Bacterial Anti-adhesives: Inhibition of Staphylococcus aureus Nasal

Aug 2, 2019 - Bacterial adhesion to the skin and mucosa is often a fundamental and early step in host colonization, the establishment of bacterial inf...
0 downloads 0 Views 794KB Size
Subscriber access provided by Gothenburg University Library

Review

Bacterial anti-adhesives: inhibition of Staphylococcus aureus nasal colonization Allison C Leonard, Laurenne E Petrie, and Georgina Cox ACS Infect. Dis., Just Accepted Manuscript • DOI: 10.1021/acsinfecdis.9b00193 • Publication Date (Web): 02 Aug 2019 Downloaded from pubs.acs.org on August 3, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Bacterial anti-adhesives: inhibition of Staphylococcus aureus nasal colonization

Allison C. Leonard1, Laurenne E. Petrie1 & Georgina Cox1*

1College

of Biological Sciences, Department of Molecular and Cellular Biology, University of

Guelph, 50 Stone Rd E, Guelph, Ontario, Canada N1G 2W1. *Corresponding author:

[email protected]

ACS Paragon Plus Environment

1

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 45

Bacterial adhesion to the skin and mucosa is often a fundamental and early step in host colonization, the establishment of bacterial infections, and pathology. This process is facilitated by adhesins on the surface of the bacterial cell that recognize host cell molecules. Interfering with bacterial host cell adhesion—so-called anti-adhesive therapeutics—offers promise for the development of novel approaches to control bacterial infections. In this review, we focus on the discovery of anti-adhesives targeting the high priority pathogen Staphylococcus aureus. This organism remains a major clinical burden and S. aureus nasal colonization is associated with poor clinical outcomes. We describe the molecular basis of nasal colonization and highlight potentially efficacious targets for the development of novel nasal decolonization strategies.

Keywords Anti-adhesives; anti-virulence; Staphylococcus aureus; MRSA; antibiotic resistance; prophylactic therapeutics; colonization; ClfB; IsdA; WTA

ACS Paragon Plus Environment

2

Page 3 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Antibiotics are life-saving molecules that have been relied upon for >70 years to control bacterial infections. Unfortunately, we are now experiencing a global ‘antibiotic resistance crisis’ that seriously threatens our ability to treat infections with these molecules1. This crisis has been spurred by the emergence of multi-drug resistant pathogenic bacteria and is compounded by our failure to discover new classes of antibiotics2. Due to the selective pressure imparted by antibacterial chemotherapy, antibiotic use and antibiotic resistance are intricately connected. Despite initiatives to encourage antibiotic stewardship, there continues to be a steady increase in global antibiotic consumption, which is expected to increase a further 200% by 20303-4. In addition to the burden of resistance, antibiotics negatively disrupt the ecology of our microbiota, which has been associated with a wide variety of health problems5. In particular, antibiotics are extensively administered to the pediatric population and up to 50% of infants under 1 year of age will have been exposed6. Early life antibiotic exposure has been linked to a number of negative health outcomes later in life such as metabolic syndrome, obesity, and asthma7. Addressing the antibiotic resistance crisis requires a multi-faceted approach involving the identification of new antibiotic classes and/or vaccines, improved antibiotic stewardship, and global surveillance programs. The development of therapeutic alternatives to antibiotics is also prominent among the solutions to this crisis8. One novel alternative approach is the attenuation of bacterial virulence, which has gained increased attention over the last 10 years and there is a growing pipeline for such drug candidates9. The rationale behind this strategy is to disarm a pathogen’s infectious capability without impacting growth, rendering the bacteria more susceptible to the host’s immune response. An attractive and validated anti-virulence approach involves inhibiting the attachment of bacteria to the host’s skin and mucosa10-11, which is enabled by bacterial adhesins12. Bacterial attachment to the skin and mucosa is often a fundamental and early step in host colonization, the establishment of infections, and host pathology10. By adhering to the host, bacteria prevent physical clearance by cleansing mechanisms and attain proximity to the host cell, which allows

ACS Paragon Plus Environment

3

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 45

for the delivery of toxins, access to nutrients, colonization, biofilm formation and host cell invasion13. Adhesion is a common prerequisite for pathogenesis; initially, bacteria reversibly associate with host cells via weak non-specific interactions14. These transient interactions are reinforced by high-affinity interactions mediated by host cell specific bacterial adhesins. Bacteria possess a wide repertoire of these appendages, ranging from glycopolymers to monomeric proteins and complex multimeric proteinaceous assemblies12. From a drug discovery perspective, numerous stages of bacterial host cell adhesion could be targeted for the development of anti-adhesives and there are a number of factors to consider in the development of such compounds. Primarily, anti-adhesives are ‘precision therapeutics’, whereby they target bacterial specific virulence factors, leaving the remainder of the microbiota intact15. Indeed, the appendages used by bacteria to adhere to host tissues vary considerably, even amongst species. For example, virulent strains of Escherichia coli can cause a wide range of diseases including gastrointestinal and urinary tract infections (UTIs), which is dependent on the strain’s repertoire of virulence factors16. Some of the most promising examples of small molecule anti-adhesives target the carbohydrate recognition domains of uropathogenic E. coli fimbriae17-21, which enable adherence to the uroepithelium15, 22. These anti-adhesives form the basis of an orthogonal, or complementary, group of therapeutics to control UTIs15, 22. Clearly, anti-adhesives need to be tailored for specific pathogens and for use in different clinical contexts. A major advantage of anti-adhesive therapeutics is that they could alleviate some of the negative side effects associated with antibiotic use. Namely, antibiotics are intended to inhibit the growth of bacteria. As such, when a mixed population of bacteria are exposed, antibiotics naturally select for mutations that confer an advantage. The antibiotic resistant population of bacteria can then proliferate and outnumber the susceptible organisms. In contrast, antiadhesives are not intended to inhibit bacterial growth. Therefore, in principle, this approach ought to be less likely to promote and give rise to bacterial resistance due to reduced selective

ACS Paragon Plus Environment

4

Page 5 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

pressure11, 23. While it is conceivable (and likely) that bacteria could mutate, becoming resistant to anti-adhesives, resistant isolates will likely not infect the host and will be expelled by cleansing mechanisms10-11. Therefore, these organisms will not be continually exposed to a sustained selective pressure that disrupts the balance between resistant and susceptible organisms24. In addition, it is anticipated that the spread of anti-adhesive resistance amongst bacterial populations will occur at much lower frequencies10. Finally, since anti-adhesives target specific pathogens, their therapeutic use should not cause dysbiosis of our microbiota15. However, these potentially promising factors need to be experimentally validated in vivo, under physiologically relevant conditions. There are a number of well-described strategies for the development of anti-adhesive therapeutics 10-11, 24. Anti-adhesives can be broadly grouped into major categories depending on their mechanism of action (Figure 1)10-11, 24: competitive inhibitors that can mimic bacterial adhesins17-21 (Class I) and host cell receptors (Class II), molecules that inhibit the biosynthesis or surface presentation of adhesins25-26 (Class III) or host receptors (Class IV)27. Finally, the use of antibodies targeting bacterial adhesins has been well described (Class V)10-11, 24. This review will discuss clinical targets for the development of small molecule antiadhesive therapeutics aimed at the Gram-positive pathogen Staphylococcus aureus. Antibiotic resistant S. aureus is a major clinical burden, which is exacerbated by the commensal lifestyle of this organism28. Despite efforts to develop a S. aureus vaccine, none have progressed past phase III clinical trials29, signifying a critical need for alternative innovative approaches. Adhesion is an important facet of S. aureus pathogenesis and the adhesins facilitating this process are well understood29-30. Targeting adhesion could offer an orthogonal and efficacious approach to control S. aureus infections.

ACS Paragon Plus Environment

5

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 45

Figure 1: Schematic overview of bacterial anti-adhesives. The first panel shows a simplistic representation of S. aureus successfully adhering to host cells using cell wall-anchored (CWA) adhesins. The following panels depict the different classes of anti-adhesives. Methicillin-resistant S. aureus (MRSA): the need for new treatment options S. aureus is a colonizing opportunistic pathogen causing infections when the skin is breached and/or the host’s immune system is weakened. This organism is capable of causing a variety of diseases ranging from skin and soft tissue infections to severe systemic infections31. Antibiotic resistant S. aureus isolates, in particular methicillin-resistant S. aureus (MRSA), are associated with poorer clinical outcomes32. The World Health Organization reported global antibiotic resistance statistics showing 31-50% of S. aureus isolates are methicillin resistant33. The same report identified MRSA as a high priority for new antibiotic research and development efforts33. High proportions of MRSA-associated infections require treatment with second-line antibiotics, which are expensive and need monitoring during treatment due to undesirable side effects34. S. aureus persistently colonizes ~20% of the adult population and intermittently colonizes a further 30%; the nasal cavity is the natural niche/primary S. aureus reservoir in the human body35-36. S. aureus is considered to favour a dispersed—rather than biofilm-like—mode of growth in the nose37-38. It is important to note that the majority (~80%39) of invasive

ACS Paragon Plus Environment

6

Page 7 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

nosocomial infections arise from endogenous S. aureus isolates40-41. The association between S. aureus nasal carriage and wound infections was observed as early as the 1920s, which led investigations into ‘chemotherapeutic nasal snuffs’ to reduce colonization36. Currently, preoperative nasal decolonization with the antibiotic mupirocin is routinely used in the hospital setting and reduces the incidence of infections36, 42. However, mupirocin resistance is increasing and in some instances levels as high as 80% have been reported, threatening decolonization efficacy42. Indeed, there is a direct correlation between resistance emerging following exposure to mupirocin; increased use predisposes patients to resistance42. This observation is problematic since patients eradicated of S. aureus often become recolonized with isolates exhibiting decreased sensitivity to mupirocin36, 42. A strategy to decolonize that does not provide a strong selection for resistance is needed and anti-adhesives could be well-suited to this application. In summary, asymptomatic nasal carriage of S. aureus is a major risk factor for infections43-46 and for transmission in both the community and hospital settings. This review will discuss the molecular basis of this phenomenon, followed by an in-depth discussion of clinical targets to eradicate colonization. We will conclude with a review of promising known inhibitors of S. aureus adhesion.

The molecular basis of S. aureus nasal colonization: the host perspective Given that the nose is an important ecological niche, S. aureus tightly adheres to avoid mucociliary clearance, cell shedding and to compete with other commensal bacteria that make up the nasal microbiota28. There are two distinct S. aureus niches: the nasal vestibule (anterior nasal cavity; the nostril opening) and the inner nasal cavity, also known as the posterior nasal cavity (Figure 2). The epithelium of the nasal cavity in these two regions is distinct, which alters the molecular basis of S. aureus host cell adhesion, as described below.

