powerpoint presentation · 1961. he then joined the laboratory of angus graham at the wistar...

39
BCID 2018 May 23rd to 27th Banff Centre for Arts and Creativity Banff Conference on Infectious Diseases Program and Abstracts Cover photo of Banff Avenue credit – Diane E Armstrong

Upload: others

Post on 05-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

BCID 2018

May 23rd to 27th

Banff Centre for Arts and Creativity

Banff Conference on Infectious Diseases Program and Abstracts

Cover photo of Banff Avenue credit – Diane E Armstrong

Page 2: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

2

BCID Sponsors

Biological sciences Microbiology, Immunology and Infectious Diseases

Microbial Research Group

Page 3: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

Banff Conference on Infectious Diseases

Initiated in 1982 by Dr. William Paranchych, Dr. Don Woods and their colleagues from the Universities of Alberta and Calgary, the Banff Conference on Infectious Diseases (formerly the UA-UC Conference on Infectious Diseases) represents a unique forum for internationally recognized experts to present their research on pathogenesis and other aspects of infectious diseases and host defenses. Informal poster sessions also allow registrants the opportunity to present their work. Nestled in the Canadian Rocky Mountains, the informal setting is conducive to discussing science with investigators at the forefront of their research areas. The conference includes scientific sessions in the mornings and evenings. The afternoons present an opportunity to explore the impressive surroundings with your scientific colleagues. The nineteenth biennial Banff Conference on Infectious Diseases will be held May 23rd to 27th, 2018 at the Banff Centre for Arts and Creativity located in Banff, Alberta, Canada. Created as Canada’s first national park, Banff and its famous hot springs have been a tourist attraction for the past 130 years. The town offers a host of excellent restaurants, shops, and museums. Hiking, golfing, trail riding, skiing, fly-fishing, or soaking in the sulfur hot springs are a few of Banff’s many attractions.

Acknowledgements

The organizers gratefully acknowledge the very capable assistance of Taunya Shewciw for administrative support, Wei Chong (Maritime Travel), Kristen Trueman (University of Calgary Conference and Event Management), Julie Pecsi-Miller (Banff Centre Conference Services manager), and Vashti Communications.

3

Page 4: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

BCID 2018 Organizing Committee Dr. Glen Armstrong (Chair), Dr. Nathan Peters (Co-Chair), Dr. George Chaconas, Dr. John Conly, Dr. Jeroen De Buck, Dr. Rebekah DeVinney, Dr. Tao Dong, Dr. Matthias Gotte (U of Alberta), Dr. Joe Harrison, Dr. Chris Mody, Dr. Dylan Pillai, Dr. Tracy Raivio (U of Alberta), Dr. Tony Schryvers, Dr. Alexei Savchenko, Dr. Doug Storey, Dr. Guido van Marle, Dr. Kunyan Zhang.

Conference General Information

Hotel check in and the BCID Registration desk are located in the Main Lobby Lounge of the Professional Development Centre (see map). All oral presentations will be held in the Max Bell Auditorium. Poster Sessions and Breaks will be in the Elder Tom Crane Bear Lounge and Max Bell Central Foyer. Poster abstracts available online at: www.ucalgary.ca/bcid Breakfast and lunch (name tags required) will be served in the Vistas Dining Room on Thursday May 24th through Saturday May 26th. Breakfast only will be available in the Vistas Dining room on Sunday morning, May 27th. There will be a welcome reception in the Max Bell Central Foyer immediately following the Opening Session on Wednesday May 23rd. Conference participants are responsible for their own dinners on Thursday May 24th and Saturday May 26th. Dinner and an evening of socializing will be held on Friday May 25th in the Kinnear Centre, room 101. Speakers are asked to provide their PowerPoint presentations on a USB stick 30 minutes prior to their session. All posters can be put up on the morning of Thursday, May 24th and taken down at the end of Poster Session 2 on Friday, May 25th. Presenters of even numbered posters should be available to answer questions during Poster Session 1 on Thursday, May 24th. Presenters of odd numbered posters should be available to answer questions during Poster Session 2 on Friday, May 25th. Poster sessions will run from 12:30 – 2:30 p.m. A Sponsors Translational and Industry session will be held from 2:30 to 4:30 p.m. on Friday, May 25th in the Max bell Auditorium.

4

Page 5: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

5

Hotel check in and BCID registration

Max Bell Auditorium

ETCB Lounge

Vistas Restaurant Breakfast/Lunch

Kinnear Centre

Banff Centre for Arts and Creativity

Parking

Page 6: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

BCID Founders

William Paranchych Dr. William Paranchych was born in Drumheller, Alberta in 1933 where he also grew up. Bill came to the University of Alberta to study chemistry in 1950. Bill’s first scientific paper was published in 1957 during his M.Sc. in biochemistry under Professor Jules Tuba. He completed his Ph.D. at McGill in 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science, the discovery of the ssRNA bacteriophage R17, isolated from the sewers of Philadelphia. John Coulter, the chair of the Department of Biochemistry at the University of Alberta, hired Bill to join the department in 1963. In following the R17 story where curiosity would lead him, Bill eventually came to study its mode of infection in bacteria, which involves attachment to the F pilus of E. coli and from thence moved on to the study of pili more generally, focusing for the last 15+ years on Pseudomonas aeruginosa. Bill was a member of the Department of Biochemistry from 1963 until 1989 when he was lured away to become Chair of Microbiology in the University of Alberta’s Faculty of Science. In 1994 he became the first chair of the newly formed Department of Biological Sciences, which incorporated his department as well as several others. There are many things one could say about Bill the academic and scientist to illustrate his abilities, perhaps the most telling is that he continually attracted some of the best students and led them to research careers. In recruiting to the research effort, there was no one better. Bill was an enthusiast who was able to sweep people into his enthusiasm by his warmth, intelligence, and genuine interest in others. Bill’s greatest exuberance was for his students and he yielded to no one in his loyalty, encouragement, and helpfulness to them. Bill passed away at the age of 62 in July 1995. The William Paranchych Lecture has been named in honor of his great contributions to microbiology both nationally and internationally (adapted from attribute to Bill Paranchych by Vern Paetkau, U of A Sept. 1995)

