastitis in Egypt. Pathogens 2020, 9, 381. [Google Scholar] [CrossRef]
  12. NMC (National Mastitis Council (U.S.)). Microbiological Procedures for the Diagnosis of Udder Infection and Determination of Milk Quality, 4th ed.; National Mastitis Council Inc.: Verona, WI, USA, 2004. [Google Scholar]
  13. Bizzini, A.; Greub, G. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, a revolution in clinical microbial identification. Clin. Microbiol. Infect. 2010, 16, 1614–1619. [Google Scholar] [CrossRef] [Green Version]
  14. CLSI. Performance standards for antimicrobial susceptibility testing: Twenty-fifth informational supplement M100-S25. Clin. Lab. Stand. Inst. 2015, 35, 64–70. [Google Scholar]
  15. Pedroso, S.H.S.P.; Sandes, S.H.C.; Filho, R.A.T.; Nunes, A.C.; Serufo, J.C.; Farias, L.M.; Carvalho, M.A.R.; Bomfim, M.R.Q.; Santos, S.G. Coagulase-negative staphylococci isolated from human bloodstream infections showed multidrug resistance profile. Microb. Drug Resist. 2018, 24, 635–647. [Google Scholar] [CrossRef] [PubMed]
  16. Becker, K.; van Alen, S.; Idelevich, E.A.; Schleimer, N.; Seggewiß, J.; Mellmann, A.; Kaspar, U.; Peters, G. Plasmid-encoded transferable mecB-mediated methicillin resistance in Staphylococcus aureus. Emerg. Infect. Dis. 2018, 24, 242–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Vesterholm-Nielsen, M.; Olhom Larsen, M.; Elmerdahl Olsen, J.; Moller Aarestrup, F. Occurrence of the blaZ gene in penicillin resistant Staphylococcus aureus isolated from bovine mastitis in Denmark. Acta Vet. Scand. 1999, 40, 279–286. [Google Scholar] [CrossRef] [PubMed]
  18. Getachew, Y.; Hassan, L.; Zakaria, Z.; Zaid, C.Z.; Yardi, A.; Shukor, R.A.; Marawin, L.T.; Embong, F.; Aziz, S.A. Characterization and risk factors of vancomycin-resistant enterococci (VRE) among animal-affiliated workers in Malaysia. J. Appl. Microbiol. 2012, 113, 1184–1195. [Google Scholar] [CrossRef] [PubMed]
  19. Ünal, N.; Askar, S.; Yildirim, M. Antibiotic resistance profile of Enterococcus faecium and Enterococcus faecalis isolated from broiler cloacal samples. Turk. J. Vet. Anim. Sci. 2017, 41, 199–203. [Google Scholar] [CrossRef]
  20. Sutcliffe, J.; Grebe, T.; Tait-Kamradt, A.; Wondrack, L. Detection of erythromycin-resistant determinants by PCR. Antimicrob. Agents Chemother. 1996, 40, 2562–2566. [Google Scholar] [CrossRef] [Green Version]
  21. Werner, G.; Hildebrandt, B.; Witte, W. The newly described msrC gene is not equally distributed among all isolates of Enterococcus faecium. Antimicrob. Agents Chemother. 2001, 45, 3672–3673. [Google Scholar] [CrossRef] [Green Version]
  22. Ng, L.K.; Martin, I.; Alfa, M.; Mulvey, M. Multiplex PCR for the detection of tetracycline resistant genes. Mol. Cell. Probes 2001, 15, 209–215. [Google Scholar] [CrossRef]
  23. van Asselt, G.J.; Vliegenthart, J.S.; Petit, P.L.; van de Klundert, J.A.; Mouton, R.P. High-level aminoglycoside resistance among enterococci and group A streptococci. J. Antimicrob. Chemother. 1992, 30, 651–659. [Google Scholar] [CrossRef]