Composition of the nasal vestibule and the inner nasal cavity

ACS Paragon Plus Environment

7

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 45

The nasal vestibule is lined with the same stratified and keratinized epithelium of the skin (Figure 2)47. During desquamation (skin shedding), proliferating keratinocytes within the basal layer begin their migration to the surface of the skin. Through this process they change morphology, increase keratinization and lose their nucleus, eventually forming the stratum corneum (the outermost layer of the skin) (Figure 2). The keratins are the major structural proteins on the surface of these epithelial cells; the K1 and K10 cytokeratin molecules are present in large quantities on the both the interior and exterior surface of these dead keratinized cells (Figure 2)48. The stratum corneum is surrounded by a proteinaceous structure referred to as the cornified envelope, whereby the cytoplasmic membrane of the mature squamous cells is replaced by the protein loricrin (~80% of the total protein content)49 and other proteins such as involucrin (Figure 2). This enveloping structure is strengthened by transglutination and decorated by the covalent linkage of ceramides and fatty acids50; the latter exhibits antimicrobial activity and is a component of the innate immune response51-52. S. aureus has the highest affinity for the mature squames on the outermost layer of the epithelium (i.e., the stratum corneum)53-54. Indeed, recent studies investigating the location of S. aureus in the nasal vestibule of healthy individuals confirmed the stratum corneum as the primary source (Figure 2)47. However, lower quantities of bacterial cells were also identified in both the upper and lower layers of the epidermis and were found intracellular and in the different layers of the skin47. Overall, cytokeratin K1, K10 and loricrin proteins within the stratum corneum are integral S. aureus host ligands enabling colonization of the nasal vestibule48, 55. The second S. aureus niche resides within the inner cavity vestibule (Figure 2). The epithelium within this region transitions toward the morphology of ciliated and pseudostratified upper airway epithelium, losing the skin’s characteristic keratinization. These cells have been shown to express the scavenger receptor expressed by endothelial cells I (SREC-I; also termed SR-F1), an F-type scavenger receptor56. This host cell membrane protein has been identified as a key receptor for S. aureus adhesion within the inner nasal cavity56.

ACS Paragon Plus Environment

8

Page 9 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Interestingly, nasal colonization is considered to be a multifactorial process involving these two different S. aureus niches (Figure 2)54, 57. SREC-I mediated adhesion to the inner nasal cavity is thought to be important in the initial stages of colonization38, 57. In contrast, longterm persistent colonization, which is observed in ~20% of the adult population, implicates the squamous keratinized epithelium within the nasal vestibule38, 54, 57.

frontal sinus

sphenoid sinus

nasal turbinates

nasopharynx

nasal cavity

vestibulum nasi

eustachian tube orifice

anterior naris

hard palate

IsdA

ClfB WTA

Mucus

loricrin, involucrin and filaggrin

N N

N

N

N

N

N

SREC-1

stratum corneum

Goblet cell

stratum granulosum

cytokeratin K1/K10

stratum spinosa

cytokeratin K5/K14

stratum basal

ClfB and IsdA CWA proteins facilitate adhesion to the nasal vestibule

WTAs mediate adhesion to the host cell receptor SREC-I within the inner nasal cavity

Figure 2: S. aureus colonization of the human nose. S. aureus adheres to the anterior vestibulum nasi using the CWA proteins ClfB and IsdA, which bind to host cell proteins within the cornified envelope of the stratum corneum. S. aureus also colonizes the inner nasal cavity due to the interaction of WTAs with the host cell receptor SREC-I, which is expressed on the surface of the ciliated epithelial cells lining this region of the nose. The molecular basis of S. aureus nasal colonization: the bacterial perspective

ACS Paragon Plus Environment

9

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 45

Given the differences of the epithelium in these two different S. aureus niches (Figure 2), S. aureus adhesins facilitating colonization are also distinct. The CWA proteins clumping factor B (ClfB)48, 58-59 and the iron-regulated surface determinant A (IsdA)38, 60-61 have been identified as the major S. aureus adhesins that enable persistent colonization of the nasal vestibule, via adhesion to proteinaceous host molecules within the squamous epithelium (Figure 2). On the other hand, the S. aureus glycopolymer wall teichoic acid (WTA) mediates adhesion to ciliated epithelial cells expressing SREC-I within the inner nasal cavity (Figure 2).

Cell wall-anchored (CWA) proteins mediate adhesion to the nasal vestibule There are four major staphylococcal cell wall-anchored (CWA) protein families, comprising 25 different proteins30. These surface proteins play various roles in pathogenesis30 and are covalently anchored to the cell wall by sortases (see Foster et al., 2014 for a comprehensive review30). CWA proteins significantly contribute to the virulence of S. aureus by facilitating immune evasion, cell invasion, nutrient acquisition, and adhesion to host cells30. Historically, the names of CWA proteins were assigned in regard to the function they were first associated. However, this nomenclature can be misleading, and some have been re-named since many of these proteins display numerous functions and possess different ligand binding sites. The CWA proteins ClfB and IsdA are the primary adhesins facilitating colonization of the nasal vestibule via interaction with the host cell receptor molecules cytokeratin K10 and/or loricrin. The genes encoding these determinants are highly conserved amongst S. aureus isolates, indicating that anti-adhesives targeting these proteins could enjoy clinical success62. While additional CWA proteins—namely SdrC, SdrD, and SasG60, 63—have been implicated in nasal colonization, ClfB and IsdA are considered the major determinants of S. aureus colonization of the nasal vestibule. Specifically, IsdA and ClfB devoid S. aureus strains are defective in colonizing the nares of rodents61, 64. In human volunteers, a ClfB mutant was also shown to be impaired in nasal colonization over an extended time period, highlighting the

ACS Paragon Plus Environment

10

Page 11 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

importance of this protein in persistent colonization59. Of note, the latter study used the S. aureus strain 8325-4 defective in the alternative sigma factor B (σB) 59. Therefore, this study has limitations since this global regulator is involved in the upregulation of clfB expression, which is described in more detail below. Finally, elevated expression of isdA and clfB has been observed in a cotton rat model of nasal colonization (a common and efficacious model to study S. aureus nasal colonization65) several days after bacterial instillation38. ClfB was originally identified as a fibrinogen-binding protein (hence the designation clumping factor B) but has since been shown to be susceptible to proteolytic cleavage by the S. aureus metalloprotease aureolysin66. The truncated form is unable to bind fibrinogen but has been shown to retain affinity for cytokeratin K1048. ClfB is the predominant keratin-binding adhesin expressed in the exponential phase of growth58. This adhesin interacts with repeats of the amino acid sequence Y[GS]nY in the C-terminal tail region of cytokeratin K10, which is a highly variable region of the keratins58. The Y[GS]nY motifs form omega loops within the keratin molecule: the GS sequences project out due to hydrophobic interactions between the Y residues55. Cytokeratin K10 binds in a hydrophobic trench between the N2 and N3 subdomains of ClfB (Figure 3A & B)67-68 via the ‘dock, lock and latch’ binding mechanism described for other staphylococcal adhesins69. Specifically, by interacting with the hydrophobic ClfB trench, a conformational change is induced in the C-terminal extension of the N3 domain, covering the peptide ligand, while also interacting with residues located in the N2 domain. This interaction effectively locks the peptide ligand in place and the strength of the interaction has been shown to be enhanced by force70. The latter indicates S. aureus adheres more firmly under high shear stress induced by physical stresses, such as mucociliary clearance70. It was originally assumed that cytokeratin K10 is the primary ClfB ligand. However, subsequent studies revealed that ClfB also binds loricrin, which has been shown to be critical for nasal colonization55. Loricrin is composed of cytokeratin K10 similar omega loops mediating this interaction. Loricrin is the major component of the cornified envelope; the interaction of ClfB

ACS Paragon Plus Environment

11

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 45

with loricrin is also crucial to the establishment of S. aureus skin infections71. In summary, loricrin is considered the major ClfB ligand in the nose, signifying that the loricrin-ClfB interaction is integral to nasal colonization and is a key target for the development of nasal decolonization strategies.

A

N

SP

N

B

197 63

SD repeats 542

MS 812

C 927

C

NEAT

SP 1

N3

N2

N1

1

180

350

C

Figure 3: Domain organization and structures of the nasal cell adhesins ClfB and IsdA. (A) Schematic model depicting the domain organization of S. aureus ClfB (top) and IsdA (bottom). For both proteins, the N and C terminus are labelled, and the amino acid numbering indicates the domain boundaries. SP corresponds to the signal peptides targeting the proteins for secretion. For ClfB, the cytokeratin K10/loricrin binding interface is located within the N2 and N3 region, followed by the serine-aspartate (SD) repeat region and finally, the membrane spanning portion of the protein (MS). Of note, there is an aureolysin66 protease cleavage site located between the N1 and N2 region that truncates the protein, abolishing fibrinogen binding. The heme binding NEAr iron Transporter (NEAT) domain of IsdA is proposed to also mediate binding to host cell ligands such as cytokeratin and loricrin. (B) Ribbon diagram of the N2 and N3 domains of ClfB (PDB accession number: 4F1Z) bound to a cytokeratin K10 peptide (shown as green sticks). ClfB is coloured blue and residues that are critical for ligand binding—the ClfB ‘hotspot’—are colored red. (C) Ribbon diagram of the IsdA NEAT domain (shown in dark blue) in complex with heme (shown as sticks and colored green). The NEAT domain is proposed to mediate affinity to the nasal epithelium (PDB accession number: 2ITF).

ACS Paragon Plus Environment

12

Page 13 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

The iron-regulated CWA IsdA protein is the second adhesin implicated in nasal cell adhesion55. The nose is a unique habitat characterized by low nutrient and iron availability72. Iron is essential for bacterial growth and vertebrates have evolved systems to sequester iron intracellularly to prevent the proliferation of pathogenic bacteria73. In S. aureus, the NEAr iron Transporter (NEAT) domain family encompasses four iron-regulated surface determinants (Isd): IsdA, IsdB, IsdC and IsdH. The expression of these CWA proteins is induced when iron is deficient and these proteins have been shown to be key components of a system facilitating iron acquisition via a relay system involving heme binding and subsequent degradation in the cytoplasm of the bacterial cell74. As the nose represents an iron-limited environment, there is increased expression of Isd CWA proteins in nasal isolates72. Expression of IsdA has been shown to maximize nasal colonization in cotton rats and promote adherence to human nasal epithelial cells in vitro61. IsdA has broad ligand binding capacity, including components of the extra cellular matrix (ECM) such as fibrinogen and fibronectin75, serum proteins76 and heme77. However, the role of these interactions in nasal colonization remain unknown. The IsdA NEAT domain spans ~120 amino acids forming an immunoglobulin-like βsandwich fold78. Heme binds in a large hydrophobic pocket (Figure 3C) formed between one sheet and a loop, residing in a region of the protein opposite the chain termini78. The NEAT domain is responsible for the broad ligand binding capacity of IsdA and this domain can only interact with one ligand at a time, indicating that the different ligands likely have overlapping binding sites79. The ability of the NEAT domain to bind cytokeratin K10, loricrin, and involucrin, is thought to underlie the proteins ability to promote S. aureus nasal colonization79. The precise binding interface and the molecular details underlying the proteins ability to bind cytokeratin K10, loricrin and involucrin is unknown. In analogy to ClfB, loricrin is likely the major ligand for IsdA in the nose. Finally, in addition to host cell ligand binding mediated by the NEAT domain, the hydrophilic C-terminal domain of IsdA (Figure 3A) provides resistance against hydrophobic

ACS Paragon Plus Environment

13

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 45

fatty acids present on the surface of the cornified envelope80. In addition, IsdA was shown to protect S. aureus from the antibacterial activities of the protease apolactoferrin, an abundant polypeptide in nasal secretions81. Therefore, this protein promotes adhesion to the nasal epithelium whilst also mediating resistance to the host’s innate defences80-81. To summarize, ClfB and IsdA are integral for long-term nasal colonization. Based on these findings and the observation that the genes encoding ClfB and IsdA are present in the vast majority of S. aureus isolates62, interfering with these elements offers promise for the development of novel anti-adhesive nasal decolonization strategies.

