Open access peer-reviewed chapter

The Evolution and Dissemination of Methicillin Resistance Determinant in Staphylococcus aureus

Written By

Abdul Rahim Abdul Rachman, Zarizal Suhaili and Mohd Nasir Mohd Desa

Submitted: 21 April 2016 Reviewed: 05 September 2016 Published: 08 March 2017

DOI: 10.5772/65514

From the Edited Volume

The Rise of Virulence and Antibiotic Resistance in Staphylococcus aureus

Edited by Shymaa Enany and Laura E. Crotty Alexander

Chapter metrics overview

2,157 Chapter Downloads

View Full Metrics

Abstract

Staphylococcus aureus is an opportunistic pathogen and is frequently associated with the antimicrobial resistance. There has been horizontal gene transfer of Staphylococcus chromosome cassette mec (SCCmec) among the staphylococcal species that colonize a similar colonization niche, which eventually results in emergence of new variant with enhanced survival ability in terms of antimicrobial resistance and virulence level in S. aureus. Evolution and dissemination of SCCmec structure resulted in the emergence of methicillin-resistant S. aureus (MRSA) clones around the world covering hospital, community, and livestock settings. MRSA also has the ability to resist different antibiotic profiles known as multidrug-resistant S. aureus (MDR S. aureus).

Keywords

  • Staphylococcus aureus
  • SCCmec
  • MRSA clones
  • multi-drug-resistant S. aureus

1. Introduction

Staphylococcus aureus is an opportunistic pathogen and lives as part of the animal normal flora of skin and nasopharynx. Favorably, it resides in the nasal mucosal environment posing infection threat to human as well as in domestic animals [1, 2]. In human, it is the leading agent of infection involving bloodstream, skin, and soft tissue to the lower respiratory tract [35]. S. aureus can easily colonize certain part of the body, especially the exposed area on skin due to ulcers, burns, and surgical wounds [6].

Methicillin-resistant S. aureus (MRSA) has been well known for being resistant to β-lactam antibiotics, which are the most common antimicrobial agents used to fight staphylococcal infection. Previous studies reported that methicillin resistance in staphylococci was carried by a specific mobile genetic element (MGE) called staphylococcal chromosome cassette mec (SCCmec), which carries with it several virulence factors as well [7]. SCCmec contains mecA gene which encodes for a low affinity penicillin-binding protein (pbp2a or pbp2′), which is currently exploited as the methicillin resistance marker in Staphylococcus species including S. aureus [8]. SCCmec contains several elements that can be categorized into several types. Genetic events such as point mutation, recombination, acquisition, and deletion, coupled with host and environmental selective pressures, make the structure evolve and disseminate in the population [9]. The emergence of certain MRSA clones, which have been disseminating worldwide since 1960, was closely related to the continuous evolution of SCCmec structure in S. aureus.

Multidrug-resistant MRSAs have also been reported that make the antibiotic regiment limited. Prevalence of MRSA is of a growing concern, particularly due to the more recent increased frequency of community-associated MRSA (CA-MRSA) and livestock-associated MRSA (LA-MRSA). Thus, this pathogen becomes a major concern in public health as well as livestock industry [1013].

Many studies have been looking at the mechanism and dissemination pattern of MRSA and its genetic characteristics, but due to the potential, geographical, and temporal differences, a comprehensive review is needed to put the whole picture connected.

Advertisement

2. Staphylococcus aureus

S. aureus is a Gram-positive bacterium with a grape-like cluster morphology and can usually be found in skin or mucous membrane, especially in nasal of healthy person [14]. Kluytmans et al. reported that approximately 20–30% of human population carries S. aureus [15]. Morphologically, S. aureus can be observed as a ’golden’ medium-size colony on solid media such as nutrient agar and can cause β-hemolysis on sheep blood agar [16].

The production of golden pigmentation of S. aureus colonies is closely related to the presence of carotenoids which is previously reported as virulence factor protecting S. aureus from the immune system [17]. Among Staphylococcus species, only S. aureus has the ability to ferment mannitol leading to the production of lactic acid on mannitol salt agar with yellow zones around the colonies [18]. S. aureus is also classified as a halophilic bacterium for being able to live in the presence of salt (sodium chloride) up to 1.7 molar. It also produces coagulase that causes blood to clot [14].

Generally, 20–30% of individuals are persistent carriers of S. aureus and 30% are transient or intermittent carriers [19]. S. aureus may live in human without any clinical symptoms, but it may infect the host when the host defense system is compromised. Individuals may acquire infection by S. aureus that they previously carry as commensal [15].

Immunocompromised patients with S. aureus infection may suffer several diseases such as bacteremia, ventilator-assisted pneumonia (VAP), endocarditis, and osteomyelitis, especially when the patients are frequently exposed to injections and catheter insertions [20, 21]. S. aureus can also cause toxin-mediated disease such as toxic shock syndrome, scalded skin syndrome, and Staphylococcal foodborne diseases (SFD) [21]. Frequently, S. aureus is the main cause of skin and soft tissue infection (SSTI) in human [22].

Advertisement

3. Methicillin-resistant S. aureus (MRSA)

MRSA has the ability to resist almost all available β-lactam antibiotics. Statistics showed about 40–70% of S. aureus nosocomial infections worldwide are caused by MRSA. MRSA was first reported in a hospital in the United Kingdom in 1961 after the introduction of methicillin to treat patient with penicillin-resistant Staphylococcus infection [23].

Generally, MRSA can be categorized into two major groups known as hospital-associated MRSA (HA-MRSA) and community-associated MRSA (CA-MRSA). Globally, the majority of MRSA infections are HA-MRSA that are acquired from healthcare facilities. Currently, MRSA isolates are subdivided into three major groups known as hospital-associated MRSA (HA-MRSA), community-associated MRSA (CA-MRSA), and livestock-associated MRSA (LA-MRSA). Previous reports revealed that both HA-MRSA and CA-MRSA isolates differ distinctly from each other, with HA-MRSA showing high antimicrobial resistance but less virulence and lack of capabilities as colonizers [2326]. Meanwhile, CA-MRSA isolates exhibit a low antimicrobial resistance but a high virulence harboring PVL gene and numerous pathogenicity factors, as well as good colonizers [2629].

MRSA spread in population since 1990 and become the major cause of community-associated infection [27]. The scenario worsens when multidrug MRSA emerges, in which it can resist more than two antibiotics of different classes that reduce the option for available treatment of Staphylococcal infection [30, 31].

Methicillin resistance characteristic in S. aureus is due to the presence of altered penicillin-binding protein (PBP2a) in the cell wall that has a reduced binding affinity to β-lactam antibiotics. PBP2a is encoded by mecA gene that is located in the large chromosomal cassette called staphylococcal chromosome cassette mec element (SCCmec) [3235]. The mecA gene expression is controlled by mecI-mecRI regulatory genes encoding repressor and inducer protein, respectively [36].

Advertisement

4. SCCmec structure

Staphylococcal cassette chromosome mec (SCCmec) has a size of about 20–60 kb. The structure is unique as it carries various mobile genetic elements that are integrated in it [37]. To date, more than 80 SCCmec elements have been identified in several staphylococci species [38]. SCCmec disseminates among Staphylococcal species by horizontal gene transfer and integrates at a specific site called attB or ISS (integration site sequence) at the 3′ end of orfX gene that encodes for unknown function [39].

A single SCCmec carries mec complex and cassette chromosome recombinase (ccr) flanked by direct inverted repeat (DR) and inverted repeat (IR) sequences; mec complex consists of mecA gene (methicillin resistance determinant), mecRI (sensor inducer), and mecI (mec repressor). Both mecRI and mecI are recognized as mec regulator elements, while ccr genes encode serine recombinases (ccrA, ccrB, ccrC) responsible for site and orientation of specific integration and excision of SCCmec. In addition, SCCmec also harbors other elements such as insertion sequences (IS), plasmids, and transposons [24, 40, 41].

4.1. Complete SCCmec

To date, the International Working Group-SCCmec (IWG-SCCmec) identified eleven SCCmec types based on complete nucleotide sequences in Staphylococcal databases, and each SCCmec type is named using a roman numeral based on the unique combination of ccr complex and mec complex [4042]. A complete SCCmec structure in S. aureus contains a mec complex (mecA, mecRI and mecI), a ccr complex (ccrA, ccrB, ccrC), and a J region (region other than mec and ccr complexes) [40].