6

Page 7: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

Donald E. Woods Dr. Donald (Don) Woods had a distinguished career as a research scientist investigating the pathogenesis of disease caused by bacterial organisms. He earned his Ph.D. in Microbiology from the University of Texas Health Sciences Centre, San Antonio, Texas then joined the Department of Microbiology and Infectious Diseases at the University of Calgary in 1982 after completing a Postdoctoral Research Fellowship in the Department of Microbiology and Immunology at the Oregon Health Sciences University in Portland, Oregon. Initially appointed as an Assistant Professor and Research Scholar of the Alberta Heritage Foundation for Medical Research, he then became a Professor in 1991. Dr. Woods was the Director of the Research Development Program II of the Canadian Cystic Fibrosis Foundation from 1988-1993, Chairman of the Department of Microbiology and Infectious Diseases from 1991 to 1996, then became Scientific Director of the Canadian Bacterial Diseases Network (CBDN), a national network of scientists associated with the Network of Centres of Excellence Program of Canada. He and his laboratory have made significant contributions to the understanding of the pathogenesis of disease due to Pseudomonas and Burkholderia spp. Dr. Woods was one of the first recipients of a Canada Research Chair and is a Fellow of the Canadian Academy of Health Sciences. The Donald E. Woods Young Investigator Lecture was initiated in 2012 to honour his contribution to microbiology and BCID.

7

Page 8: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

BCID Program

May 23rd to 27th, 2018 – Banff Centre Wednesday – May 23 3:00 to 6:30 p.m. – Check-in and registration. ______________________________________________________________ 6:30 to 9:40 p.m. – Session 1 – New Horizons Chair: Dr. Olivia Steele-Mortimer 6:30 to 6:40 p.m. – Opening remarks 6:40 to 7:40 p.m. – Donald E. Woods Young Investigator Lecture - Jennifer Gaddy (Vanderbilt University) - Zinc homeostasis in C. jejuni. 7:40 to 8:20 p.m. - Alexei Savchenko (U of Calgary) - Hijacking of the host ubiquitination processes by bacterial pathogens. 8:20 to 9:00 p.m. – Keith Pardee (U of Toronto) - Using Cell-free Synthetic Biology to Build Tools for Human Health. 9:00 p.m. – Opening reception. ______________________________________________________________ Thursday – May 24 8:30 – 11:50 a.m. – Session 2 – The Microbiome Chair: Dr. Tao Dong 8:30 to 9:10 a.m. – Gregor Reid – Western University – Probiotics to counter infectious diseases. 9:10 to 9:50 a.m. - Marie-Claire Arrieta (U of Calgary) - The early-life microbiome influences host immune dysregulation. 9:50 to 10:10 a.m. – Break. 10:10 to 10:50 a.m. - Cara Haney (UBC) - Manipulation and evasion of plant immunity by beneficial microbes.

8

Page 9: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

10:50 to 11:30 a.m. – Emma Allen-Vercoe (Guelph) – Microbial ecosystem therapeutics: bugs as drugs. 11:30 to 11:50 a.m. – Sonu Subudhi - Stressors in persistently infected bats may influence the severity of the disease and/or increase virus shedding. 11:50 a.m. to 1:00 p.m. – Lunch. 12:30 – 2:30 p.m. – Poster session 1. ______________________________________________________________ 7:00 – 9:40 p.m. – Session 3 – Microbial Diagnostics and Therapeutics Chair: Dr. Bryan Yipp 7:00 to 7:40 p.m. – Cezar Khursigara (U of Guelph) - Quantitative Proteomics to Study Antimicrobial Resistance. 7:40 to 8:20 p.m. - Tom Louie (U of Calgary) – Fecal bugs down the hatch: unpalatable, unacceptable and unsafe or passport to the lower GI tract? 8:20 to 8:40 p.m. – Break. 8:40 to 9:20 p.m. – Ian Lewis (U of Calgary) - Harnessing metabolomics to fight infections. 9:20 to 9:40 p.m. – Brent Weber - Developing new therapies to treat bloodstream infections caused by Klebsiella pneumoniae. ______________________________________________________________ Friday – May 25 8:30 – 11:50 a.m. – Session 4 – Infection and Immunity. Chair: Dr. Tony Schryvers 8:30 to 9:10 a.m. - David Marchant (U of Alberta) - The Transmission, Replication and Treatment of Respiratory Syncytial Virus. 9:10 to 9:50 a.m. - Bryan Yipp (U of Calgary) - Lung vascular neutrophils mediate host defense in vivo. 9:50 to 10:10 a.m. – Break. 10:10 to 10:50 a.m. - Don Sheppard (McGill) – Fungal biofilms at the host pathogen interphase.

9

Page 10: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

10:50 to 11:30 a.m. - Dawn Bowdish (McMaster) - Age-related inflammation increases susceptibility to Streptococcus pneumoniae. 11:30 to 11:50 a.m. – Hong Bing Yu - Vasoactive Intestinal Peptide (VIP) Promotes Intestinal Host Defense by Modulating Group 3 Innate Lymphoid Cells. 11:50 a.m. to 1:00 p.m. – Lunch. 12:30 – 2:30 p.m. – Poster session 2. 2:30 – 4:30 p.m. – Translational Research and Industry. Chair: Dr. Dylan Pillai 2:30 – 3:10 p.m. - Dr. Angel Chu – Alberta Health Services - Evaluating influenza vaccines and treatment options in older adults. 3:10 – 3:50 p.m. - Dr. Erik Wendlandt, Integrated DNA Technologies – Navigating qPCR assay design, What is the best chemistry for my application? 3:50 – 4:30 p.m. – Dr. Patrick R Murray, PhD, Sr Director WW Scientific Affairs BD Life Sciences - Future Directions in Clinical Microbiology. 4:30 p.m. – 5:30 p.m. – William Paranchych Lecture. Chair: Dr. Glen Armstrong Dr. Gerry Wright (McMaster University) – Using resistance and natural history to find new antibiotics. 6:00 – 10:30 p.m. – Social evening. ______________________________________________________________ Saturday – May 26 8:30 – 11:50 a.m. – Session 5 – Pathogenesis I Chair: Dr John Conly 8:30 to 9:10 a.m. - Tracy Raivio (U of Alberta) – The Role of a Bacterial Envelope Stress Response in Surface Sensing, Pathogenesis, and Antibiotic Resistance.