  24. Fan, R.; Li, D.; Feßler, A.T.; Wu, C.; Schwarz, S.; Wang, Y. Distribution of optrA and cfr in florfenicol-resistant Staphylococcus sciuri of pig origin. Vet. Microbiol. 2017, 210, 43–48. [Google Scholar] [CrossRef] [PubMed]
  25. Haenni, M.; Saras, E.; Chaussière, S.; Treilles, M.; Madec, J.Y. ermB-Mediated erythromycin resistance in Streptococcus uberis from bovine mastitis. Vet. J. 2011, 189, 356–358. [Google Scholar] [CrossRef] [PubMed]
  26. Lina, G.; Quaglia, A.; Reverdy, M.E.; Leclercq, R.; Vandenesch, F.; Etienne, J. Distribution of genes encoding resistance to macrolides, lincosamides, and streptogramins among staphylococci. Antimicrob. Agents Chemother. 1999, 43, 1062–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Rebelo, A.R.; Bortolaia, V.; Kjeldgaard, J.S.; Pedersen, S.K.; Leekitcharoenphon, P.; Hansen, I.M.; Guerra, B.; Malorny, B.; Borowiak, M.; Hammerl, J.A.; et al. Multiplex PCR for detection of plasmid-mediated colistin resistance determinants, mcr-1, mcr-2, mcr-3, mcr-4 and mcr-5 for surveillance purposes. Eurosurveillance 2018, 23, 17-00672. [Google Scholar] [CrossRef]
  28. Borowiak, M.; Fischer, J.; Hammerl, J.A.; Hendriksen, R.S.; Szabo, I.; Malorny, B. Identification of a novel transposon-associated phosphoethanolamine transferase gene, mcr-5, conferring colistin resistance in d-tartrate fermenting Salmonella enterica subsp. enterica serovar Paratyphi B. J. Antimicrob. Chemother. 2017, 72, 3317–3324. [Google Scholar] [CrossRef] [Green Version]
  29. Braun, S.D.; Monecke, S.; Thürmer, A.; Ruppelt, A.; Makarewicz, O.; Pletz, M.; Reiβig, A.; Slickers, P.; Ehricht, R. Rapid identification of carbapenemase genes in Gram-negative bacteria with an oligonucleotide microarray-based assay. PLoS ONE. 2014, 9, e102232. [Google Scholar] [CrossRef]
  30. Hamzah, A.M.; Kadim, H.K. Isolation and identification of Enterococcus faecalis from cow milk samples and vaginal swab from human. J. Entomol. Zool. St. 2018, 6, 218–222. [Google Scholar]
  31. Kuyucouğlu, Y. Antibiotic resistance of enterococci isolated from bovine subclinical mastitis. Eurasian J. Vet. Sci. 2011, 27, 231–234. [Google Scholar]
  32. Cameron, M.; Saab, M.; Heider, L.; McClure, J.T.; Rodriguez-Lecompte, J.C.; Sanchez, J. Antimicrobial susceptibility patterns of environmental streptococci recovered from bovine milk samples in the Maritime province of Canada. Front. Vet. Sci. 2016, 3, 79. [Google Scholar] [CrossRef] [Green Version]
  33. Elhadidy, M.; Elsayyad, A. Uncommitted role of enterococcal surface protein, Esp, and origin of isolates on biofilm production by Enterococcus faecalis isolated from bovine mastitis. J. Microbiol. Immunol. Infect. 2013, 46, 80–84. [Google Scholar] [CrossRef] [Green Version]
  34. Rysanek, D.; Zouharova, M.; Babak, V. Monitoring major mastitis pathogens at the population level based on examination of bulk tank milk samples. J. Dairy Res. 2009, 76, 117–123. [Google Scholar] [CrossRef] [PubMed]
