Next Article in Journal
Risk Factors for Exposure of Wild Birds to West Nile Virus in A Gradient of Wildlife-Livestock Interaction
Previous Article in Journal
qPCR Detection and Quantification of Aggregatibacter actinomycetemcomitans and Other Periodontal Pathogens in Saliva and Gingival Crevicular Fluid among Periodontitis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Determination of Virulence-Associated Genes and Antimicrobial Resistance Profiles in Brucella Isolates Recovered from Humans and Animals in Iran Using NGS Technology

1
Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj 3197619751, Iran
2
Friedrich-Loeffler-Institute, Institute of Bacterial Infections and Zoonoses (IBIZ), 96a, D-07743 Jena, Germany
3
Laboratory of Veterinary Pharmacology, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
4
Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
5
Internal Medicine III, Tropical Medicine, Tanta University Hospital, Tanta 31527, Egypt
*
Authors to whom correspondence should be addressed.
Submission received: 29 November 2022 / Revised: 25 December 2022 / Accepted: 29 December 2022 / Published: 3 January 2023
(This article belongs to the Section Bacterial Pathogens)

Abstract

:
Brucellosis is a common zoonotic disease in Iran. Antimicrobial-resistant (AMR) Brucella isolates have been reported from different developing countries, posing an imminent health hazard. The objective of this study was to evaluate AMR and virulence-associated factors in Brucella isolates recovered from humans and animals in different regions of Iran using classical phenotyping and next generation sequencing (NGS) technology. Our findings revealed that B. melitensis is the most common species in bovines, small ruminants and camels. B. abortus was isolated only from one human case. Probable intermediate or resistant phenotype patterns for rifampicin, trimethoprim-sulfamethoxazole, ampicillin-sulbactam and colistin were found. Whole genome sequencing (WGS) identified mprF, bepG, bepF, bepC, bepE, and bepD in all isolates but failed to determine other classical AMR genes. Forty-three genes associated with five virulence factors were identified in the genomes of all Brucella isolates, and no difference in the distribution of virulence-associated genes was found. Of them, 27 genes were associated with lipopolysaccharide (LPS), 12 genes were related to a type IV secretion system (virB1-B12), two were associated with the toll-interleukin-1 receptor (TIR) domain-containing proteins (btpA, btpB), one gene encoded the Rab2 interacting conserved protein A (ricA) and one was associated with the production of cyclic β-1,2 glucans (cgs). This is the first investigation reporting the molecular-based AMR and virulence factors in brucellae isolated from different animal hosts and humans in Iran. Iranian B. abortus and B. melitensis isolates are still in vitro susceptible to the majority of antibiotics used for the treatment of human brucellosis. WGS failed to determine classical AMR genes and no difference was found in the distribution of virulence-associated genes in all isolates. Still, the absence of classical AMR genes in genomes of resistant strains is puzzling, and investigation of phenotypic resistance mechanisms at the proteomic and transcriptomic levels is needed.

1. Introduction

Brucellosis is a zoonotic bacterial infection causing significant economic losses in livestock populations and severely debilitating disease in humans worldwide [1,2]. Brucellosis can be transmitted from infected animals to humans mainly by consuming unpasteurized dairy and undercooked meat/meat products, or through close contact with an infected animal [3]. Four species of the genus Brucella (B.) are pathogenic for humans: B. melitensis (main hosts: sheep and goat), B. suis (swidae), B. abortus (bovidae), and B. canis (canidae) [4]. However, the cross-species infection of hosts with different Brucella spp. has also been documented [5]. The clinical symptoms of human brucellosis are non-specific and variable, ranging from severe acute or irregular febrile illness, headache, anorexia, fatigue, weight loss, generalized aching and arthralgia to chronic disease with severe complications [6]. The treatment of brucellosis demands prolonged and appropriate antimicrobial therapy. The antibiotic treatment of intracellular bacteria such as Brucella is hampered by handicapped intracellular diffusion of antibiotics as well as the development of resistance to antibiotics [7,8,9]. Different mechanisms can induce antibiotic resistance, e.g., efflux pumps, enzymatic inactivation, horizontal gene transfer and modification of drug targets [10]. In several studies, antibiotic resistance to commonly used antimicrobial drugs, including trimethoprim/sulfamethoxazole and rifampicin, has been reported [8,11,12]. Therefore, susceptibility testing and detection of genetic resistance determinants have been strongly proposed as effective methods to monitor the efficiency of antibiotics for the advised treatment of human brucellosis [13,14,15]. However, antimicrobial susceptibility testing is not carried out routinely due to the traditional approach to therapy and technical challenges. It has to be noted that laboratory infections with aerosolized Brucella spp. often occur; thus, a specialized BSL3 laboratory needs to be established [16]. In Iran, several studies on brucellosis showed that B. abortus and B. melitensis were the most isolated Brucella species in livestock and humans [17,18]. Most human brucellosis cases have been caused by B. melitensis, and a few by B. abortus [19]. Different studies have reported a relapse rate of 13–18% in Iranian patients as one of the most critical complications, even following an appropriate treatment [20,21,22]. Several studies reported AMR in a few isolates [12,23] and an increasing trend in the number of Brucella isolates with a resistant phenotype was noted [24,25]. However, it is unclear whether this is due to the development of intrinsic or acquired resistance against antibiotic compounds, or whether the results are a consequence of the intracellular nature of brucellae and thus the inability of antimicrobial compounds to penetrate the infected site, e.g., bone tissue or reticuloendothelial cells. Furthermore, identification and characterization of virulent genes are essential to implementing efficient disease control and prevention approaches, and evaluating the pathogenicity of brucellae [26]. However, very few investigations have been performed applying whole-genome sequencing (WGS) to evaluate the antimicrobial resistance and investigate virulence genes in Brucella worldwide [27,28]. No data on the resistance genes of Brucella isolates based on WGS exist from Iran. The current study aimed to examine the sensitivity of Brucella isolates recovered from humans and animals in Iran against most of the antibiotics used for brucellosis treatment in human patients to verify the adequacy of current treatment guidelines. Moreover, NGS technology was applied to investigate AMR and virulence-associated genes.

2. Materials and Methods

2.1. Ethics Committee

This survey was part of the national surveillance plan for brucellosis, 2015–2020, and all the activities follow the ethics requirement of the plan. This study was approved by the ethics committee of the Razi Vaccine and Serum Research Institute, Karaj, Iran (IR.RVSRI.REC.2015.001) in 2015, confirming that all experiments were performed following relevant guidelines and regulations. The patients gave informed consent for sampling and for participating in the survey/questionnaire.

2.2. Brucella Isolates

Forty Brucella isolates (23 of human origin and 17 of animal origin) from culture-positive human and animal cases of brucellosis collected at the Razi Vaccine and Serum Research Institute, Karaj, Iran from 2015 to 2020 were analyzed. For this study, we selected the exanimated isolates according to the various species, various biovars and different geographical locations that Brucella spp. isolated in Iran. The isolates were recovered from different specimens. Human isolates (n = 23) were recovered from blood samples (n = 22) and one cerebrospinal fluid (CSF) sample. Animal isolates (n = 17) were recovered from milk (six cows, one camel, and one sheep), four lymph nodes (three cows and one camel), and five aborted fetuses (four sheep and one goat). All camels were apparently healthy and had not received any Brucella vaccine. Goats, cows, and sheep had an abortion history on the farm. Animal samples from aborted fetuses (abomasum content, liver, spleen, and kidneys) and milk were gathered in a sterile falcon tube and kept at −20 °C until further evaluation. Human cases were patients referred with clinical complaints of brucellosis to Razi laboratory with positive Rose Bengal, Wright, and 2ME tests.