10

Page 11: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

9:10 to 9:50 a.m. - David Heinrichs (Western U) – Staphylococcus aureus pathogenesis. 9:50 to 10:10 a.m. – Break. 10:10 to 10:50 a.m. - Brian Coombes (McMaster) - Harnessing immunity to combat superbugs. 10:50 to 11:30 a.m. – Peter Billingsley (Sanaria Inc.) - Malaria vaccination. 11:30 to 11:50 a.m. – Sahar Bagheri - Identification of unique genomic elements in Prevotella bivia strains capable of ascending the urogenital tract. 11:50 a.m. to 1:00 p.m. – Lunch. 1:00 p.m. – 4:40 p.m. – Session 6 – Pathogenesis II Chair: Dr. Alexei Savchenko 1:00 p.m. – 1:40 p.m. – George Chaconas (U of Calgary) - Hematogenous dissemination of the Lyme disease spirochete. 1:40 p.m. – 2:20 p.m. – Jeroen De Buck (U of Calgary) - A spectrum of pathogenicity in twenty-seven Staphylococcus spp. in intramammary infections. 2:20 p.m. – 2:40 p.m. – Break 2:40 p.m. – 3:20 p.m. - Ted Hackstadt (RML) – Strain dependent variation in virulence of Rocky Mountain spotted fever rickettsia. 3:20 p.m. – 3:40 p.m. – Peter Stogios - Tracking the source of antimicrobial resistance determinants: structural characterization of the intrinsic colistin resistance enzyme from Moraxella catarrhalis. ______________________________________________________________ Sunday – May 27 7:00 – 9:30 a.m. – Breakfast. 11:00 a.m. – Checkout. ______________________________________________________________

11

Page 12: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

Poster Abstracts

P1. Type VI Secretion System Dynamics and Modeling. Maria Silvina Stietz1, Xiaoye Liang1, Megan Wong1, Steven Hersch1, Tao G. Dong1,2,3,4

1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine; 2 Snyder Institute for Chronic Diseases; 3 Department of Biochemistry and Molecular Biology; 4

Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, T2N4Z6, Canada. Bacteria overtake their competitors utilizing a diverse range of strategies. The type VI secretion system (T6SS) is a contractile needle-like organelle that propels toxin effectors directly into the cytosol of surrounding cells. The T6SS needle is anchored to the cell envelope by a cytosolic baseplate and a membrane-spanning complex. Conformational changes in the baseplate are believed to initiate sheath contraction. However, the signals that define polymerization dynamics are unknown. Here we investigated the mechanisms that regulate T6SS polymerization in the Gram-negative bacterium Vibrio cholerae. Using a spheroplast model to image the dynamics of the T6SS contraction, we found that the T6SS could undergo similar contraction and assembly process as in untreated cells. However, the T6SS needles displayed high variation in lengths that is likely dependent on the orientation of the initial polymerization. This was also observed using non-contractile needle mutants. Our results highlight the flexibility of the T6SS length and lead us to propose a conceptual model for the dynamic process of T6SS firing and recycling.

P2. Signal transduction of Type VI Secretion System Effectors in Vibrio cholerae. Fatima Kamal1, Xiaoye Liang1, Steven Hersch1, Tao G. Dong1,2,3,4

1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine; 2 Snyder Institute for Chronic Diseases; 3 Department of Biochemistry and Molecular Biology; 4 Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, T2N4Z6, Canada. Type VI secretion system (T6SS) is a multi-component system which allows bacteria to compete with other microorganisms in the environment. The T6SS structure is analogous to a bacteriophage tail and is composed of an inner tube made up of Hcp proteins, a puncturing tip consisting of VgrG and PAAR proteins, a membrane-bound baseplate and an outer sheath that wraps around the Hcp tube. The contraction of the sheath propels the inner Hcp tube and delivers the toxic effectors which are carried in the tube or loaded on VgrG/PAAR into the nearby cells. H1-T6SS of Pseudomonas aeruginosa has been shown to be involved in counterattack of T6SS positive Vibrio cholerae and Acinetobacter baylyi. More killing is observed for T6SS active V. cholerae cells compared to the T6SS defective cells. However, it is not clear whether effectors contribute to such Tit-for-Tat response. In our study, we investigated the role of V. cholera effectors in the counter attack. Our findings indicate effectors contribute to eliciting attack by P. aeruginosa. We report that membrane damage caused by effector activities transduces a signal to activate the retaliatory response by P. aeruginosa.

12

Page 13: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P3. Probucol attenuates complete Freund adjuvant-induced inflammatory hyperalgesia by targeting the NF-κB pathway in paw and spinal cord.