The glycopolymer WTA mediates adhesion to the inner nasal cavity during the initial stages of colonization WTAs are situated within the S. aureus cell wall and extend beyond the peptidoglycan of the cell; the polymer is composed of ~40 repeats of glycosylated and D-alanylated ribitol-phosphate units82-83. For some time, WTAs have been implicated in nasal colonization84. SREC-I was identified as the S. aureus WTA receptor expressed at the surface of ciliated epithelial cells lining the inner nasal cavity (Figure 2)56. SREC-I is a membrane protein composed of a large extracellular domain comprising multiple epidermal growth factor (EGF) and EGF-like repeats, followed by transmembrane and cytoplasmic domains (Figure 4)85. WTA possesses high affinity for the SREC-I receptor and the binding site was located to domains 3 and 4 of the EFG-like repeats (Figure 4). This interaction is charge-dependent, relying on the free amino groups of the D-alanine moieties decorating the WTA polymer56. The alanine residues counter the predominant negative charge of the phosphate groups within WTA, rendering the polymer more zwitterionic in nature. More recently, glycosylation of WTA (Figure 4) has also been shown to be a key factor in nasal colonization of cotton rats86. However, the receptor for WTA glycosyl residues is unknow86. Glycosylation is mediated by the enzymes TarM87 and TarS88, which

ACS Paragon Plus Environment

14

Page 15 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

decorate WTA with -N-acetylglucosamine (-GlcNAc) and -GlcNAc, respectively. The gene encoding TarS is highly conserved amongst S. aureus, however, tarM is less prevalent86. Considering the importance of WTA in the early stages of S. aureus nasal colonization, inhibiting the WTA-SREC-I interaction could offer promise in the development of anti-adhesive nasal decolonizing strategies. Given the multifactorial process of S. aureus nasal colonization, it is conceivable that such therapeutics could be combined with anti-adhesives targeting the interaction of ClfB and/or IsdA with the nasal vestibule, providing a combinatorial therapy that removes S. aureus from both nasal niches. Taking these factors into consideration, here we highlight candidate anti-adhesive targets (Figure 1) to enable the development of novel S. aureus nasal decolonization strategies. Table 1 summarises the anti-adhesive targets discussed in this review.

Figure 4: Schematic diagram of the interaction between S. aureus wall teichoic acid (WTA) and the nasal cell scavenger receptor expressed by endothelial cells I (SREC-I; also termed SR-F1). S. aureus WTA is covalently linked to the peptidoglycan (shown as yellow circles and blue hexagons, representing repeating units of N-acetylglucosamine (GlcNAc) and N-acetylmuramic acid, respectively). The polymer is composed of ~40-60 ribitol-phosphate repeats (yellow triangles) that can be decorated with cationic D-alanine esters (shown as green circles) and glycosylated with /-GlcNAc (blue hexagons). The interaction between S. aureus WTA and SREC-I is charge dependent, involving the free amines of D-alanine residues (green

ACS Paragon Plus Environment

15

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 45

circles) decorating the polymer. SREC-I is a transmembrane protein (as shown), comprising an extracellular region of epidermal growth factor (EFG) and EFG-like repeats. WTA has been shown to interact with the EFG 3 and 4 repeats of the receptor. Loss of WTA glycosylation (blue hexagons), mediated by the enzymes TarS and TarM, also interferes with WTA-mediated adhesion to the inner nasal cavity. Inhibiting S. aureus nasal colonization: the development of anti-adhesives targeting ClfB Anti-adhesives targeting ClfB would interfere with the protein’s ability to bind loricrin. ClfB interacts with G/S-rich omega loops within proteinaceous ligands using the ‘dock, lock and latch’ binding mechanism67-68 (Figure 3B). The molecular details of this interaction were revealed by crystallization of the protein in complex with its peptide ligands, identifying a GSSGXGXXG binding motif67-68. Importantly, ClfB residues implicated in binding to this motif are highly conserved amongst S. aureus lineages67-68. The development of a direct, ClfB-targeting, antiadhesive requires the identification of a molecule interfering with the ClfB-loricrin protein-protein interaction. Small-molecule inhibition of protein-protein interactions, which is generally more cost effective than other therapeutics89, is challenging. In contrast to the deep cavities often recognized by small molecules, protein-protein interfaces are frequently large and flat90. However, not all residues within these interfaces are critical for binding and there has been significant progress into small molecule inhibitors of protein-protein interactions, many of which have been found to bind their targets with high affinity89-90. In particular, fragment-based lead discovery (FBLD) offers promise in this field89-90. This approach uses biophysical methods to identify molecules that bind weakly to protein surfaces89-90. These molecules can be evolved to bind with a higher affinity, increasing potency. The ClfB-ligand binding interface is 1463 Å2 and ligand recognition resides within amino acid residues (526-542) in the ‘lock’ region67. Despite this large contact surface area, substitution of either N526Q, Q235A, S236Q, F328A, and W522V (Figure 3B) residues completely abrogate the ability of ClfB to bind protein ligands67; this ‘hotspot’ could be the target of small molecules. Despite the challenges associated with the

ACS Paragon Plus Environment

16

Page 17 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

inhibition of protein-protein interactions, the ClfB hotspot could be an efficacious target for the development of anti-ClfB therapeutics. An alternate approach involves the indirect inhibition of adhesion by interfering with the biosynthesis or surface assembly/presentation of ClfB (Figure 1). The vast majority of CWA adhesins are anchored to the cell surface by the enzyme sortase A (SrtA). Deletion of the gene encoding this enzyme inhibits the presentation of cell surface proteins and significantly reduces the virulence of S. aureus, without impacting growth74, 91-92. Therefore, SrtA has been the focus of a number of drug discovery endeavours, resulting in the identification of promising pre-clinical inhibitors9. Since this has been an active area of research for some time, SrtA inhibitors are described in more detail later in this review. Here we will discuss alternate ClfB anti-adhesive targets. A number of CWA proteins—including ClfB—harbour a long stretch of Ser-Asp dipeptide repeats (referred to as the SD region) shown to be glycosylated by the SD-repeat glycosyltransferases (Sdg) A and B 93 (Figure 5A). The Ser residues in these repeats are modified with O-linked GlcNAc by the SdgB enzyme94. SdgA glycosylates sites that have already been modified by SdgB94. Glycosylation of the SD region prevents proteolytic degradation of adhesins, such as ClfB, by human neutrophil-derived cathepsin93 (Figure 5A). Indeed, the virulence of SdgB defective S. aureus is significantly impaired in a murine model of sepsis94. Inhibiting SdgB-mediated glycosyltransferase activity could render ClfB more susceptible to proteolytic cleavage in the host environment, reducing the affinity of S. aureus for the nasal epithelium (Figure 5A). However, whether deletion of the gene encoding SdgB, and thus inhibition of ClfB glycosylation, impacts nasal decolonization has not been investigated. Furthermore, the discovery of bacterial glycosyltransferase inhibitors can be challenging95-96. With the exception of pyranose scaffold compounds inhibiting glycosyltransferases involved in peptidoglycan polymerization97, there is a lack of potent and specific bacterial

ACS Paragon Plus Environment

17

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 45

glycosyltransferase inhibitors98. The efficacy of SdrB as an anti-adhesive target awaits to be established and would be enhanced by increased mechanistic and structural insight. Another tactic in the search for indirect anti-adhesives entails the inhibition of global regulators of clfB expression (Figure 5B). S. aureus virulence factors are controlled by complex networks of regulators including two-component systems, sigma factors, regulatory RNAs and DNA-binding proteins99-101. Indeed, a single virulence gene can be influenced by several regulators that ‘cross talk’; regulators can impact expression directly via promoter recognition or indirectly through the action of additional regulators102. There is a biphasic distinction between the expression of CWA proteins earlier in the growth cycle followed by the expression of secreted toxins in the stationary phase100. In regard to ClfB, a subset of studies show production is positively regulated by the global regulator Rot (the repressor of toxins)102-103. Analysis of Rotmediated global regulation revealed that the only other S. aureus surface proteins positively regulated by Rot are SdrC and protein A102. Of note, this study was conducted with S. aureus harvested at the late-exponential phase of growth, which could impart a limitation since ClfB is maximally expressed at the mid-exponential phase66. An additional study using S. aureus strain 8325-4, describes Rot directly binding to the clfB promoter, enhancing expression103. However, as described, the use of this strain in the study of virulence gene expression is significantly compromised due to impaired σB (also referred to as SigB) activity104. Rot exhibits homology with the global transcriptional regulator SarA (the staphylococcal accessory regulator), which recognizes a specific motif in the promoter of its target genes105. All members of the SarA family share an KXRXXXDER amino acid motif implicated in this recognition99. However, it has been suggested that rot expression is repressed by SarA binding to the rot promoter106. Rot is a dimer with subunits forming the characteristic winged helix-turn-helix DNA-binding motif107. Mutagenesis of specific Rot residues inhibit the proteins activity, highlighting the DNA binding site and thus, the target of potential anti-adhesives107. Transcription factors have long been considered unsuitable targets for drug discovery, partly due to the lack of appropriate methods

ACS Paragon Plus Environment

18

Page 19 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

to screen for small molecule inhibitors108-109. However, approaches have now been described that could enable higher throughput screening of transcriptional targets108. Indeed, the development of approaches to target transcription factors in the treatment of cancer has seen a revival (referred to as transcription therapy) 110. Additionally, structure based drug design has been described for the identification of SarA inhibitors111 and this approach could be applied to Rot. More recently, the RNA-binding protein SpoVG was implicated in clfB expression112. The spoVG locus encodes an RNA binding regulatory protein positively regulated by the σB (Figure 5B). σB is a subunit of RNA polymerase providing promoter recognition specificity, enabling differential gene expression in response to changes in environmental conditions113. SpoVG was recently shown to positively regulate clfB expression by directly interacting with the clfB promoter and/or in a Rot-mediated fashion112. Therefore, inhibiting the action of SpoVG or the σB would interfere with the expression of ClfB, making these elements additional candidates for the development of indirect anti-adhesives (Figure 5B). The gene encoding σB is located within the sigB operon (composed of rsbU-V-W-sigB genes)114. The activity of this global regulator is controlled by so-called ‘partner-switching’ between the anti-σB kinase (encoded by rsbW), which associates and sequesters σB, and the non-phosphorylated form of RsbV (anti-anti-σB). RsbV competes for anti-σB binding and triggers release of σB 115. To add a further layer of detail, the rsbU gene in the sigB operon encodes a phosphatase that dephosphorylates RsbV, triggering the release of σB. Therefore, another attractive target for inhibition is the phosphatase RsbU (Figure 4B). Inhibition of this enzyme would result in a higher concentration of phosphorylated RsbV (anti-anti-σB), reducing the affinity of the protein for anti-σB. Thus, phosphorylated RsbV maintains anti-σB-SigB complexes, inhibiting the interaction of σB with RNA polymerase. Indeed, the overexpression of rsbU in S. aureus strongly activates σB 114 and the S. aureus 8325-4 strain has compromised σB activity due to mutation in the rsbU gene104. However, evidence supporting the efficacy of σB as an anti-virulence target are conflicting, with a subset of studies

ACS Paragon Plus Environment

19

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 45

describing no impact to pathogenicity in animal models of infection when σB is compromised104, 116-117.