2.3. Brucella Isolation, Biotyping, and Molecular Confirmation

For bacterial isolation, milk samples were centrifuged for 15 min at 3000× g. Subsequently, the sediment and the creamy upper layer of samples were spread on cultivation media. Primary cultivation of all samples was carried out by streaking of 10 μL samples on a Brucella selective agar [Brucella agar (Himedia, Mumbai, India) supplemented with 5% inactivated horse serum, nystatin (50,000 IU), nalidixic acid (2.5 mg), vancomycin (10 mg), bacitracin (12,500 IU), polymyxin B (2500 IU), and cycloheximide (50 mg) (Oxoid, Basingstoke, UK)]. Plates were kept for 14 days at 37 °C under 10% CO2. Suspected colonies, i.e., round, pinpoint, translucent, and pearly white colonies were selected for further analysis. A panel of classical biotyping tests was performed, i.e., H2S production, dependence on carbon dioxide (CO2), lysis by specific phages, agglutination by acriflavine, growth characteristics on dye-agar media containing thionin and basic fuchsin and agglutination with specific monospecific Brucella antisera of A and M [29]. The interpretation of results was performed according to the WOAH (OIE) manual (http://www.oie.int/en/animal-health-in-the-world/animal-diseases/Brucellosis/ (accessed on 15 October 2022)). Extraction of genomic DNA was carried out using the Exgene Cell SV kit (GeneAll, Seoul, Republic of Korea) according to the manufacturers protocol. Nanodrop (Thermo Scientific, Waltham, MA, USA) was used to evaluate DNA concentration. Furthermore, the DNA integrity was analyzed with 1% agarose gel. Samples were stored at −20 °C for later analysis. AMOS-PCR and Bruce-Ladder PCR were done as previously described [30,31]. 1% agarose gel electrophoresis was used to resolve the PCR products.

2.4. Antibiotic Susceptibility Testing (AST)

All identified isolates were subjected to AST using disk diffusion susceptibility tests and minimum inhibitory concentrations (MICs) tests for nine antibiotics. Disk diffusion susceptibility tests were performed for the antibiotics gentamicin (10 μg), streptomycin (10 μg), rifampin (5μg), doxycycline (30 μg), ceftriaxone (30 μg), ampicillin-sulbactam (10 + 10 μg) and trimethoprim/sulfamethoxazole (1.25/23.75 μg). The criteria for choosing these nine antibiotics was based on WHO guidelines on treatment of human brucellosis and the recommendation of used antibiotics in different studies [25]. The results of all antimicrobial tests were read after 48 h. The thresholds of antibiotic tests were set using the guidelines for Haemophilus spp. [8,32,33]. The MICs were evaluated by E-test. Briefly, a solution (0.5 McFarland standard) was prepared and spread onto the Muller-Hinton agar plates enriched with 5% sheep’s blood. The bacterial plates were kept under 10% CO2 at 37 °C, and AST was recorded after 48 h. MICs of bacterial isolates for rifampin (0.016–256 μg/mL), gentamicin (0.064–1024 μg/mL), doxycycline (0.016–256 μg/mL), ceftriaxone (0.016–256 μg/mL), streptomycin (0.064–1024 μg/mL), trimethoprim/sulfamethoxazole (0.002–32 μg/mL), imipenem (0.002–32 μg/mL), ampicillin (0.016–256 μg/mL), and colistin (0.016–256 μg/mL) were evaluated according to the manufacturer’s protocol (liofilchem/Italy) and the Clinical and Laboratory Standards Institute (CLSI) guidelines (2020) [34]. Reference strains, Pseudomonas aeruginosa (ATCC 27853), Escherichia coli (ATCC 25922), Staphylococcus aureus (ATCC 29213), Streptococcus pneumonia (ATCC 49619), and Enterococcus faecalis (ATCC 29212) were used to confirm the AST results. All ASTs were performed in duplicates for all Brucella isolates [35,36].

2.5. WGS and in Silico Detection of AMR and Virulence-Associated Genes

After DNA extraction through the Exgene Cell SV kit (GeneAll, South Korea), the sequencing library was prepared. The samples were sequenced on an Illumina MiSeq (Illumina, San Diego, CA, USA) using paired-end sequencing. The analysis and assembly of raw sequencing data were carried out as previously described [27]. Detection of the gene and protein sequences for AMR-associated genes was carried out through several databases, including the Resistance Gene Identifier (RGI) according to the ResFinder database [37], the Comprehensive Antibiotic Resistance Database (CARD) [38], and the NCBI AMR Finder Plus (https://github.com/ncbi/amr/wiki/Running-AMRFinderPlus, accessed on 15 October 2022) [39]. Potential virulence-associated genes were identified via the virulence factor database using the core dataset “VFDB, http://www.mgc.ac.cn/VFs/ (accessed on 22 June 2022)” [27,40].

3. Results

3.1. Brucella Identification and Characterization

In the current study, 40 Brucella isolates (3 B. abortus and 37 B. melitensis) strains were characterized by cultivation-dependent and molecular methods (Bruce-Ladder PCR, AMOS PCR, and WGS). The results of Bruce-Ladder PCR and AMOS PCR were consistent with the results of WGS. B. melitensis was detected in human blood (n = 21), and human CSF (n = 1), bovine milk (n = 4), bovine lymph nodes (n = 3), camel milk (n = 1), camel lymph nodes (n = 1), and ovine aborted fetus (n = 4), ovine milk (n = 1) and caprine aborted fetus (n = 1) samples. These isolates showed identical results in the Bruce-Ladder PCR and AMOS PCR, and were also identified as B. melitensis through WGS. One isolate from a human patient and two isolates from milk of seropositive cows were confirmed as B. abortus by PCR and WGS using the Kraken program (Table 1).

3.2. Phenotypic AMR Profiles of Brucella Strains

The obtained MIC and disk diffusion values for all tested antibiotics are shown in Table 2 and Table 3. According to MIC measurements, all tested Brucella isolates were susceptible to doxycycline (MIC90 = 0.094 μg/mL), rifampicin (MIC90 = 0.5 μg/mL), gentamycin (MIC90 = 0.75 μg/mL), imipenem (MIC90 = 4 μg/mL), ceftriaxone (MIC90 = 0.75 μg/mL), streptomycin (MIC90 = 0.5 μg/mL) and trimethoprim-sulfamethoxazole (MIC90 = 0.064 μg/mL). Non-susceptible isolates (resistant and intermediate) were observed only for colistin and ampicillin-sulbactam. All B. abortus and B. melitensis isolates (n = 40) showed resistance to colistin, while most B. melitensis isolates (n = 23) had intermediate resistance for ampicillin-sulbactam (MIC90 = 2 μg/mL) (Table 2). In disk diffusion assays, 12 B. melitensis isolates (32.5%) showed an inhibition zone of 19–17 mm for discs with 5 μg rifampicin. They were classified as intermediate rifampin-resistant according to the slow-growing bacteria standards of CLSI. In contrast, all B. abortus isolates (n = 3) and 17 B. melitensis isolates exhibited an inhibition zone of ≥20 mm (20 and 33 mm, respectively) and were considered susceptible. Resistance to rifampicin was seen in 8 B. melitensis (21.6%) isolates by disk diffusion values of ≤16 μg/mL for incubation at 10% CO2. In contrast, only 13 B. melitensis (35%) isolates showed resistance to ampicillin-sulbactam according to disk diffusion values and breakpoints of between 13 and 19 mm (≤19 mm) (Table 3).