Zucoloto, A. Z.1,2,3; Manchope, M. F.3; Fattori, V.3; Borghi, S. M.3; Garcia, S. B.3, Casagrande, R.4; Verri, W. A. Jr.3 1Snyder Institute for Chronic Diseases, University of Calgary, HRIC 4C51, 3230 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada; 2Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, HRIC 4C51, 3230 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada; 3Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil 4Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Estadual de Londrina, Londrina, Brazil This study aimed to evaluate the effect of probucol, a lipid-lowering agent, in a model of inflammatory hyperalgesia induced by complete Freund ajudvant (CFA). CFA is a water-in-oil emulsion made of non-metabolizable oils and heat killed Mycobacterium tuberculosis (MTB). MTB cell wall structures are sensed by at least 3 different toll-like receptors (TLRs) as well as NOD2 and Dectin-1. Triggering of these receptors by MTB was previously shown to induce the activation of the NF-κB pathway and subsequent production of TNF-α, IL-6 and IL-10. These mechanisms underlie CFA-mediated long-lasting nociception, which is characterized by glial activation and spinal production of pro-inflammatory mediators. To evaluate the analgesic and anti-inflammatory activity of probucol and the mechanisms involved, probucol at 0.3-3 mg/kg was administrated p.o. daily 24 h after CFA intraplantar injection. Mechanical and thermal hyperalgesia induced by CFA were determined over 7 days using an electronic anesthesiometer and hot plate apparatus, respectively. Paw edema was measured using a dial thickness gauge and overt pain-like behaviors were determined by the number of flinches and time spent licking paw. Cytokine production and NF-кB activation were evaluated 3 days post-stimulus by ELISA and Iba-1 and Gfap expression by immunofluorescence, as parameters of glial activation. Over 7 days, daily treatment with probucol reduced CFA-induced mechanical and thermal hyperalgesia and paw edema. Our results suggest that inhibition of the NF-κB pathway may underlie these effects, as peripheral and central cytokines levels as well as glial activation were reduced in probucol-treated animals.

P4. Characterization of Type IV Pili in Klebsiella pneumoniae strains from neonatal sepsis. LÓPEZ REYNOSO CÉSAR ISRAEL1,2, LOZANO ZARAÍN PATRICIA2, MARÍA CRISTINA GONZÁLEZ VÁZQUEZ2, MARTÍNEZ DE LA PEÑA CLAUDIA FABIOLA2 1Maestría en Ciencias, 2Centro de Investigaciones en Ciencias Microbiológicas – Instituto de Ciencias, BUAP, Puebla, México. Bacteria are endowed with a wide repertoire of filamentous structures on their surface. The type IV pili (T4P) is an exceptional example since they are used in various functions ranging from adherence, aggregation, DNA uptake, twitching motility, bacteriophages uptake and electrical conductance. T4P are formed by pilin proteins, a basal body composed by inner membrane proteins, two ATPases, and a porin. Klebsiella pneumoniae has been considered a non-motile bacterium, however, our lab characterized strains that present motility mediated by flagellum that was identified by electron microscopy. In the same micrographs we also observed putative T4P structures. Therefore, we are investigating if K. pneumoniae strains are capable to express a functional T4P. In the present work, we identified by PCR the pilin gene on 26 K. pneumoniae strains isolated from neonatal sepsis. In order to identify if the T4P is involved in the twitching motility we tested for it on three different 2% agar media, Luria-Bertani, brain-heart infusion and blood agar. We incubated at different times and temperatures. Our results showed a greater motility on BHI plates at 37ºC and after 96 hours of incubation, observing the characteristic twitching phenotype. To know if there is a relationship between the twitching phenotype and the expression of T4P, qRT-PCR was performed to quantify the expression levels of the pilin. For this purpose, we extracted RNA from the cells of the motility halos every 24 hours until reaching 96 hours of incubation. The RNA was used to obtain the cDNA that will be used in the relative quantification of the pilin gene. 13

Page 14: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P5. Platelet-mediated activation of neutrophils contributes Neutrophil Extracellular Trap release and to increased lung pathogenesis during influenza infection. Agostina Carestia, Seok-Joo Kim, Heidi Grosjean, Victor Naumenko, Silvio Antoniak, Nigel Mackman, Mohamed Sarjoon Abdul-Cader, Mohamed Faizal Abdul-Careem, Morley Hollenberg, and Craig Jenne. Influenza A virus (IAV) is a common cause of respiratory tract infection in humans with approximately 10% of the population infected by the virus each year. The hallmark of severe IAV infection is excessive inflammation and tissue pathology in the lungs. Although platelets and neutrophils are associated with IAV infection and its pathogenesis, the specific mechanisms involved have not yet been clarified. To address this issue, mice were infected intranasal with A/PR/8/34 (PR8, H1N1, 250 PFU) and confocal intravital microscopy was conducted to analyze platelets and neutrophils behavior after 5 days of infection. We observed that IAV infection induced a significant increase in wet-to-dry ratio of mouse lungs (pneumonia), the number of cells in the bronchoalveolar space and MPO levels in the lung, indicating that this model of severe IAV infection induces significant inflammation in the lung. Using intravital microscopy, we also found that IAV infection induced an augment in neutrophils’ recruitment, and a profound platelet aggregation and neutrophil extracellular trap (NET) production in vivo. In concert with cell recruitment, an increase in intravascular thrombin activity was observed within the lung microvasculature of IAV-infected mice. The blockade of protease-activated receptor 4 (PAR4), the primary thrombin receptor on platelets, reduced platelet aggregation and NET production as well as protected mice from virus-induced lung tissue damage and edema. Similar results were observed using PAR4-deficient animals. Together, these data imply thrombin-stimulated platelets play a critical role in the activation/recruitment of blood neutrophils, contributing to IAV pathogenesis in the lung.

P6. Enzymatic and functional assays of YbgC from E. coli to determine role of the protein. Zinaida Eltsova*, Nobuhiko Watanabe, Alexei Savchenko. Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1 The Tol-Pal gene cluster of Gram-negative bacteria encodes proteins involved in interaction between outer and inner membranes and critical for the outer-membrane stability. The components of this system were focus of intensive research (J. N. Sturgis, 2001), however, the molecular function of cytoplasmic protein YbgC also encoded in this cluster remains elusive. E. coli DybgC strain demonstrates higher permeability for hydrophobic dyes and antibiotics (Paradis-Bleau et al., 2014; Liu A. et al., 2010) making it an attractive target for antibiotic adjuvant therapies. We have previously determined the crystal structure on YbgC from E. coli, revealing the general fold associated with thioester hydrolase activity. To gain further understanding into function of YbgC we have determined the crystal structure of this protein in complex with Co-A as well as with hexanoyl and decanoyl-CoA, capturing the interactions of this protein with its co-substrates. Our functional analysis demonstrated that the E. coli YbgC thioesterase demonstates higher activity towards decanoyl-CoA (C10) rather than butyryl-CoA (C4) in contrast to Haemophilus influenza homologue (Zhuang Z. et al., 2002) with which it shares 53% of sequence identity. This analysis allowed us to probe the role of individual active site residues in substrate specificity of YbgC and shed new light on the function of this protein in gram negative bacteria’s cell envelop phospholipid metabolism.