Furthermore, many different SCCmec subtypes have also been described containing the same ccr and mec gene combination but vary in the J regions [40]. Among the eleven SCCmec types (I–XI) that have been reported so far, five of them (SCCmec I, II, III IV, and V) are globally distributed, while others only distributed in certain countries [38, 43]. Three (SCCmec IVa, SCCmec IVc, SCCmec V) from the 11 SCCmec types have been detected in MRSA isolated from animals called LA-MRSA [42]. In general, SCCmec type IV and V are more widely found among CA-MRSA, and the other three types (SCCmec I, II, III) are frequently found among HA-MRSA [44, 45, 46]. An early study by Ito et al. detected only three types of SCCmec structures (SCCmec type I, II, III) isolated from human [37], and a recent finding showed that MRSA with SCCmec type I, II, III is originated from animals [41].

Different types of SCCmec in MRSA are also observed to be geographically distributed. For example, SCCmec type III or IIIA was most commonly found in Asian countries, but Korea and Japan had more type II while Taiwan had more type IV [47]. SCCmec type IV was also commonly found in Latin and European countries [48, 49]. Similarly, in African countries, SCCmec type III was also predominant with SCCmec types II, IV, and V found in selected countries such as Egypt, Niger, Nigeria, Algeria, Tunisia, and South Africa [50].

4.2. Pseudo-SCCmec

Pseudo-SCCmec is recognized as SCCmec that does not carry ccr complexes but has mecA gene. Although this element is different from the complete SCCmec in terms of gene or operon organization, it still has some similarities in certain parts in both pseudo-SCCmec and complete SCCmec structure. Deletion is the major event as inferred by the absence of certain genes or operon in pseudo-SCCmec structure. For example, regions within mec complex and J region are absent in both pseudo-SCCmec II.5 and pseudo-SCCmec16691. It was observed that pseudo-SCCmec16691 lacks J1, J2 regions, and ccr genes, whereas missing parts were detected in pseudo-SCCmec II.5 and replaced by transposable elements called Tn6012 [51, 52].

However, certain pseudo-SCCmec does not carry both mec and ccr complexes. An example is the arginine catabolic mobile element (ACME) for having SCC-like elements but lack in mecA and ccr genes. This could be the remnant of SCCmec structure that had gone through multiple mutational events. Lindqvist et al. discovered first remnant of pseudo-SCCmec structure in methicillin susceptible S. aureus (MSSA) that caused clonal outbreak in Sweden. They suggested that this pseudo-SCCmec structure could be derived from SCCmec type II [52].

ACME is found in both MRSA and MSSA, especially with sequence type ST8 (ST8), and has been disseminated in virulent S. aureus by horizontal gene transfer [3, 53]. Nevertheless, ACME was frequently associated with MRSA-IVa with sequence type 8 (ST8-MRSA-IVa), which was also known as CA-MRSA USA300 [3, 53]. ACME has been associated with the ability of CA-MRSA to colonize on other parts of human body such as skin and mucosal membranes rather than limited to only nostril. The acquisition of ACME may enhance the ability of CA-MRSA to survive in acidic environment of human skin by driving production of polyamine-resistant enzyme that combats excess host polyamine (toxic compound on human skin for S. aureus) [54].

Advertisement

5. Origin of SCCmec structure

The origin of SCCmec in MRSA is still in debate; mecA gene was believed to be originated from Staphylococcus fleurettii due to a high sequence similarity (>99%) with mecA gene of a MRSA strain N315. It was proposed that SCCmec is a combination of SCC elements without mec complex, and the mec gene complex was derived from S. fleurettii since no evidence showed that S. fleurettii contained SCCmec structure in its chromosome [55].

Several studies described coagulase-negative staphylococci (CoNS) as the primary reservoir of the SCCmec structure in S. aureus, which was considered as the recipient strain due to some reasons; a very similar SCCmec structure and organization was observed in both S. aureus and CoNS [56, 57], and the prevalence of methicillin-resistant coagulase-negative staphylococci (MRCoNS) in human is higher as compared to MRSA [35, 5659]. Although a study discovered other non-staphylococci species called Macrococcus to also carry SCCmec-like elements, those were different with SCCmec in MRSA in terms of nucleotide sequences and genetic organization of the mec complex [55].

5.1. From coagulase-negative staphylococci species to MRSA

The existence of various forms of SCCmec in MRCoNS as compared to MRSA becomes the main argument why MRCoNS is suggested as the main reservoir of SCCmec for S. aureus leading to the emergence of MRSA [40, 57]. In a rapid genetic typing, polymerase chain reaction (PCR) technique is used to characterize the SCCmec types instead of nucleotide sequencing analysis. Consequently, SCCmec from MRCoNS is frequently defined as non-typeable due to a diverse combination of ccr and mec complexes that could not be assigned based on current SCCmec structure databases used against S. aureus [7]. Nevertheless, Zong et al. successfully assigned 10 SCCmec elements with a new combination of ccr and mec complexes in various species of MRCoNS. They assigned these untypeable SCCmec elements as UT1–UT10 [35]. In addition, another study also described new SCCmec types in Staphylococcus hominis and described those as NT1till NT4 [60].

Staphylococcus epidermidis, Staphylococcus haemolyticus, and S. hominis were found to carry a diverse SCCmec structure among CoNS. SCCmec type IV is the common structure found in S. epidermidis, while other SCCmec types I, II, III, V, VI and non-typeable SCCmec were also detected at a lower rate [61, 62]. For S. haemolyticus, SCCmec type V predominated in combination with other novel SCCmec types [60, 63]. In S. hominis, SCCmec types contained a combination of novel non-typeable SCCmec, SCCmec types VI, VIII, III, and other elements [35, 61].

5.2. From MSSA to MRSA

MRSA emerges when MSSA receives SCCmec structure elements from other MRSA or MRCoNS via horizontal gene transfer [64]. In a specific condition (high vancomycin concentration), SCCmec is unstable in certain MRSA that can lead to complete or partial deletion of SCCmec structure, which may result in the presence of certain SCCmec DNA fragment to remain in S. aureus chromosome [6467].

Wong et al. [64] identified SCCmec type II with internal deletion in MSSA isolates from different geographical areas. This happened during in vitro exposure to vancomycin [64]. Furthermore, Vandendriessche et al. [67] described MSSA CC398 as the precursor for emergence of MRSA CC398 in livestock. They found non-SCCmec elements in MSSA CC398 harboring czrC and tet(K) genes generated during partial excision of SCCmec elements [67].

Advertisement

6. Clonal dissemination of MRSA

Nowadays, the dissemination of MRSA has become a major global problem that threatens human health [27]. However, only limited clones of MRSAs could be inferred to disseminate in different countries and continents through genotypic analysis using several DNA typing methods such as SCCmec typing, PFGE, MLST, and spa typing [27, 68, 69]. For example, more than 3000 MRSA isolates from certain continents (Europe, USA, and South America) were described to belong to only five major pandemic clones or clonal complexes (CC5, CC8, CC22, CC30, and CC45) [70]. To date, 11 clonal complexes (CC1, CC5, CC8, CC12, CC15, CC22, CC25, CC30, CC45, CC51, and CC121) have been detected in which 5 of them (CC8, CC15, CC22, CC30, and CC45) were isolated from human [71, 72]. These successful clones may transmit their genetic elements into other S. aureus, which are well adapted to hospital environment [73].

MRSA strain COL was the first MRSA clone detected carrying SCCmec type I with sequence type 250 (ST 250) and belonged to clonal complex 8 (CC8). Then, other MRSA clones with SCCmec type II and III were reported and recognized as EMRSA-1 (ST239), EMRSA-5 (ST247), and New York/Japan clone (ST5, USA100) [74]. Certain MRSA clones were originated from community setting. For example, Wang and co-workers (2007) detected the spread of community-associated SCCmec type IV and V MRSA in hospital setting in Taiwan between 1999 and 2005. They concluded that SCCmec types IV and V are carried by both CA-MRSA and HA-MRSA [7577].