3.3. Whole-Genome Sequencing and Data Availability

Sequencing of 40 Iranian Brucella strains yielded an average of 1,645,251 reads per isolate (range 1,217,718–2,835,032) with an average length of 275 bp. The mean coverage of genome sequences was 105.9, ranging from 99 to 202. Moreover, the Kraken2 software was applied to classify each read and assembled contig to evaluate the accurate species identification and detection of potential contaminations [41]. The first match for all isolates at the genus level was “Brucella”, on average 99.7% of the reads (minimum 99.5%, maximum 99.8%). At the level of species detection, the first match for 37 isolates was “B. melitensis”, and three isolates were confirmed as “B. abortus”. From these reads, genomes were assembled for all isolates with an average genome size of 3288,126 bp, minimum of 3243,150 bp and a mean N50 of 336.295 bp (range 251,030–462,204 bp). The GC ratio was on average 57.24% (Table S1). All study data are included in the article and supporting information (Table S2). The data have also been submitted to the European Nucleotide Archive (ENA). The project accession number is PRJEB50179.

3.4. In Silico Identification of AMR and Virulence-Associated Genes

The in silico analysis of AMR genes in 40 genomes of Iranian Brucella isolates using several databases yielded only the multiple peptide resistance factors (Brucella_suis_mprF) protein and efflux-related genes bepC, bepD, bepE, bepF, bepG by the CARD and AMRFinderPlus, respectively (Table S2). Those genes were found in all Brucella genomes, either susceptible or resistant, except one strain lacked bepG and bepF. The virulence factor database (VFDB) revealed the presence of forty-three virulence and pathogenicity factors in all tested Brucella strains (Table 4). The majority of these were associated with the LPS (lipopolysaccharide) operon (n = 27), followed by genes encoding the type IV secretion system (virB1-B12). Furthermore, two genes (btpA, btpB) code for TIR domain-containing proteins that inhibit dendritic cell maturation and proinflammatory cytokines’ production, increasing immune evasion. One gene, ricA, encoded the Rab2 interacting conserved protein A that specifically interacts with the GDP-bound form of Rab2 and may play an influential role in the maturation of the Brucella-containing vacuole, as it could slow down intracellular replication and thus increase evasion from the innate immune system. Finally, one gene (cgs) is associated with the production of cyclic β-1,2-glucans that increase intracellular survival (Table 4). It is significant to highlight that Iranian B. abortus and B. melitensis isolates showed no difference in the distribution of virulence-associated genes, even from different hosts.

4. Discussion

Brucellosis remains a notorious zoonotic infection causing significant damage to the farming industry and public health. The disease is prevalent in Mediterranean and Middle Eastern countries including Iran [42,43,44]. According to the WHO, the brucellosis burden specifically on developing and low-income countries substantiates its classification as a serious zoonotic disease. In Iran, B. melitensis and B. abortus are the dominant Brucella species [19]. Our study confirmed the accurate diagnosis of Brucella spp. isolated from animals and humans through classical and molecular typing. The current study used classical biotyping and bacterial culture as the gold standard combined with DNA-based tools (multiplex PCR and WGS) to identify Brucella strains isolated from various hosts. The diagnosis of the different methods used in this study was consistent at the genus and species levels. Multiplex PCR tests and WGS appeared to be a reliable and rapid approach for the accurate classification of Brucella strains [27,31] with the potential to replace classical biotyping. Brucella identification by molecular methods has been reported as a fast and precise test in diagnostic laboratories. However, for improving in silico Brucella identification, global reference databases are required to identify different species accurately. In this way, WGS provides a powerful method for accurate typing of Brucella spp. because of the evaluation of the entire bacterial genome, thus improving discriminatory power [45,46] and replacing or overcoming the classical approach. Although WGS enables detailed strain typing, PCR should still be considered an essential method. It quickly provides information on genus and species identity and thus helps to take appropriate safety measures to decrease the risk of laboratory-acquired infections. These results improve our knowledge on the current species of Brucella in ruminant and human reservoirs of Iran and confirm a significant burden of B. melitensis. In livestock, B. melitensis is common in bovines, camels, and small ruminants [44]. The increasing number of cases of B. melitensis in cattle are also reported from other Middle East and African countries [47,48,49,50] and Iran [51]. The results of this study confirm that human brucellosis in Iran is mainly caused by B. melitensis, which is the predominant species causing human disease globally [52,53,54].
In Iran, brucellosis treatment follows the World Health Organization (WHO) guidelines [6]. The WHO recommends a combination of streptomycin and doxycycline or gentamicin and doxycycline for patients younger than 60 years, and rifampicin and doxycycline for patients older than 60 years and children [55,56]. For pregnant women, trimethoprim/sulfamethoxazole (cotrimoxazole) plus rifampin has been used [57]. For pregnant women <36 weeks gestation, the treatment depends on a combination of trimethoprim/sulfamethoxazole and rifampicin, and after 36 weeks gestation, rifampicin is administered as a monotherapy [58]. Our results show that all Brucella isolates were susceptible to ceftriaxone, imipenem, doxycycline and gentamicin. The findings that Brucella isolates were susceptible to tetracycline, doxycycline, streptomycin, ciprofloxacin, gentamicin, levofloxacin and trimethoprim-sulfamethoxazole are in accordance with reports from Egypt [27], Turkey [59], Saudi Arabia [60], China [61] and Norway [28]. Intermediate or resistant phenotypes for rifampicin, trimethoprim-sulfamethoxazole, ampicillin-sulbactam and colistin were, however, found. Indeed, antimicrobial resistance is frequently observed in brucellae [62]. This increase in AMR may be responsible for the increasing number of relapses which was reported over the last few years in different studies [20,35,61]. However, AST is often not practised before the start of the treatment due to the notorious serological diagnosis and the lack of suitable samples. A considerable number of brucellae were found resistant to rifampin in Iran [23,63] and several countries in the Middle East, such as Turkey [9,15], Saudi Arabia [64], Qatar [65] and Egypt [27]. However, it is still an essential antibiotic in the treatment regimens of brucellosis. Trimethoprim-sulfamethoxazole was found to be an effective antimicrobial compound in the treatment of human brucellosis [66]. However, a decreased susceptibility was reported in Iran previously [32,63,67,68].
The analysis of WGS data of 40 Iranian isolates revealed two AMR genes, multiple peptide resistance factors (mprF) and the outer membrane efflux protein bep G, F, C, E and D in all strains. It is known that mprF plays an essential role in resistance to cationic antibiotics such as gentamycin, moenomycin and vancomycin [69]. However, the results from the disk diffusion assays in this study showed no resistance to gentamycin. It is known that bep proteins increase resistance to some antibiotic compounds such as tetracycline, doxycycline, chloramphenicol and ciprofloxacin in B. suis [70]. Hence, none of these B. abortus and B. melitensis isolates showed resistance to those antibiotics. The inability to detect classical resistance genes in the genome of brucellae is puzzling. This finding might be explained by the presence of other factors, like mutations in housekeeping genes or regulatory mechanisms [71], or till now unknown AMR genes in Brucella, which are not yet registered in public AMR databases. Furthermore, the intracellular lifestyle of brucellae that prohibits the penetration of different antimicrobials into the cells may play a role in the resistance development of these bacteria. There are only a few molecular-based investigations on the genetic determinants of antibiotic resistance in brucellae [28,61,72,73].
In the present study, we have also analyzed the virulence genes of different Brucella isolates in silico. Brucellae, as facultative intracellular bacteria, do not use “classic” virulence factors such as proteases, cytolysins, exotoxins, capsules, exoenzymes, virulence plasmids and pili or fimbriae [74]. In the current study, most identified pathogenicity-associated genes are involved in LPS production and type IV secretion systems. Until now, there exist few reports on the detection of virulence-associated genes in Brucella strains isolated from humans and livestock from Iran [75,76]. Examination of B. abortus and B. melitensis strains isolated from animal and human hosts in Iran revealed the presence of virB5, btpA, btpB, vceC, bpe275, bspB, and virB2 genes in all strains, while betB was found in 97% and prpA in 86% of the strains [75]. Another study also reported the presence of ure, wbkA, omp19, manA, mviN, and perA genes in B. melitensis and B. abortus using multiplex-PCR tests [77]. Although several PCR methods have been reported to identify virulence- and resistance-associated genes in Brucella isolates, these methods are limited by the species-specificity of used primers.