14

Page 15: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P7. Dispersal of the trans-Golgi network by the effector RARP2 of Rickettsia rickettsii inhibits host cell protein transport to the cell surface. Karin Aistleitner, Tina Clark and Ted Hackstadt. Rocky Mountain Laboratories, NIH, Hamilton, MT The trans-Golgi network is the major secretory pathway sorting station that directs newly synthesized proteins to different subcellular destinations. We show here that virulent strains of the obligate intracellular bacterium R. rickettsii cause dispersal of the trans Golgi, but not the cis Golgi network in host cells. The trans-Golgi is dispersed shortly after entry of R. rickettsii into host cells and stays dispersed throughout the whole infection process. While dispersal occurs in both Vero and human dermal microvascular endothelial cells infected with virulent R. rickettsii strains, the Golgi is not affected in tick cells which represent the reservoir of R. rickettsii in nature. The dispersal is caused by the secretion of the rickettsial Ankyrin repeat protein 2 (RARP2), a recently identified type IV secreted effector protein. RARP2 is one of the few genes showing differences in genomic comparisons between R. rickettsii strains differing in virulence, being truncated in the avirulent Iowa strain compared to virulent strains. The dispersal also interferes with both functions of the Golgi: glycosylation of host cell proteins is incomplete as shown by weight shifts in Western blot analysis and dispersal of lectin staining in infected host cells. In addition, protein transport to the cell surface is severely impaired in cells infected with virulent strains of R. rickettsii. This includes trafficking of MHC I and might thereby help virulent strains of R. rickettsii to evade the host immune system.

P8. IRF3 signaling is critical to prevent Middle East respiratory syndrome (MERS) coronavirus propagation in big brown bat cells. Arinjay Banerjee1, Darryl Falzarano2, Noreen Rapin1, Jocelyne Lew2, Ankita Tulangekar1, Scott Napper2,3 and Vikram Misra1. 1 Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Canada. 2 Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), Saskatoon, Canada. 3Department of Biochemistry, University of Saskatchewan, Saskatoon, Canada. Insectivorous bats are speculated to be ancestral hosts of Middle-East respiratory syndrome (MERS) coronavirus (CoV). MERS-CoV causes disease in humans with a thirty-five percent mortality largely due to its ability to counteract human antiviral responses. Since bats experimentally infected with MERS-CoV do not develop visible signs of disease, we tested the hypothesis that MERS-CoV cannot subvert antiviral interferon responses in bat cells. We infected human and bat (Eptesicus fuscus) cells with MERS-CoV and observed that the virus propagated significantly more in human cells. MERS-CoV also effectively suppressed antiviral interferon beta (IFN) response in human cells, unlike in bat cells. By studying the response of IRF3 to poly(I:C), a synthetic analogue of viral double-stranded RNA, we observed that bat IRF3 responded to poly(I:C) by phosphorylation and nuclear translocation; hallmarks of IRF3 activation. By knocking down IRF3 in bat cells by small interfering RNA (siRNA), we demonstrated that IRF3 is critical in poly(I:C) and MERS-CoV mediated IFN gene expression. We performed a kinome analysis on poly(I:C) treated bat and human cells and observed that similar pathways were up-regulated. To identify the role of IRF3 in controlling virus replication, we infected IRF3 knocked-down bat and human cells with MERS-CoV and observed that MERS-CoV propagated to significantly higher levels in these bat cells. Our study identified that IRF3 mediated antiviral signaling process in bat cells is resistant to subversion by MERS-CoV. We will identify these strategies to restore antiviral signaling in coronavirus infected human cells.

15

Page 16: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P9. Livestock-Associated Staphylococcus aureus (LA-SA) ST398 Virulence Is Associated with Factors Carried on ϕSa3 and/or Staphylococcal Protein A (spa). Ayesha Kashif1, Michael Pharm1, Jo-Ann M. McClure1, Sidong Chen 2, John M. Conly1, Kunyan Zhang1. Centre for Antimicrobial Resistance, Alberta Health Services/Calgary Laboratory Services /University of Calgary, Calgary, Alberta, Canada 1; Guangdong Pharmaceutical University, Guangzhou, Guangdong, P.R. China2 Objective: The virulence of emerging ST398 strains may differ. We collected strains from China and Canada to test in a Caenorhabditis elegans infection model and compared their whole genome sequences (WGS) to explore potential insights into their virulence. Methods: 10 (Guangdong), 4 (Calgary) and 1 (Canada) strains were collected, molecularly characterized (PFGE, MLST, spa, agr, SCCmec typing), and sequenced (PacBio, Illumina). Virulence was assessed using the C. elegans survival assay. SNP WGS phylogenetic analysis was performed using CSI Phylogeny v1.4 and FigTree v1.4.3, BLAST ring images by BRIG v0.95, Mauve-alignment by DNAStar-SeqMan Ngen L15, and Prophage identification/annotation/comparison by PHAST software and Easyfig. Results: All isolates belonged to ST398-MSSA except 1 ST398-MRSA (Canada) with variant spa types (t034, t571, t1451, t1250). PFGE and SNP-WGS analyses showed that the 15 isolates clustered into 3 groups, GD1 (including ST398-MRSA), CAL, and GD2 with high-, moderate-, and low-nematocidal activities (killing 100%, 73-95%, and 40-73%, respectively). Genomic comparison revealed striking genetic similarities in their genomes among these 3 groups. However, GD1 carried ϕSa3 (containing virulence-associated genes: chp, sak, sea, scn), CAL carried ϕSa3 (missing chp and sea) with a truncated spa, and GD2 lacked ϕSa3. Interestingly, ST398-MRSA genetically clustered within the GD1 group but only had a 40% killing rate and lacked ϕSa3. Conclusion: This study demonstrated that different ST398 sub-lineages possess variant virulence capacities depending on their possession of the mobile genetic elements that carry virulence factors, such as ϕSa3. It also suggests differential strategies for treatments of infections due to LA-SA.