  35. Nam, H.M.; Lim, S.K.; Moon, J.S.; Kang, H.M.; Kim, J.M.; Jang, K.C.; Kim, J.M.; Kang, M.I.; Joo, Y.S.; Jung, S.C. Antimicrobial resistance of enterococci isolated from mastitic bovine milk samples in Korea. Zoonoses Public Health 2010, 57, e59–e64. [Google Scholar] [CrossRef] [PubMed]
  36. Chajęcka-Wierzchowska, W.; Zadernowska, A.; Łaniewska-Trokenheim, Ł. Antibiotic resistance of Enterococcus strains present in food. Kosmos 2017, 314, 67–79. [Google Scholar]
  37. Erbas, G.; Parin, U.; Turkyilmaz, S.; Ucan, N.; Ozturk, M.; Kaya, O. Distribution of antibiotic resistance genes in Enterococcus spp. isolated from mastitis bovine milk. Acta Vet. 2016, 66, 336–346. [Google Scholar] [CrossRef] [Green Version]
  38. Petersen, A.; Dalsgaard, A. Species composition and antimicrobial resistance genes of Enterococcus spp., isolated from integrated and traditional fish farms in Thailand. Environ. Microbiol. 2003, 5, 395–402. [Google Scholar] [CrossRef] [PubMed]
  39. Yang, F.; Zhang, S.; Shang, X.; Wang, X.; Yan, Z.; Li, H.; Li, J. Short communication: Antimicrobial resistance and virulence genes of Enterococcus faecalis isolated from subclinical bovine mastitis cases in China. J. Dairy Sci. 2019, 102, 140–144. [Google Scholar] [CrossRef] [Green Version]
  40. Diarra, M.S.; Rempel, H.; Champagne, J.; Masson, L.; Pritchard, J.; Topp, E. Distribution of antimicrobial resistance and virulence genes in Enterococcus spp. and characterization of isolates from broiler chickens. Appl. Environ. Microbiol. 2010, 76, 8033–8043. [Google Scholar] [CrossRef] [Green Version]
  41. Šeputienė, V.; Bogdaitė, A.; Ružauskas, M.; Sužiedėlienė, E. Antibiotic resistance genes and virulence factors in Enterococcus faecium and Enterococcus faecalis from diseased farm animals: Pigs, cattle and poultry. Pol. J. Vet. Sci. 2012, 15, 431–438. [Google Scholar]
  42. Lamey, A.E.; Ammar, A.M.; Zaki, E.R.; Khairy, N.; Moshref, B.S.; Refai, M.K. Virulence factors of Escherichia coli isolated from recurrent cases of clinical and subclinical mastitis in buffaloes. Int. J. Microbiol. Res. 2013, 4, 86–94. [Google Scholar]
  43. Galal, H.M.; Hakim, A.S.; Dorgham, S.M. Phenotypic and virulence genes screening of Escherichia coli strains isolated from different sources in delta Egypt. Life Sci. J. 2013, 10, 352–361. [Google Scholar]
  44. Elbably, M.A.; Emeash, H.H.; Asmaa, N.M. Risk factors associated with mastitis occurrence in dairy herds in Benisuef, Egypt. World’s Vet. J. 2013, 3, 05–10. [Google Scholar] [CrossRef]
  45. Aly, R.G.O. Coliform Mastitis in Farm Animals. Master’s Thesis (Microbiology), Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt, 2006. [Google Scholar]
  46. Sabry, H.; Deutz, A.; Awad-Masalmeh, M. Virulence factors, O-serogroups and antibiotic resistance of Escherichia coli isolates from cases of bovine acute mastitis in Austria. Wien. Tierarztl. Wschr. 2006, 93, 136–144. [Google Scholar]