5. Conclusions

The implementation of high-throughput WGS allowed for more comprehensive detection of virulence- and resistance-associated genes. No clear difference in the distribution of the AMR and virulence genes among both resistant and sensitive B. abortus and B. melitensis strains was found, even for those recovered from different hosts. Therefore, further investigations of antibiotic susceptibility have to be continued on Brucella isolates. Although the study of resistance and virulence mechanisms based on the genome was helpful and provided a comprehensive explanation in several microorganisms, it is of little value in the case of brucellae. Thus, resistance and virulence mechanisms at the proteomic and transcriptomic levels have to be considered in brucellae in future research.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/pathogens12010082/s1, Table S1: The WGS and genome assembly data including the average read length and the coverage of 40 Brucella isolates from ruminants and humans from Iran. Table S2: The results of in silico AMR and virulence-associated genes of 40 Brucella isolates from ruminants and humans from Iran.

Author Contributions

Conceptualization, M.D. and G.W.; methodology, M.D. and S.A.; software, H.B.; validation, M.E., A.R.E. and H.N.; formal analysis, A.R.E.; investigation, M.D. and S.A.; resources, M.D. and G.W.; data curation, H.B.; writing—original draft preparation, M.D. and G.W.; writing—review and editing, M.D. and G.W.; visualization, M.D. and G.W.; supervision, M.D. and G.W.; project administration, M.D. and S.A.; funding acquisition, M.D. and G.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work is based upon research funded by Iran National Science Foundation (INFS), Iran, under project No: 99030922.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board (or Ethics Committee) of the Razi Vaccine and Serum Research Institute, Karaj, Iran (IR.RVSRI.REC.2015.001) in 2015 confirming that all experiments were performed following relevant guidelines and regulations.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All study data are included in the article and Supplementary Materials. The data have also been submitted to the European Nucleotide Archive (ENA). The project accession number is PRJEB50179.