P10. Identification of unique genomic elements in Prevotella bivia strains capable of ascending the urogenital tract. Sahar Bagheri1, MSc and Laura Sycuro1,2,3, PhD. Depts. of Microbiology, Immunology & Infectious Diseases1, Biochemistry & Molecular Biology2, and Obstetrics & Gynecology3, Cumming School of Medicine, University of Calgary, Alberta. Background: In women experiencing preterm birth with intact membranes, Prevotella/Bacteroides species are the third most frequently detected taxa in amniotic fluid, but the identities of the invading species remain poorly described. One candidate is Prevotella bivia, which has been detected in amniotic fluid and shown to incur a two-fold higher risk of PTB when present at ≥104 cfu/mL in vaginal fluid. The type strain of P. bivia, ATCC 29303, was isolated from the endometrium along with closely related strains from other women experiencing ascending infections; more distantly related P. bivia strains came from other body sites/infections. Objective: We hypothesized that P. bivia strains closely related to the type strain carry specific genes that enable them to colonize the upper genital tract (UGT). Results: We compared the genome of the P. bivia type strain to those of 4 vaginal P. bivia isolates available in IMG/ER. JCVIHMP010 was highly similar to the type strain (98% of genes aligning with 100% identity), suggesting it is the same strain. pBLAST identified 33 genes unique to the ‘ascending’ type strain, 25 of which lacked functional information. One unique locus encoded a sulfur-based DNA modification system, possibly representing a bacterial defense system contributing to the strain’s genomic isolation. We also identified two potential virulence loci encoding a putative serine protease and serine O-acetyltransferase, proteins that have been shown in other ascending pathogens to assist in evading the immune system. Conclusion: One P. bivia strain encodes genes that could provide evolutionarily stable protection from UGT immune surveillance.

16

Page 17: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P12. Liposomal therapy attenuates bacterial virulence by targeting secreted toxins of community-associated methicillin-resistant Staphylococcus aureus and Pseudomonas aeruginosa. Heidi Wolfmeier 1*, Sarah C. Mansour 1, Daniel Pletzer 1, Leo T. Liu 1, Annette Draeger 2, Eduard B. Babiychuk 2, Robert E.W. Hancock 1. 1 Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2 Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3000 Bern 9, Switzerland In the wake of growing antibiotic resistance, novel strategies are urgently needed for the treatment of bacterial infections. Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), typified by the pulse-field type USA300, is an emerging endemic pathogen that is spreading rapidly among healthy people. CA-MRSA causes skin and soft tissue infections but also other life-threatening diseases such as necrotizing pneumonia and sepsis, and is remarkably resistant to many antibiotics. Here we show that engineered liposomes composed of naturally occurring sphingomyelin were able to sequester cytolytic toxins secreted by USA300 and prevent necrosis of human erythrocytes, peripheral blood mononuclear cells and bronchial epithelial cells. Mass spectrometric analysis revealed the presence of phenol soluble modulins, α-hemolysin, leukocidins and other liposome-bound toxins. Sphingomyelin liposomes prevented hemolysis induced by pure phenol soluble modulin-α3, one of the main cytolytic components in the USA300 secretome. In a murine cutaneous abscess model of USA300 infection, a single dose of sphingomyelin liposomes was sufficient to significantly decrease tissue dermonecrosis. Furthermore we show that engineered liposomes neutralized cytolytic virulence factors secreted by three different strains of the potent and difficult-to-treat Gram-negative pathogen Pseudomonas aeruginosa (PAO1, PA14, and LESB58). Sphingomyelin liposomes with a high cholesterol content (66 mol/%) efficiently reduced bacterial secretome-induced cytotoxicity of human cells. We identified the virulence-promoting exotoxin hemolytic phospholipase C to be sequestered by the liposomes. Our results provide further insight into the promising potential of tailored liposomal therapy in the battle against infectious diseases.

P11. Chlamydia trachomatis inclusion membrane microdomain proteins interact with host proteins to regulate extrusion formation. Phu Hai Nguyen and Ted Hackstadt. Host-Parasite Interactions Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America. Chlamydia trachomatis is an obligate intracellular bacterium that replicates within a vacuole termed an inclusion. At the end of their intracellular developmental cycle, chlamydiae are released either by lysis of the host cell or by extrusion of the intact inclusion. The inclusion membrane is modified by the insertion of multiple proteins known as Incs. Certain of the Incs are enriched in specialized microdomains of the chlamydial inclusion membrane that are also enriched in active Src-family kinases (SFK). An interaction was identified between the inclusion membrane protein, MrcA, with the Ca2+ channel inositol-1,4,5- trisphosphate receptor (ITPR3). Disruption of MrcA by directed mutagenesis resulted in loss of ITPR3 recruitment and simultaneous reduction of chlamydial release by extrusion. Complementation of MrcA restored ITPR3 recruitment and extrusion. Chlamydial extrusion was also inhibited by ITPR3 siRNA and by the calcium chelator BAPTA-AM treatment. Each of these treatments resulted in a reduction in phosphorylation of the myosin regulatory light chain (MLC2) and a loss of myosin motor activity at the end of the developmental cycle. Other host proteins were also recruited and localized in inclusion membrane microdomains: pY421-cortactin (pY421-CTTN) and alpha actinin 4 (αACTN4) interacted with Inc C and Inc B, respectively. Inhibition of extrusion was observed following siRNA depletion of host CTTN or ACTN4. Targeted knockout of Inc C reduced pY421-CTTN recruitment that was restored with complementation of Inc C. CTTN and ACTN4 are known to be involved in actin polymerization. Taken together these studies suggest that Ca2+ signaling pathways play an important role in regulation of release mechanisms by C. trachomatis and that extrusion requires an active actin-myosin complex.