  47. Nadine, G.; Roger, S.; Herbert, H. Occurrence and characteristics of extended-spectrum β-lactamase (ESBL) producing Enterobacteriaceae in food producing animals, minced meat and raw milk. BMC Vet. Res. 2012, 8, 21. [Google Scholar]
  48. Eisenberger, D.; Carl, A.; Balsliemke, J.; Kämpf, P.; Nickel, S.; Schulze, G.; Valenza, G. Molecular characterization of extended-spectrum β-lactamase-producing Escherichia coli isolates from milk samples of dairy cows with mastitis in Bavaria, Germany. Microb. Drug Resist. 2018, 24, 505–510. [Google Scholar] [CrossRef] [PubMed]
  49. D’Andrea, M.M.; Arena, F.; Pallecchi, L.; Rossolini, G.M. CTX-M-type β-lactamases: A successful story of antibiotic resistance. Int. J. Med. Microbiol. 2013, 303, 305–317. [Google Scholar] [CrossRef]
  50. Lahlaoui, H.; Ben Haj Khalifa, A.; Ben Moussa, M. Epidemiology of Enterobacteriaceae producing CTX-M type extended spectrum β-lactamase (ESBL). Med. Mal. Infect. 2014, 44, 400–404. [Google Scholar] [CrossRef]
  51. Xu, G.; An, W.; Wang, H.; Zhang, X. Prevalence and characteristics of extended-spectrum β-lactamase genes in Escherichia coli isolated from piglets with post-weaning diarrhea in Heilongjiang province, China. Front. Microbiol. 2015, 6, 1103. [Google Scholar] [CrossRef]
  52. Cavaco, L.M.; Abatih, E.; Aarestrup, F.M.; Guardabassi, L. Selection and persistence of CTX-M-producing Escherichia coli in the intestinal flora of pigs treated with amoxicillin, ceftiofur or cefquinome. Antimicrob. Agents Chemother. 2008, 52, 3612–3616. [Google Scholar] [CrossRef] [Green Version]
  53. Cantòn, R.; Coque, T.M. The CTX-M β-lactamase pandemic. Curr. Opin. Microbiol. 2006, 9, 466–475. [Google Scholar] [CrossRef]
  54. Timofte, D.; Maciuca, I.E.; Evans, N.J.; Williams, H.; Wattret, A.; Fick, J.C.; Williams, N.J. Detection and molecular characterization of Escherichia coli CTX-M-15 and Klebsiella pneumoniae SHV-12 β-lactamases from bovine mastitis isolates in the United Kingdom. Antimicrob. Agents Chemother. 2014, 58, 789–794. [Google Scholar] [CrossRef] [Green Version]
  55. Ombarak, R.A.; Zayda, M.G.; Awasthi, S.P.; Hinenoya, A.; Yamasaki, S. Serotypes, pathogenic potential, and antimicrobial resistance of Escherichia coli isolated from subclinical bovine mastitis milk samples in Egypt. Jpn. J. Infect. Dis. 2019, 72, 337–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Kar, D.; Bandyopadhyay, S.; Bhattacharyya, D.; Samanta, I.; Mahanti, A.; Nanda, P.K.; Mondal, B.; Dandapat, P.; Das, A.K.; Dutta, T.K.; et al. Molecular and phylogenetic characterization of multidrug resistant extended spectrum beta-lactamase producing Escherichia coli isolated from poultry and cattle in Odisha, India. Infect. Genet. Evol. 2015, 29, 82–90. [Google Scholar] [CrossRef] [PubMed]
  57. Gu, B.; Pan, S.; Wang, T.; Zhao, W.; Mei, Y.; Huang, P.; Tong, M. Novel cassette arrays of integrons in clinical strains of Enterobacteriaceae in China. Int. J. Antimicrob. Agents 2008, 32, 529–533. [Google Scholar] [CrossRef] [PubMed]
