Submit manuscript...
eISSN: 2373-6372

Gastroenterology & Hepatology: Open Access

Review Article Volume 13 Issue 6

MRSA infections: priorities and future approaches for research

Manal ME Ahmed

Department of Pharmacology, Medical Research and Clinical Studies Institute, Egypt

Correspondence: Manal ME Ahmed, Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt, Tel +20-10936-27027

Received: September 12, 2022 | Published: November 4, 2022

Citation: Ahmed MME. MRSA infections: priorities and future approaches for research. Gastroenterol Hepatol Open Access. 2022;13(6):200-208. DOI: 10.15406/ghoa.2022.13.00523

Download PDF

Abstract

Methicillin-resistant Staphylococcus aureus (MRSA) has arisen since the 1960s, spread around the world, and become a major cause of bacterial infections in both healthcare and community settings. However, the prevalence of MRSA varies significantly by region due to a number of reasons, including variations in local infection control procedures and pathogen-specific traits of the circulating clones. The independent acquisition of staphylococcal cassette chromosome mec (SCCmec), which contains genes encoding proteins that render the bacterium resistant to the majority of β-lactam antibiotics (such as methicillin), by several S. aureus clones, has led to the emergence of various MRSA clones. The abundance of virulence factors that S. aureus produces, along with β-lactam resistance and, for the majority of clones, resistance to other antibiotic classes, contribute to MRSA’s success. Clinical signs of MRSA can include asymptomatic nasal mucosa colonization, moderate skin and soft tissue infections, or fulminant invasive illness with a high fatality rate. Although there aren’t many choices for treating MRSA, numerous new antibiotics are in the works.

It will be possible to optimise strategies to effectively control MRSA if you have a thorough understanding of colonisation dynamics, transmission pathways, risk factors for progressing to infection, and environmental factors that encourage the evolution of resistance. Additionally, vaccine candidates are being developed and may eventually be useful as a preventative approach.

Keywords: MRSA, Staphylococcus aureus, Methicillin-resistant Staphylococcus aureus, staphylococcal cassette chromosome mec, β-lactam

Abbreviations

MRSA, methicillin-resistant Staphylococcus aureus; SCCmec, staphylococcal cassette chromosome mec, ClfB, clumping factor B; PMNs, polymorphonuclear leukocytes; PBP2a, penicillin-binding protein 2a; IgG, immunoglobulin G; CP5, capsular polysaccharide 5; WTA, wall teichoic acid; WGS, whole-genome sequencing

Introduction

The Coccoid bacteria Staphylococcus aureus belongs to the Firmicutes phylum and is Gram-positive, nonmotile, and coagulase-positive. S. aureus is by far the most clinically relevant of the 52 species and 28 subspecies that make up the Staphylococcus genus (List of Prokaryotic names with Standing in Nomenclature). 20–40% of the general population has S. aureus in their nasal mucosal commensal microbiota.1,2  Due to variations in the size and demography of the study populations, the quality of the sample, and the culture techniques used,3 the stated prevalence varies. S. aureus can enter underlying tissues or the bloodstream and spread infection when the cutaneous and mucosal barriers are compromised, such as due to chronic skin conditions, wounds, or surgical intervention. S. Aureus infection is more likely to occur in people with invasive medical devices (like peripheral and central venous catheters) or weakened immune systems.4 The first case of methicillin-resistant S. aureus (MRSA) was reported in England in 1961,5 not long after the drug was first used in clinical settings. Methicillin was once widely used, but due to its toxicity, it is no longer marketed for human use. Instead, similar, more stable penicillins like oxacillin, flucloxacillin, and dicloxacillin6 have largely taken its place. However, the phrase “methicillin-resistant S. Aureus ” is still in use. MRSA was the cause of hospital epidemics (healthcare-associated MRSA, or HA-MRSA) in various regions of the world in the decade after its initial description.7 The occurrence of community-associated MRSA (CA-MRSA), also known as MRSA, marked a significant shift in the epidemiology of the bacteria. It has also been linked to exposure to livestock since the middle of the 2000s (livestock-associated MRSA, or LA-MRSA).8 Several S. aureus clones have evolved into MRSA through horizontal gene transfer of staphylococcal cassette chromosome mec (SCCmec), a mobile genetic element that encodes the genes mecA and mecB. Clones are bacteria that cannot be distinguished from one another by a variety of genetic tests (for example, pulsed-field gel electrophoresis, multilocus enzyme electrophoresis, or ribotyping), or that are so similar that they.9,10 MRSA frequently exhibits resistance to antibiotics with β-lactams and numerous other antibiotic groups. The remarkable capacity of S. aureus to develop resistance to any antibiotic7 has significant ramifications for both existing and potential future treatments for this pathogen.

People who have MRSA colonisation or carriage—i.e., the presence of bacteria that do not result in cellular damage, clinical signs and symptoms of infection, or detectable host immune responses—have a higher risk of contracting the infection again and are a significant source of person-to-person transmission. People who are susceptible to illness are housed in healthcare institutions, settings with strong antibiotic selection pressure (which might lead to the selection of antimicrobial resistance in bacteria, for instance, because to invasive procedures and/or immunological impairment) and frequent human contact. MRSA is now endemic in many healthcare facilities around the world, and as a result, it has become a major focus for infection control efforts globally. These conditions have facilitated the epidemic spread of MRSA in hospitals.

Future research priorities are covered in this primer, which also addresses the mechanisms, pathophysiology, prevention and management of MRSA (Figure 1).