17

Page 18: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

18

P14. Characterizing the virulence effectors of the emerging pathogen Aeromonas dhakensis. Le Tang, Xiaoye Liang, Tao G. Dong. Ecosystem and Public Health, University of Calgary, AB, Canada. Aeromonas dhakensis is a ubiquitous waterborne pathogen that can cause gastrointestinal or systemic infections in humans. During infection, A. dhakensis translocate virulence effectors to target cells through protein secretion systems to cope with the hostile host environment. The type 6 secretion system (T6SS) is one of the six protein secretion systems found in Gram-negative bacteria. Although T6SS has been reported to be important for virulence in A. dhakensis, its delivery mechanism is unclear. Especially, T6SS effectors remain to be identified. Our preliminary data show that A. dhakensis employs the T6SS to kill Escherichia coli, suggesting that T6SS plays a role in competing with other bacterial species. Here we report two new antimicrobial effectors, TseI and TseP, and their cognate immunity proteins that confer self-protection. We demonstrate a substrate selection mechanism that dictates effector delivery depending on cognate VgrG and chaperone proteins.

P13. Suppressing Salmonella enterica SspH1 activity to gain insights into Novel E3 ubiquitin Ligase effectors. Jan W. Stoepel1,*, Karolynn J. Hsu1,*, Adithya S. Shankara2, Ivan K. Domingo2, Wei Zhang3, Kevin Mehr4, Peter C. Stirling5, Michel Roberge4, Sachdev Sidhu3, Philip Hieter1, B. Brett Finlay1 and Amit P. Bhavsar2. 1 Michael Smith Laboratories, The University of British Columbia, 2 Medical Microbiology and Immunology Department, University of Alberta, 3 Donnelly CCBR, University of Toronto, 4 Life Sciences Centre, The University of British Columbia, 5 Terry Fox Laboratory, BC Cancer Agency *These authors contributed equally Salmonella enterica encodes two Type 3 secretion systems that deliver effector proteins directly into the host. S. enterica expresses three effectors that are members of the novel E3 ubiquitin ligase (NEL) family, including SspH1 and SspH2 that have been shown to modulate host innate immunity. We utilized a cross-kingdom approach for studying NEL effector function and discovered that expression of active SspH1 is profoundly toxic to yeast. The objective of our study is to suppress SspH1 toxicity in yeast to identify the mechanism underlying this phenotype. We have used genetic and non-genetic strategies to suppress SspH1 activity. Genetic strategies included overexpression of PKN1, the human substrate of SspH1, as well as employing a yeast multicopy suppression library (MoBY-ORF v2.0). We established a high-throughput yeast-based selection to identify small molecule inhibitors of SspH1 and interrogated the Prestwick library of FDA-approved drugs. Finally, we have used a binding screen of recombinant SspH1 against a library of ubiquitin variants (Ubvs) to identify protein inhibitors of SspH1. Our results showed that PKN1 was able to partially restore yeast growth compared to a catalytically inactive SspH1 variant. Multicopy suppression analysis identified two genes, TAD3 and RAM2 that fully restored yeast growth that was determined to be the result of abrogated SspH1 expression. The small molecule selection was robust with a z-score of 0.8. Three putative hits were identified but this was due to their intrinsic colourimetric character. Two Ubvs were identified that showed highly similar sequence variants that will now be screened in yeast.

Page 19: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P16. Engineering and evaluating novel protein vaccines against porcine respiratory bacterial pathogens. Rafael Frandoloso1, Somshukla Chaudhuri2, Gabriela Paraboni Frandoloso1, Rong-hua Yu2, Anthony Bernard Schryvers2. 1 Laboratory of Microbiology and Advanced Immunology, Faculty of Agronomy and Veterinary Medicine, University of Passo Fundo, Brazil, 2 Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Canada Actinobacillus pleuropneumoniae, Actinobacillus suis, and Haemophilus parasuis are upper respiratory residing pathogens, which cause fatal respiratory illnesses, meningitis, and septicaemia in pigs. Prevention through immunizations is key, however, prevention based on vaccination is a major concern due to the limited cross protection conferred by the inactivated whole cell vaccines currently used. These pathogens rely on surface transferrin-binding proteins A and B (TbpA and TbpB) for acquiring iron from the host iron-binding protein transferrin, a process essential for survival and causing disease. We previously demonstrated that a recombinant form of TbpB defective in binding transferrin, Y167A-TbpB, provides superior protection against infection by H. parasuis compared to the wildtype and is cross-protective. Recently, we evaluated the ability of different vaccine formulations and administration routes based on the mutant Y167A-TbpB against a H. parasuis challenge in a conventional pig model. The vaccines were all immunogenic in pigs, however, differences in terms of antigenicity, immune response, and clinical symptoms were observed. We observed that some vaccines not only prevented infection but also appeared to eliminate natural colonization by H. parasuis during the experiment. In conjunction with previous studies, our results demonstrate that the Y167A-TbpB antigen is a promising antigen for developing a broad-spectrum vaccine against H. parasuis infection and colonization and brings us closer to identifying a limited set of TbpB based antigens that are capable of inducing a cross-protective response against all three pathogens. 19