  58. Effendi, M.H.; Harijani, N.; Budiarto, N.; Triningtya, N.P.; Tyasningsih, W.; Plumeriastuti, H. Prevalence of pathogenic Escherichia coli isolated from subclinical mastitis in East Java Province, Indonesia. Indian Vet. J. 2019, 96, 22–25. [Google Scholar]
  59. Ahmed, A.M.; Younis, E.E.; Osman, S.A.; Ishida, Y.; El-Khodery, S.A.; Shimamoto, T. Genetic analysis of antimicrobial resistance in Escherichia coli isolated from diarrheic neonatal calves. Vet. Microbiol. 2009, 136, 397–402. [Google Scholar] [CrossRef] [PubMed]
  60. Ahmed, A.M.; Shimamoto, T. Molecular characterization of antimicrobial resistance in Gram-negative bacteria isolated from bovine mastitis in Egypt. Microbiol. Immunol. 2011, 55, 318–327. [Google Scholar] [CrossRef]
  61. Fazel, F.; Jamshidi, A.; Khoramian, B. Phenotypic and genotypic study on antimicrobial resistance patterns of E. coli isolates from bovine mastitis. Microb. Pathog. 2019, 132, 355–361. [Google Scholar] [CrossRef]
  62. Robicsek, A.; Jacoby, G.A.; Hooper, D.C. The worldwide emergence of plasmid-mediated quinolone resistance. Lancet Infect. Dis. 2006, 6, 629–640. [Google Scholar] [CrossRef]
  63. Srinivasan, V.; Gillespie, B.E.; Lewis, M.J.; Nguyen, L.T.; Headrick, S.I.; Schukken, Y.H.; Oliver, S.P. Phenotypic and genotypic antimicrobial resistance patterns of Escherichia coli isolated from dairy cows with mastitis. Vet. Microbiol. 2007, 124, 319–328. [Google Scholar] [CrossRef]
  64. Jamali, H.; Krylova, K.; Aïder, M. Identification and frequency of the associated genes with virulence and antibiotic resistance of Escherichia coli isolated from cow´s milk presenting mastitis pathology. Anim. Sci. J. 2018, 89, 1701–1706. [Google Scholar] [CrossRef]
Table 1. Primers and their sequences used for the detection of antibiotic resistance-associated genes in Enterococcus species and Escherichia coli isolates.
Table 1. Primers and their sequences used for the detection of antibiotic resistance-associated genes in Enterococcus species and Escherichia coli isolates.
AntibioticTarget GenePrimer Sequences
(5′–3′)
Expected Amplicon Size (bp)Reference
Methicillin/
oxacillin
mecAF: TCC AGA TTA CAA CTT CAC CAG G
R: CCA CTT CAT ATC TTG TAA CG
161[15]
mecBF: TTA ACA TAT ACA CCC GCT TG
R: TAA AGT TCA TTA GGC ACC TCC
2263[16]
mecCAL3: TCA AAT TGA GTT TTT CCA TTA TCA
AL4: AAC TTG GTT ATT CAA AGA TGA CGA
1931[16]
PenicillinblaZF: AAG AGA TTT GCC TAT GCT TC
R: GCT TGA CCA CTT TTA TCA GC
517[17]
VancomycinvanAF: ATG AAT AGA ATA AAA GTT GCA ATA
R: CCC CTT TAA CGC TAA TAC GAT CAA
1030[18]
vanBF: AAG CTA TGC AAG AAG CCA TG
R: CCG ACA AAA TCA TCC TC
536[18]
vanC1F: GGA ATC AAG GAA ACC TC
R: CTT CCG CCA TCA TAG CT
822[19]
Erythromycinerm(B)F: GAA AAG GTA CTC AAC CAA ATA
R: AGT AAC GGT ACT TAA ATT GTT TAC
639[20]
erm(A)F: TAT CTT ATC GTT GAG AAG GGA TT
R: CTA CAC TTG GCT TAG GAT GAA A
138[15]
erm(C)F: CTT CTT GAT CAC GAT AAT TTC C
R: ATC TTT TAG CAA ACC CGT ATTC
189[15]
MacrolidemsrCF: AAG GAA TCC TTC TCT CTC CG
R: GTA AAC AAA ATC GTT CCC G
342[21]