Figure 1 Most frequent MRSA clones. Sequence types (STs) of methicillin-sensitive Staphylococcus aureus (MSSA) are grouped into clonal complexes (CCs) by their similarity to a founder allelic profile (genotype)226. STs have different molecular properties that enable monitoring of the geographical spread of different clones. STs of MSSA can evolve into MRSA by acquiring staphylococcal cassette chromosome mec (SCCmec), of which there are different types (represented by roman numerals). Commonly used clone names are within parentheses. Numbers in the names of MRSA USA clones are ased on pulsed- field gel electrophoresis analysis.
ACME, arginine catabolic mobile element; EMRSA, epidemic MRSA; PVL, Panton–Valentine leukocidin; sek2 and seq2 are staphylococcal genes encoding enterotoxin.80

Mechanisms/pathophysiology

Colonisation by Staphylococcus aureus

Most of the time, S. aureus colonisation occurs before an infection occurs.11 Less frequently, infection can happen when S. aureus colonisation is unknown, for instance, when catheters or wounds are contaminated due to subpar infection control procedures by healthcare professionals. Although colonisation occurs at other sites, most notably in the throat and perineum,12 the nose is the primary site of S. aureus colonization. Three temporal patterns of S. aureus (including MSSA an MRSA) colonisation have been identified by longitudinal studies.13 Only 15% of people were found to have continuous S. aureus colonisation (known as persistent carriers), while 70% of people had intermittent colonization, which indicates that most people can repeatedly acquire S. aureus and spontaneously clear it., and 15% of people (known as non-carriers) had no evidence of S. aureus.13 Other studies14 found comparable outcomes. There are underlying host factors that determine the carriage status, according to studies examining particular host polymorphisms in genes related to the inflammatory response. These studies, which examined these genes,15 However, it is not quite clear what these underlying variables exactly are. The length of colonisation can vary, especially for MRSA, and quoted estimates may be skewed by antibiotic use, which can cut the length of colonisation. The median time of colonisation in one research of patients with MRSA colonisation at the time of hospital release was 282 days,16 and 81% of those in this sample had persistent skin lesions, which is a known risk factor for MRSA colonisation.that might have caused the protracted carriage. Along with host variables, the nasal microbiota and factors related to the virus itself might affect the host carrier status.

Colonisation dynamics

Teichoic acid on the cell wall serves as a mediator during the early stages of S. aureus colonisation while microbial surface components that recognise adhesive matrix molecules are involved during the later stages of nasal colonization.17,18

S. aureus clumping factor B (ClfB), one of these components, has been researched in vitro and on human volunteers.19 The nose was injected with both the wild-type strain and its single locus clfB knockout variation; the knockout version cleared much more quickly than the wild-type strain.ClfB-deficient strains, however, may still interact with nasal cells, suggesting that there are a number of distinct microbial surface elements that contribute to colonization.20 It should be noted that this study only used one strain.

Along with host and pathogen factors, S. aureus interacts with other species that colonise the nose, such as Corynebacterium spp., Propionibacterium acnes, Staphylococcus lugdunensis, and Staphylococcus epidermidis. According to research on the nasal microbiota, certain species (such as S. Epidermidis, which has been positively correlated with S. aureus presence) are associated with the presence or absence of S. aureus.21,22 The organisms that make up the nasal microbiota compete with one another in a number of ways. For instance, they struggle for adhesion sites and nutrition because the human nose has few of both.

S. aureus is more adapted to the human nose because it can survive in environments with nutrient levels that are lower than coagulase-negative staphylococci can23 (possibly due to differences in metabolism). However, there hasn’t been any evidence of a difference in nutritional levels between carriers and non-carriers.23 Species of the microbiota can also engage in antibiosis, or the production of chemicals that prevent their competitors from growing. For instance, S. Lugdunensis creates lugdunin, an antimicrobial compound that inhibits and kills S. aureus (including MRSA) in a mouse model and in vitro, possibly by causing a rapid breakdown of bacterial energy resources.24 A sixfold lower risk of nasal colonisation with S. aureus has been linked to nasal colonisation with S. lugdunensis in humans. Although these findings are intriguing, they only partially account for carriage patterns since S. lugdunensis colonisation has only been documented in 9–26% of the general population.24,25 Last but not least, S. aureus also engages in competitive host defences, causing the host to produce antimicrobial proteins that are less toxic to S. aureus than to other commensal bacteria.26 Numerous studies back up the involvement of these mechanisms in the interactions between S. aureus and the commensal microbiota, but only one mechanism can fully account for all carriage patterns that have been seen (Figure 2).

Figure 2 Stages of Staphylococcus aureus infection. A | Bacteria obtain access to sterile tissues via open wounds and use adhesin proteins, such as fibronectin-binding protein A (FnBPA), FnBPB, iron-regulated surface determinant protein A (IsdA), clumping factor A (ClfA), ClfB and collagen adhesin (Cna), for specific attachment to extracellular matrix proteins, such as fibronectin, cytokeratin and collagen, among others. Staphylococcus aureus can also in part regulate polymorphonuclear leukocyte (PMN) influx in subtle ways involving activators (formylated peptides and phenol-soluble modulin (PSM) peptides) and inhibitors (for example, chemotaxis inhibitory protein of S. Aureus (CHIPS) and FPRL1 inhibitory protein (FLIPr)) of PMN chemotaxis. PSM peptides also promote the release of pro-inflammatory lipoproteins, the major S. Aureus microorganism-associated molecular pattern (MAMP) molecules, which activate Toll-like receptor 2 (TLR2) and contribute to local inflammation.  B | S. Aureus produces coagulases to polymerize fibrin and form an encapsulated abscess around the infection site. The capacity of PMNs, which are found in high numbers in an abscess, to eliminate S. Aureus is limited by leukocidins and by virulence factors interfering with opsonophagocytosis and PMN killing. S. Aureus can compromise effective opsonization by antibodies using a polysaccharide microcapsule and surface proteins (Staphylococcus protein A (SpA) and immunoglobulin-binding protein Sbi) binding immunoglobulin G (IgG) via the crystallizable fragment (Fc) domain in a futile way66. The bacteria can also inhibit the complement signalling pathway by small secreted inhibitors such as staphylococcal complement inhibitor (SCIN), fibrinogenbinding protein (Efb), extracellular complement- binding protein (Ecb) or staphylococcal superantigen-like protein 7 (SSL7), among others. Phagocytosed bacteria can survive within the PMNs by producing catalase, superoxide dismutase [Mn] 1 (SodA), staphylococcal peroxidase inhibitor (SPIN),staphyloxanthin (against the bactericidal oxidative burst generated by the PMNs)72 and extracellular adherence protein (Eap) (against elastase), and the cell envelope modifications mediated by multiple peptide resistance factor (MprF) and the d-alanine transfer proteins DltA, DltB, DltC and DltD protect against defensins. S. Aureus also secretes cytolytic toxins that can kill PMNs; S. aureus leukocidins include large pore-forming proteins (α-toxin and several two-component leukocidins, such as Panton–Valentine-Leukocidin (PVL)) and small peptide (PSM peptides) toxins. Superantigen toxins (toxic shock syndrome toxin 1 (TSST1), enterotoxin type A (SEA), staphylococcal enterotoxin-like X (SEIX) and several others) contribute to exuberant inflammation by nonspecific T cell activation. C | Abscesses can release live bacteria to the surface of the skin and/or the bloodstream at later stages; the plasminogen-activating protein staphylokinase might contribute to bacterial dissemination. APC, antigen-presenting cell.81