Acknowledgments

The authors would like to thank the Brucellosis department staff whose support and collaboration made this study possible. All individuals included in this section have consented to the acknowledgement.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ghanbari, M.K.; Gorji, H.A.; Behzadifar, M.; Sanee, N.; Mehedi, N.; Bragazzi, N.L. One health approach to tackle brucellosis: A systematic review. Trop. Med. Health 2020, 48, 86. [Google Scholar] [CrossRef] [PubMed]
  2. Dadar, M.; Fakhri, Y.; Shahali, Y.; Mousavi Khaneghah, A. Contamination of milk and dairy products by Brucella species: A global systematic review and meta-analysis. Food Res. Int. 2020, 128, 108775. [Google Scholar] [CrossRef] [PubMed]
  3. Memish, Z.A.; Balkhy, H.H. Brucellosis and international travel. J. Travel Med. 2004, 11, 49–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hull, N.C.; Schumaker, B.A. Comparisons of brucellosis between human and veterinary medicine. Infect. Ecol. Epidemiol. 2018, 8, 1500846. [Google Scholar] [CrossRef] [PubMed]
  5. Richomme, C.; Gauthier, D.; Fromont, E. Contact rates and exposure to inter-species disease transmission in mountain ungulates. Epidemiol. Infect. 2006, 134, 21–30. [Google Scholar] [CrossRef] [PubMed]
  6. Corbel, M.J. Brucellosis in Humans and Animals; World Health Organization: Geneva, Switzerland, 2006. [Google Scholar]
  7. Biswas, S.; Raoult, D.; Rolain, J.-M. A bioinformatic approach to understanding antibiotic resistance in intracellular bacteria through whole genome analysis. Int. J. Antimicrob. Agents 2008, 32, 207–220. [Google Scholar] [CrossRef]
  8. Abdel-Maksoud, M.; House, B.; Wasfy, M.; Abdel-Rahman, B.; Pimentel, G.; Roushdy, G.; Dueger, E. In vitro antibiotic susceptibility testing of Brucella isolates from Egypt between 1999 and 2007 and evidence of probable rifampin resistance. Ann. Clin. Microbiol. Antimicrob. 2012, 11, 24. [Google Scholar] [CrossRef] [Green Version]
  9. Etiz, P.; Kibar, F.; Ekenoglu, Y.; Yaman, A. Characterization of antibiotic susceptibility of Brucella spp isolates with E-Test method. Arch. Clin. Microbiol. 2015, 6, 1–5. [Google Scholar]
  10. Munita, J.M.; Arias, C.A. Mechanisms of Antibiotic Resistance. Microbiol. Spectr. 2016, 4, 2–4. [Google Scholar] [CrossRef] [Green Version]
  11. Alamian, S.; Dadar, M.; Wareth, G. Role of Brucella abortus Biovar 3 in the Outbreak of Abortion in a Dairy Cattle Herd Immunized with Brucella abortus Iriba Vaccine. Arch. Razi Inst. 2020, 75, 377–384. [Google Scholar]
  12. Razzaghi, R.; Rastegar, R.; Momen-Heravi, M.; Erami, M.; Nazeri, M. Antimicrobial susceptibility testing of Brucella melitensis isolated from patients with acute brucellosis in a centre of Iran. Indian J. Med. Microbiol. 2016, 34, 342–345. [Google Scholar] [CrossRef] [PubMed]
  13. Dadar, M.; Bazrgari, N.; Garosi, G.A.; Hassan, S. Investigation of Mutations in the Rifampin-Resistance-Determining Region of the rpoB Gene of Brucella melitensis by Gene Analysis. Jundishapur J. Microbiol. 2021, 14, e115526. [Google Scholar] [CrossRef]
  14. Hashim, R.; Ahmad, N.; Zahidi, M.; Tay, B.; Mohd Noor, A.; Zainal, S.; Hamzah, H.; Hamzah, S.; Chow, T.; Wong, P. Identification and in vitro antimicrobial susceptibility of Brucella species isolated from human brucellosis. Int. J. Microbiol. 2014, 2014, 596245. [Google Scholar] [CrossRef] [Green Version]
  15. Baykam, N.; Esener, H.; Ergönül, Ö.; Eren, Ş.; Çelïkbas, A.K.; Dokuzoǧuz, B. In vitro antimicrobial susceptibility of Brucella species. Int. J. Antimicrob. Agents 2004, 23, 405–407. [Google Scholar] [CrossRef]
  16. Siengsanan-Lamont, J.; Blacksell, S.D. A review of laboratory-acquired infections in the Asia-Pacific: Understanding risk and the need for improved biosafety for veterinary and zoonotic diseases. Trop. Med. Infect. Dis. 2018, 3, 36. [Google Scholar] [CrossRef] [Green Version]
  17. Dadar, M.; Shahali, Y.; Fakhri, Y. Brucellosis in Iranian livestock: A meta-epidemiological study. Microb. Pathog. 2021, 155, 104921. [Google Scholar] [CrossRef] [PubMed]
  18. Dadar, M.; Wareth, G.; Neubauer, H. Brucellosis in Iranian buffalo: Prevalence and diagnostic methods. Ger. J. Vet. Res 2021, 1, 13–16. [Google Scholar] [CrossRef]
  19. Dadar, M.; Alamian, S.; Behrozikhah, A.M.; Yazdani, F.; Kalantari, A.; Etemadi, A.; Whatmore, A.M. Molecular identification of Brucella species and biovars associated with animal and human infection in Iran. Vet. Res. Forum. 2019, 10, 315–321. [Google Scholar] [PubMed]
  20. Alavi, S.M.; Alavi, S.M.R.; Alavi, L. Relapsed human brucellosis and related risk factors. Pak. J. Med. Sci. 2009, 25, 46–50. [Google Scholar]
  21. Hadadi, A.; Rasoulinejad, M.; HajiAbdolbaghi, M.; Mohraz, M.; Khashayar, P. Clinical profile and management of brucellosis in Tehran–Iran. Acta Clin. Belg. 2009, 64, 11–15. [Google Scholar] [CrossRef]
  22. Sasan, M.-S.; Nateghi, M.; Bonyadi, B.; Aelami, M.-H. Clinical features and long term prognosis of childhood brucellosis in northeast Iran. Iran. J. Pediatr. 2012, 22, 319. [Google Scholar] [PubMed]
  23. Alamian, S.; Dadar, M.; Etemadi, A.; Afshar, D.; Alamian, M.M. Antimicrobial susceptibility of Brucella spp. isolated from Iranian patients during 2016 to 2018. Iran. J. Microbiol. 2019, 11, 363. [Google Scholar] [CrossRef] [PubMed]
  24. Yuan, H.-T.; Wang, C.-L.; Liu, L.-N.; Wang, D.; Li, D.; Li, Z.-J.; Liu, Z.-G. Epidemiologically characteristics of human brucellosis and antimicrobial susceptibility pattern of Brucella melitensis in Hinggan League of the Inner Mongolia Autonomous Region, China. Infect. Dis. Poverty 2020, 9, 79. [Google Scholar] [CrossRef]
  25. Skalsky, K.; Skalsky, K.; Yahav, D.; Bishara, J.; Pitlik, S.; Leibovici, L.; Paul, M. Treatment of human brucellosis: Systematic review and meta-analysis of randomised controlled trials. BMJ 2008, 336, 701–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Awwad, E.; Adwan, K.; Farraj, M.; Essawi, T.; Rumi, I.; Manasra, A.; Baraitareanu, S.; Gurau, M.R.; Danes, D. Cell envelope virulence genes among field strains of Brucella melitensis isolated in West Bank part of Palestine. Agric. Agric. Sci. Procedia 2015, 6, 281–286. [Google Scholar]
  27. Wareth, G.; El-Diasty, M.; Abdel-Hamid, N.H.; Holzer, K.; Hamdy, M.E.; Moustafa, S.; Shahein, M.A.; Melzer, F.; Beyer, W.; Pletz, M.W. Molecular characterization and antimicrobial susceptibility testing of clinical and non-clinical Brucella melitensis and Brucella abortus isolates from Egypt. One Health 2021, 13, 100255. [Google Scholar] [CrossRef]
  28. Johansen, T.B.; Scheffer, L.; Jensen, V.K.; Bohlin, J.; Feruglio, S.L. Whole-genome sequencing and antimicrobial resistance in Brucella melitensis from a Norwegian perspective. Sci. Rep. 2018, 8, 8538. [Google Scholar] [CrossRef]