TetracyclinetetKF: TCG ATA GGA ACA GCA GTA
R: CAG CAG ATC CTA CTC CTT
169[22]
tetLF: TCG TTA GCG TGC TGT CAT
R: GTA TCC CAC CAA TGT AGC CG
267[22]
tetMF: GTG GAC AAA GGT ACA ACG AG
R: CGG TAA AGT TCG TCA CAC AC
406[22]
tetOF: AACTTA GGC ATT CTG GCT CAC
R: TCC CAC TGT TCC ATA TCG TCA
515[22]
Aminoglycosideaac6-aph2F: CCA AGA GCA ATA AGG GCA TA
R: CAC TAT CAT AAC CAC TAC CG
219[23]
aac-aphDF: TAA TCC AAG AGC AAT AAG GGC
R: GCC ACA CTA TCA TAA CCA CTA
227[15]
Linezolid, chloramphenicoloptrAF: AGG TGG TCA GCG AAC TCA
R: ATC AAC TGT TCC CAT TCA
1400[24]
OxazolidinonecfrF: TGA AGT ATA AAG CAG GTT GGG AGT CA
R: ACC ATA TAA TTG ACC ACA AGC AGC
400[24]
LincosamidelnuDF: ACG GAG GGA TCA CAT GGT AA
R: TCT CTC GCA TAA TAA CCT TAC GTC
475[25]
lnuAF: GGT GGC TGG GGG GTA GAT GTA TTA ACT GG
R: GCT CTC TTT GAA ATA CAT GGT ATT TTT CGA TC
323[26]
Colistinmcr-1F: AGT CCG TTT GTT CTT GTG GC
R: AGA TCC TTG GTC TCG GCT TG
320[27]
mcr-2F: CAA GTG TGT TGG TCG CAG TT
R: TCT AGC CCG ACA AGC ATA CC
715[27]
mcr-3F: AAA TAA AAA TTGTTC CGC TTA TG
R: AAT GGA GAT CCC CGT TTT T
929[27]
mcr-4F: TCA CTT TCA TCA CTG CGT TG
R: TTG GTC CAT GAC TAC CAA TG
1116[27]
mcr-5F: ATG CGG TTG TCT GCA TTT ATC
R: TCA TTG TGG TTG TCC TTT TCT G
1644[28]
Table 2. Prevalence of Enterocococcus and coliform isolates in milk samples.
Table 2. Prevalence of Enterocococcus and coliform isolates in milk samples.
Origin of MilkNumber of Milk SamplesEnterococcus faecalisEnterococcus casseliflavusEnterococcus hiraeEscherichia coliEnterobacter cloacae
n%n%n%n%n%
Clinical mastitisCattle22522.714.514.529.100.0
Buffalo10330.000.0110.0110.0110.0
Subclinical mastitisCattle5360.0120.000.0240.000.0
Buffalo13215.400.000.0215.400.0
Total501326.024.024.0714.012.0
Table 3. Antimicrobial resistance in Enterococcus isolates.
Table 3. Antimicrobial resistance in Enterococcus isolates.
AntibioticClassEnterococcus faecalis (n = 13)Other Enterococcus Species (n = 4)
SIRResistance Rate (%)SIRResistance Rate (%)
Ampicillinβ-Lactam80538.420250.0
Cefoxitinβ-Lactam; cephamycin101292.3004100
CeftarolineCephalosporin 5th generation101292.312125.0
ClindamycinLincosamide0013100004100
DaptomycinCyclic lipopeptide0013100004100
ErythromycinMacrolide011292.3004100
Erythromycin/
clindamycin
0013100004100
FosfomycinEpoxide antibiotic101292.3004100
Fusidic acidSteroide antibiotic101292.3004100
GentamicinAminoglysides0013100004100
Gentamicin high levelAminoglysides101292.301375.0
LinezolidOxazolidinone101292.301375.0
MoxifloxacinFluorchinolone 4th generation101292.3004100
Mupirocin 121076.902250.0
Oxacillinbeta-Lactam201184.6004100
Penicillin Gbeta-Lactam15753.812125.0
RifampicinAnsamycine0013100004100
SynercidStreptogramine001310002250.0
TeicoplaninGlycopeptide301076.930125
TigecyclineGlycylcycline201184.610375.0
Trimethoprim/
sulphamethoxazole
Dihdrofolatreductase/
Sulfonamide
0013100004100
VancomycinGlycopeptide211076.921125.0
Table 4. Phenotypic resistance detected by MICRONAUT system and resistance-associated genes found in of Escherichia coli isolates.