The popular human MRSA pandemic clones, the EMRSA-15 and EMRSA-16, were identified in the United Kingdom (UK) around early 1990s. Since then, the clones become predominant healthcare-associated MRSA in UK [78, 79] and several European countries such as Denmark [80], Sweden [81], Belgium [79], and Spain [82]. Studies in Kuwait [83] and USA [84] also reported the spread of EMRSA-15 and 16 clones in hospital setting in the countries. To date, these clones have already been widespread in 15 countries around the world [85]. Both MRSAs belong to SCCmec type IV with sequence type 22 (ST 22) for EMRSA-15 and sequence type 30 (ST 30) for EMRSA-16 and originated from hospital setting. EMRSA-15 and 16 have high surviving and spreading rate in hospital compared to other EMRSA in UK [78]. In 2013, MRSA clone with a rare sequence type, ST 779, was identified in eleven Irish hospitals from 2006 until 2011 harboring a novel pseudo (SCCmec)-SCC-SCCCRISPR composite element. This clone contained novel mec class region, a fusidic acid resistance gene (fusC), and two copper resistance genes (copB and copC) but lacking ccr genes [86].

CA-MRSA clones have also been observed to disseminate worldwide particularly with sequence types ST80 and ST30. MRSA clone with ST80 is the most common CA-MRSA clone in European countries and usually carries PVL genes. Moreover, ST80 clone also showed resistance toward fluoroquinolones, tetracyclines, and fusidic acid [87]. CA-MRSA clone with sequence type ST30 was observed to disseminate in Asian and Oceanic countries. An example is the multidrug USA300 clone, known as West Pacific clone. It was first identified in the USA and carried plasmid that encodes several antibiotic resistance genes [88]. Enany et al. identified novel clones with sequence types ST1010 (121)c and ST1009 (1153)c isolated from Egypt after they analyzed different genetic patterns of PVL+CA-MRSA isolates from different countries [89].

In certain countries, it was found that MRSA can also spread among livestock, known as LA-MRSA. LA-MRSA CC398 is the popular clonal complex among livestock and has already been reported to spread in several European farms in Netherland, Denmark, Germany, France, and Italy [90]. MRSA CC398 was originated from pigs and spread among dairy cattle and turkey [91, 92]. In Netherlands, MRSA contamination on meat was reported after 2217 meat samples were analyzed covering 35.3% turkey, 15.2% beef, 15.2% veal, 10.7% pork, and 6.2% lamb meat [93]. LA-MRSA can be transmitted to human by physical contact with livestock contaminated with MRSA [94]. LA-MRSA may have equal virulence ability as compared to CA-MRSA and HA-MRSA toward human. Therefore, persons with continuous exposure to livestock carrying LA-MRSA are at high risk [95]. Other than meat, LA-MRSA can also be found in dairy milk. Recently, 11 sequence types were detected from LA-MRSA isolated from 15 Brazilian dairy farms (n = 552) with four of them contain novel sequence types (ST1622, ST1623, ST1624, and ST1625) [96].

Advertisement

7. Multidrug-resistant (MDR) MRSA

Antibiotic or antimicrobial drugs are the most effective therapeutic agents used in treating microbial infections through either one or both bactericidal and bacteriostatic effects. Nevertheless, antibiotic or antimicrobial drug resistance has been a major problem worldwide, with incidence of MRSA reported in healthcare facilities in Asia to reach its peak in late 1990s, and stayed at plateau level during 2000s [97]. The heavy usage of drugs in treatment hastens the selection of bacteria that harbor multidrug resistance genes particularly S. aureus to proliferate and dominate [98, 99]. Moreover, over-crowded community creates environment that is suitable for the rapid spread of numerous multidrug-resistant pathogens, particularly the airborne organisms such as S. aureus.

The emergence of multidrug-resistant S. aureus in both hospitals and community invokes a tremendous financial burden due to the persistence of hard-to-treat infections [97, 100102]. Until present, it was reported that <90% of S. aureus strains are resistant to penicillin as well as ordinary antimicrobial agents such as drug from categories of aminoglycosides, ansamycins, anti-staphylococcal β-lactams (or cephamycins), chloramphenicols, fusidanases, fluoroquinolones, glycopeptides, lincosamides, macrolides, phenicols, and tetracyclines [103105]. We are now observing the emergence of multidrug-resistant S. aureus and MDR-MRSA with broad spectrum of resistance with a distinct ability to survive and spread in the hospital environment, community setting, as well as livestock sectors.

There has been a dramatic increase in the incidence of nosocomial infections as well as community-associated MRSA and livestock-associated MRSA caused by strains of S. aureus that are resistant to multiple antibiotics [106]. At present, there have been reports that some strains demonstrate resistance to as many as 20 antimicrobial compound types, including antiseptics and disinfectants [107, 108]. Central Asian surveillance studies found that the prevalence of MRSA infection in tertiary hospital was reported in 10 among 1000 hospital admissions [109] and incidence reported previously in Japan was between 0.7 and 0.8 per 100 admission from 1999 to 2003 with a total rate among hospitalized patients in the Asia-Pacific region at 45.9% [110, 111]. Previous surveillance also reported that Asia is among the highest for the incidence of MRSA in the world, and interestingly a novel MRSA strain with glycopeptides resistance had spread in livestock animals making it as a potential human pathogen in this region [112].

Several studies attempted to profile all possible multidrug-resistant MRSA since 1987, encompassing samples from hospitals, community, as well as veterinary settings [113, 114]. Lim et al. (2013) carried out temporal comparative surveillance of antibiograms from clinical samples in 2003–2008 and showed a significant increase in resistance rates (from 1 to 96%), as well as multidrug-resistant phenotypes (96%). This study also indicated the prevalence of multidrug-resistant MRSA with SCCmec type III and ST239 [99]. Another study also reported the prevalence of resistance against other important antibiotics such as mupirocin, whose resistance rate in Malaysia is still low, but still higher than previous reports in Malaysia [107]. Another cross-sectional studies at a few major medical centers in Malaysia found that the occurrence of MRSA infection increased gradually with years, from 25.7 to 28.7% in 1996, 27.9% in 1998, and 33% in 2000 [115, 117, 118]. Meanwhile, a study done at a single Malaysian hospital found a gradual reduction in MRSA prevalence from 2002 to 2006, most likely due to the improvement in the quality of healthcare systems [103, 109, 116 118].

The first international surveillance study on epidemiology of CA-MRSA in Asian countries revealed important findings with regard to the current epidemiology of MRSA infections in the community and hospitals within Asia with multidrug-resistance rates at 73.1 and 83.7% for CA-MRSA and HA-MRSA, respectively [119]. At least, 357 isolates of CA-MRSA were analyzed with resistance rates of gentamicin, ciprofloxacin, and trimethoprim/sulfamethoxazole being significantly lower than those of HA-MRSA isolates, whereas resistance rates of clindamycin, erythromycin, and tetracycline were similarly high in both CA-MRSA and HA-MRSA [119, 120].

Advertisement

8. Conclusion

S. aureus and MRSA evolve and adapt the changing environment. Therefore, dissemination of MRSA should be continuously monitored for the antibiotic susceptibility pattern and molecular epidemiology comprising hospital, community, and livestock settings. The origin and dissemination of SCCmec are also important to be tracked in the diverse staphylococcal population. With the advancement in molecular methods such as next-generation sequencing, the pattern of the genetic evolution, spread of the bacteria, and the resistance determinants can be further explored and understood.