  29. Alton, G.; Jones, L.; Angus, R.; Verger, J. Techniques for the Brucellosis laboratory Paris Institute National de la Recherdie Agrononique. J. Clin. Microbiol. 1988, 33, 3198–3200. [Google Scholar]
  30. Ewalt, D.R.; Bricker, B.J. Validation of the Abbreviated BrucellaAMOS PCR as a Rapid Screening Method for Differentiation ofBrucella abortus Field Strain Isolates and the Vaccine Strains, 19 and RB51. J. Clin. Microbiol. 2000, 38, 3085–3086. [Google Scholar] [CrossRef] [Green Version]
  31. Lopez-Goñi, I.; Garcia-Yoldi, D.; Marin, C.; De Miguel, M.; Munoz, P.; Blasco, J.; Jacques, I.; Grayon, M.; Cloeckaert, A.; Ferreira, A. Evaluation of a multiplex PCR assay (Bruce-ladder) for molecular typing of all Brucella species, including the vaccine strains. J. Clin. Microbiol. 2008, 46, 3484–3487. [Google Scholar] [CrossRef] [Green Version]
  32. Asadi, F.T.; Hashemi, S.H.; Alikhani, M.Y.; Moghimbeigi, A.; Naseri, Z. Clinical and diagnostic aspects of brucellosis and antimicrobial susceptibility of Brucella isolates in Hamedan, Iran. Jpn. J. Infect. Dis. 2017, 70, 235–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Cooke, R.; Perrett, L. Antimicrobial susceptibility data for Brucella melitensis isolates cultured from UK patients. J. Infect. 2014, 68, 401. [Google Scholar] [CrossRef] [PubMed]
  34. Weinstein, M.P.; Lewis, J.S. The clinical and laboratory standards institute subcommittee on antimicrobial susceptibility testing: Background, organization, functions, and processes. J. Clin. Microbiol. 2020, 58, e01864-19. [Google Scholar] [CrossRef] [PubMed]
  35. Pauletti, R.B.; Stynen, A.P.R.; da Silva Mol, J.P.; Dorneles, E.M.S.; Alves, T.M.; Souto, M.d.S.M.; Minharro, S.; Heinemann, M.B.; Lage, A.P. Reduced susceptibility to Rifampicin and resistance to multiple antimicrobial agents among Brucella abortus isolates from cattle in Brazil. PLoS ONE 2015, 10, e0132532. [Google Scholar]
  36. Wayne, P. Clinical and laboratory standards institute. Perform. Stand. Antimicrob. Susceptibility Test. 2007, 17, 100–121. [Google Scholar]
  37. Zankari, E.; Hasman, H.; Cosentino, S.; Vestergaard, M.; Rasmussen, S.; Lund, O.; Aarestrup, F.M.; Larsen, M.V. Identification of acquired antimicrobial resistance genes. J. Antimicrob. Chemother. 2012, 67, 2640–2644. [Google Scholar] [CrossRef] [PubMed]
  38. Jia, B.; Raphenya, A.R.; Alcock, B.; Waglechner, N.; Guo, P.; Tsang, K.K.; Lago, B.A.; Dave, B.M.; Pereira, S.; Sharma, A.N. CARD 2017: Expansion and model-centric curation of the comprehensive antibiotic resistance database. Nucleic Acids Res. 2016, 45, gkw1004. [Google Scholar] [CrossRef]
  39. Feldgarden, M.; Brover, V.; Haft, D.H.; Prasad, A.B.; Slotta, D.J.; Tolstoy, I.; Tyson, G.H.; Zhao, S.; Hsu, C.-H.; McDermott, P.F. Validating the AMRFinder tool and resistance gene database by using antimicrobial resistance genotype-phenotype correlations in a collection of isolates. Antimicrob. Agents Chemother. 2019, 63, e00483-19. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, B.; Zheng, D.; Jin, Q.; Chen, L.; Yang, J. VFDB 2019: A comparative pathogenomic platform with an interactive web interface. Nucleic Acids Res. 2019, 47, D687–D692. [Google Scholar] [CrossRef]
  41. Bankevich, A.; Nurk, S.; Antipov, D.; Gurevich, A.A.; Dvorkin, M.; Kulikov, A.S.; Lesin, V.M.; Nikolenko, S.I.; Pham, S.; Prjibelski, A.D. SPAdes: A new genome assembly algorithm and its applications to single-cell sequencing. J. Comput. Biol. 2012, 19, 455–477. [Google Scholar] [CrossRef] [Green Version]
  42. Bagheri Nejad, R.; Krecek, R.C.; Khalaf, O.H.; Hailat, N.; Arenas-Gamboa, A.M. Brucellosis in the Middle East: Current situation and a pathway forward. PLoS Negl. Trop. Dis. 2020, 14, e0008071. [Google Scholar] [CrossRef] [PubMed]
  43. Wareth, G.; El-Diasty, M.; Melzer, F.; Schmoock, G.; Moustafa, S.A.; El-Beskawy, M.; Khater, D.F.; Hamdy, M.E.R.; Zaki, H.M.; Ferreira, A.C.; et al. MLVA-16 Genotyping of Brucella abortus and Brucella melitensis Isolates from Different Animal Species in Egypt: Geographical Relatedness and the Mediterranean Lineage. Pathogens 2020, 9, 498. [Google Scholar] [CrossRef]
  44. El-Diasty, M.; Salah, K.; El-Hofy, F.I.; Abd El Tawab, A.A.; Soliman, E.A. Investigation of an outbreak of brucellosis in a mixed dairy farm and evaluation of a test and slaughter strategy to release the herd out of the quarantine. Ger. J. Vet. Res. 2022, 2, 1–9. [Google Scholar] [CrossRef]
  45. Georgi, E.; Walter, M.C.; Pfalzgraf, M.-T.; Northoff, B.H.; Holdt, L.M.; Scholz, H.C.; Zoeller, L.; Zange, S.; Antwerpen, M.H. Whole genome sequencing of Brucella melitensis isolated from 57 patients in Germany reveals high diversity in strains from Middle East. PLoS ONE 2017, 12, e0175425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Foster, J.T.; Beckstrom-Sternberg, S.M.; Pearson, T.; Beckstrom-Sternberg, J.S.; Chain, P.S.; Roberto, F.F.; Hnath, J.; Brettin, T.; Keim, P. Whole-genome-based phylogeny and divergence of the genus Brucella. J. Bacteriol. 2009, 191, 2864–2870. [Google Scholar] [CrossRef] [Green Version]
  47. El-Diasty, M.; Wareth, G.; Melzer, F.; Mustafa, S.; Sprague, L.D.; Neubauer, H. Isolation of Brucella abortus and Brucella melitensis from Seronegative Cows is a Serious Impediment in Brucellosis Control. Vet. Sci. 2018, 5, 28. [Google Scholar] [CrossRef] [Green Version]
  48. Musallam, I.; Abo-Shehada, M.; Hegazy, Y.; Holt, H.; Guitian, F. Systematic review of brucellosis in the Middle East: Disease frequency in ruminants and humans and risk factors for human infection. Epidemiol. Infect. 2016, 144, 671–685. [Google Scholar] [CrossRef] [PubMed]
  49. El-Hady, A.M.; Sayed-Ahmed, M.; Saleh, M.E.; Younis, E.E. Seroprevalence and molecular epidemiology of brucellosis in cattle in Egypt. Adv. Dairy Res. 2016, 4, 1–4. [Google Scholar]
  50. Kolo, F.B.; Fasina, F.O.; Ledwaba, B.; Glover, B.; Dogonyaro, B.B.; van Heerden, H.; Adesiyun, A.A.; Katsande, T.C.; Matle, I.; Gelaw, A.K. Isolation of Brucella melitensis from cattle in South Africa. Vet. Rec. 2018, 182, 668. [Google Scholar] [CrossRef]
  51. Zowghi, E.; Ebadi, A.; Yarahmadi, M. Isolation and identification of Brucella organisms in Iran. Iran. J. Clin. Infect. Dis. 2008, 3, 185–188. [Google Scholar]
  52. Jiang, H.; Fan, M.; Chen, J.; Mi, J.; Yu, R.; Zhao, H.; Piao, D.; Ke, C.; Deng, X.; Tian, G. MLVA genotyping of Chinese human Brucella melitensis biovar 1, 2 and 3 isolates. BMC Microbiol. 2011, 11, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Xiao, P.; Yang, H.; Di, D.; Piao, D.; Zhang, Q.; Hao, R.; Yao, S.; Zhao, R.; Zhang, F.; Tian, G. Genotyping of human Brucella melitensis biovar 3 isolated from Shanxi Province in China by MLVA16 and HOOF. PLoS ONE 2015, 10, e0115932. [Google Scholar] [CrossRef]