Virulence

The wide range of infections that S. aureus can cause27,28 is a reflection of its extensive arsenal of virulence factors, which include adhesive, host-cell damaging, and immuno-modulatory molecules and can vary in their presence or specificity between clones.29,30 Since a large number of virulence genes are present on mobile genetic elements, their combination varies greatly between clones and even between strains that are distantly related. Because many of these virulence factors have redundant or partially overlapping functions, it is still unclear whether they are associated with particular types or levels of aggressiveness in S. aureus infections. Additionally, because they are human-specific, many virulence variables cannot be studied in animal models.31 The most notable virulence mechanisms and common entry points are the main topics of this section (Table 1).

Type of virulence factors

Virulence factors

Corresponding host ligands

MAMPs

Chemotactic MAMPs

*Formylated peptides

* N-Formyl-peptide receptor (FPR) 1 and FPR2

*Phenol-soluble modulins (PSMs)

Non-chemotactic MAMPs

* Lipoproteinsb

* Toll-like receptor (TLR) 2 and TLR9

* DNA

* Nucleotide-binding oligomerization

* Peptidoglycan

domain-containing protein 2 (NOD2)

Adhesins

Surface proteins

* Fibronectin-binding protein A (FnBPA) and FnBPB

* Fibronectin

* Collagen adhesin (Cna)b

* Collagen

* Iron-regulated surface determinant protein A (IsdA)

* Cytokeratin 10 (also known as KRT10)

* Loricrin

Glycopolymers

* Wall teichoic acid (WTA)

Scavenger receptors

Evasins

MAMP receptor inhibitors

* Chemotaxis inhibitory protein of S. aureus (CHIPS)b

FPR1

* FPR-like 1 (FPRL1) inhibitory protein (FLIPr)b

•C5a anaphylatoxin chemotactic receptor (C5aR1)

* FLIPr-likeb

• FPR2

* Staphylococcal superantigen-like protein 3 (SSL3)b

• TLR2

* SSL5b

Chemokine receptor inhibitors

* SSL5b

* Several chemokine receptors

* SSL10b

PMN extravasation inhibitors

* SSL5b

* P-Selectin glycoprotein ligand 1 (PSGL1)

* Extracellular adherence protein (Eap)b

* Intercellular adhesion molecule 1(ICAM1)

Coagulation factors

* Coagulase (Coa)

* Prothrombin

* Secreted von Willebrand factor binding protein (vWbp)

* Fibrinogen

* Clumping factor A (ClfA)

* ClfB

Anticoagulants

* Staphylokinaseb

* Plasmin

Complement inhibitors

* Zinc metalloproteinase aureolysin

* Complement proteins C3, C3b, C3bBb and C5a

* Staphylococcal complement inhibitor (SCIN)b

* Complement factor H

* Fibrinogen-binding protein (Efb)b

* Extracellular complement-binding protein (Ecb, also known    

as extracellular fibrinogen-binding protein)b

* SSL7b

* Immunoglobulin-binding protein Sbib

Opsonophagocytosis inhibitors

* Staphylococcus protein A (SpA)

* Immunoglobulin G (IgG)

* Immunoglobulin-binding protein Sbib

* Immunoglobulin-γ receptor (FcγR)

* Microcapsuleb

* FLIPrb

Synthases of anti-phagocytic mediators    

* Adenosine synthase (AdsA)

* Adenosine monophosphate

Inhibitors of PMN killing

* Catalase

* Reactive oxygen species

* Superoxide dismutase [Mn] 1 (SodA)

* Elastase

* Staphyloxanthin

* Lysozyme

* Eapb

* Defensins

* Staphylococcal peroxidase inhibitor (SPIN)

* Neutrophil extracellular traps (NETs)

* O-Acetyltransferase A (OatA)

* Multiple peptide resistance factor (MprF)

* d-alanine transfer protein A (DltA), DltB, DltC and DltD

* Thermonuclease (Nuc)

Toxins

Pore-forming protein toxins

* α-Toxin

* Disintegrin and metalloproteinase

* Bi-component γ-Haemolysin (Hlg) AB

domain-containing protein 10 (ADAM10)

* Bi-component HlgCB

* Several chemokine receptors

* Leukocidin (Luc) EDb

* Duffy antigen/chemokine receptor (DARC,

* LucAB

also known as atypical chemokine receptor 1)

* Panton–Valentine leukocidin (PVL)b

* C5aR1

Pore-forming peptide toxins

* PSMα1–PSMα4

* Host cell membranes

* PSMβ1 and PSMβ2

* SCCmec-encoded PSM (PSMmec)b

* δ-Toxin (also known as δ-haemolysin, Hld)

Toxins (cont.)

Superantigen toxins

* Toxic shock syndrome toxin 1 (TSST1)b

* Major histocompatibility complex (MHC) class II

* Enterotoxins types (SE) A-Qb

* T cell receptor

* Staphylococcal enterotoxin-like X (SEIX)b

Sphingomyelinase

* β-Haemolysin (Hlb)b

* Sphingomyelin

Proteolytic toxins

* Exfoliative toxins (Etx)b

* Desmoglein 1

Table 1 Major Staph. aureus virulence factors and corresponding host ligands83

Starting of infection

S. aureus SSTIs are typically started by bacterial transfer from the main reservoir in the nose to open micro-lesions and wounds on the skin.32,33 This transfer is most likely caused by hand contact. S. aureus surface proteins that bind to extracellular matrix proteins, such as fibronectin-binding protein A (FnBPA), FnBPB, clumping factor A (ClfA), ClfB, and collagen adhesin (Cna), allow the bacteria to adhere to and grow on damaged tissues.34 S. aureus is a common cause of catheter-associated infections, joint replacement infections, and ventilator-associated pneumonia due to its ability to adhere to and form biofilms (sticky agglomerations of microorganisms embedded in an extracellular matrix that facilitate resistance to mechanical interference, host defences, and antibiotic treatment) on artificial plastic or metal surfaces.35 S. aureus manipulates the subsequent influx of polymorphonuclear leukocytes (PMNs),36 which shapes local inflammation.37