References

  1. 1. Azis MA, AB Hamid A, Pung H., Abdul PA, Suhaili Z, Mohd Nasir MD: Staphylococcus aureus infection risk in a population of health sciences students at a public university. Iranian Journal of Public Health, 2014; 43(3): 112–116.
  2. 2. ChengAG, Kim HK, Burts ML, Krausz T, Schneewind O, Missiakas DM: Genetic requirements for Staphylococcus aureus abscess formation and persistence in host tissues. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 2009; 23(10): 3393–3404. doi:10.1096/fj.09-135467
  3. 3. Diep BA, Sensabaugh GF, Somboonna N, Somboona NS, Carleton HA, Perdreau-Remington F: Widespread skin and soft-tissue infections due to two methicillin-resistant Staphylococcus aureus strains harboring the genes for Panton-Valentine leucocidin. Journal of Clinical Microbiology, 2004; 42(5): 2080–2084. doi:10.1128/JCM.42.5
  4. 4. Forbes, Forbes GB: Infection with penicillin-resistant staphylococci in hospital and general practice. British Medical Journal, 1949; 2(4627): 569–571. doi:10;2(4627)
  5. 5. Gehanno JF, Louvel A, Nouvellon M, Caillard JF,Pestel-Caron M: Aerial dispersal of methicillin-resistant Staphylococcus aureus in hospital rooms by infected or colonised patients. The Journal of Hospital Infection, 2009; 71(3): 256–262. doi:10.1016/j.jhin.2008.11.015
  6. 6. Dryden MS: Skin and soft tissue infection: microbiology and epidemiology. International Journal of Antimicrobial Agents, 2009; 34 Suppl 1(1872–7913 (Electronic): S2–S7. doi:10.1016/S0924-8579(09)70541-2
  7. 7. Shore AC, Coleman DC: Staphylococcal cassette chromosome mec: Recent advances and new insights. International Journal of Medical Microbiology, 2003; 303(6–7): 350–359. doi:10.1016/j.ijmm.2013.02.002
  8. 8. Al-Abbas MA: Antimicrobial susceptibility of Enterococcus faecalis and a novel Plano-microbium isolate of bacteraemia. International Journal of Medicine and Medical Sciences, 2012; 4: 19–27. doi:10.5897/IJMMS11.130
  9. 9. Deurenberg RH, Vink C, Kalenic S, Friedrich AW, Bruggeman CA, Stobberingh EE: The molecular evolution of methicillin-resistant Staphylococcus aureus. Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases, 2007; 13(3): 222–235. doi: 10.1111/j.1469-0691.200 6.01573.x
  10. 10. Graveland H, Duim B, van Duijkeren E, Dick Heederik JAW: Livestock-associated methicillin-resistant in animals and humans. International Journal of Medical Microbiology, 2011; 301(8): 630–634. doi:10.1016/j.ijmm.2011.09.004
  11. 11. Michalopoulos AS, Livaditis IG, Gougoutas V: The revival of fosfomycin. International Journal of Infectious Diseases: IJID: Official Publication of the International Society for Infectious Diseases, 2011; 15(11): e732–739. doi:10.1016/j.ijid.2011.07.007
  12. 12. Roberts MC, Soge OO, No D. Comparison of multi-drug resistant environmental methicillin-resistant Staphylococcus aureus isolated from recreational beaches and high touch surfaces in built environments. Frontiers in Microbiology, 2013; 4: 74. doi:10.3389/fmicb.2013.00074.
  13. 13. Uzunovi S, Ibrahimagi A, Kamberovi F, Rijnders MIA, Stobberingh EE: Molecular characterization of methicillin-susceptible and methicillin-resistant Staphylococcus aureus in food handlers in Bosnia and Herzegovina. The Open Infectious Diseases Journal, 2013; 7: 15–20. doi:10.1007/s10354-012-0142-8
  14. 14. Crossley KB, Archer GL. Jefferson KK: Staphylococci in Human Disease, Hoboken, NJ: John Wiley & Sons, Inc., 2009; doi:10.1002/9781444308464
  15. 15. Kluytmans J, van Belkum A, Verbrugh H: Nasal carriage of Staphylococcus aureus: epidemiology, underlying mechanisms, and associated risks. Clinical Microbiology Reviews, 1997; 10(3): 505–520.
  16. 16. Sasaki T, Kikuchi K, Tanaka Y, Takahashi N, Kamata S. Hiramatsu K: reclassification of phenotypically identified Staphylococcus intermedius strains. Journal of Clinical Microbiology, 2007; 45: 2770–2778. doi:10.1128/JCM.00360-07
  17. 17. Liu D, Chai T, Xia X, Gao Y, Cai Y, Li X., Miao Z, Hao H, Roesler U, Wang, J: Formation and transmission of Staphylococcus aureus (including MRSA) aerosols carrying antibiotic-resistant genes in a poultry farming environment. Science of the Total Environment, 2012; 426: 139–145. doi:10.1016/j.scitotenv.2012.03.060
  18. 18. Pai V, Rao VI, Rao SP: Prevalence and antimicrobial susceptibility pattern of methicillin-resistant Staphylococcus aureus (MRSA) isolates at a tertiary care hospital in Mangalore, South India. Journal of Laboratory Physicians, 2010; 2(2): 82–84. doi:10.4103/0 974-27 27.72155
  19. 19. Wertheim HFL, Melles DC, Vos MC, van Leeuwen W, van Belkum A, Verbrugh HA, Nouwen JL: The role of nasal carriage in Staphylococcus aureus infections. The Lancet. Infectious Diseases, 2005; 5(12): 751–762. doi:10.1016/S1473-3099(05)70295-4
  20. 20. Schito GC. The importance of the development of antibiotic resistance in Staphylococcus aureus. Clinical Microbiology and Infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases, 2006; 12 Suppl 1: 3–8. doi:10.1111/j.1469-0691.2006.01343.x
  21. 21. Lindsay JA, Holden MTG: Understanding the rise of the superbug: Investigation of the evolution and genomic variation of Staphylococcus aureus. Functional and Integrative Genomics, 2006; 6: 186–201. doi:10.1007/s10142-005-0019-7
  22. 22. Diekema DJ, Pfaller MA, Schmitz FJ, Smayevsky J, Bell J, Jones RN, Beach M: Survey of infections due to staphylococcus species: frequency of occurrence and antimicrobial susceptibility of isolates collected in the United States, Canada, Latin America, Europe, and the Western Pacific Region for the SENTRY Antimicrobial Surveillance. Clinical Infectious Diseases, 2001; 32(s2): S114–S132. doi:10.1086/320184
  23. 23. Chambers HF. DeLeo FR: Waves of resistance: Staphylococcus aureus in the antibiotic era. Nature Revision Microbiology. 2009; 7: 629–641. doi:10.1038/nrmicro2200
  24. 24. Chongtrakool P, Ito T, Ma XX, Kondo Y, Trakulsomboon S, Tiensasitorn C, Jamklang M, Chavalit T, Song JH, Hiramatsu K: Staphylococcal cassette chromosome mec (SCCmec) typing of methicillin-resistant Staphylococcus aureus strains isolated in 11 Asian countries: a proposal for a new nomenclature for SCCmec elements. Antimicrobial Agents and Chemotherapy, 2006; 50(3): 1001–1012. doi:10.1128/AAC.50.3.1001-10 12.2006
  25. 25. Afroz S, Kobayashi N, Nagashima S, Alam MM, Hossain AB, Rahman MA, Islam MR, Lutfor AB, Muazzam N, Khan MA, Paul SK, AK, Mahmud MC, Musa AK, Hossain, MA: Genetic characterization of Staphylococcus aureus isolates carrying Panton-Valentine leukocidin genes in Bangladesh. Japanese Journal of Infectious Diseases, 2008; 61(5): 393–396.
  26. 26. Berglund C, Prévost G, Laventie BJ, Keller D, Söderquist B: The genes for Panton Valentine leukocidin (PVL) are conserved in diverse lines of methicillin-resistant and methicillin-susceptible Staphylococcus aureus. Microbes and Infection, 2008; 10(8): 878–884. doi:10.1016/j.micinf.2008.04.018
  27. 27. Deleo FR, Chambers HF: Review series re-emergence of antibiotic-resistant Staphylococcus aureus in the genomics era. Journal of Clinical Investigation, 2009; 119(9): 2464–2474. doi:10.1172/JCI38226
  28. 28. Gonzalez BE, Rueda AM, Shelburne SA, Musher DM, Hamill RJ, Hulten KG: Community-associated strains of methicillin-resistant Staphylococccus aureus as the cause of healthcare-associated infection. Infection Control and Hospital Epidemiology: The Official Journal of the Society of Hospital Epidemiologists of America, 2006; 27(10): 1051–1056. doi:10.1086/507923
  29. 29. Suhaili Z, Lean SS, Yahya A, Mohd Desa MN, AliAM, Yeo CC: Draft genome sequence of methicillin-resistant Staphylococcus aureus KT/Y21, a sequence type 772 (ST772) strain isolated from a pediatric blood sample in Terengganu, Malaysia. Genome Announcements, 2014; 2(2): 2–3. doi:10.1128/genomeA.00271-14
  30. 30. Oliveira DC, de Lencastre H: Methicillin-resistance in Staphylococcus aureus is not affected by the overexpression in trans of the mecA gene repressor: a surprising observation. PLoS One, 2011; 6(8): e23287. doi:10.1371/journal.pone.0023287
  31. 31. Tong SYC, Steer AC, Jenney AW, Carapetis JR: Community-associated methicillin-resistant Staphylococcus aureus skin infections in the tropics. Dermatologic Clinics, 2011; 29(1): 21–32. doi:10.1016/j.det.2010.09.005
  32. 32. Song MD, Wachi M, Doi M, Ishino F, Matsuhashi M: Evolution of an inducible penicillin target protein in MRSA by gene fusion. FEBS Letters, 1987; 221(I): 167. doi:10.1016/0014-5793(87)80373-3
  33. 33. Hartmann FA, Trostle SS, Klohnen AAO: Isolation of methicillin-resistant Staphylococcus aureus from a postoperative wound infection in a horse. Journal of the American Veterinary Medical Association, 1997; 211(5): 590–592.
  34. 34. Ito T, Katayama Y, Hiramatsu K: Cloning and nucleotide sequence determination of the entire mec DNA of pre-methicillin-resistant Staphylococcus aureus N315. Antimicrobial Agents and Chemotherapy, 1999; 43(6): 1449–1458.
  35. 35. Zong Z, Peng C, Lü X: Diversity of SCCmec elements in methicillin-resistant coagulase-negative staphylococci clinical isolates. PLoS One, 2011; 6(5): 1–6. doi:10.1371/journal.pone.0020191
  36. 36. Hiramatsu K, Asada K, Suzuki E, Okonogi K, Yokota T: Molecular cloning and nucleotide sequence determination of the regulator region of mecA gene in methicillin-resistant Staphylococcus aureus (MRSA). FEBS Letters. 1992; 298: 133–136. doi:10.1016/0014-5793(92)80039-J
  37. 37. Ito T, Katayama Y, Asada K, Mori N, Tsutsumimoto K, Tiensasitorn C, Hiramatsu K: Structural comparison of three types of staphylococcal cassette chromosome mec integrated in the chromosome in methicillin-resistant Staphylococcus aureus. Antimicrobial Agents and Chemotherapy, 2001; 45(5): 1323–1336. doi:10.1128/AAC.45.5.1323-1336.2001
  38. 38. Ito T, Kuwahara-Arai K, Katayama Y, Uehara Y, Han X, Yoko Kondo KH: Staphylococcal cassette chromosome mec (SCCmec) analysis of MRSA. Methods in Molecular Biology, 2014; 1085: 131–148. doi:10.1007/978-1-62703-664-1_8
  39. 39. Boundy S, Safo MK, Wang L, Musayev FN, O’Farrell HC, Rife JP, Archer GL: Characterization of the Staphylococcus aureus rRNA methyltransferase encoded by orfX, the gene containing the staphylococcal chromosome Cassette mec (SCCmec) insertion site. The Journal of Biological Chemistry, 2013; 288: 132–140. doi:10.1074/jbc.M112.385138
  40. 40. International Working Group on the Classification of Staphylococcal Cassette Chromosome Elements (IWG-SCC), & IWG: CLASSIFICATION of Staphylococcal Cassette Chromosome mec (SCCmec): Guidelines for reporting novel SCCmec Elements. Antimicrobial Agents and Chemotherapy, 2009; 53(12): 4961–4967. doi:10.1128/AAC.00579-09
  41. 41. Shore AC, Deasy EC, Slickers P, Brennan G, O’Connell B, Monecke S, Ehricht R, Coleman DC: Detection of staphylococcal cassette chromosome mec type XI carrying highly divergent mecA, mecI, mecR1, blaZ, and ccr genes in human clinical isolates of clonal complex 130 methicillin-resistant Staphylococcus aureus. Antimicrobial Agents and Chemotherapy, 2011; 55(8): 3765–3773. doi:10.1128/AAC.00187-11
  42. 42. García-Álvarez L, Holden MTG, Lindsay H, Webb CR, Brown DFJ, Curran MD, Walpole E, Brooks K, Pickard DJ, Teale C, Parkhill J, Bentley SD, Edwards GF, Girvan EK, Kearns AM, Pichon B, Hill RL, Larsen AR, Skov RL, Peacock SJ, Maskell DJ, Holmes MA: Methicillin-resistant Staphylococcus aureus with a novel mecA homologue in human and bovine populations in the UK and Denmark: a descriptive study. The Lancet Infectious Diseases, 2011; 11(8): 595–603. doi:10.1016/S1473-3099(11)70126-8
  43. 43. Ghaznavi-Rad E, Nor Shamsudin M, Sekawi Z, Khoon LY, Aziz MN, Hamat RA, Othman N, Chong PP, van Belkum A, Ghasemzadeh-Moghaddam H, Neela V: Predominance and emergence of clones of hospital-acquired methicillin-resistant Staphylococcus aureus in Malaysia. Journal of Clinical Microbiology, 2010; 48(3): 867–872. doi:10.1128/JCM.01112-09
  44. 44. Oliveira DC, Milheiriço C, de Lencastre H: Redefining a structural variant of staphylococcal cassette chromosome mec, SCCmec type VI. Antimicrobial Agents and Chemotherapy, 2006; 50(10): 3457–3459. doi:10.1128/AAC.00629-06
  45. 45. Hiramatsu K, Okuma K, Ma XX, Yamamoto M, Hori S, Kapi M: New trends in Staphylococcus aureus infections: glycopeptide resistance in hospital and methicillin resistance in the community. Current Opinion in Infectious Diseases, 2002; 15(4): 407–413.
  46. 46. Ma XX, Ito T, Tiensasitorn C, Jamklang M, Chongtrakool P, Boyle-Vavra S, Daum RS, Hiramatsu K: Novel type of staphylococcal cassette chromosome mec identified in community-acquired methicillin-resistant Staphylococcus aureus strains. Antimicrobial Agents and Chemotherapy, 2002; 46(4): 1147–1152. doi:10.1128/AAC.46.4.1147-1152. 2002
  47. 47. Ko KS, Lee Ji-Y, Suh JY, Oh WS, Peck KR, Lee NY, Song Jae-H: Distribution of major genotypes among methicillin-resistant Staphylococcus aureus clones in Asian countries. Journal of Clinical Microbiology, 2005; 43(1), 421–426. doi:10.1128/JCM.43.1.42 1-426. 2005
  48. 48. Jiménez JN, Ocampo AM, Vanegas JM, Rodriguez EA, Mediavilla JR, Chen L, Muskus CE, Vélez LA, Rojas C, Restrepo AV, Ospina S, Garcés C, Franco L, Bifani P, Kreiswirth BN, Correa MM: CC8 MRSA strains harboring SCCmec type ivc are predominant in Colombian hospitals. 2012; 7(6), e38576. doi: 10.1371/journal.pone.003857631
  49. 49. Berglund, C. Mölling, P. Sjöberg, L. Söderquist, Bo: Predominance of staphylococcal cassette chromosome mec (SCCmec) type IV among methicillin-resistant Staphylococcus aureus (MRSA) in a Swedish county and presence of unknown SCCmec types with Panton-Valentine leukocidin genes. Clinical Microbiology and Infection, 2005; 11(6),447–456. doi:10.1111/j.1469-0691.2005.01150.x
  50. 50. Abdulgader SM, Shittu AO, Nicol MP, Kaba M: Molecular epidemiology of methicillin-resistant Staphylococcus aureus in Africa: a systematic review. Frontier Microbiology, 2015; 6, 348. doi:10.3389/fmicb.2015.00348
  51. 51. Shore AC, Rossney AS, O’Connell B, Herra CM, Sullivan DJ, Humphreys H, Coleman DC: Detection of staphylococcal cassette chromosome mec-associated DNA segments in multiresistant methicillin-susceptible Staphylococcus aureus (MSSA) and identification of Staphylococcus epidermidisccrAB4 in both methicillin- resistant S. aureus and MSSA. Antimicrobial Agents and Chemotherapy, 2008; 52: 4407–4419. doi:10.1128/AAC.00447-08
  52. 52. Lindqvist M, Isaksson B, Grub C, Jonassen TO, Hällgren A: Detection and characterisation of SCCmec remnants in multiresistant methicillin-susceptible Staphylococcus aureus causing a clonal outbreak in a Swedish county. European Journal of Clinical Microbiology and Infectious Diseases, 2012; 31: 141–147. doi:10.1007/s10096-011-12 86-y
  53. 53. Ghasemzadeh-Moghaddam H, Neela V, Goering R, Mariana NS: Methicillin sensitive Staphylococcus aureus (MSSA) isolates as a potential source for the emergence of USA 300 methicillin resistant Staphylococcus aureus (MRSA) in Malaysia. Tropical Biomedicine, 2012; 29(3): 429–433.
  54. 54. Thurlow LR, Joshi GS., Clark JR., Spontak JS., Neely CJ, Maile R., Richardson AR.: Functional modularity of the arginine catabolic mobile element contributes to the success of USA300 methicillin-resistant Staphylococcus aureus. Cell Host and Microbe, 2013; 13(1): 100-107. doi:10.1016/j.chom.2012.11.012
  55. 55. Tsubakishita S, Kuwahara-Arai K, Sasaki T, Hiramatsu K: Origin and molecular evolution of the determinant of methicillin resistance in staphylococci. Antimicrobial Agents and Chemotherapy, 2010; 54(10): 4352–4359. doi:10.1128/AAC.00356-10
  56. 56. Hanssen AM, Ericson Sollid JU: SCCmec in staphylococci: Genes on the move. FEMS Immunology and Medical Microbiology, 2006; 46(1): 8–20. doi:10.1111/j.1574-695X.2005.00009.x.
  57. 57. Tulinski P, Fluit AC, Wagenaar JA, Mevius D, van de Vijver L, Duim B: Methicillin-resistant coagulase-negative staphylococci on pig farms as a reservoir of heterogeneous staphylococcal cassette chromosome mec elements. Applied and Environmental Microbiology, 2012; 78(2):, 299–304. doi:10.1128/AEM.05594-11
  58. 58. McGavin MJ: Genome comparisons of diverse Staphylococcus aureus strains. Bacterial Genomes and Infectious Diseases, 2006; 5: 191–212. doi:10.1007/978-1-59745-152-9_11
  59. 59. Wielders CL, Vriens MR, Brisse S, Al, E: In-vivo transfer of mecA DNA to Staphylococcus aureus. Lancet, 2001; 357: 1674–1675. doi: 10.1016/S0140-6736(00)04832-7
  60. 60. Bouchami O, Ben Hassen A, de Lencastre H, Miragaia M: Molecular epidemiology of methicillin-resistant Staphylococcus hominis (MRSHo): low clonality and reservoirs of SCCmec structural elements. PLoS One, 2011; 6(7): e21940. doi:10.1371/journal.pone.0021940
  61. 61. Lebeaux D, Barbier F, Angebault C, Benmahdi L, Ruppé E, Felix B, Gaillard K, Djossou F, Epelboin L, Dupont C, Renard M, Peroz G, Vandenesch F, Wolff M, Andremont A, Ruimy R: Evolution of nasal carriage of methicillin-resistant coagulase-negative staphylococci in a remote population. Antimicrobial Agents and Chemotherapy, 2012; 56(1): 315–323. doi:10.1128/AAC.00547-11
  62. 62. Rolo J, de Lencastre H, Miragaia M: Strategies of adaptation of Staphylococcus epidermidis to hospital and community: amplification and diversification of SCCmec. The Journal of Antimicrobial Chemotherapy, 2012; 67(6): 1333–1341. doi:10.1093/jac/dks068
  63. 63. Ruppé E, Barbier F, Mesli Y, Maiga A, Cojocaru R, Benkhalfat M, Benchouk S, Hassaine H, Maiga I, Diallo A, Koumaré AK, Ouattara K, Soumaré S, Dufourcq JB, Nareth C, Sarthou JL, Andremont A, Ruimy R: Diversity of staphylococcal cassette chromosome mec structures in methicillin-resistant Staphylococcus epidermidis and Staphylococcus haemolyticus strains among outpatients from four countries. Antimicrobial Agents and Chemotherapy, 2009; 53(2): 442–449. doi:10.1128/AAC.00724-08
  64. 64. Wong H, Louie L, Lo RYC, Simor AE: Characterization of Staphylococcus aureus isolates with a partial or complete absence of staphylococcal cassette chromosome elements. Journal of Clinical Microbiology, 2010; 48(10): 3525–3531. doi:10.1128/JCM.00775-10
  65. 65. Donnio PY, Février F, Bifani P, Dehem M, Kervégant C, Wilhelm N, Gautier-Lerestif AL, Lafforgue N, Cormier M; MR-MSSA study group of the Collège de Bactériologie-Virologie-Hygiène des Hôpitaux de France, Le Coustumier A. Molecular and epidemiological evidence for spread of multiresistant methicillin-susceptible Staphylococcus aureus strains in hospitals. Antimicrobial Agents and Chemotherapy, 2007; 51(12): 4342–4350. doi:10.1128/AAC.01414-06
  66. 66. Chlebowicz MA, Nganou K, Kozytska S, Arends JP, Engelmann S, Grundmann H, Ohlsen K, van Dijl JM, Buist G: Recombination between ccrC genes in a type V (5C2&5) staphylococcal cassette chromosome mec (SCCmec) of Staphylococcus aureus ST398 leads to conversion from methicillin resistance to methicillin susceptibility in vivo. Antimicrobial Agents and Chemotherapy, 2010; 54(2): 783–791. doi:10.1128/AAC.00696-09
  67. 67. Vandendriessche S, Vanderhaeghen W, Larsen J, de Mendonça R, Hallin M, Butaye P, Hermans K, Haesebrouck F, Denis O: High genetic diversity of methicillin-susceptible Staphylococcus aureus (MSSA) from humans and animals on livestock farms and presence of SCCmec remnant DNA in MSSA CC398. The Journal of Antimicrobial Chemotherapy, 2014; 69(2): 355–362. doi:10.1093/jac/dkt366
  68. 68. Aires de Sousa M, de Lencastre H: Bridges from hospitals to the laboratory: genetic portraits of methicillin-resistant Staphylococcus aureus clones. FEMS Immunology and Medical Microbiology, 2004; 40(2): 101–111. doi:10.1016/S0928-8244(03)00370-5
  69. 69. Crisóstomo MI, Westh H, Tomasz A, Chung M, Oliveira DC, de Lencastre H: The evolution of methicillin resistance in Staphylococcus aureus: similarity of genetic backgrounds in historically early methicillin-susceptible and resistant isolates and contemporary epidemic clones. Proceedings of the National Academy of Sciences of the United States of America, 2001; 98(17): 9865–9870. doi:10.1073/pnas.161272898
  70. 70. Oliveira DC, de Lencastre H: Multiplex PCR strategy for rapid identification of structural types and variants of the mec element in methicillin-resistant Staphylococcus aureus. Antimicrobial Agents and Chemotherapy, 2002; 46(7): 2155–2161. doi:10.1128/AAC.46.7.2155-2161
  71. 71. Witte W, Strommenger B, Stanek C, Cuny C: Methicillin-resistant Staphylococcus aureus ST398 in Humans and Animals, Central Europe. Emerging Infectious Diseases, 2007; 13(2): 255–258. doi:10.3201/eid1302.060924
  72. 72. Cuny C, Wieler L, Witte W: Livestock-associated MRSA: the impact on humans. Antibiotics, 2015; 4(4): 521–543. doi:10.3390/antibiotics4040521
  73. 73. Blanc DS, Petignat C, Moreillon P, Entenza JM, Eisenring M, Kleiber H, Wenger A, Troillet N, Blanc C, Francioli P: Unusual spread of a penicillin-susceptible methicillin-resistant Staphylococcus aureus clone in a geographic area of low incidence. Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America, 1999; 29(6): 1512–1518. doi:10.1086/313522
  74. 74. Otto M: MRSA virulence and spread. Cellular Microbiology, 2012; 14(10): 1513–1521. doi:10.1111/j.1462-5822.2012.01832.x
  75. 75. Wang JT, Fang CT, Chen YC, Wu CL, Chen ML, Chang SC: Staphylococcal cassette chromosome mec in MRSA, Taiwan. Emerging Infectious Diseases, 2007; 13(3): 494–497.
  76. 76. Ahmad N, Ruzan IN, Abd Ghani MK, Hussin A, Nawi S, Aziz MN, Maning N, Eow VL: Characteristics of community and hospital-acquired methicillin-resistant Staphylococcus aureus strains carrying SCCmec type IV isolated in Malaysia. Journal of Medical Microbiology, 2009; 58(9): 1213–1218. doi:10.1099/jmm.0.011353-0
  77. 77. Brennan GI, Shore A C, Corcoran S, Tecklenborg S, Coleman DC, O’Connell B: Emergence of hospital and community-associated Panton-valentine leukocidin-positive methicillin-resistant Staphylococcus aureus genotype ST772-MRSA-V in Ireland and detailed investigation of an ST772-MRSA-V cluster in a neonatal intensive care unit. Journal of Clinical Microbiology, 2012; 50(3): 841–847. doi:10.1128/JCM.06354-11
  78. 78. Moore PCL, Lindsay JA: Molecular characterisation of the dominant UK methicillin-resistant Staphylococcus aureus strains, EMRSA-15 and EMRSA-16. Journal of Medical Microbiology, 2002; 51(6): 516–521. doi:10.1099/0022-1317-51-6-516
  79. 79. Murchan S, Kaufmann ME, Deplano A, de Ryck R, Struelens M, Zinn CE, Fussing V, Salmenlinna S, Vuopio-Varkila J, El Solh N, Cuny C, Witte W, Tassios PT, Legakis N, van Leeuwen W, van Belkum A, Vindel A, Laconcha I, Garaizar J, Haeggman S, Olsson-Liljequist B, Ransjo U, Coombes G, Cookson B: Harmonization of pulsed-field gel electrophoresis protocols for epidemiological typing of strains of methicillin-resistant Staphylococcus aureus: a single approach developed by consensus in 10 European laboratories and its application for tracing the spread of related strains. Journal of Clinical Microbiology, 2009; 41(4): 1574–1585. doi:10.1128/JCM.41.4.1574-1585.2003
  80. 80. Faria NA, Oliveira DC, Westh H, Monnet DL, Larsen AR, Skov R, de Lencastre H: Epidemiology of emerging methicillin-resistant Staphylococcus aureus (MRSA) in Denmark: a nationwide study in a country with low prevalence of MRSA infection. Journal of Clinical Microbiology, 2005; 43(4): 1836–1842. doi:10.1128/JCM.43.4.1836-18 42.2005
  81. 81. Seeberg S, Larsson L, Welinder-Olsson C, Sandberg T, Skyman E, Bresky B, Lindqvist A, van Raalte M: How an outbreak of MRSA in gothenburg was eliminated: by strict hygienic routines and massive control-culture program. Lȧkartidningen, 2002; 99(32–33): 3198–3204.
  82. 82. Montesinos I, Delgado T, Riverol D, Salido E, Miguel MA, Jimenez A, Sierra A: Changes in the epidemiology of methicillin-resistant Staphylococcus aureus associated with the emergence of EMRSA-16 at a university hospital. Journal of Hospital Infection, 2006; 64(3): 257–263. doi:10.1016/j.jhin.2006.07.004
  83. 83. Udo EE, Al-Sweih N, Noronha B: Characterisation of non-multiresistant methicillin-resistant Staphylococcus aureus (including EMRSA-15) in Kuwait hospitals. Clinical Microbiology and Infection, 2006; 12(3): 262–269. doi:10.1111/j.1469-0691.2005.01350.x
  84. 84. McDougal LK, Steward CD, Killgore GE, Chaitram JM, McAllister SK, Tenover FC: Pulsed-field gel electrophoresis typing of oxacillin-resistant Staphylococcus aureus isolates from the United States: establishing a national database. Journal of Clinical Microbiology, 2003; 41(11): 5113–5120. doi:10.1128/JCM.41.11.5113-5120.2003
  85. 85. Grundmann H, Aanensen DM, van den Wijngaard CC, Spratt BG, Harmsen D, Friedrich AW: Group the European staphylococcal reference laboratory working group. geographic distribution of Staphylococcus aureus causing invasive infections in Europe: A molecular-epidemiological analysis. PLoS Medicine, 2010;7(1): e1000215. doi:10.1371/journal.pmed.1000215
  86. 86. Kinneveya PM., Shore AC., Brennan GI, Sullivana DJ, Ehrichtd R, Monecke S, Slickers P., Coleman DC: Emergence of sequence type 779 methicillin-resistant Staphylococcus aureus harboring a novel pseudo staphylococcal cassette chromosome mec (SCCmec)-SCC-SCCCRISPR composite element in irish hospitals. Antimicrobial Agents and Chemotherapy, 2013;57(1), 524–531. doi:10.1128/AAC.01689-12
  87. 87. Vandenesch F, Naimi T, Enright MC, et al: Community-acquired methicillin-resistant Staphylococcus aureus carrying Panton-Valentine leukocidin genes: worldwide emergence. Emerging Infectious Diseases, 2003;9:978–984. doi:10.3201/eid0908.030089
  88. 88. Y. Takizawa, I. Taneike, S. Nakagawa, T. Oishi, Y. Nitahara, N. Iwakura: A Panton-Valentine leucocidin (PVL)-positive community-acquired methicillin-resistant Staphylococcus aureus (MRSA) strain, another such strain carrying a multiple-drug resistance plasmid, and other more-typical PVL-negative MRSA strains found in Japan. Journal of Clinical Microbiology, 2005; 43, 3356–3363. doi:10.1128/JCM.43.7.3356-33 63.2005
  89. 89. Enany S., Yaoita E., Yoshida Y., Enany M., Yamamoto T: Molecular characterization of Panton-Valentine leukocidin-positive community-acquired methicillin-resistant Staphylococcus aureus isolates in Egypt. Microbiological Research. 2010; 165(2); 152–162. doi:10.1016/j.micres.2009.03.005
  90. 90. Olivier Andreoletti, Herbert Budka, Sava Buncic, Pierre Colin, John D Collins, Aline De Koeijer, John Griffin, Arie Havelaar, James Hope, Günter Klein, Hilde Kruse, Simone Magnino, Antonio, Martínez López, James McLauchlin, Christophe Nguyen-The, Karsten Noeckler, Birgit Noerrung, Miguel Prieto Maradona, Terence Roberts, Ivar Vågsholm, Emmanuel Vanopdenbosch: Assessment of the public health significance of methicillin resistant Staphylococcus aureus (MRSA) in animals and foods. scientific opinion of the panel on biological hazards adopted on 5 March 2009. The EFSA Journal, 2009; 993: 1–73. doi:10.2903/j.efsa.2009.993
  91. 91. Vanderhaeghen W, Cerpentier T, Adriaensen C, Vicca J, Hermans K, Butaye P: Methicillin-resistant Staphylococcus aureus (MRSA) ST398 associated with clinical and subclinical mastitis in Belgian cows. Veterinary Microbiology, 2010; 144(1–2): 166–171. doi:10.1016/j.vetmic.2009.12.044
  92. 92. Richter A, Sting R, Popp C, Rau J, Tenhagen BA, Guerra B, Hafez HM, Fetsch A: Prevalence of types of methicillin-resistant Staphylococcus aureus in turkey flocks and personnel attending the animals. Epidemiology and Infection, 2012; 140(12): 2223–2232. doi:10.1017/S095026881200009X
  93. 93. de Boer E, Zwartkruis-Nahuis JT, Wit B, Huijsdens XW, de Neeling AJ, Bosch T, van Oosterom RA, Vila A, Heuvelink AE: Prevalence of methicillin-resistant Staphylococcus aureus in meat. International Journal of Food Microbiology, 2009; 134(1–2): 52–56. doi:10.1016/j.ijfoodmicro.2008.12.007
  94. 94. Schulz J, Friese A, Klees S, Tenhagen BA, Fetsch A, Rösler U, Hartung J: Longitudinal study of the contamination of air and of soil surfaces in the vicinity of pig barns by livestock-associated methicillin-resistant Staphylococcus aureus. Applied and Environmental Microbiology, 2012; 78(16): 5666–5671. doi:10.1128/AEM.00550-12
  95. 95. Layer F, Cuny C, Strommenger B, Werner G, Witte W: Current data and trends on methicillin-resistant Staphylococcus aureus (MRSA). Bundesgesundheitsblatt, Gesundheitsforschung, Gesundheitsschutz, 2012; 55(11–12):1377–1386. doi:10.1007/s00103-0 12-1560-x