  54. Zhong, Z.; Yu, S.; Wang, X.; Dong, S.; Xu, J.; Wang, Y.; Chen, Z.; Ren, Z.; Peng, G. Human brucellosis in the People’s Republic of China during 2005–2010. Int. J. Infect. Dis. 2013, 17, e289–e292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Hasanjani, R.M.; ESMAEILNEZHAD, G.S.; Janmohammadi, N. Update on the treatment of adult cases of human brucellosis. Iran. J. Clin. Infect. Dis. 2008, 3, 167–173. [Google Scholar]
  56. Alavi, S.M.; Alavi, L. Treatment of brucellosis: A systematic review of studies in recent twenty years. Casp. J. Intern. Med. 2013, 4, 636. [Google Scholar]
  57. Roushan, M.R.H.; Baiani, M.; Asnafi, N.; Saedi, F. Outcomes of 19 pregnant women with brucellosis in Babol, northern Iran. Trans. R. Soc. Trop. Med. Hyg. 2011, 105, 540–542. [Google Scholar] [CrossRef]
  58. Bosilkovski, M.; Arapović, J.; Keramat, F. Human brucellosis in pregnancy-an overview. Bosn. J. Basic Med. Sci. 2020, 20, 415–422. [Google Scholar] [CrossRef]
  59. Dal, T.; Durmaz, R.; Ceylan, A.; Bacalan, F.; Karagoz, A.; Celebi, B.; Yasar, E.; Kilic, S.; Acikgoz, C. Molecular investigation of the transmission dynamics of brucellosis observed among children in the province of South-East Anatolia, Turkey. Jundishapur J. Microbiol. 2018, 11, e58857. [Google Scholar] [CrossRef] [Green Version]
  60. Qadri, H.; Ueno, Y. Susceptibility of Brucella melitensis to the new fluoroquinolone PD 131628: Comparison with other drugs. Chemotherapy 1993, 39, 128–131. [Google Scholar] [CrossRef]
  61. Liu, Z.G.; Di, D.D.; Wang, M.; Liu, R.H.; Zhao, H.Y.; Piao, D.R.; Zhao, Z.Z.; Hao, Y.Q.; Du, Y.N.; Jiang, H.; et al. In vitro antimicrobial susceptibility testing of human Brucella melitensis isolates from Ulanqab of Inner Mongolia, China. BMC Infect. Dis. 2018, 18, 43. [Google Scholar] [CrossRef] [Green Version]
  62. Wareth, G.; Dadar, M.; Ali, H.; Hamdy, M.E.R.; Al-Talhy, A.M.; Elkharsawi, A.R.; Tawab, A.; Neubauer, H. The perspective of antibiotic therapeutic challenges of brucellosis in the Middle East and North African countries: Current situation and therapeutic management. Transbound. Emerg. Dis. 2022, 69, e1253–e1268. [Google Scholar] [CrossRef]
  63. Farazi, A.; Hoseini, S.; Ghaznavirad, E.; Sadekhoo, S. Antibiotic Susceptibility of Brucella Melitensis in Markazi Province of Iran. Int. J. Infect. Dis. 2018, 73, 124. [Google Scholar] [CrossRef]
  64. Al Shaalan, M.; Memish, Z.A.; Al Mahmoud, S.; Alomari, A.; Khan, M.Y.; Almuneef, M.; Alalola, S. Brucellosis in children: Clinical observations in 115 cases. Int. J. Infect. Dis. 2002, 6, 182–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Deshmukh, A.; Hagen, F.; Al Sharabasi, O.; Abraham, M.; Wilson, G.; Doiphode, S.; Al Maslamani, M.; Meis, J.F. In vitro antimicrobial susceptibility testing of human Brucella melitensis isolates from Qatar between 2014–2015. BMC Microbiol. 2015, 15, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Parlak, M.; Güdücüoğlu, H.; Bayram, Y.; Çıkman, A.; Aypak, C.; Kılıç, S.; Berktaş, M. Identification and determination of antibiotic susceptibilities of Brucella strains isolated from patients in van, Turkey by conventional and molecular methods. Int. J. Med. Sci. 2013, 10, 1406. [Google Scholar] [CrossRef] [Green Version]
  67. Hashemi, S.; Alikhani, M.-Y.; Asadi, F.T.; Naseri, Z. Antibiotic susceptibility pattern of Brucella melitensis clinical isolates in Hamedan, West of Iran. Int. J. Infect. Dis. 2016, 45, 91–92. [Google Scholar] [CrossRef] [Green Version]
  68. Irajian, G.R.; Jazi, F.M.; Mirnejad, R.; Piranfar, V. Species-specific PCR for the diagnosis and determination of antibiotic susceptibilities of brucella strains isolated from Tehran, Iran. Iran. J. Pathol. 2016, 11, 238. [Google Scholar]
  69. Nishi, H.; Komatsuzawa, H.; Fujiwara, T.; McCallum, N.; Sugai, M. Reduced content of lysyl-phosphatidylglycerol in the cytoplasmic membrane affects susceptibility to moenomycin, as well as vancomycin, gentamicin, and antimicrobial peptides, in Staphylococcus aureus. Antimicrob. Agents Chemother. 2004, 48, 4800–4807. [Google Scholar] [CrossRef] [Green Version]
  70. Martin, F.A.; Posadas, D.M.; Carrica, M.C.; Cravero, S.L.; O’Callaghan, D.; Zorreguieta, A. Interplay between two RND systems mediating antimicrobial resistance in Brucella suis. J. Bacteriol. 2009, 191, 2530–2540. [Google Scholar] [CrossRef] [Green Version]
  71. Wareth, G.; Neubauer, H.; Sprague, L.D. A silent network’s resounding success: How mutations of core metabolic genes confer antibiotic resistance. Signal Transduct. Target. Ther. 2021, 6, 301. [Google Scholar] [CrossRef]
  72. Liu, Z.G.; Cao, X.A.; Wang, M.; Piao, D.R.; Zhao, H.Y.; Cui, B.Y.; Jiang, H.; Li, Z.J. Whole-genome sequencing of a Brucella melitensis strain (BMWS93) isolated from a bank clerk and exhibiting complete resistance to rifampin. Microbiol. Resour. Announc. 2019, 8, e01645-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Singh, B.; Singh, K.; Singh, S.; Agrawal, R.; Agri, H. Antimicrobial susceptibility pattern of Brucella Isolates from abortion cases in animals in Northern India. Austin J. Vet. Sci. Anim. Husb. 2019, 6, 1062. [Google Scholar]
  74. Głowacka, P.; Żakowska, D.; Naylor, K.; Niemcewicz, M.; Bielawska-Drózd, A. Brucella-virulence factors, pathogenesis and treatment. Pol. J. Microbiol. 2018, 67, 151–161. [Google Scholar] [CrossRef]
  75. Hashemifar, I.; Yadegar, A.; Jazi, F.M.; Amirmozafari, N. Molecular prevalence of putative virulence-associated genes in Brucella melitensis and Brucella abortus isolates from human and livestock specimens in Iran. Microb. Pathog. 2017, 105, 334–339. [Google Scholar] [CrossRef] [PubMed]
  76. Naseri, Z.; Alikhani, M.Y.; Hashemi, S.H.; Kamarehei, F.; Arabestani, M.R. Prevalence of the most common virulence-associated genes among Brucella Melitensis isolates from human blood cultures in Hamadan Province, West of Iran. Iran. J. Med. Sci. 2016, 41, 422. [Google Scholar]
  77. Mirnejad, R.; Jazi, F.M.; Mostafaei, S.; Sedighi, M. Molecular investigation of virulence factors of Brucella melitensis and Brucella abortus strains isolated from clinical and non-clinical samples. Microb. Pathog. 2017, 109, 8–14. [Google Scholar] [CrossRef] [PubMed]
Table 1. Molecular characterization of B. melitensis and B. abortus isolates from humans and ruminants in Iran.
Table 1. Molecular characterization of B. melitensis and B. abortus isolates from humans and ruminants in Iran.
IDHostSourceYearLocationDescriptionBiotypingPCRWGS
RAZI20Y0140humanblood2015Alborz♂, farmerB. abortusB. abortusB. abortus