Abscess formation

Infiltrating PMNs and bacteria are surrounded by a fibrin pseudo-capsule that formed by the S. aureus coagulase proteins, which inhibits further leukocyte influx.38 For example, S. aureus can prevent opsonization by producing a polysaccharide microcapsule and blocking the complement cascade.39 However, crucial MRSA clones like USA300 lack the microcapsule (REF.63). In addition to resisting PMN death mechanisms.40,41 bacteria that are phagocytosed by PMNs can also survive killing over time with the use of cytolytic toxins. For instance, many CA-MRSA clones produce α-haemolysin, pore-forming peptides (phenol soluble modulins, or PSMs) and a number of bi-component leukocidins, such the Panton-Valentine leukocidin (PVL), that are host species-specific and bind to host leukocyte membranes, causing holes to develop and lytic cell death to occur.42 so boosting bacterial virulence. S. aureus superantigen toxins, which bind to MHC class II antigen-presenting cells and activate a significant portion of T cells non-specifically, exacerbate the severe inflammation induced by activated or necrotic PMNs. This systemic hyper-inflammation is known as “cytokine storms.”43

Fulminant systemic inflammation

At a later stage, abscesses may rupture, releasing pus and live bacteria either onto the skin’s surface to aid in the spread of pathogens or into the bloodstream to result in bacteremia. Endovascular S. aureus has the ability to stick to endothelial surfaces and platelets.44,45 This adhesion can cause endocarditis, encourage the growth of metastatic abscesses, or cause bacterial uptake into endothelial cells, where the bacteria are difficult for antibiotics and host defence molecules to reach.46 Systemic blood coagulation is thought to be aided by coagulases’ agglutinating activity, and if the endovascular spread of the bacteria is not controlled, massive releases of cytokines and molecules associated with microorganisms result in sepsis, multi-organ failure, and fulminant systemic inflammation.47

Adaptation and regulation

The quorum-sensing system of the accessory gene regulator (Agr) and other regulatory networks48 are responsible for differentially regulating the majority of S. aureus virulence factors. Many CA-MRSA clones, including USA300, have extremely active Agr systems, which promote the profuse expression of toxins and are associated with a high propensity to infect even healthy people with SSTIs and invasive infections.49 In contrast, many HA-MRSA clones also have a phenol-soluble modulin that is encoded by the SCCmec (PSM; PSMmec), whose mRNA inhibits the expression of Agr.50 As a result, Agr is not highly active in many HA- MRSA clones, which produce more adhesins than toxins and frequently lead to bacteraemia through contaminated catheters or implanted medical devices. High virulence may even be harmful for S. aureus in bacteremia, as evidenced by the discovery of many isolates from bloodstream infections carrying point mutations that render Agr inactive.51 For the development of novel preventive and therapeutic approaches against MRSA, it will be essential to elucidate the virulence mechanisms whose inhibition would make S. aureus most vulnerable.

Methicillin resistance mechanisms

The independent acquisition of the SCCmec complex by several multidrug-resistant strains (resistant to penicillin, streptomycin, tetracycline, and erythromycin) in the early 1960s, which made S. aureus resistant to the majority of members of the β- lactam family of antibiotics,52 was a significant event in the evolution of S. aureus. The cassette chromosome recombinase (ccr) complex type and the class of the mec complex are used to categorise the twelve known SCCmec kinds (I-XII) (Table 2). Large SCCmec elements of Types I, II, and III, which are typically seen in HA- MRSA,53 include genes that confer resistance to various antibiotic classes. CA- MRSA, such as USA300 and USA400, contains smaller elements, such as kinds IV and V SCCmec, as well as in a few well-known HA-MRSA clones, as ST22-MRSA-IV, ST45-MRSA-IV, and ST5-MRSA-VI. The line separating the two epidemiological groups (HA-MRSA and CA-MRSA) has, however, blurred over time (Table 2).54

SCCmec types

mec determinant

ccr gene complexesa

mec gene complexes

High-prevalence setting

I

mecA

1 (A1B1)

B

HA-MRSA

II

mecA

2 (A2B2)

A

HA-MRSA

III

mecA

3 (A3B3)

A

HA-MRSA

IV

mecA

2 (A2B2)

B

CA-MRSA and HA-MRSA

V

mecA

5 (C1)

C2

CA-MRSA and HA-MRSA

VI

mecA

4 (A4B4)

B

HA-MRSA

VII

mecA

5 (C1)

C1

NAb

VIII

mecA

4 (A4B4)

A

NAb

IX

mecA

1 (A1B1)

C2

NAb

X

mecA

7 (A1B6)

C1

NAb

XI

mecC

8 (A1B3)

E

LA-MRSA

XII

mecA

9 (C2)

C2

NAb

Table 2 Currently identified SCCmec types in Staphylococcus aureus strains
(Parentheses indicate the ccr gene(s) in the ccr gene complex. B Not possible to assign this SCCmec type because there is insufficient information concerning its occurrence. CA-MRSA, Community-associated methicillin-resistant Staphylococcus aureus; HA-MRSA, health-care-associated methicillin-resistant Staphylococcus aureus; LA-MRSA, livestock-associated Methicillin-resistant Staphylococcus aureus; NA, not applicable. Adapted with permission From International Working Group on the Staphylococcal Cassette Chromosome elements).

All SCCmec types have mecA, which encodes penicillin-binding protein 2a (PBP2a), a peptidoglycan transpeptidase, with the exception of type XI, which contains the homologue mecC.55 PBP2a can take over the transpeptidase function of peptidoglycan biosynthesis in the presence of β-lactam antibiotics that inhibit the activity of the four native S. aureus penicillin-binding proteins (PBP1, PBP2, PPB3, and PBP4). PBP2a has a very low affinity for most β -lactam antibiotics. MecA encodes PBP2aLGA, which is named after the MRSA strain LGA251 from which it was first isolated. MecC, a variant of mecA, was discovered in several S. aureus clones from animal and human isolates.56 When compared to MRSA strains that carry the mecA gene, the LGA251 strain’s control of β-lactam resistance has a different mechanism.57,58 The level of methicillin resistance in the LGA251 strain is dependent on the mecC gene as well as genes in the strain’s genetic background.