Table 4. Phenotypic resistance detected by MICRONAUT system and resistance-associated genes found in of Escherichia coli isolates.
IsolatePhenotypic Antimicrobial ResistanceDetected Resistance-Associated Genes
19CS0095-1PEN, STR, CAZ, CIP, LEV, GEN, AMK, TET, ERY, CMP, RAM, T/Serm(B), tetK
19CS0065PEN, STR, AMC, CAZ, IMP, ERY, CMP, RAM, T/Serm(B)
19CS0080-1PEN, STR, AMC, CAZ, IMP, CIP, LEV, GEN, TET, ERY, CMP, RAM, T/StetL, tetK
19CS0092-1PEN, STR, AMC, CAZ, IMP, CIP, LEV, GEN, AMK, TET, ERY, CMP, RAM, T/Serm(B), msrC, tetL
19CS0078-1PEN, STR, CIP, LEV, GEN, AMK, TET, ERY, CMP, RAM, T/Serm(B), aac6-aph2, tetL
19CS0069PEN, STR, CAZ, CIP, LEV, GEN, AMK, TET, ERY, CMP, AM, T/SmsrC
19CS0098-1PEN, STR, AMP, CAZ, CIP, LEV, GEN, TET, ERY, CMP, RAM, T/Serm(B), tetL
AMK (amikacin), AMC (amoxicillin/clavulanic acid), CAZ (ceftazidime), CMP (chloramphe-nicol), CIP (ciprofloxacin), ERY (erythromycin), GEN (gentamicin), IMP (imipenem), LEV (levofloxacin), PEN (penicillin G), RAM (rifampicin), STR (streptomycin), TET (tetracycline), T/S (trimethoprim/sulfamethoxazole).
Table 5. Results of antimicrobial resistance test for 7 E. coli isolates using the VITEK 2 system.
Table 5. Results of antimicrobial resistance test for 7 E. coli isolates using the VITEK 2 system.
IsolatePIPTZPCTXCAZFEBATMIMPMEMAMKGENTOBCIPMXFTGCFOSCTT/S
19CS0095-1RIRRRRSSSRRRRSSSRESBL
19CS0065RIRRRRSSSSSSSSSSRESBL
19CS0080-1RIRRRRSSSRSSSSSSRESBL
19CS0092-1RIRRRRSSSRRSRSRSRESBL
19CS0078-1RIRRRRSSSRRRRRRRRESBL
19CS0069RIRRRRSSSRRSRSRSRESBL
19CS0098-1RIRRRRSSSRRRRSSSRESBL
PIP—piperacillin, TZP—piperacillin/tazobactam, CTX—cefotaxime, CAZ—ceftazidime, FEB—cefepime, ATM—aztreonam, IMP—imipenem, MEM—meropenem, AMK—amikacin, GEN—gentamicin, TOB—tobramycin, CIP—ciprofloxacin, MXF—moxifloxacin, TGC—tigecycline, FOS—fosfomycin, CT—colistin, T/S—trimethoprim/sulfamethoxazole.
Table 6. Antibiotic resistance-associated genes detected in Enterococcus isolates.
Table 6. Antibiotic resistance-associated genes detected in Enterococcus isolates.