For this reason, decolonization strategies targeting σB and σB-related elements ought to

be postponed until there is more information supporting the efficacy of this target in relevant infection models. In summary, the development of anti-ClfB nasal decolonization strategies should focus on the identification of anti-adhesives that interfere with ClfB-mediated recognition of loricrin within the nasal vestibule (Figure 2). Such approaches are facilitated by a wealth of structural and molecular information that highlight the ClfB ligand binding ‘hotspot’ as a target for the development of protein-protein inhibitors. On the other hand, the development of indirect antiadhesive therapeutics would be enhanced by studies investigating the in vivo efficacy of the aforementioned targets.

A

B

Figure 5: Examples of indirect anti-adhesive targets to reduce the surface presentation of ClfB. (A) Schematic model of the CWA protein ClfB (as seen in Figure 3A). The glycosyltransferase SdgB covalently links N-acetylglucosamine (GlcNAc; shown as blue squares) to serine residues within the serine-aspartate repeat (SDR) region of ClfB. A second glycosyltransferase, SdgA, then adds an additional GlcNAc moiety. Glycosylation of the SDR region has been shown to protect SDR containing proteins from proteolytic cleavage by human neutrophil-derived cathepsin G. Inhibition of SdgB could render ClfB more susceptible to host proteolytic cleavage, reducing the surface presentation of the protein and thus, nasal

ACS Paragon Plus Environment

20

Page 21 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

colonization. (B) Schematic diagram of the regulatory cascades impacting clfB expression. The arrows represent positive interactions and the bars indicate repression. The red crosses depict favourable targets for anti-adhesives. Inhibiting S. aureus nasal colonization: the development of IsdA anti-adhesives The identification of directly acting IsdA anti-adhesives necessitates the need for molecules interfering with the adhesin’s affinity for loricrin. As described, the broad ligand binding capabilities of IsdA reside in its single-copy NEAT domain (spanning amino acids I69 to E177)75, which is solely composed of -sheets78 (Figure 3C). Non-specific electrostatic interactions are proposed to underlie the proteins ligand promiscuity78. With the exception of heme, the structures of IsdA bound to other ligands have not been reported and the IsdA-loricrin binding interface remains unknown. Therefore, we are unable to provide a detailed description of approaches to inhibit the IsdA-loricrin interaction. In the absence of structural and mechanistic knowledge, FBLD—as described for ClfB—could be used to identify IsdA-loricrin protein-protein interaction inhibitors using the NEAT domain as a target. Indirect IsdA anti-adhesives intend to interfere with the expression or presentation of IsdA on the cell surface. The isdA gene is negatively regulated by iron through the iron responsive regulatory protein Fur (ferric uptake regulator)75. Fur is a dimeric protein requiring metal cofactors, which binds a palindromic 19 base pair consensus sequence (the ‘Fur box’) within the promoters of target genes118. This interaction inhibits the transcription of downstream genes; in low iron conditions Fur dissociates allowing transcription to proceed118. In contrast to inhibition of ClfB mediated by interference with global regulators, inhibiting the action of Fur would result in increased isdA expression, and thus nasal colonization. Overall, the identification of IsdA anti-adhesives would be expedited by more insight into the molecular details underlying the broad ligand binding specificity of this protein. Akin to ClfB, the identification of a ligand binding ‘hotspot’ would enable the development of small molecule protein-protein interaction inhibitors. Furthermore, with the exception of Fur (and CWA protein

ACS Paragon Plus Environment

21

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 45

common targets such as SrtA), it is unknown if other genetic and cellular elements are implicated in the presentation of this protein. Such studies could expand the target space for the development of anti-IsdA therapeutics.

Inhibiting S. aureus nasal colonization: interfering with WTA mediated nasal colonization Anti-adhesives targeting WTA mediated nasal colonization would interfere with the interaction of this polymer with the host cell receptor SREC-I. One interesting avenue of attack could be the inhibition of WTA biosynthesis. WTA biosynthesis is mediated by an enzyme cluster: the teichoic acid ribitol pathway (tar enzymes; referred to as tag enzymes in other organisms such as Bacillus subtilis)82. In contrast to the late stages of WTA biosynthesis, which are conditionally essential, the early-stages (involving the membrane-associated glycosyltransferases TarO and TarA) are non-essential for in vitro S. aureus growth119-121. However, deficiencies in these enzymes leads to an avirulent phenotype, highlighting these early steps as attractive antivirulence targets83, 119. Indeed, one of the first accounts of WTA-mediated virulence was observed in a cotton rat model of nasal colonization; tarO deficient S. aureus was unable to colonize the nares of rodents84. TarO reversibly catalyzes the transfer of GlcNAc phosphate—from UDP-GlcNAc—to a molecule of undecaprenyl phosphate in the bacterial membrane121. TarA then transfers NAcetyl-D-mannosamine (ManNAc)—from UDP-ManNAc—to the terminal GlcNAc, which serves as a substrate for the next enzyme in the pathway121. Inhibition of either of these enzymes renders the cell devoid of WTA. Therefore, TarO and/or TarA represent potentially efficacious anti-adhesive targets. Such approaches are supported by structural knowledge of the two proteins, which could facilitate the identification of enzyme-specific inhibitors. The crystal structure of TarO remains unknown, however, a recent study describes a three-dimensional model of the protein based on a structurally and functionally similar plasma membrane associated glycosyltransferase122. The crystal structure of a S. aureus TarA homolog from

ACS Paragon Plus Environment

22

Page 23 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Thermoanaerobacter italicus was also recently reported (PDB accession codes: 5WB4 & 5WFG), highlighting a unique glycosyltransferase fold. Finally, since WTA is an attractive antivirulence therapeutic target, there are a number of known TarO-specific inhibitors that could be promising anti-adhesives122-124. A series of potent and recently described TarO inhibitors compounds are discussed in more detail in the proceeding section122. In addition to inhibiting WTA biosynthesis, a complementary approach could involve the inhibition of the enzymes that tailor the polymer. As described, TarS is a highly conserved WTA glycosyltransferase integral to nasal colonization in cotton rats86. While a proportion of strains do harbour both tarM and tarS, which would necessitate the need to compromise both enzymes, the majority only possess TarS86. Importantly, compromising TarS does not induce growth defects in vitro88. Combined, these factors make TarS an attractive anti-adhesive target (Figure 4). Such efforts could be enabled by a recent study depicting the structure and mechanism of action of this enzyme (PDB accession codes: 5TZ8 and 5TZE)125.

Promising known inhibitors of S. aureus host cell adhesion Inhibiting nasal colonization mediated by CWA proteins An attractive strategy to prevent the surface presentation of the majority of CWA proteins— including ClfB and IsdA—is the inhibition of the anchoring enzyme SrtA. As such, the identification of SrtA inhibitors has been an active research area for >15 years126. Such efforts involved a number of approaches including structure based mimetic substrate design, in silico virtual screening and the profiling of naturally derived and synthetic small molecules (see Guo et al. 2015 and Cascioferro et al. 2015 for detailed reviews). The in silico virtual screening approach was particularly successful, identifying one of the most promising classes of known SrtA inhibitors: the triazolothiadiazoles (Figure 6)127. This approach involved virtual screening of ~300,000 compounds to identify molecules that bind the SrtA active site, using a known natural product inhibitor (topsentin A128) as a model ligand127.

ACS Paragon Plus Environment

23

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 45

The most efficacious compounds were 3,6-disubstituted triazolothiadiazoles (Figure 6), which lack antibacterial activity, exhibit potent and reversible inhibition of SrtA, and had efficacy in an animal model of sepsis127. These compounds could be potentially efficacious anti-adhesives and it would be advantageous to investigate their use in a cotton rat model of nasal colonization. An additional study, published simultaneously, describe a class of benzo[d]isothiazol3(2H)-one-adamantane amine derivatives (Figure 6) identified from an in vitro screen of >50,000 compounds against recombinant SrtA129. In contrast to the triazolothiadiazole compounds, these inhibitors show irreversible inhibition of SrtA by reaction with the active site cysteine residue, forming a covalent linkage between the cysteine thiol and the compounds benzothiazole129. However, unlike the triazolothiadiazoles, these compounds do exhibit modest S. aureus antibacterial activity (the most promising compounds have a minimum inhibitory concentration of 8-16 g/mL) and modest cytotoxicity129. Therefore, the triazolothiadiazoles may be more promising for the development of anti-adhesives that do not impart selective pressure.

Inhibiting WTA-mediated nasal colonization As indicated above, interfering with the biosynthesis and/or tailoring of WTAs could have potential for the development of anti-adhesives targeting the inner nasal cavity. Inhibiting the early stages of WTA biosynthesis renders S. aureus devoid of WTA, while not impacting growth in vitro. As such, this process has been the target of a recent large-scale (2.8 million compounds) drug discovery endeavour by Merck Research Laboratories122. Overall, two chemically distinct TarO inhibitor series were identified: tarocin A (oxazolidinone series) and tarocin B (benzimidazole series)122 (Figure 6). Within the tarocin A and B series, the most promising compounds did not inhibit the growth of S. aureus and/or other microorganisms tested, lacked cytotoxicity, and exhibited potent inhibition of WTA biosynthesis122. The inhibitors were shown to target TarO and it was proposed that they inhibit the interaction of this enzyme with its bactoprenol substrate122. To summarise, these compounds are strong therapeutic leads

ACS Paragon Plus Environment

24

Page 25 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

taking aim at WTA biosynthesis. Future studies could assess their ability to decolonize the nares of rodents to investigate their efficacy as potential anti-adhesive therapeutics. One potential limitation regarding anti-adhesives targeting the inner nasal cavity could be the presence of mucous within this region of the nose. Compounds targeting this niche will need to be experimentally investigated to determine if this is a physiochemical barrier that needs to be overcome.

Figure 6: Structures of sortase A (SrtA) and TarO inhibitors. The first two panels are SrtA inhibitors (A) 3,6-disubstituted triazolothiadiazoles and (B) SrtA benzo[d]isothiazol-3(2H)-oneadamantane amine derivatives. The bottom two panels are TarO inhibitors (C) tarocin A and (D) tarocin B. With the exception of the abovementioned compounds, there is a lack of known inhibitors of S. aureus adhesion. While the 3,6-disubstituted triazolothiadiazoles are promising pre-clinical candidates, other previously identified SrtA inhibitors often suffer from off-target effects and thus cytotoxicty126. Principally, CWA proteins contain an LPXTG motif that is cleaved by SrtA via an active site cysteine residue, prior to covalent linkage to the peptidoglycan. Previous studies have relied upon an in vitro assay that detects inhibition by monitoring

ACS Paragon Plus Environment

25

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 45

cleavage of this motif126. Sortase inhibitors often react with the thiols of cysteine residues within other proteins, causing non-specific cytotoxicity. A whole-cell based assay would rule out such inhibitors at the beginning of the drug discovery process, circumventing this problem. However, one bottleneck in the discovery of anti-adhesives has been the lack of a high-throughput whole cell-based assay. Existing methods to study S. aureus adhesion are often not high-throughput, involving the laborious determination of viable remaining bacteria by culture-based methods. Additional methods describe the use of crystal violet to stain adhered bacteria. However, due to the lack of specificity of this dye, high-throughput assays are often hindered by poor sensitivity. Therefore, future efforts should include the development of innovative high-throughput assays to enable the rapid profiling of chemical libraries and the identification of small molecule inhibitors of S. aureus adhesion.

Concluding remarks Anti-virulence approaches have received increased attention over the last decade; as such, there have been a subset of drugs approved for clinical use that block exotoxins produced by bacteria9. Furthermore, it has been rigorously established that small molecule inhibition of myriad virulence factors attenuates and/or prevents disease9, which signifies that this research area offers significant clinical potential. Indeed, the numerous S. aureus virulence factors offers the promise for multiple avenues of attack. Given the high prevalence and increased risk factor associated with S. aureus nasal colonization, targeting the adhesins—namely ClfB, IsdA and WTA—facilitating nasal colonization could offer promise in reducing the health burden imposed by S. aureus. Prophylactic antibiotic use (e.g., mupirocin) in nasal decolonization has proven success in the clinic. However, the emergence of mupirocin-resistant isolates is comprising the efficacy of this approach. Anti-adhesives could be well-suited to correct this shortfall, providing an orthogonal and innovative approach to control bacterial infections that does not exacerbate the antibiotic resistance crisis. Overall, the development of high-throughput approaches to

ACS Paragon Plus Environment

26

Page 27 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

detect such molecules, in combination with increased insight into the molecular underpinnings of nasal colonization, would provide substantial benefits in the search for new ways to control S. aureus infections. Finally, we highlight the potential of a combinatorial anti-adhesive therapy targeting the two S. aureus niches within the nose; the nasal vestibule and the inner nasal cavity. Indeed, as discussed in the preceding section, there are a number of promising pre-clinical compounds that could be used as anti-adhesive decolonizing therapeutics. In particular, the 3,6-disubstituted triazolothiadiazole SrtA inhibitors (impacting ClfB/IsdA mediated adhesion) and the tarocins (inhibiting WTA biosynthesis and thus SREC-I adhesion) could represent a novel combinatorial therapy to eradicate S. aureus from the two respective niches. Establishing their anti-adhesive efficacy in vivo—in relevant animal models of nasal colonization—could highlight their potential as novel decolonizing agents, reducing the clinical burden of S. aureus infections.

ACS Paragon Plus Environment

27

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 45

Table 1. Clinical targets for the development of S. aureus anti-adhesive nasal decolonizing strategies Target

Description

References

ClfB

Antiadhesive Group Class II

Nasal cell adhesin that binds loricrin and cytokeratin proteins

48, 55, 58, 61, 64, 66-68

IsdA

Class II

Nasal cell adhesin—expressed in low iron environments—that binds loricrin and cytokeratin proteins

38, 60, 61, 72, 78, 79

SrtA

Class III

Transpeptidase that covalently anchors cell-wall associated proteins to the peptidoglycan

74, 91, 92

SdgB

Class III

SD-repeat glycosyltransferase that protects ClfB from proteolytic cleavage within the host

93, 94

SigB (σB)

Class III

Subunit of RNA polymerase providing promoter recognition specificity, enhancing the expression of clfB

112-114

RsbU

Class III

Phosphatase targeting RsbV, triggering the release of σB from inhibitory proteins

104, 114

Rot

Class III

Global regulator enhancing the expression of clfB

102, 103

SpoVG

Class III

RNA binding regulatory protein that is positively regulated by the σB. SpoVG positively regulates clfB expression by directly interacting with the clfB promoter and/or in a Rot-mediated fashion

112, 113

TarO

Class III

Glycosyltransferase catalyzing the first step in WTA biosynthesis

83, 84, 119-124

TarA

Class III

Glycosyltransferase catalyzing the second step in WTA biosynthesis

120, 121

TarS

Class III

Glycosyltransferase catalyzing β-OGlcNAcylation of the WTA polymer

86, 88, 125

ACS Paragon Plus Environment

28

Page 29 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Acknowledgements This work was supported by funding through the New Frontiers in Research Fund-Exploration Grant (NFRFE-2018-01058) and a Medical Research Grant from the J.P. Bickell Foundation.

ACS Paragon Plus Environment

29

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1.

Page 30 of 45

Perry, J.; Waglechner, N.; Wright, G., The Prehistory of Antibiotic Resistance. Cold

Spring Harb Perspect Med 2016, 6 (6). 2.

Brown, E. D.; Wright, G. D., Antibacterial drug discovery in the resistance era. Nature

2016, 529 (7586), 336-43. 3.

Blaskovich, M. A. T., The Fight Against Antimicrobial Resistance Is Confounded by a

Global Increase in Antibiotic Usage. ACS Infect Dis 2018. 4.

Klein, E. Y.; Van Boeckel, T. P.; Martinez, E. M.; Pant, S.; Gandra, S.; Levin, S. A.;

Goossens, H.; Laxminarayan, R., Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc Natl Acad Sci U S A 2018, 115 (15), E3463-E3470. 5.

Langdon, A.; Crook, N.; Dantas, G., The effects of antibiotics on the microbiome

throughout development and alternative approaches for therapeutic modulation. Genome Med 2016, 8 (1), 39. 6.

Anderson, H.; Vuillermin, P.; Jachno, K.; Allen, K. J.; Tang, M. L.; Collier, F.; Kemp, A.;

Ponsonby, A. L.; Burgner, D.; Barwon Infant Study Investigator, G., Prevalence and determinants of antibiotic exposure in infants: A population-derived Australian birth cohort study. J Paediatr Child Health 2017, 53 (10), 942-949. 7.

Lynn, M. A.; Tumes, D. J.; Choo, J. M.; Sribnaia, A.; Blake, S. J.; Leong, L. E. X.; Young,

G. P.; Marshall, H. S.; Wesselingh, S. L.; Rogers, G. B.; Lynn, D. J., Early-Life Antibiotic-Driven Dysbiosis Leads to Dysregulated Vaccine Immune Responses in Mice. Cell Host Microbe 2018, 23 (5), 653-660 e5. 8.

Allen, H. K., Alternatives to antibiotics: Why and how. NAM Perspectives. 2017.

9.

Dickey, S. W.; Cheung, G. Y. C.; Otto, M., Different drugs for bad bugs: antivirulence

strategies in the age of antibiotic resistance. Nat Rev Drug Discov 2017, 16 (7), 457-471. 10.

Ofek, I.; Hasty, D. L.; Sharon, N., Anti-adhesion therapy of bacterial diseases: prospects

and problems. FEMS Immunol Med Microbiol 2003, 38 (3), 181-91.

ACS Paragon Plus Environment

30

Page 31 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

11.

Kelly, C. G.; Younson, J. S., Anti-adhesive strategies in the prevention of infectious

disease at mucosal surfaces. Expert Opin Investig Drugs 2000, 9 (8), 1711-21. 12.

Kline, K. A.; Falker, S.; Dahlberg, S.; Normark, S.; Henriques-Normark, B., Bacterial

adhesins in host-microbe interactions. Cell Host Microbe 2009, 5 (6), 580-92. 13.

JW, P., Bacterial Pathogenesis. In Medical Microbiology, 4 ed.; S, B., Ed. Galveston TX,

1996. 14.

Yuehuei, H.; Dickinson, R. B.; Doyle, R. J., Molecular Basis of Bacterial Adhesion. In

Handbook of Bacterial Adhesion Principles, Methods, and Applications, Springer, Ed. 2000. 15.

Spaulding, C. N.; Klein, R. D.; Schreiber, H. L. t.; Janetka, J. W.; Hultgren, S. J.,

Precision antimicrobial therapeutics: the path of least resistance? NPJ Biofilms Microbiomes 2018, 4, 4. 16.

Kaper, J. B.; Nataro, J. P.; Mobley, H. L., Pathogenic Escherichia coli. Nat Rev Microbiol

2004, 2 (2), 123-40. 17.

Cusumano, C. K.; Pinkner, J. S.; Han, Z.; Greene, S. E.; Ford, B. A.; Crowley, J. R.;

Henderson, J. P.; Janetka, J. W.; Hultgren, S. J., Treatment and prevention of urinary tract infection with orally active FimH inhibitors. Sci Transl Med 2011, 3 (109), 109ra115. 18.

Kalas, V.; Hibbing, M. E.; Maddirala, A. R.; Chugani, R.; Pinkner, J. S.; Mydock-

McGrane, L. K.; Conover, M. S.; Janetka, J. W.; Hultgren, S. J., Structure-based discovery of glycomimetic FmlH ligands as inhibitors of bacterial adhesion during urinary tract infection. Proc Natl Acad Sci U S A 2018, 115 (12), E2819-E2828. 19.

Maddirala, A. R.; Klein, R.; Pinkner, J. S.; Kalas, V.; Hultgren, S. J.; Janetka, J. W.,

Biphenyl Gal and GalNAc FmlH Lectin Antagonists of Uropathogenic E. coli (UPEC): Optimization through Iterative Rational Drug Design. J Med Chem 2019, 62 (2), 467-479. 20.

Spaulding, C. N.; Klein, R. D.; Ruer, S.; Kau, A. L.; Schreiber, H. L.; Cusumano, Z. T.;

Dodson, K. W.; Pinkner, J. S.; Fremont, D. H.; Janetka, J. W.; Remaut, H.; Gordon, J. I.;

ACS Paragon Plus Environment

31

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 45

Hultgren, S. J., Selective depletion of uropathogenic E. coli from the gut by a FimH antagonist. Nature 2017, 546 (7659), 528-532. 21.

Totsika, M.; Kostakioti, M.; Hannan, T. J.; Upton, M.; Beatson, S. A.; Janetka, J. W.;

Hultgren, S. J.; Schembri, M. A., A FimH inhibitor prevents acute bladder infection and treats chronic cystitis caused by multidrug-resistant uropathogenic Escherichia coli ST131. J Infect Dis 2013, 208 (6), 921-8. 22.

Sattin, S.; Bernardi, A., Glycoconjugates and Glycomimetics as Microbial Anti-

Adhesives. Trends Biotechnol 2016, 34 (6), 483-495. 23.

Gordon, C. P.; Williams, P.; Chan, W. C., Attenuating Staphylococcus aureus virulence

gene regulation: a medicinal chemistry perspective. J Med Chem 2013, 56 (4), 1389-404. 24.

Krachler, A. M.; Orth, K., Targeting the bacteria-host interface: strategies in anti-

adhesion therapy. Virulence 2013, 4 (4), 284-94. 25.

Aberg, V.; Almqvist, F., Pilicides-small molecules targeting bacterial virulence. Org

Biomol Chem 2007, 5 (12), 1827-34. 26.

Svensson, A.; Larsson, A.; Emtenas, H.; Hedenstrom, M.; Fex, T.; Hultgren, S. J.;

Pinkner, J. S.; Almqvist, F.; Kihlberg, J., Design and evaluation of pilicides: potential novel antibacterial agents directed against uropathogenic Escherichia coli. Chembiochem 2001, 2 (12), 915-8. 27.

Svensson, M.; Platt, F. M.; Svanborg, C., Glycolipid receptor depletion as an approach

to specific antimicrobial therapy. FEMS Microbiol Lett 2006, 258 (1), 1-8. 28.

Krismer, B.; Weidenmaier, C.; Zipperer, A.; Peschel, A., The commensal lifestyle of

Staphylococcus aureus and its interactions with the nasal microbiota. Nat Rev Microbiol 2017, 15 (11), 675-687. 29.

Redi, D.; Raffaelli, C. S.; Rossetti, B.; De Luca, A.; Montagnani, F., Staphylococcus

aureus vaccine preclinical and clinical development: current state of the art. New Microbiol 2018, 41 (3), 208-213.

ACS Paragon Plus Environment

32

Page 33 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

30.

Foster, T. J.; Geoghegan, J. A.; Ganesh, V. K.; Hook, M., Adhesion, invasion and

evasion: the many functions of the surface proteins of Staphylococcus aureus. Nat Rev Microbiol 2014, 12 (1), 49-62. 31.

Tong, S. Y.; Davis, J. S.; Eichenberger, E.; Holland, T. L.; Fowler, V. G., Jr.,

Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management. Clin Microbiol Rev 2015, 28 (3), 603-61. 32.

Hassoun, A.; Linden, P. K.; Friedman, B., Incidence, prevalence, and management of

MRSA bacteremia across patient populations-a review of recent developments in MRSA management and treatment. Crit Care 2017, 21 (1), 211. 33.

Prioritization of pathogens to guide discovery, research and development of new

antibiotics for drug-resistant bacterial infections, including tuberculosis; Geneva: World Health Organization: 2017. 34.

Coates, R.; Moran, J.; Horsburgh, M. J., Staphylococci: colonizers and pathogens of

human skin. Future Microbiol 2014, 9 (1), 75-91. 35.

Wertheim, H. F.; Melles, D. C.; Vos, M. C.; van Leeuwen, W.; van Belkum, A.; Verbrugh,

H. A.; Nouwen, J. L., The role of nasal carriage in Staphylococcus aureus infections. Lancet Infect Dis 2005, 5 (12), 751-62. 36.

Bornstein, E., Eradication of Staphylococcus aureus and MRSA in the Nares: A

Historical Perspective of the Ecological Niche, with Suggestions for Future Therapy Considerations. Adv Microbiol 2017, 7 (6), 420-449. 37.

Krismer, B.; Peschel, A., Does Staphylococcus aureus nasal colonization involve biofilm

formation? Future Microbiol 2011, 6 (5), 489-93. 38.

Burian, M.; Rautenberg, M.; Kohler, T.; Fritz, M.; Krismer, B.; Unger, C.; Hoffmann, W.

H.; Peschel, A.; Wolz, C.; Goerke, C., Temporal expression of adhesion factors and activity of global regulators during establishment of Staphylococcus aureus nasal colonization. J Infect Dis 2010, 201 (9), 1414-21.

ACS Paragon Plus Environment

33

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

39.

Page 34 of 45

Wertheim, H. F.; Vos, M. C.; Ott, A.; van Belkum, A.; Voss, A.; Kluytmans, J. A.; van

Keulen, P. H.; Vandenbroucke-Grauls, C. M.; Meester, M. H.; Verbrugh, H. A., Risk and outcome of nosocomial Staphylococcus aureus bacteraemia in nasal carriers versus noncarriers. Lancet 2004, 364 (9435), 703-5. 40.

Kluytmans, J. A.; Wertheim, H. F., Nasal carriage of Staphylococcus aureus and

prevention of nosocomial infections. Infection 2005, 33 (1), 3-8. 41.

von Eiff, C.; Becker, K.; Machka, K.; Stammer, H.; Peters, G., Nasal carriage as a

source of Staphylococcus aureus bacteremia. Study Group. N Engl J Med 2001, 344 (1), 11-6. 42.

Poovelikunnel, T.; Gethin, G.; Humphreys, H., Mupirocin resistance: clinical implications

and potential alternatives for the eradication of MRSA. J Antimicrob Chemother 2015, 70 (10), 2681-92. 43.

Perl, T. M.; Cullen, J. J.; Wenzel, R. P.; Zimmerman, M. B.; Pfaller, M. A.; Sheppard, D.;

Twombley, J.; French, P. P.; Herwaldt, L. A.; Mupirocin; The Risk Of Staphylococcus aureus Study, T., Intranasal mupirocin to prevent postoperative Staphylococcus aureus infections. N Engl J Med 2002, 346 (24), 1871-7. 44.

Bode, L. G.; Kluytmans, J. A.; Wertheim, H. F.; Bogaers, D.; Vandenbroucke-Grauls, C.

M.; Roosendaal, R.; Troelstra, A.; Box, A. T.; Voss, A.; van der Tweel, I.; van Belkum, A.; Verbrugh, H. A.; Vos, M. C., Preventing surgical-site infections in nasal carriers of Staphylococcus aureus. N Engl J Med 2010, 362 (1), 9-17. 45.

Kluytmans, J. A.; Manders, M. J.; van Bommel, E.; Verbrugh, H., Elimination of nasal

carriage of Staphylococcus aureus in hemodialysis patients. Infect Control Hosp Epidemiol 1996, 17 (12), 793-7. 46.

Garrouste-Orgeas, M.; Timsit, J. F.; Kallel, H.; Ben Ali, A.; Dumay, M. F.; Paoli, B.;

Misset, B.; Carlet, J., Colonization with methicillin-resistant Staphylococcus aureus in ICU patients: morbidity, mortality, and glycopeptide use. Infect Control Hosp Epidemiol 2001, 22 (11), 687-92.

ACS Paragon Plus Environment

34

Page 35 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

47.

Hanssen, A. M.; Kindlund, B.; Stenklev, N. C.; Furberg, A. S.; Fismen, S.; Olsen, R. S.;

Johannessen, M.; Sollid, J. U., Localization of Staphylococcus aureus in tissue from the nasal vestibule in healthy carriers. BMC Microbiol 2017, 17 (1), 89. 48.

O'Brien, L. M.; Walsh, E. J.; Massey, R. C.; Peacock, S. J.; Foster, T. J., Staphylococcus

aureus clumping factor B (ClfB) promotes adherence to human type I cytokeratin 10: implications for nasal colonization. Cell Microbiol 2002, 4 (11), 759-70. 49.

Steven, A. C.; Steinert, P. M., Protein composition of cornified cell envelopes of

epidermal keratinocytes. J Cell Sci 1994, 107 ( Pt 2), 693-700. 50.

Lopez, O.; Cocera, M.; Wertz, P. W.; Lopez-Iglesias, C.; de la Maza, A., New

arrangement of proteins and lipids in the stratum corneum cornified envelope. Biochim Biophys Acta 2007, 1768 (3), 521-9. 51.

Wille, J. J.; Kydonieus, A., Palmitoleic acid isomer (C16:1delta6) in human skin sebum is

effective against gram-positive bacteria. Skin Pharmacol Appl Skin Physiol 2003, 16 (3), 176-87. 52.

Takigawa, H.; Nakagawa, H.; Kuzukawa, M.; Mori, H.; Imokawa, G., Deficient production

of hexadecenoic acid in the skin is associated in part with the vulnerability of atopic dermatitis patients to colonization by Staphylococcus aureus. Dermatology 2005, 211 (3), 240-8. 53.

Bibel, D. J.; Aly, R.; Shinefield, H. R.; Maibach, H. I.; Strauss, W. G., Importance of the

keratinized epithelial cell in bacterial adherence. J Invest Dermatol 1982, 79 (4), 250-3. 54.

Mulcahy, M. E.; McLoughlin, R. M., Host-Bacterial Crosstalk Determines Staphylococcus

aureus Nasal Colonization. Trends Microbiol 2016, 24 (11), 872-886. 55.

Mulcahy, M. E.; Geoghegan, J. A.; Monk, I. R.; O'Keeffe, K. M.; Walsh, E. J.; Foster, T.

J.; McLoughlin, R. M., Nasal colonisation by Staphylococcus aureus depends upon clumping factor B binding to the squamous epithelial cell envelope protein loricrin. PLoS Pathog 2012, 8 (12), e1003092. 56.

Baur, S.; Rautenberg, M.; Faulstich, M.; Grau, T.; Severin, Y.; Unger, C.; Hoffmann, W.

H.; Rudel, T.; Autenrieth, I. B.; Weidenmaier, C., A nasal epithelial receptor for Staphylococcus

ACS Paragon Plus Environment

35

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 45

aureus WTA governs adhesion to epithelial cells and modulates nasal colonization. PLoS Pathog 2014, 10 (5), e1004089. 57.

Weidenmaier, C.; Kokai-Kun, J. F.; Kulauzovic, E.; Kohler, T.; Thumm, G.; Stoll, H.;

Gotz, F.; Peschel, A., Differential roles of sortase-anchored surface proteins and wall teichoic acid in Staphylococcus aureus nasal colonization. Int J Med Microbiol 2008, 298 (5-6), 505-13. 58.

Walsh, E. J.; O'Brien, L. M.; Liang, X.; Hook, M.; Foster, T. J., Clumping factor B, a

fibrinogen-binding MSCRAMM (microbial surface components recognizing adhesive matrix molecules) adhesin of Staphylococcus aureus, also binds to the tail region of type I cytokeratin 10. J Biol Chem 2004, 279 (49), 50691-9. 59.

Wertheim, H. F.; Walsh, E.; Choudhurry, R.; Melles, D. C.; Boelens, H. A.; Miajlovic, H.;

Verbrugh, H. A.; Foster, T.; van Belkum, A., Key role for clumping factor B in Staphylococcus aureus nasal colonization of humans. PLoS Med 2008, 5 (1), e17. 60.

Corrigan, R. M.; Miajlovic, H.; Foster, T. J., Surface proteins that promote adherence of

Staphylococcus aureus to human desquamated nasal epithelial cells. BMC Microbiol 2009, 9, 22. 61.

Clarke, S. R.; Brummell, K. J.; Horsburgh, M. J.; McDowell, P. W.; Mohamad, S. A.;

Stapleton, M. R.; Acevedo, J.; Read, R. C.; Day, N. P.; Peacock, S. J.; Mond, J. J.; Kokai-Kun, J. F.; Foster, S. J., Identification of in vivo-expressed antigens of Staphylococcus aureus and their use in vaccinations for protection against nasal carriage. J Infect Dis 2006, 193 (8), 1098108. 62.

McCarthy, A. J.; Lindsay, J. A., Genetic variation in Staphylococcus aureus surface and

immune evasion genes is lineage associated: implications for vaccine design and hostpathogen interactions. BMC Microbiol 2010, 10, 173. 63.

Roche, F. M.; Meehan, M.; Foster, T. J., The Staphylococcus aureus surface protein

SasG and its homologues promote bacterial adherence to human desquamated nasal epithelial cells. Microbiology 2003, 149 (Pt 10), 2759-67.

ACS Paragon Plus Environment

36

Page 37 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

64.

Schaffer, A. C.; Solinga, R. M.; Cocchiaro, J.; Portoles, M.; Kiser, K. B.; Risley, A.;

Randall, S. M.; Valtulina, V.; Speziale, P.; Walsh, E.; Foster, T.; Lee, J. C., Immunization with Staphylococcus aureus clumping factor B, a major determinant in nasal carriage, reduces nasal colonization in a murine model. Infect Immun 2006, 74 (4), 2145-53. 65.

Kokai-Kun, J. F., The Cotton Rat as a Model for Staphylococcus aureus nasal

colonization in humans: cotton rat S. aureus nasal colonization model. Methods Mol Biol 2008, 431, 241-54. 66.

McAleese, F. M.; Walsh, E. J.; Sieprawska, M.; Potempa, J.; Foster, T. J., Loss of

clumping factor B fibrinogen binding activity by Staphylococcus aureus involves cessation of transcription, shedding and cleavage by metalloprotease. J Biol Chem 2001, 276 (32), 2996978. 67.

Ganesh, V. K.; Barbu, E. M.; Deivanayagam, C. C.; Le, B.; Anderson, A. S.; Matsuka, Y.

V.; Lin, S. L.; Foster, T. J.; Narayana, S. V.; Hook, M., Structural and biochemical characterization of Staphylococcus aureus clumping factor B/ligand interactions. J Biol Chem 2011, 286 (29), 25963-72. 68.

Xiang, H.; Feng, Y.; Wang, J.; Liu, B.; Chen, Y.; Liu, L.; Deng, X.; Yang, M., Crystal

structures reveal the multi-ligand binding mechanism of Staphylococcus aureus ClfB. PLoS Pathog 2012, 8 (6), e1002751. 69.

Ponnuraj, K.; Bowden, M. G.; Davis, S.; Gurusiddappa, S.; Moore, D.; Choe, D.; Xu, Y.;

Hook, M.; Narayana, S. V., A "dock, lock, and latch" structural model for a staphylococcal adhesin binding to fibrinogen. Cell 2003, 115 (2), 217-28. 70.

Vitry, P.; Valotteau, C.; Feuillie, C.; Bernard, S.; Alsteens, D.; Geoghegan, J. A.;

Dufrene, Y. F., Force-Induced Strengthening of the Interaction between Staphylococcus aureus Clumping Factor B and Loricrin. MBio 2017, 8 (6).

ACS Paragon Plus Environment

37

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

71.

Page 38 of 45

Lacey, K. A.; Mulcahy, M. E.; Towell, A. M.; Geoghegan, J. A.; McLoughlin, R. M.,

Clumping factor B is an important virulence factor during Staphylococcus aureus skin infection and a promising vaccine target. PLoS Pathog 2019, 15 (4), e1007713. 72.

Krismer, B.; Liebeke, M.; Janek, D.; Nega, M.; Rautenberg, M.; Hornig, G.; Unger, C.;

Weidenmaier, C.; Lalk, M.; Peschel, A., Nutrient limitation governs Staphylococcus aureus metabolism and niche adaptation in the human nose. PLoS Pathog 2014, 10 (1), e1003862. 73.

Skaar, E. P., The battle for iron between bacterial pathogens and their vertebrate hosts.

PLoS Pathog 2010, 6 (8), e1000949. 74.

Mazmanian, S. K.; Liu, G.; Jensen, E. R.; Lenoy, E.; Schneewind, O., Staphylococcus

aureus sortase mutants defective in the display of surface proteins and in the pathogenesis of animal infections. Proc Natl Acad Sci U S A 2000, 97 (10), 5510-5. 75.

Clarke, S. R.; Wiltshire, M. D.; Foster, S. J., IsdA of Staphylococcus aureus is a broad

spectrum, iron-regulated adhesin. Mol Microbiol 2004, 51 (5), 1509-19. 76.

Taylor, J. M.; Heinrichs, D. E., Transferrin binding in Staphylococcus aureus:

involvement of a cell wall-anchored protein. Mol Microbiol 2002, 43 (6), 1603-14. 77.

Vermeiren, C. L.; Pluym, M.; Mack, J.; Heinrichs, D. E.; Stillman, M. J., Characterization

of the heme binding properties of Staphylococcus aureus IsdA. Biochemistry 2006, 45 (42), 12867-75. 78.

Grigg, J. C.; Vermeiren, C. L.; Heinrichs, D. E.; Murphy, M. E., Haem recognition by a

Staphylococcus aureus NEAT domain. Mol Microbiol 2007, 63 (1), 139-49. 79.

Clarke, S. R.; Andre, G.; Walsh, E. J.; Dufrene, Y. F.; Foster, T. J.; Foster, S. J., Iron-

regulated surface determinant protein A mediates adhesion of Staphylococcus aureus to human corneocyte envelope proteins. Infect Immun 2009, 77 (6), 2408-16. 80.

Clarke, S. R.; Mohamed, R.; Bian, L.; Routh, A. F.; Kokai-Kun, J. F.; Mond, J. J.;

Tarkowski, A.; Foster, S. J., The Staphylococcus aureus surface protein IsdA mediates resistance to innate defenses of human skin. Cell Host Microbe 2007, 1 (3), 199-212.

ACS Paragon Plus Environment

38

Page 39 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

81.

Clarke, S. R.; Foster, S. J., IsdA protects Staphylococcus aureus against the bactericidal

protease activity of apolactoferrin. Infect Immun 2008, 76 (4), 1518-26. 82.

Brown, S.; Santa Maria, J. P., Jr.; Walker, S., Wall teichoic acids of Gram-positive

bacteria. Annu Rev Microbiol 2013, 67, 313-36. 83.

Sewell, E. W.; Brown, E. D., Taking aim at wall teichoic acid synthesis: new biology and

new leads for antibiotics. J Antibiot (Tokyo) 2014, 67 (1), 43-51. 84.

Weidenmaier, C.; Kokai-Kun, J. F.; Kristian, S. A.; Chanturiya, T.; Kalbacher, H.; Gross,

M.; Nicholson, G.; Neumeister, B.; Mond, J. J.; Peschel, A., Role of teichoic acids in Staphylococcus aureus nasal colonization, a major risk factor in nosocomial infections. Nat Med 2004, 10 (3), 243-5. 85.

Schade, J.; Weidenmaier, C., Cell wall glycopolymers of Firmicutes and their role as

nonprotein adhesins. FEBS Lett 2016, 590 (21), 3758-3771. 86.

Winstel, V.; Kuhner, P.; Salomon, F.; Larsen, J.; Skov, R.; Hoffmann, W.; Peschel, A.;

Weidenmaier, C., Wall Teichoic Acid Glycosylation Governs Staphylococcus aureus Nasal Colonization. MBio 2015, 6 (4), e00632. 87.

Xia, G.; Maier, L.; Sanchez-Carballo, P.; Li, M.; Otto, M.; Holst, O.; Peschel, A.,

Glycosylation of wall teichoic acid in Staphylococcus aureus by TarM. J Biol Chem 2010, 285 (18), 13405-15. 88.

Brown, S.; Xia, G.; Luhachack, L. G.; Campbell, J.; Meredith, T. C.; Chen, C.; Winstel,

V.; Gekeler, C.; Irazoqui, J. E.; Peschel, A.; Walker, S., Methicillin resistance in Staphylococcus aureus requires glycosylated wall teichoic acids. Proc Natl Acad Sci U S A 2012, 109 (46), 18909-14. 89.

Wells, J. A.; McClendon, C. L., Reaching for high-hanging fruit in drug discovery at

protein-protein interfaces. Nature 2007, 450 (7172), 1001-9. 90.

Arkin, M. R.; Tang, Y.; Wells, J. A., Small-molecule inhibitors of protein-protein

interactions: progressing toward the reality. Chem Biol 2014, 21 (9), 1102-14.

ACS Paragon Plus Environment

39

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

91.

Page 40 of 45

Kharat, A. S.; Tomasz, A., Inactivation of the srtA gene affects localization of surface

proteins and decreases adhesion of Streptococcus pneumoniae to human pharyngeal cells in vitro. Infect Immun 2003, 71 (5), 2758-65. 92.

Weiss, W. J.; Lenoy, E.; Murphy, T.; Tardio, L.; Burgio, P.; Projan, S. J.; Schneewind,

O.; Alksne, L., Effect of srtA and srtB gene expression on the virulence of Staphylococcus aureus in animal models of infection. J Antimicrob Chemother 2004, 53 (3), 480-6. 93.

Hazenbos, W. L.; Kajihara, K. K.; Vandlen, R.; Morisaki, J. H.; Lehar, S. M.;

Kwakkenbos, M. J.; Beaumont, T.; Bakker, A. Q.; Phung, Q.; Swem, L. R.; Ramakrishnan, S.; Kim, J.; Xu, M.; Shah, I. M.; Diep, B. A.; Sai, T.; Sebrell, A.; Khalfin, Y.; Oh, A.; Koth, C.; Lin, S. J.; Lee, B. C.; Strandh, M.; Koefoed, K.; Andersen, P. S.; Spits, H.; Brown, E. J.; Tan, M. W.; Mariathasan, S., Novel staphylococcal glycosyltransferases SdgA and SdgB mediate immunogenicity and protection of virulence-associated cell wall proteins. PLoS Pathog 2013, 9 (10), e1003653. 94.

Thomer, L.; Becker, S.; Emolo, C.; Quach, A.; Kim, H. K.; Rauch, S.; Anderson, M.;

Leblanc, J. F.; Schneewind, O.; Faull, K. F.; Missiakas, D., N-acetylglucosaminylation of serineaspartate repeat proteins promotes Staphylococcus aureus bloodstream infection. J Biol Chem 2014, 289 (6), 3478-86. 95.

Bella, M.; Koos, M.; Lin, C. H., Towards inhibitors of glycosyltransferases: A novel

approach to the synthesis of 3-acetamido-3-deoxy-D-psicofuranose derivatives. Beilstein J Org Chem 2015, 11, 1547-52. 96.

Gao, Z.; Ovchinnikova, O. G.; Huang, B. S.; Liu, F.; Williams, D. E.; Andersen, R. J.;

Lowary, T. L.; Whitfield, C.; Withers, S. G., High-Throughput "FP-Tag" Assay for the Identification of Glycosyltransferase Inhibitors. J Am Chem Soc 2019, 141 (6), 2201-2204. 97.

Zuegg, J.; Muldoon, C.; Adamson, G.; McKeveney, D.; Le Thanh, G.; Premraj, R.;

Becker, B.; Cheng, M.; Elliott, A. G.; Huang, J. X.; Butler, M. S.; Bajaj, M.; Seifert, J.; Singh, L.; Galley, N. F.; Roper, D. I.; Lloyd, A. J.; Dowson, C. G.; Cheng, T. J.; Cheng, W. C.; Demon, D.;

ACS Paragon Plus Environment

40

Page 41 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Meyer, E.; Meutermans, W.; Cooper, M. A., Carbohydrate scaffolds as glycosyltransferase inhibitors with in vivo antibacterial activity. Nat Commun 2015, 6, 7719. 98.

Vaideira, P. A.; Marcelo, F.; Grewal, R. K., Glycosyltransferase inhibitors: a promising

strategy to pave a path from laboratory to therapy. In Carbohydrate Chemistry: Chemical and Biological Approaches, 2018; Vol. 43, pp 135-158. 99.

Arvidson, S.; Tegmark, K., Regulation of virulence determinants in Staphylococcus

aureus. Int J Med Microbiol 2001, 291 (2), 159-70. 100.

Bronner, S.; Monteil, H.; Prevost, G., Regulation of virulence determinants in

Staphylococcus aureus: complexity and applications. FEMS Microbiol Rev 2004, 28 (2), 183200. 101.

Cheung, A. L.; Zhang, G., Global regulation of virulence determinants in Staphylococcus

aureus by the SarA protein family. Front Biosci 2002, 7, d1825-42. 102.

Said-Salim, B.; Dunman, P. M.; McAleese, F. M.; Macapagal, D.; Murphy, E.;

McNamara, P. J.; Arvidson, S.; Foster, T. J.; Projan, S. J.; Kreiswirth, B. N., Global regulation of Staphylococcus aureus genes by Rot. J Bacteriol 2003, 185 (2), 610-9. 103.

Xue, T.; You, Y.; Shang, F.; Sun, B., Rot and Agr system modulate fibrinogen-binding

ability mainly by regulating clfB expression in Staphylococcus aureus NCTC8325. Med Microbiol Immunol 2012, 201 (1), 81-92. 104.

Horsburgh, M. J.; Aish, J. L.; White, I. J.; Shaw, L.; Lithgow, J. K.; Foster, S. J., sigmaB

modulates virulence determinant expression and stress resistance: characterization of a functional rsbU strain derived from Staphylococcus aureus 8325-4. J Bacteriol 2002, 184 (19), 5457-67. 105.

Chien, Y.; Manna, A. C.; Projan, S. J.; Cheung, A. L., SarA, a global regulator of

virulence determinants in Staphylococcus aureus, binds to a conserved motif essential for sardependent gene regulation. J Biol Chem 1999, 274 (52), 37169-76.

ACS Paragon Plus Environment

41

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

106.

Page 42 of 45

Manna, A. C.; Ray, B., Regulation and characterization of rot transcription in

Staphylococcus aureus. Microbiology 2007, 153 (Pt 5), 1538-45. 107.

Killikelly, A.; Benson, M. A.; Ohneck, E. A.; Sampson, J. M.; Jakoncic, J.; Spurrier, B.;

Torres, V. J.; Kong, X. P., Structure-based functional characterization of repressor of toxin (Rot), a central regulator of Staphylococcus aureus virulence. J Bacteriol 2015, 197 (1), 188-200. 108.

Alonso, N.; Guillen, R.; Chambers, J. W.; Leng, F., A rapid and sensitive high-throughput

screening method to identify compounds targeting protein-nucleic acids interactions. Nucleic Acids Res 2015, 43 (8), e52. 109.

Yan, C.; Higgins, P. J., Drugging the undruggable: transcription therapy for cancer.

Biochim Biophys Acta 2013, 1835 (1), 76-85. 110.

Lambert, M.; Jambon, S.; Depauw, S.; David-Cordonnier, M. H., Targeting Transcription

Factors for Cancer Treatment. Molecules 2018, 23 (6). 111.

Arya, R.; Ravikumar, R.; Santhosh, R. S.; Princy, S. A., SarA based novel therapeutic

candidate against Staphylococcus aureus associated with vascular graft infections. Front Microbiol 2015, 6, 416. 112.

Zhu, Q.; Wen, W.; Wang, W.; Sun, B., Transcriptional regulation of virulence factors Spa

and ClfB by the SpoVG-Rot cascade in Staphylococcus aureus. Int J Med Microbiol 2018. 113.

Schulthess, B.; Meier, S.; Homerova, D.; Goerke, C.; Wolz, C.; Kormanec, J.; Berger-

Bachi, B.; Bischoff, M., Functional characterization of the sigmaB-dependent yabJ-spoVG operon in Staphylococcus aureus: role in methicillin and glycopeptide resistance. Antimicrob Agents Chemother 2009, 53 (5), 1832-9. 114.

Senn, M. M.; Giachino, P.; Homerova, D.; Steinhuber, A.; Strassner, J.; Kormanec, J.;

Fluckiger, U.; Berger-Bachi, B.; Bischoff, M., Molecular analysis and organization of the sigmaB operon in Staphylococcus aureus. J Bacteriol 2005, 187 (23), 8006-19. 115.

Kazmierczak, M. J.; Wiedmann, M.; Boor, K. J., Alternative sigma factors and their roles

in bacterial virulence. Microbiol Mol Biol Rev 2005, 69 (4), 527-43.

ACS Paragon Plus Environment

42

Page 43 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

116.

Depke, M.; Burian, M.; Schafer, T.; Broker, B. M.; Ohlsen, K.; Volker, U., The alternative

sigma factor B modulates virulence gene expression in a murine Staphylococcus aureus infection model but does not influence kidney gene expression pattern of the host. Int J Med Microbiol 2012, 302 (1), 33-9. 117.

Nicholas, R. O.; Li, T.; McDevitt, D.; Marra, A.; Sucoloski, S.; Demarsh, P. L.; Gentry, D.

R., Isolation and characterization of a sigB deletion mutant of Staphylococcus aureus. Infect Immun 1999, 67 (7), 3667-9. 118.

Fillat, M. F., The FUR (ferric uptake regulator) superfamily: diversity and versatility of key

transcriptional regulators. Arch Biochem Biophys 2014, 546, 41-52. 119.

Swoboda, J. G.; Meredith, T. C.; Campbell, J.; Brown, S.; Suzuki, T.; Bollenbach, T.;

Malhowski, A. J.; Kishony, R.; Gilmore, M. S.; Walker, S., Discovery of a small molecule that blocks wall teichoic acid biosynthesis in Staphylococcus aureus. ACS Chem Biol 2009, 4 (10), 875-83. 120.

D'Elia, M. A.; Pereira, M. P.; Chung, Y. S.; Zhao, W.; Chau, A.; Kenney, T. J.; Sulavik,

M. C.; Black, T. A.; Brown, E. D., Lesions in teichoic acid biosynthesis in Staphylococcus aureus lead to a lethal gain of function in the otherwise dispensable pathway. J Bacteriol 2006, 188 (12), 4183-9. 121.

Swoboda, J. G.; Campbell, J.; Meredith, T. C.; Walker, S., Wall teichoic acid function,

biosynthesis, and inhibition. Chembiochem 2010, 11 (1), 35-45. 122.

Lee, S. H.; Wang, H.; Labroli, M.; Koseoglu, S.; Zuck, P.; Mayhood, T.; Gill, C.; Mann,

P.; Sher, X.; Ha, S.; Yang, S. W.; Mandal, M.; Yang, C.; Liang, L.; Tan, Z.; Tawa, P.; Hou, Y.; Kuvelkar, R.; DeVito, K.; Wen, X.; Xiao, J.; Batchlett, M.; Balibar, C. J.; Liu, J.; Xiao, J.; Murgolo, N.; Garlisi, C. G.; Sheth, P. R.; Flattery, A.; Su, J.; Tan, C.; Roemer, T., TarO-specific inhibitors of wall teichoic acid biosynthesis restore beta-lactam efficacy against methicillin-resistant staphylococci. Sci Transl Med 2016, 8 (329), 329ra32.

ACS Paragon Plus Environment

43

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

123.

Page 44 of 45

Farha, M. A.; Leung, A.; Sewell, E. W.; D'Elia, M. A.; Allison, S. E.; Ejim, L.; Pereira, P.

M.; Pinho, M. G.; Wright, G. D.; Brown, E. D., Inhibition of WTA synthesis blocks the cooperative action of PBPs and sensitizes MRSA to beta-lactams. ACS Chem Biol 2013, 8 (1), 226-33. 124.

Farha, M. A.; Koteva, K.; Gale, R. T.; Sewell, E. W.; Wright, G. D.; Brown, E. D.,

Designing analogs of ticlopidine, a wall teichoic acid inhibitor, to avoid formation of its oxidative metabolites. Bioorg Med Chem Lett 2014, 24 (3), 905-10. 125.

Sobhanifar, S.; Worrall, L. J.; King, D. T.; Wasney, G. A.; Baumann, L.; Gale, R. T.;

Nosella, M.; Brown, E. D.; Withers, S. G.; Strynadka, N. C., Structure and Mechanism of Staphylococcus aureus TarS, the Wall Teichoic Acid beta-glycosyltransferase Involved in Methicillin Resistance. PLoS Pathog 2016, 12 (12), e1006067. 126.

Cascioferro, S.; Raffa, D.; Maggio, B.; Raimondi, M. V.; Schillaci, D.; Daidone, G.,

Sortase A Inhibitors: Recent Advances and Future Perspectives. J Med Chem 2015, 58 (23), 9108-23. 127.

Zhang, J.; Liu, H.; Zhu, K.; Gong, S.; Dramsi, S.; Wang, Y. T.; Li, J.; Chen, F.; Zhang, R.;

Zhou, L.; Lan, L.; Jiang, H.; Schneewind, O.; Luo, C.; Yang, C. G., Antiinfective therapy with a small molecule inhibitor of Staphylococcus aureus sortase. Proc Natl Acad Sci U S A 2014, 111 (37), 13517-22. 128.

Oh, K. B.; Mar, W.; Kim, S.; Kim, J. Y.; Oh, M. N.; Kim, J. G.; Shin, D.; Sim, C. J.; Shin,

J., Bis(indole) alkaloids as sortase A inhibitors from the sponge Spongosorites sp. Bioorg Med Chem Lett 2005, 15 (22), 4927-31. 129.

Zhulenkovs, D.; Rudevica, Z.; Jaudzems, K.; Turks, M.; Leonchiks, A., Discovery and

structure-activity relationship studies of irreversible benzisothiazolinone-based inhibitors against Staphylococcus aureus sortase A transpeptidase. Bioorg Med Chem 2014, 22 (21), 5988-6003.

ACS Paragon Plus Environment

44

Page 45 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

For Table of Contents Use Only

ACS Paragon Plus Environment

45