S. aureus has been found to have plasmid-borne methicillin resistance based on mecB in 2018, but the mechanism of resistance encoded by mecB has not yet been elucidated.59 MecA is primarily regulated by the regulators encoded by the genes mecI, mecR1 and mecR2,60,61 as well as by the regulators of the expression of the genes blaZ, blaI, and blaRI.62 A surprising number of auxiliary or accessory genes also have a significant impact on the resistance phenotype.63 The degree of methicillin resistance cannot be predicted by the level of mecA transcription, according to three lines of data. First, the antibiotic mupirocin induces a stringent stress response, or the bacterial response to various stress conditions such as amino acid, fatty acid, and iron limitation as well as heat shock, which results in an increase in PBP2a activity without affecting mecA transcription.64 Second, inactivating vraS (a component of the two-component regulatory system including sensor protein VraS and response regulator protein VraR (VraS-VraR) involved in the control of the cell wall peptidoglycan biosynthesis) increased mecA transcription but not PBP2a activation.65 Thirdly, PrsA, a chaperone foldase protein, modifies the amount of correctly folded PBP2a in the membrane and, consequently, methicillin resistance without influencing the transcription of mecA.66 Utilizing various experimental techniques, it has been demonstrated that the severe stress response is essential for mecA expression.67 Finding inhibitors of the stringent stress response that interact with -lactam antibiotics is the subject of a new line of research.67 Notably, since the 1960s, vancomycin, the first-line treatment for invasive MRSA infections in hospitalised patients, has developed resistance in several MRSA clones (Figure 3).68

Figure 3 Bacterial targets of antibiotics active against MRSA. Antibiotics have diverse mechanisms of action and target different bacterial structures or metabolic pathways. Existing antibiotic options are in green, new antibiotics approved and on the market are in blue and antibiotics in the pipeline are in orange.
DHFA, dihydrofolic acid; PABA, para-aminobenzoic acid; PBP, penicillin-binding protein; S. Aureus, Staphylococcus aureus; THFA, tetrahydrofolic acid.82

Vaccines

The incidence and course of MRSA infections could be significantly impacted by developing vaccines. Infections do occur more frequently in S. aureus carriers, but they are also less severe69 than infections that develop in non-carriers, suggesting that sustained exposure to S. aureus antigens can result in protective immunity. A vaccine would ideally also hinder S. aureus colonisation in order to prevent infections from occurring in the first place, greatly reducing the need for antibiotic treatment and intensive infection control measures.70 Therapeutic monoclonal antibodies, such as the passive vaccination produced by Medimmune (Gaithersburg, Maryland, USA) against α-haemolysin, may open up new therapeutic avenues, either by themselves or in conjunction with antibiotics. The next few years won’t see the development of a S. aureus protective vaccine, despite intensive research and development efforts. In early clinical testing, two monovalent vaccine candidates were unsuccessful in producing sufficient protection. Both the StaphVax (Nabi Biopharmaceuticals, Rockville, Maryland, USA) and the V710 (Merck, Kenilworth, New Jersey, USA) vaccines, which contain the capsular polysaccharide 5 (CP5) and CP8 antigens, have been shown to be protective in animal models but not in placebo-controlled phase III trials.71,72 The lack of CP expression in several significant S. aureus clones, such as the significant MRSA clone USA300, the absence of adjuvants in the antigen preparations, and inconsistent immune responses to the used antigens may all have contributed to the failure. There is also widespread concern that the many immune evasion mechanisms used by S. aureus, including immunoglobulin G (IgG)-binding protein A, may reduce the effectiveness of antibodies, that opsonizing antibodies may not be sufficient to promote protection and that toxin-neutralizing antibodies may be just as important as or even more than previously believed.73

Basic scientific developments offer hints for new, perhaps more effective immunisation strategies.The cell wall glycopolymer wall teichoic acid (WTA) has been identified as a dominant surface antigen,74 and immunoproteomics studies have assisted in elucidating the most immunogenic and protective S. aureus antigens.75 There have been a number of novel toxins discovered, whose neutralisation by antibodies may help provide protection. It is also now more obvious which T cell subsets are necessary for anti-S. immunity.76

Pharmaceutical companies are still working on polyvalent anti-S. aureus vaccines based on the bacteria’s surface proteins (ClfA) and polysaccharides (CP5 and CP8), secreted toxins (-toxin, LukS-PV, ESAT-6 secretion system extracellular protein A (EsxA) and EsxB), and membrane-bound lipoproteins that are involved in nutrient uptake (manganese transport system membrane). In preclinical infection studies, a novel WTA-targeting monoclonal antibody coupled to a rifampicin-related antibiotic shown protection.77 There is optimism that certain current vaccine development initiatives may result in the successful conclusion of clinical trials.

Priorities and new approaches for research:

MRSA and humanity will probably always cohabit. The biomedical research community would be well advised to continue its varied activities in the field of MRSA research notwithstanding the present emphasis on multidrug-resistant Gram-negative bacteria and the fall of HA-MRSA infections in some areas. MRSA continues to be a high-priority multidrug-resistant bacterium that requires continued efforts for the research and development of new antibiotics and cutting- edge prevention measures, as noted by a 2017 WHO report.78 In addition to vaccinations, bacteriophages or lytic proteins produced from bacteriophages may be employed for novel preventive measures, such as nasal MRSA decontamination in a time of rising mupirocin resistance.79 Overall, there are still many unanswered questions and significant problems to solve, which necessitate continued attention from academics, decision-makers in government policies, funders, and those in charge of MRSA treatment and management. In the 60 years after it was originally identified, MRSA has proven to have a remarkable capacity for change and widespread dissemination. Control of this extremely successful pathogen will ultimately be made easier by a number of factors, including improved understanding of the pathogenesis of infection, accurate and speedy diagnostics, assurance of the availability of effective treatment options, and optimization of the prevention of transmission and infection.