Enterococcus faecalis
(n = 13)
Other Enterococcus Species
(n = 4)
Positive (n)%Positive (n)%
Vancomycin resistance genesvanA753.8125.0
vanB00.0375.0
vanC100.0125.0
Erythromycin resistance geneserm(A)17.700.0
erm(B)131004100
erm(C)00.000.0
Penicillin resistance geneblaZ1184.6125.0
Linezolide resistance genesoptrA215.400.0
cfr00.000.0
Macrolide resistance genemsrC323.100.0
Aminoglycoside resistance genesaac-aphD13100375.0
Tetracycline resistance genestetK00.000.0
tetM1184.6125.0
tetL131004100
tetO00.000.0
Lincosamide resistance geneslnuA215.400.0
lnuD17.7250.0
Table 7. Results of DNA microarray analysis of E. coli isolates including genoserotyping and detection of genes associated with virulence and antibiotic resistance.
Table 7. Results of DNA microarray analysis of E. coli isolates including genoserotyping and detection of genes associated with virulence and antibiotic resistance.
19CS006519CS006919CS0078-119CS0080-119CS0092-119CS0095-119CS0098-1
Escherichia coli+++++++
dnaE a+++++++
gada+++++++
gapA a-+-++++
ihfA a+++++++
rrsa+++++++
O-serotyping08086--086-O157
H-serotypingH11H12H21H11H12H16H21
lpfA b--+---+
tshc-+--+--
astA d-++-+-+
cbad-+--+--
celB d-+--+--
cmad-++++++
hemL e+++++++
intl1/2 e+++++++
iroN e-----+-
isse-----+-
blaCTX-M1,M15 f-+--+--
blaTEM f-++-+++
blaCTX-M9 f+-++-++
Aminoglycosides gaadA1, aadA4aadA1, aphA, strA, strBaadA1, aphA, strAaadA1, aadA4, aphAaadA1, aphA, strA, strBstrA, strBaadA1, aphA, strA
Chloramphenicol gcmlA1cmlA1, floR, catA1cmlA1, floRcmlA1, floRcmlA1, floR, catA1floRcmlA1, floR
Macrolides g-mphA--mphAmphA-
Quinolones g-qnrA1, qnrS----qnrA1, qnrS
Tetracycline g- tetAtetA--tetA-
Sulphonamides gsul3sul1, sul2, sul3sul1, sul3sul3sul3sul2sul3
Trimethoprim gdfrA17dfrA1dfrA12dfrA17dfrA1, dfrA14dfrA12dfrA12, dfrA14
a Family, genus and species-specific marker, b Genes encoding virulence factors—fimbriae, c Genes encoding virulence factors—SPATE, d Genes encoding virulence factors—toxins, e Genes encoding virulence factors—miscellaneous, f ESBL genes, g Genes associated with antimicrobial resistance.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ahmed, W.; Neubauer, H.; Tomaso, H.; El Hofy, F.I.; Monecke, S.; Abd El-Tawab, A.A.; Hotzel, H. Characterization of Enterococci- and ESBL-Producing Escherichia coli Isolated from Milk of Bovides with Mastitis in Egypt. Pathogens 2021, 10, 97. https://doi.org/10.3390/pathogens10020097

AMA Style

Ahmed W, Neubauer H, Tomaso H, El Hofy FI, Monecke S, Abd El-Tawab AA, Hotzel H. Characterization of Enterococci- and ESBL-Producing Escherichia coli Isolated from Milk of Bovides with Mastitis in Egypt. Pathogens. 2021; 10(2):97. https://doi.org/10.3390/pathogens10020097

Chicago/Turabian Style

Ahmed, Wedad, Heinrich Neubauer, Herbert Tomaso, Fatma Ibrahim El Hofy, Stefan Monecke, Ashraf Awad Abd El-Tawab, and Helmut Hotzel. 2021. "Characterization of Enterococci- and ESBL-Producing Escherichia coli Isolated from Milk of Bovides with Mastitis in Egypt" Pathogens 10, no. 2: 97. https://doi.org/10.3390/pathogens10020097

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop