Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Amity L. Roberts is active.

Publication


Featured researches published by Amity L. Roberts.


BMC Pediatrics | 2012

Detection of group A Streptococcus in tonsils from pediatric patients reveals high rate of asymptomatic streptococcal carriage

Amity L. Roberts; Kristie L. Connolly; Daniel J. Kirse; Adele K. Evans; Katherine A. Poehling; Timothy R. Peters; Sean D. Reid

BackgroundGroup A Streptococcus (GAS) causes acute tonsillopharyngitis in children, and approximately 20% of this population are chronic carriers of GAS. Antibacterial therapy has previously been shown to be insufficient at clearing GAS carriage. Bacterial biofilms are a surface-attached bacterial community that is encased in a matrix of extracellular polymeric substances. Biofilms have been shown to provide a protective niche against the immune response and antibiotic treatments, and are often associated with recurrent or chronic bacterial infections. The objective of this study was to test the hypothesis that GAS is present within tonsil tissue at the time of tonsillectomy.MethodsBlinded immunofluorescent and histological methods were employed to evaluate palatine tonsils from children undergoing routine tonsillectomy for adenotonsillar hypertrophy or recurrent GAS tonsillopharyngitis.ResultsImmunofluorescence analysis using anti-GAS antibody was positive in 11/30 (37%) children who had tonsillectomy for adenotonsillar hypertrophy and in 10/30 (33%) children who had tonsillectomy for recurrent GAS pharyngitis. Fluorescent microscopy with anti-GAS and anti-cytokeratin 8 & 18 antibodies revealed GAS was localized to the tonsillar reticulated crypts. Scanning electron microscopy identified 3-dimensional communities of cocci similar in size and morphology to GAS. The characteristics of these communities are similar to GAS biofilms from in vivo animal models.ConclusionOur study revealed the presence of GAS within the tonsillar reticulated crypts of approximately one-third of children who underwent tonsillectomy for either adenotonsillar hypertrophy or recurrent GAS tonsillopharyngitis at the Wake Forest School of Medicine.Trial RegistrationThe tissue collected was normally discarded tissue and no patient identifiers were collected. Thus, no subjects were formally enrolled.


Microbiology | 2009

Biofilm formation by group A Streptococcus: a role for the streptococcal regulator of virulence (Srv) and streptococcal cysteine protease (SpeB)

Christopher D. Doern; Amity L. Roberts; Wenzhou Hong; Jessica Nelson; Slawomir Lukomski; William E. Swords; Sean D. Reid

Recently, biofilms have become a topic of interest in the study of the human pathogen group A Streptococcus (GAS). In this study, we sought to learn more about the make-up of these structures and gain insight into biofilm regulation. Enzymic studies indicated that biofilm formation by GAS strain MGAS5005 required an extracellular protein and DNA component(s). Previous results indicated that inactivation of the transcriptional regulator Srv in MGAS5005 resulted in a significant decrease in virulence. Here, inactivation of Srv also resulted in a significant decrease in biofilm formation under both static and flow conditions. Given that production of the extracellular cysteine protease SpeB is increased in the srv mutant, we tested the hypothesis that increased levels of active SpeB may be responsible for the reduction in biofilm formation. Western immunoblot analysis indicated that SpeB was absent from MGAS5005 biofilms. Complementation of MGAS5005Deltasrv restored the biofilm phenotype and eliminated the overproduction of active SpeB. Inhibition of SpeB with E64 also restored the MGAS5005Deltasrv biofilm to wild-type levels.


PLOS ONE | 2011

Dispersal of Group A streptococcal biofilms by the cysteine protease SpeB leads to increased disease severity in a murine model.

Kristie L. Connolly; Amity L. Roberts; Robert C. Holder; Sean D. Reid

Group A Streptococcus (GAS) is a Gram-positive human pathogen best known for causing pharyngeal and mild skin infections. However, in the 1980s there was an increase in severe GAS infections including cellulitis and deeper tissue infections like necrotizing fasciitis. Particularly striking about this elevation in the incidence of severe disease was that those most often affected were previously healthy individuals. Several groups have shown that changes in gene content or regulation, as with proteases, may contribute to severe disease; yet strains harboring these proteases continue to cause mild disease as well. We and others have shown that group A streptococci (MGAS5005) reside within biofilms both in vitro and in vivo. That is to say that the organism colonizes a host surface and forms a 3-dimensional community encased in a protective matrix of extracellular protein, DNA and polysaccharide(s). However, the mechanism of assembly or dispersal of these structures is unclear, as is the relationship of these structures to disease outcome. Recently we reported that allelic replacement of the streptococcal regulator srv resulted in constitutive production of the streptococcal cysteine protease SpeB. We further showed that the constitutive production of SpeB significantly decreased MGAS5005Δsrv biofilm formation in vitro. Here we show that mice infected with MGAS5005Δsrv had significantly larger lesion development than wild-type infected animals. Histopathology, Gram-staining and immunofluorescence link the increased lesion development with lack of disease containment, lack of biofilm formation, and readily detectable levels of SpeB in the tissue. Treatment of MGAS5005Δsrv infected lesions with a chemical inhibitor of SpeB significantly reduced lesion formation and disease spread to wild-type levels. Furthermore, inactivation of speB in the MGAS5005Δsrv background reduced lesion formation to wild-type levels. Taken together, these data suggest a mechanism by which GAS disease may transition from mild to severe through the Srv mediated dispersal of GAS biofilms.


Infection and Immunity | 2010

Loss of the Group A Streptococcus Regulator Srv Decreases Biofilm Formation In Vivo in an Otitis Media Model of Infection

Amity L. Roberts; Kristie L. Connolly; Christopher D. Doern; Robert C. Holder; Sean D. Reid

ABSTRACT Group A Streptococcus (GAS) is a common causative agent of pharyngitis, but the role of GAS in otitis media is underappreciated. In this study, we sought to test the hypothesis that GAS colonizes the middle ear and establishes itself in localized, three-dimensional communities representative of biofilms. To test this hypothesis, the middle ears of chinchillas were infected with either a strain of GAS capable of forming biofilms in vitro (MGAS5005) or a strain deficient in biofilm formation due to the lack of the transcriptional regulator Srv (MGAS5005 Δsrv). Infection resulted in the formation of large, macroscopic structures within the middle ears of MGAS5005- and MGAS5005 Δsrv-infected animals. Plate counts, scanning electron microscopy, LIVE/DEAD staining, and Gram staining revealed a difference in the distributions of MGAS5005 versus MGAS5005 Δsrv in the infected samples. High numbers of CFU of MGAS5005 Δsrv were isolated from the middle ear effusion, and MGAS5005 Δsrv was found randomly distributed throughout the excised macroscopic structure. In contrast, MGAS5005 was found in densely packed microcolonies indicative of biofilms within the excised material from the middle ear. CFU levels of MGAS5005 from the effusion were significantly lower than that of MGAS5005 Δsrv early during the course of infection. Allelic replacement of the chromosomally encoded streptococcal cysteine protease (speB) in the MGAS5005 Δsrv background restored biofilm formation in vivo. Interestingly, our results suggest that GAS naturally forms a biofilm during otitis media but that biofilm formation is not required to establish infection following transbullar inoculation of chinchillas.


BioMed Research International | 2015

Diagnostic Options and Challenges for Dengue and Chikungunya Viruses.

Stacey K. Mardekian; Amity L. Roberts

Dengue virus (DENV) and Chikungunya virus (CHIKV) are arboviruses that share the same Aedes mosquito vectors and thus overlap in their endemic areas. These two viruses also cause similar clinical presentations, especially in the initial stages of infection, with neither virus possessing any specific distinguishing clinical features. Because the outcomes and management strategies for these two viruses are vastly different, early and accurate diagnosis is imperative. Diagnosis is also important for surveillance, outbreak control, and research related to vaccine and drug development. Available diagnostic tests are aimed at detection of the virus, its antigenic components, or the host immune antibody response. In this review, we describe the recent progress and continued challenges related to the diagnosis of DENV and CHIKV infections.


Diagnostic Microbiology and Infectious Disease | 2013

Susceptibility of enterococci to daptomycin is dependent upon testing methodology

Kendall A. Bryant; Amity L. Roberts; Mark E. Rupp; James R. Anderson; Elizabeth Lyden; Paul D. Fey; Trevor C. Van Schooneveld

An increase in daptomycin nonsusceptible enterococci (DNSE) was noted in our institution (8.3% 2008 to 34.5% 2011) using MicroScan methods which may overestimate DNSE prevalence. DNSE (N = 150) from the clinical laboratory (2008-2011) underwent susceptibility testing using broth microdilution (BMD), Etest, Sensititire, MicroScan prompt (MSP), and MicroScan turbidity (MST) with only 20% of isolates confirmed as nonsusceptible. Categorical and essential agreement were highest with MSP and MST, but both missed the majority of resistant isolates (70% and 87% missed). Etest MIC values were statistically higher, more likely to be nonsusceptible, had the lowest very major error rate (37%), and the highest falsely nonsusceptible rate (22%). Sensititre MIC values were not statistically different from BMD, but missed 57% of DNSE. PFGE analysis did not define a clonal outbreak. These findings suggest that MicroScan methods overestimate nonsusceptibility, and the lack of correlation between methods raises questions regarding which method is most effective at confirming nonsusceptibility.


BMC Research Notes | 2010

Allelic replacement of the streptococcal cysteine protease SpeB in a Δsrv mutant background restores biofilm formation

Amity L. Roberts; Robert C. Holder; Sean D. Reid

BackgroundGroup A Streptococcus (GAS) is a Gram-positive human pathogen that is capable of causing a wide spectrum of human disease. Thus, the organism has evolved to colonize a number of physiologically distinct host sites. One such mechanism to aid colonization is the formation of a biofilm. We have recently shown that inactivation of the streptococcal regulator of virulence (Srv), results in a mutant strain exhibiting a significant reduction in biofilm formation. Unlike the parental strain (MGAS5005), the streptococcal cysteine protease (SpeB) is constitutively produced by the srv mutant (MGAS5005Δsrv) suggesting Srv contributes to the control of SpeB production. Given that SpeB is a potent protease, we hypothesized that the biofilm deficient phenotype of the srv mutant was due to the constitutive production of SpeB. In support of this hypothesis, we have previously demonstrated that treating cultures with E64, a commercially available chemical inhibitor of cysteine proteases, restored the ability of MGAS5005Δsrv to form biofilms. Still, it was unclear if the loss of biofilm formation by MGAS5005Δsrv was due only to the constitutive production of SpeB or to other changes inherent in the srv mutant strain. To address this question, we constructed a Δsrv ΔspeB double mutant through allelic replacement (MGAS5005Δsrv ΔspeB) and tested its ability to form biofilms in vitro.FindingsAllelic replacement of speB in the srv mutant background restored the ability of this strain to form biofilms under static and continuous flow conditions. Furthermore, addition of purified SpeB to actively growing wild-type cultures significantly inhibited biofilm formation.ConclusionsThe constitutive production of SpeB by the srv mutant strain is responsible for the significant reduction of biofilm formation previously observed. The double mutant supports a model by which Srv contributes to biofilm formation and/or dispersal through regulation of speB/SpeB.


Fems Microbiology Letters | 2007

Susceptibility of compound 48/80‐sensitized Pseudomonas aeruginosa to the hydrophobic biocide triclosan

Matthew L. Ellison; Amity L. Roberts; Franklin R. Champlin

Pseudomonas aeruginosa is intrinsically resistant to the hydrophobic biocide triclosan, and yet it can be sensitized to low concentrations by permeabilization of the outer membrane using compound 48/80. A selective plating assay revealed that compound 48/80-permeabilized YM64, a triclosan-recognizing efflux pump-deficient variant, was unable to initiate growth on a medium containing triclosan. Macrobroth dilution assay data revealed that treatment with compound 48/80 synergistically decreased minimal inhibitory concentrations of the hydrophobic antibacterial agents rifamycin SV and chloramphenicol for all cell envelope variant strains examined. A low concentration of triclosan exerted a transient bactericidal effect on permeabilized wild-type strain PAO1, after which exponential growth resumed within 4 h. Permeabilized strain YM64 was unable to overcome the inhibition; yet, both strains remained susceptible to chloramphenicol for as long as 6 h, thereby suggesting that the outer membrane remained permeable to nonpolar compounds. These data support the notion that the transitory nature of compound 48/80 sensitization to triclosan in P. aeruginosa does not involve obviation of the hydrophobic diffusion pathway through the outer membrane. The inability of strain YM64 to overcome the synergistic effect of compound 48/80 and triclosan strongly suggests that triclosan-recognizing efflux pumps are involved in maintaining viability in wild-type cells whose outer membranes are otherwise compromised.


Diagnostic Microbiology and Infectious Disease | 2016

Evaluation of matrix-assisted laser desorption ionization–time-of-flight mass spectrometry to differentiate between Candida albicans and Candida dubliniensis

Amity L. Roberts; Aqilah Alelew; Peter C. Iwen

Matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (MALDI-TOF-MS) analysis in conjunction with the direct formic acid (FA) sample processing method was evaluated for the ability to differentiate the closely related species of Candida albicans and Candida dubliniensis. The results showed that MALDI-TOF-MS, using the direct FA method, was reliable to differentiate between these species.


Methods of Molecular Biology | 2014

Identification of Staphylococcus epidermidis in the Clinical Microbiology Laboratory by Molecular Methods

Amity L. Roberts

Biochemical assays for the phenotypic identification of coagulase-negative staphylococci in the clinical microbiology laboratory have been well described in previous publications (Becker and Von Eiff Manual of Clinical Microbiology, ASM Press, Washington, pp. 308-330, 2011; Kloos and Wolfshohl J Clin Microbiol 16:509-516, 1982). This discussion focuses on identification of Staphylococcus epidermidis through molecular and proteomic methods. Molecular assays have been shown to be more discriminatory between the coagulase-negative staphylococcal species than are phenotypic assays (Zadoks and Watts Vet Microbiol 134:20-28, 2009; Sheraba et al. BMC Res Notes 3:278, 2010; Patteet et al. Eur J Clin Microbiol Infect Dis 31:747-751, 2012). The molecular and proteomic methods that have shown the greatest utilization potential within the clinical laboratory are as follows: PCR amplification and sequencing of discriminatory genes, real-time polymerase chain reaction with species-specific probes in conjunction with a melt-curve analysis, fluorescence in situ hybridization, and matrix-assisted laser desorption-ionization time-of-flight mass spectrometry.

Collaboration


Dive into the Amity L. Roberts's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter C. Iwen

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christopher D. Doern

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kaede V. Sullivan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Laura Biederman

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Patricia Couto

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Paul D. Fey

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Upasana Joneja

Thomas Jefferson University

View shared research outputs
Researchain Logo
Decentralizing Knowledge