Conclusion

The future research needs for MRSA can be summarized in the following points:

  1. MRSA Life cycle and survival
  2. Clones of MRSA: prediction modelling and simulation of future introduction of successful community-associated MRSA strains into the health-care environment
  3. Virulence of MRSA: development of new inhibition strategies targeted to prevention and treatment of invasive MRSA
  4. Interplay between the host, aureus and competing nasal microbiota: microbiome studies on the competition between S. aureus and other commensal microbiota.
  5. New decolonization regimens and approaches to decrease risk of MRSA infection: Development and clinical evaluation of new drugs, vaccines and other preventive
  6. Novel approaches: discovery of inhibitors of the Aureus stringent stress response or other S. aureus factors that can sensitize MRSA to β-lactam antibiotics.
  7. Whole-genome sequencing (WGS): effectiveness studies on WGS usefulness for clinical diagnostics and rapid antibiotic susceptibility
  8. Biomarkers: rapid molecular diagnostic tools and biomarkers for individual risk-profiling and treatment approaches
  9. MRSA reservoirs in animals: determinants of MRSA host tropism and host jumps.

Acknowledgments

None.

Conflicts of interest

Author declares there are no confits of interest.

Funding

None.

References

  1. Wertheim HF. et al. The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect Dis. 2005;5(12):751–762.
  2. Becker K, Schaumburg F, Fegeler C, et al. Staphylococcus aureus from the German general population is highly diverse. Int J Med Microbiol. 2017;307(1):21–27.
  3. Kuehnert, M. J. et al. Prevalence of Staphylococcus aureus nasal colonization in the United States, 2001–2002. J Infect Dis. 2006.193:172–179.
  4. Lowy FD. Staphylococcus aureus infections. N Engl J Med. 1998;339:520–532.
  5. Jevons M. Celbenin-resistant staphylococci. Ugeskr Laeger. 1961;124–125.
  6. Dzintars K. in Kucers’ The Use of Antibiotics. In: Grayson M, Editor. USA: CRC Press; 20198. p. 136–142.
  7. Chambers HF, Deleo FR. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat Rev Microbiol. 2009;7(9):629–641.
  8. Voss A, Loeffen F, Bakker J, et al. Methicillin-resistant Staphylococcus aureus in pig farming. Emerg Infect Dis. 2005;11(12):1965–1966.
  9. Tenover FC, Arbeit RD, Goering RV, et al. Interpreting chromosomal DNA restriction patterns produced by pulsed-field gel electrophoresis: criteria for bacterial strain typing. J Clin Microbiol. 1995;33(9):2233–2239.
  10. Ito T Katayama Y, Hiramatsu K. Cloning and nucleotide sequence determination of the entire mec DNA of pre-methicillin-resistant Staphylococcus aureus N315. Antimicrob. Agents Chemother. 1999;43(6):1449–1458.
  11. Kluytmans J, van Belkum A, Verbrugh H. Nasal carriage of Staphylococcus aureus: epidemiology, underlying mechanisms, and associated risks. Clin Microbiol Rev. 1997;10(3):505–520.
  12. Sim BL, McBryde E, Street AC, et al. Multiple site surveillance cultures as a predictor of methicillin-resistant Staphylococcus aureus infections. Infect Control Hosp Epidemiol. 2013;34(8):818–824.
  13. Eriksen NH, Espersen F, Rosdahl VT, et al. Carriage of Staphylococcus aureus among 104 healthy persons during a 19-month period. Epidemiol Infect. 1995;115:51–60.
  14. Hoffler U, Bulanda M, Heczko PB, et al. A comparison of staphylococcal nasal carrier rates in Germany and Poland. Med Microbiol Immunol. 1978;164(4):285–290.
  15. Emonts M, Uitterlinden AG, Nouwen JL, et al. Host polymorphisms in interleukin 4, complement factor H, and C-reactive protein associated with nasal carriage of Staphylococcus aureus and occurrence of boils. J Infect Dis. 2008;197(9):1244–1253.
  16. Lucet JC, Paoletti X, Demontpion C, et al. Carriage of methicillin-resistant Staphylococcus aureus in home care settings: prevalence, duration, and transmission to household members. Arch Intern Med. 2009;169(15):1372–1378.
  17. Burian M, Rautenberg M, Kohler T, et al. Temporal expression of adhesion Factors and activity of global regulators during Establishment of Staphylococcus aureus nasal Colonization. J Infect Dis. 2010;201(9):1414–1421.
  18. Weidenmaier C, Kokai-Kun JF, Kulauzovic E, et al. Differential roles of sortaseanchored surface proteins and wall teichoic acid in Staphylococcus aureus nasal colonization. Int J Med Microbiol. 2008;298(5-6):505–513.
  19. Wertheim HF, Walsh E, Choudhurry R, et al. Key role for clumping factor B in Staphylococcus aureus nasal colonization of humans. PloS Med. 2008.5(1):e17.
  20. O’Brien LM, Walsh EJ, Massey RC, et al. Staphylococcus aureus clumping factor B (ClfB) promotes adherence to human Type I cytokeratin 10: implications for nnasal colonization. Cell Microbiol. 2002;4(11):759– 770.
  21. Liu CM, Price LB, Hungate BA, et al. Staphylococcus aureus and the ecology of the nasal microbiome. Sci Adv. 2015;1(5):e1400216.
  22. Lemon KP, Klepac-Ceraj V, Schiffer HK, et al. Comparative analyses of the Bacterial microbiota of the human nostril and oropharynx. mBio. 2010;1(3):e00129-10 .
  23. Krismer B, Liebeke M, Janek D, et al. Nutrient limitation governs Staphylococcus aureus metabolism and niche adaptation in the human nose. PLoS Pathog. 2014;10(1):e1003862.
  24. Zipperer A, Konnerth M C, Laux C, et al. Human commensals producing a novel antibiotic impair pathogen colonization. Nature. 2016;535:511–516.
  25. Kaspar U, Kriegeskorte A, Schubert T, et al. The culturome of the human nose habitats reveals individual bacterial fingerprint patterns. Environ Microbiol. 2016;18(7):2130–2142.