  96. 96. Oliveira CJ. Tiao N, de Sousa F.G, de Moura J.F, Santos Filho L, Gebreyes WA: Methicillin-resistant Staphylococcus aureus from Brazilian dairy farms and identification of novel sequence types. Zoonoses Public Health. 2016;63(2):97–105. doi:10.1111/zph.12209
  97. 97. Chen CJ, Huang YC: New epidemiology of Staphylococcus aureus infection in Asia. Clinical Microbiology and Infection., 2014; 20(7): 605–623. doi:10.1111/1469-0691.12705
  98. 98. Levy SB, Marshall B: Antibacterial resistance worldwide: causes, challenges and responses. Nature Medicine, 2004; 10(1078–8956 (Print): S122–S129. doi:10.1038/nm1145
  99. 99. Lim KT, Hanifah YA, Mohd Yusof MY, Ito T, Thong KL: Comparison of methicillin-resistant Staphylococcus aureus strains isolated in 2003 and 2008 with an emergence of multidrug resistant ST22: SCCmec IV clone in a tertiary hospital, Malaysia. Journal of Microbiology, Immunology and Infection, 2013; 46(3): 224–33. doi:10.1016/j.jmii.2013.02.001.
  100. 100. Cosgrove SE, Sakoulas G, Perencevich EN, Schwaber MJ, Karchmer AW, Carmeli Y: Comparison of mortality associated with methicillin-resistant and methicillin-susceptible Staphylococcus aureus bacteremia: a meta-analysis. Clinical Infectious Diseases : An Official Publication of the Infectious Diseases Society of America, 2003; 36(1): 53–59. doi:10.1086/345476
  101. 101. Suhaili Z, Johari SA, Mohtar M, Abdullah ART, Ahmad A, Ali AM: Detection of Malaysian methicillin-resistant Staphylococcus aureus (MRSA) clinical isolates using simplex and duplex real-time PCR. World Journal of Microbiology and Biotechnology, 2009; 25(2): 253–258. doi:10.1007/s11274-008-9887-z
  102. 102. Lyon BR, Skurray R: Antimicrobial resistance of Staphylococcus aureus: genetic basis. Microbiological Reviews, 1987; 51(1): 88–134.
  103. 103. Al-Talib H, Alyaa Al-Khateeb HH: Antimicrobial resistance of Staphylococcus aureus isolates in Malaysian tertiary hospital. International Medical Journal, 2015; 22(1): 1–3.
  104. 104. Magiorakos AP, Srinivasan A, Carey RB, Carmeli Y, Falagas ME, Giske CG, Harbarth S, Hindler JF, Kahlmeter G, Olsson-Liljequist B, Paterson DL, Rice LB, Stelling J, Struelens MJ, Vatopoulos A, Weber JT, Monnet DL: Multidrug-resistant, extensively drug-resistant and pan-drug-resistant bacteria: an international expert proposal for interim standard definitions for acquired resistance. Clinical Microbiology and Infection, 2012; 18(3): 268–281. doi:10.1111/j.1469-0691.2011.03570.x
  105. 105. Ullah A, Qasim M, Rahman H, Khan J, Haroon M, Muhammad N, Khan A, Muhammad N: High frequency of methicillin-resistant Staphylococcus aureus in Peshawar Region of Pakistan. Springerplus, 2016; 5(1): 600. doi:10.1186/s40064-016-2277-3
  106. 106. Nickerson EK, Wuthiekanun V, Kumar V, Amornchai P, Wongdeethai N, Chheng K, Chantratita N, Putchhat H, Thaipadungpanit J, Day NP, Peacock SJ: Emergence of community-associated methicillin-resistant Staphylococcus aureus carriage in children in Cambodia. American Journal of Tropical Medicine and Hygiene, 2011; 84(2): 313–317. doi:10.4269/ajtmh.2011.10-0300
  107. 107. Lim SK, Nam HM, Park HJ, Lee HS, Choi MJ, Jung SC, Lee JY., Kim YC, Song SW, Wee SH: Prevalence and characterization of methicillin-resistant Staphylococcus aureus in raw meat in Korea. Journal of Microbiology and Biotechnology, 2010; 20(4): 775–778.
  108. 108. Lim KT, Hanifah YA, Yusof M, Thong KL: ermA, ermC, tetM and tetK are essential for erythromycin and tetracycline resistance among methicillin-resistant Staphylococcus aureus strains isolated from a tertiary hospital in Malaysia. Indian Journal of Medical Microbiology, 2012; 30(2): 203–207. doi:10.4103/0255-0857.96693
  109. 109. Al-Talib HI, Yean CY, Al-Jashamy K, Hasan H: Methicillin-resistant Staphylococcus aureus nosocomial infection trends in hospital Universiti Sains Malaysia during 2002–2007. Annals of Saudi Medicine, 2010; 30(5): 358–363. doi:10.4103/0256-4947.67077
  110. 110. Bell JM, Turnidge JD: SENTRY APAC. High prevalence of oxacillin-resistant Staphylococcus aureus isolates from hospitalized patients in Asia-Pacific and South Africa: results from SENTRY antimicrobial surveillance program, 1998–1999. Antimicrobial Agents and Chemotherapy. 2002; 46(3): 879–881. doi:10.1128/AAC.46.3.880-882.2002
  111. 111. Kobayashi H: National hospital infection surveillance on methicillin-resistant Staphylococcus aureus. The Journal of Hospital Infection, 2005; 60(2): 172–175.
  112. 112. Kang Cheol-I, Song Jae-H: Antimicrobial resistance in Asia: Current epidemiology and clinical implications. Infection and Chemotherapy, 2013; 45(1): 22–31. doi:10.3947/ic.2013.45.1.22
  113. 113. Saleha AA, Zunita Z: Methicillin resistant Staphylococcus aureus (MRSA): An emerging veterinary and zoonotic pathogen of public health concern and some studies in Malaysia. Journal of Animal and Veterinary Advances, 2010; 9(7): 1094–1098. doi:10.3923/javaa.2010.1094.1098
  114. 114. Choi CS, Yin CS, Bakar AA, Sakewi Z, Naing NN, Jamal F, Othman N: Nasal carriage of Staphylococcus aureus among healthy adults. Journal of Microbiology, Immunology and Infection, 2006; 39(6): 458–464.
  115. 115. Lim VK, Zulkifli HI: Methicillin resistant Staphylococcus aureus in a Malaysian neonatal unit. Singapore Medical Journal, 1987; 28(2): 176–179.
  116. 116. Norazah A, Lim VK, Koh YT, Rohani MY, Zuridah H, Spencer K, Ng PP, Kamel AG: Molecular fingerprinting of fusidic acid- and rifampicin-resistant strains of methicillin-resistant Staphylococcus aureus (MRSA) from Malaysian hospitals. Journal of Medical Microbiology, 2002; 51(12): 1113–1116. doi:10.1099/0022-1317-51-12-1113
  117. 117. Ong WHS, Lai QX, Azhan F, Sapiee NA, Mohd Zaidi SH, Zahir ME, Harun SN: Choices of antibiotics for MRSA infection in choices of antibiotics for MRSA infection in Malaysia. Web Medical Central, 2011; 2(12): 1–12. doi:10.9754/journal.wmc.2011.002675
  118. 118. Asmat A, Zulkifli A, Usup G: Detection of methicillin resistant Staphylococcus aureus (MRSA) isolated from Bagan Lalang recreational beach, Malaysia. Malaysian Journal of Microbiology, 2013; 9(2): 166–175.
  119. 119. Song JH, Hsueh PR, Chung DR, Ko KS, Kang CI, Peck KR, Yeom JS, Kim SW, Chang HH, Kim YS, Jung SI, Son JS, So TM, Lalitha MK, Yang Y, Huang SG, Wang H, Lu Q, Carlos CC, Perera JA, Chiu CH, Liu JW, Chongthaleong A, Thamlikitkul V, Van PH; ansorp study group. Spread of methicillin-resistant Staphylococcus aureus between the community and the hospitals in Asian countries: An ANSORP study. Journal of Antimicrobial Chemotherapy, 2011; 66(5), 1061–1069. doi:10.1093/jac/dkr024
  120. 120. Song JH, Hiramatsu K, Suh JY, Ko KS, Ito T, Kapi M, Kiem S, Kim YS, Oh WS, Peck KR, Lee NY: Asian network for surveillance of resistant pathogens study group. Emergence in Asian countries of Staphylococcus aureus with reduced susceptibility to vancomycin. Antimicrobial Agents and Chemotherapy, 2004; 48(12): 4926–4928. doi:10.1128/AAC.48.12.4926-4928.2004

Written By

Abdul Rahim Abdul Rachman, Zarizal Suhaili and Mohd Nasir Mohd Desa

Submitted: 21 April 2016 Reviewed: 05 September 2016 Published: 08 March 2017