RAZI20Y0141humanblood2015Tehran♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0142humanblood2016Alborz♀, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0143humanblood2017Tehran♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0144humanblood2018Kermanshah♀, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0145humanCNS2015Alborz♀, retiredB. melitensisB. melitensisB. melitensis
RAZI20Y0146humanblood2020Tehran♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0147humanblood2015Kerman♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0148humanblood2020Alborz♂, teacherB. melitensisB. melitensisB. melitensis
RAZI20Y0149cowmilk2015QomabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0150cowmilk2017TehranabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0151cowmilk2016QomabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0152camelmilk2017TehranseropositiveB. melitensisB. melitensisB. melitensis
RAZI20Y0153cowmilk2018YazdabortionB. abortusB. abortusB. abortus
RAZI20Y0154cowmilk2019FarsabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0155sheepmilk2018MazandaranabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0156cowmilk2019FarsabortionB. abortusB. abortusB. abortus
RAZI20Y0157humanblood2018Kermanshah♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0158humanblood2018Kermanshah♀, house-
keeper
B. melitensisB. melitensisB. melitensis
RAZI20Y0159humanblood2019Alborz♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0160humanblood2019Kermanshah♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0161humanblood2019Alborz♀, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0162humanblood2019Tehran♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0163humanblood2019Hamedan♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0164humanblood2019Hamedan♀, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0165humanblood2019Hamedan♀, house-
keeper
B. melitensisB. melitensisB. melitensis
RAZI20Y0166humanblood2019Kermanshah♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0167humanblood2019Alborz♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0168humanblood2019Alborz♀, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0169humanblood2019Kermanshah♂, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0170humanblood2019Tehran♀, farmerB. melitensisB. melitensisB. melitensis
RAZI20Y0171sheepaborted fetus2020FarsabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0172sheepaborted fetus2020YazdabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0173cowL.N2020FarsabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0174cowL.N2019SemnanabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0175cowL.N2019IsfahanabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0176sheepaborted fetus2019ZanjanabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0177goataborted fetus2019AlborzabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0178sheepaborted fetus2020FarsabortionB. melitensisB. melitensisB. melitensis
RAZI20Y0179camelL.N2020HormozganseropositiveB. melitensisB. melitensisB. melitensis
Male ♂; Female ♀.
Table 2. MIC values of antibiotics against human and ruminant Brucella isolates using E-test.
Table 2. MIC values of antibiotics against human and ruminant Brucella isolates using E-test.
Antibiotics MIC Range
(μg/mL)
MIC Range 50
(μg/mL)
MIC Range 90
(μg/mL)
MIC Interpretive Criteria (μg/mL)
SRI
Ceftriaxone0.032–10.250.75≤2--
Imipenem1.5–824≤4--
Doxycycline0.032–0.1250.0640.094≤48≥16
Rifampicin0.047–0.750.380.5≤12≥4
Streptomycin0.094–0.750.380.5≤8--
ColistinRRRND--
Trimethoprim-Sulfamethoxazole0.023–0.0640.0470.064≤0.51–2≥4
Gentamycin0.094–10.380.75≤4--
Ampicillin-sulbactam0.25–31.52≤12≥4
Standard breakpoints according to the guidelines for slowly growing bacteria (Haemophilus spp.) from CLSI S: Sensitive; I: Intermediate, and R: Resistant. ND: not described by CLSI standards.
Table 3. Antibiotic susceptibility testing of human and ruminant Brucella isolates using disk diffusion testing.
Table 3. Antibiotic susceptibility testing of human and ruminant Brucella isolates using disk diffusion testing.
Antibiotics Concentration
μg/disk
Range
(mm)
Sensitive
no (%)
Intermediate
no (%)
Resistant
no (%)
Resistance Pattern
SIR
Ceftriaxone30 μg25–6240 (100)00≥26NDND
Imipenem10 μg21–3940 (100)00≥16NDND
Doxycycline30 μg29–4840 (100)0010≥NDND
Rifampicin5 μg15–3320 (50)12 (30%)8 (20%)≥2017–19≤16
Streptomycin10 μg18–4140 (100)0080≥NDND
Colistin10 μg00040 (100%)NDNDND
Trimethoprim-Sulfamethoxazole1.25/23.75 μg15–3539 (97.5)1 (2.5%)0≥1611–15≤10
Gentamicin10 μg22–4540 (100)00≥16NDND
Ampicillin-sulbactam20 μg13–4526 (65)1 (2.5%)13 (32.5%)≥20ND≤19
ND: not determined by CLSI standards. S, Sensitive; I, Intermediate and R, Resistant.
Table 4. Associated virulence and pathogenicity factors found in all Iranian Brucella genomes.
Table 4. Associated virulence and pathogenicity factors found in all Iranian Brucella genomes.
Virulence and Pathogenicity FactorsRelated Genes
LPS (lipopolysaccharide) pathogenicity factors, entry, intracellular
survival and immunomodulatory
acpXL, fabZ, gmd, htrB, kdsA, kdsB, lpsA, lpsB. lpcC, lpxA, lpxB, lpxC, lpxD, lpxE, manAoAg, manCoAg, per, pgm, pmm, wbdA, wbkA, wbkB, wbkC, wboA, wbpL, wbpZ, wzm, wzt.
Type IV secretion system
effector secretion
virB1, virB2, virB3, virB4, virB5, virB6, virB7, virB8, virB9, virB10, virB11, virB12.
TIR domain-containing protein
immune evasion
btpA, btpB
Rab2 interacting conserved protein A
intracellular survival
RicA
CβG (cyclic β-1,2 glucan)
intracellular survival
Cgs
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dadar, M.; Alamian, S.; Brangsch, H.; Elbadawy, M.; Elkharsawi, A.R.; Neubauer, H.; Wareth, G. Determination of Virulence-Associated Genes and Antimicrobial Resistance Profiles in Brucella Isolates Recovered from Humans and Animals in Iran Using NGS Technology. Pathogens 2023, 12, 82. https://0-doi-org.brum.beds.ac.uk/10.3390/pathogens12010082

AMA Style

Dadar M, Alamian S, Brangsch H, Elbadawy M, Elkharsawi AR, Neubauer H, Wareth G. Determination of Virulence-Associated Genes and Antimicrobial Resistance Profiles in Brucella Isolates Recovered from Humans and Animals in Iran Using NGS Technology. Pathogens. 2023; 12(1):82. https://0-doi-org.brum.beds.ac.uk/10.3390/pathogens12010082

Chicago/Turabian Style

Dadar, Maryam, Saeed Alamian, Hanka Brangsch, Mohamed Elbadawy, Ahmed R. Elkharsawi, Heinrich Neubauer, and Gamal Wareth. 2023. "Determination of Virulence-Associated Genes and Antimicrobial Resistance Profiles in Brucella Isolates Recovered from Humans and Animals in Iran Using NGS Technology" Pathogens 12, no. 1: 82. https://0-doi-org.brum.beds.ac.uk/10.3390/pathogens12010082

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