  26. Krismer B, Weidenmaier C, Zipperer A, et al. The commensal lifestyle of Staphylococcus aureus and its interactions with the nasal microbiota. Nat Rev Microbiol. 2017;15:675–687.
  27. Boyle-Vavra S, Li X, Alam T, et al. USA300 and USA500 clonal lineages of Staphylococcus aureus do not produce a capsular polysaccharide due to conserved mutations in the cap5 locus. mBio. 2015;6(2):02585-14.
  28. Spaan AN, van Strijp VTorres VJ. Leukocidins: staphylococcal bi- component poreforming toxins find their receptors. Nat Rev Microbiol. 2017;15(7):435– 447.
  29. Laabei, M, Recker M, Rudkin JK, et al. Predicting the virulence of MRSA from its genome sequence. Genome Res. 2014;24(5):839–849.
  30. Thammavongsa V, Kim HK, Missiakas D, et al. Staphylococcal manipulation of host immune responses. Nat Rev Microbiol. 2015;13(9):529–543.
  31. Recker M, Laabei M, Michelle S, et al. Clonal differences in Staphylococcus aureus bacteraemia- associated mortality. Nat Microbiol. 2017;2(10):1381–1388.
  32. Kluytmans JA, MoutonJW, Ijzerman EP, et al. Nasal carriage of Staphylococcus aureus as a major risk factor for wound infections after cardiac surgery. J Infect Dis. 1995;171(1):216–219.
  33. von Eiff C, Becker K, Machka K, et al. Nasal carriage as a source of Staphylococcus aureus bacteremia. N Engl J Med. 2001;344(1):11–16.
  34. Foster TJ, Geoghegan JA, Ganesh VK, et al. Adhesion, invasion and evasion: the many functions of the surface proteins of Staphylococcus aureus. Nat Rev Microbiol. 2014;12(1):49–62.
  35. Vanhommerig E, Moons P, Pirici D, et al. Comparison of biofilm formation between major clonal lineages of methicillin resistant Staphylococcus aureus. PLoS One. 2014;9(8):e104561.
  36.  Spaan AN, Surewaard BG, Nijland R, et al. Neutrophils versus Staphylococcus aureus: a biological tug of war. Annu Rev Microbiol. 2013;67: 629–650.
  37. Hanzelmann D, Joo HS, Franz-Wachtel M, et al. Toll-like receptor 2 activation depends on lipopeptide shedding by bacterial surfactants. Nat Commun. 2016;7:12304.
  38. Cheng AG, DeDent AC, Schneewind O, et al. A play in four acts: Staphylococcus aureus abscess formation. Trends Microbiol. 2011;19(5):225–232 .
  39. Laarman A, Milder F, van Strijp J, et al. Complement inhibition by gram-positive pathogens: Molecular mechanisms and therapeutic implications. J Mol Med. 2010;88(2):115–120.
  40. Stapels DA, Ramyar KX, Bischoff M, Staphylococcus aureus secretes a unique class of neutrophil serine protease inhibitors. Proc Natl Acad Sci USA. 2014;111(36):13187–13192.
  41. Peschel A, Sahl HG. The co-evolution of host cationic antimicrobial peptides and microbial resistance. Nat Rev Microbiol. 2006;4(7):529–536.
  42. Loffler B, Muzaffar M, Grundmeier M, et al. Staphylococcus aureus pantonvalentine leukocidin is a very potent cytotoxic factor for human neutrophils. PloS Pathog. 2010;6:e1000715.
  43. Spaulding AR, W Salgado-Pabón W, Kohler PL, et al. Staphylococcal and streptococcal superantigen exotoxins. Clin Microbiol Rev. 2013;26(3):422–447.
  44. Chavakis T, Wiechmann K, Preissner KT, et al. Staphylococcus aureus interactions with the endothelium: the role of bacterial “secretable expanded repertoire adhesive molecules” (SERAM) in disturbing host defense systems. Thromb Haemost. 2005;94(2):278–285.
  45. Weidenmaier C, Peschel A, Y Xiong YQ, et al. Lack of wall teichoic acids in Staphylococcus aureus leads to reduced interactions with endothelial cells and to attenuated virulence in a rabbit model of endocarditis. J Infect Dis. 2005;191(10):1771–1777.
  46. Loffler B, Tuchscherr L, Niemann S, et al. Staphylococcus aureus persistence in non-professional phagocytes. Int J Med Microbiol. 2014;304:170–176.
  47. Thomer L, Schneewind O, Missiakas D, et al. Pathogenesis of Staphylococcus aureus bloodstream infections. Annu Rev Pathol. 2016;11:343–364.
  48. Le KY, Otto M. Quorum-sensing regulation in Staphylococci-an overview. Front Microbiol. 2015;6:1174.
  49. Cheung GY, Wang R, Khan BA, et al. Role of the accessory gene regulator agr in community-associated methicillin-resistant Staphylococcus aureus pathogenesis. Infect Immun. 2011;79(5):1927–1935.
  50. Kaito C, Saito Y, Ikuo M, et al. Mobile genetic element SCCmecencoded psm-mec RNA suppresses translation of agrA and attenuates MRSA virulence. PLoS Pathog. 2013;9:e1003269.
  51. Painter KL, Krishna A, Wigneshweraraj S, et al. What role does the quorum-sensing accessory gene regulator system play during Staphylococcus aureus bacteremia? Trends Microbiol. 2014;22:676–685.
  52. Crisostomo MI, Westh H, Tomasz A, et al. The evolution of methicillin resistance in Staphylococcus aureus: similarity of genetic backgrounds in historically early Methicillin- susceptible and -resistant isolates and contemporary epidemic clones. Proc Natl Acad Sci USA. 2001;98(17):9865–9870.
  53. International Working Group on the Classification of Staphylococcal Cassette Chromosome Elements (IWG-SCC). Classification of staphylococcal cassette chromosome mec (SCCmec): guidelines for reporting novel SCCmec elements. Antimicrob Agents Chemother. 2009;53:4961–4967.
  54. Bal AM, Coombs GW, Holden MTG, et al. Genomic insights into the emergence and spread of international clones of healthcare community and livestock-associated meticillin resistant Staphylococcus aureus: Blurring of the raditional definitions. J Glob Antimicrob Resist. 2016;6:95–101.
  55. Hartman BJ, Tomasz A. Low-affinity penicillinbinding protein associated with beta-lactam resistance in Staphylococcus aureus. J Bacteriol. 1984;158:513–516.
  56. Garcia-Alvarez L, Holden MTG, Lindsay H, et al. Meticillin-resistant Staphylococcus aureus with a novel mecA homologue in human and bovine populations in the UK and Denmark: a descriptive study. Lancet Infect Dis. 2011;11:595–603.
  57. Kim C, Milheiriço C, Gardete S, Properties of a novel PBP2A protein homolog from Staphylococcus aureus strain LGA251 and its contribution to the beta-lactam-resistant phenotype. J Biol Chem. 2012;287:36854–36863.
  58. Milheirico C, de Lencastre H, Tomasz A. Full-genome sequencing identifies in the genetic background several determinants that modulate the resistance phenotype in methicillin-resistant Staphylococcus aureus strains carrying the novel mecC gene. Antimicrob Agents Chemother. 2017;61(3):e02500–16.
  59. Becker K, van Alen S, Idelevich EA, et al. Plasmid-encoded transferable mecBmediated methicillin resistance in Staphylococcus aureus. Emerg Infect Dis. 2018;24(2):242–248.
  60. Hiramatsu K, Asada K, Suzuki E, et al. Molecular cloning and nucleotide sequence determination of the regulator region of mecA gene in methicillin-resistant Staphylococcus aureus (MRSA). FEBS Lett. 1992;298(2-3):133–136.
  61. Arede P, Milheirico C, de Lencastre H, et al. The anti-repressor MecR2 promotes the proteolysis of the mecA repressor and enables optimal expression of beta-lactam resistance in MRSA. PloS Pathog. 2012;8:e1002816.
  62. Zhang, HZ, Hackbarth CJ, Chansky KM, et al. A proteolytic transmembrane signaling pathway and resistance to beta-lactams in Staphylococci. Science. 2001;291(5510):1962–1965.
  63. De Lencastre H, Wu SW, Pinho MG, et al. Antibiotic resistance as a stress response: complete sequencing of a large number of chromosomal loci in Staphylococcus aureus strain COL that impact on the expression of resistance to methicillin. Microb Drug Resist. 1999;5(3):163–175.
  64. Kim C, Mwangi M, Chung M, et al. The mechanism of heterogeneous beta-lactam resistance in MRSA: key role of the stringent stress response. PloS ONE. 2113;8:e82814 .
  65. Boyle-Vavra S, Yin S, Daum RS. The VraS/VraR two-component regulatory system required for oxacillin resistance in community-acquired methicillin- resistant Staphylococcus aureus. FEMS Microbiol Lett. 2006;262:163–171 (2006).
  66. Jousselin A, Manzano C, Biette A. The Staphylococcus aureus chaperone PrsA is a new auxiliary factor of oxacillin resistance affecting penicillin-binding protein 2A. Antimicrob. Agents Chemother. 2015;60(3):1656–1666.
  67. Kim CK, Milheirico C, de Lencastre H, et al. Antibiotic resistance as a stress response: recovery of high-level oxacillin resistance in methicillin-resistant Staphylococcus aureus “Auxiliary” (fem) mutants by induction of the stringent stress response. Antimicrob. Agents Chemother. 2017;61:E00313–17.
  68. Gardete S, Tomasz A. Mechanisms of vancomycin resistance in Staphylococcus aureus. J Clin Invest. 2014;124(7):2836–2840.
  69. Wertheim HF, Vos MC, Ott A, et al. Risk and outcome of nosocomial Staphylococcus aureus bacteraemia in nasal carriers versus non-carriers. Lancet. 2004;364(9435):703–705.
  70. Pozzi C, Olaniyi R, Liljeroos L, et al. Vaccines for Staphylococcus aureus and target populations. Curr Top Microbiol Immunol. 2017;409:491–528.
  71. Fowler VG. Allen KB, Edson D Moreira, et al. Effect of an investigational vaccine for preventing Staphylococcus aureus infections after cardiothoracic surgery: a randomized trial. JAMA. 2013;309:1368–1378.
  72. Shinefield H, Black S, Fattom A, et al. Use of a Staphylococcus aureus conjugate vaccine in patients receiving hemodialysis. N Engl J Med. 2002;346:491–496.
  73. Fowler VG, Proctor RA. Where does a Staphylococcus aureus vaccine stand? Clin Microbiol Infect. 2014;20(Suppl. 5):66–75.
  74. Stentzel S, Sundaramoorthy N, Michalik S, et al. Specific serum IgG at diagnosis of Staphylococcus aureus bloodstream invasion is correlated with disease progression. J Proteom. 2015;128:1–7.
  75. Kurokawa K, Takahashi K, Lee BL. The staphylococcal surface- glycopolymer wall teichoic acid (WTA) is crucial for complement activation and immunological defense against Staphylococcus aureus infection. Immunobiology. 2016;221(10):1091–1101.
  76. Bekeredjian-Ding I. Deciphering the significance of the T cell response to Staphylococcus aureus. Future Microbiol. 2017;12:1023–1026.
  77. Lehar SM, Pillow T, Xu M, et al. Novel antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature. 2015;527(7578):323–328.
  78. World Health Organization. Prioritization of Pathogens to Guide Discovery, Research and Development of New Antibiotics for Drug-Resistant Bacterial Infections, Including Tuberculosis. WHO, Geneva: World Health Organization; 2017.
  79. Idelevich EA, von Eiff C, Friedrich AW, et al. In vitro activity against Staphylococcus aureus of a novel antimicrobial agent, PRF-119, a recombinant chimeric bacteria phage Endolysin. Antimicrob Agents Chemother. 2011;55:4416–4419.
  80. Thurlow LR, Joshi GS, Richardson AR. Virulence strategies of the dominant USA300 lineage of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA). FEMS Immunol Med Microbiol. 2012;65:5–22.
  81. Peschel A, Otto M. Phenol-soluble modulins and staphylococcal infection. Nat Rev Microbiol. 2013;11(10):667–673.
  82. Wright GD. Q&A: Antibiotic resistance: where does it come from and what can we do about it? BMC Biol. 2010;123.
  83. McCarthy AJ, Lindsay JA. Staphylococcus aureus innate immune evasion is lineage-specific: A bioinfomatics study. Infect Genet Evol. 2013;9:7–14.
Creative Commons Attribution License

©2022 Ahmed. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.