P15. HAND HYGIENE QUALITY IMPROVEMENT USING AN INNOVATIVE DIGITAL MONITORING PROGRAM. S. Bansaghi1, A. Saiyed2, N. Wallace3, P. Szeremy4,5, T. Haidegger5,6. 1 Department for Epidemiology, Faculty of Health Science, Semmelweis University, Hungary, 2 Rehabilitation Institute of Chicago, IL, 3 Center of Excellence for Infection Prevention and Control (COEIPAC), Buffalo, NY, 4 Faculty of Medicine, University of Szeged, Szeged, Hungary, 5 Austrian Center for Medical Innovation and Technology, Austria, 6 University Research and Innovation Center, Obuda University, Budapest, Hungary Hand hygiene (HH) is believed to be the most efficient tool in fighting hospital-acquired infections. Despite years of effort, caregivers’ HH frequency and compliance remain low. An innovative HH scanner was introduced to the critical care units to observe the results of HH quality scores and quality improvement through automated training. Caregivers were involved from 1 MICU and 1 NICU at an Acute Care Hospital in the Chicago area, in 2017. Caregivers had to perform hand rubbing with a UV labeled alcohol-based hand rub, engaged with an automated HH quality measurement device, and received objective visual feedback with missed spots at every instance. The integrated reporting system provided real-time statistics about HH at a personal and institutional level. The system computed the Semmelweis Hand Hygiene Quality (SHHQ) score (0–100), which indicated the quality of HH. 92 caregivers took part in the quality improvement program. The average SHHQ achieved by the MICU was 85.38±17.71% (mean±STD) and 89.48±13.81% by the NICU. Most frequently missed areas were the thumbs and the fingertips. HH quality improved (people with 95+ SHHQ) at both units: only 28.4% of people passed on the first 4 days of the trial, while 81.1% passed in the last 4 days. Digital HH training and evaluation allows for systematic, personalized data collection, and it makes it possible to reach a larger fraction of the staff. This technique can reveal typical week points in the caregivers’ HH practices and can be used efficiently to improve HH quality.

Page 20: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P17. Title: The Association between Food Insecurity and Acute Gastroenteritis in the United States (NHANES 2013-2014). Arneja, Jasleen S. and Pokarowski, Martha. Both authors contributed equally and are Master of Public Health students at the Dalla Lana School of Public Health at the University of Toronto. Household (HH) food insecurity is associated with serious adverse health outcomes due to inadequate nutrition, causing poor physical and mental health. Acute gastroenteritis (AG) is caused by a variety of infectious agents, many of which are acquired by the consumption of contaminated food. We aim to examine the relationship between HH food security and acute AG and investigate the interaction between sex and HH food security. Cross-sectional data from the 2013-2014 cycle of the National Health and Nutrition Examination Survey (NHANES) provided data on HH food security (full, marginal, low and very low), acute AG for 4,265 respondents. A log-binomial regression model was generated, adjusting for age, sex, survey period, birthplace, income, alcohol consumption. The adjusted prevalence of AG was 1.62 (95% CI 1.17, 2.25) times greater for those in households with very low food security compared to those in households with full food security. Women in households with very low food security had a significantly higher prevalence of AG (PR=2.01, 95% CI: 1.36, 3.00) compared to women in households with full food security, whereas the same relationship was not observed in men (PR=1.04 95% CI: 0.57, 1.90). HH food security status was associated with acute gastroenteritis among US adults controlling for age, sex, alcohol consumption, income, and survey period. This finding can help identify effective public health policy to address household food insecurity and establish interventions for adults living in food-insecure households.

P18. Small molecule screening for cholera disease treatment. Brianne J. Burkinshaw1, Linh Lam1 and Tao G. Dong1*. 1Ecosystem and Public Health, Faculty of Veterinary Medicine; Biochemistry and Molecular Biology, Cumming School of Medicine; Snyder Institute for Chronic Diseases, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N4Z6, Canada. Vibrio cholerae is a Gram-negative bacterium that causes cholera, an acute diarrhoeal disease that affects up to 4 million per year. The on-going Yemen cholera outbreak has affected more than 1 million with over 2000 death cases. In this study, we screened a set of small molecules that have low toxicity against mammalian cells for toxicity against V. cholerae. We first spotted each compound on a lawn of V. cholerae and compared zones of growth inhibition. Our initial screens identified two hit compounds, C1 and C2, with antimicrobial activities against V. cholerae. We then determined that C1 and C2 have minimum inhibitory concentrations of 2.5 uM and 50 nM, respectively. Further investigation revealed C1 is bactericidal and C2 is bacteriostatic. To determine the mode of action of each compound, we looked for changes in cell morphology after co-incubation with the compound and we are analyzing differential gene expression after exposure to each compound using RNA-Seq analysis. The hit compounds were also effective against other V. cholerae clinical strains. These molecules have the potential to be the chemical chassis for small-molecule inhibitor drug development.

20

Page 21: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P19. A novel aadA Aminoglycoside resistance gene in bovine and porcine pathogens. Sara Andres-Lasheras1, Andrew Cameron1,2, Cassidy L. Klima1,3, Reuben Ha1, Robert J. Gruninger1, Rahat Zaheer1 , Tim A. McAllister1. 1 Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada, 2 Faculty of Veterinary Medicine, University of Calgary, Alberta, Canada, 3 Feedlot Health Management Services, Okotoks, Alberta, Canada. The broad-spectrum drug class aminoglycoside belongs to category II (high importance) based on its importance in human medicine. Aminoglycosides are the fifth-most-used drug class in veterinary medicine. The synthesis of aminoglycoside-modifying enzymes (AMEs) is one of the aminoglycoside-resistance strategies found in bacteria. AAD(3’’)-class AMEs are O-adenylyltransferases which are known to confer resistance to spectinomycin and streptomycin. In this study, a new variant of the AAD(3’’)-class of AMEs was identified in Pasteurella multocida and Histophilus somni, both isolated from mortality-associated clinical cases of Bovine Respiratory Disease (BRD) in Alberta. Genomic DNA cosmid-based libraries were constructed to functionally screen for the spectinomycin ARGs and to determine their genomic context. The new gene, named aadA31 (spectinomycin/streptomycin adenylytransferase), was found in a variant of the integrative and conjugative element ICEMh1, a mobile genetic element with self-transmissibility potential among members of the family Pasteurellaceae. This gene was also detected by nr BLAST in Mannheimia haemolytica from a case of porcine pneumonia and in Moraxella bovoculi from a case of bovine keratoconjunctivitis. Widespread multi-drug resistance in BRD pathogens may have important consequences for the health of the beef and dairy industries. These findings highlight the ARG diversity and the prospects of horizontal movement of ARGs via ICE.

P20. Administration of metaphylactic antimicrobials to feedlot cattle alters the fecal and nasopharyngeal microbiota. Trevor W. Alexander1, Devin B. Holman2, Wenzhu Yang3. 1.Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, Alberta, Canada, 2. Lacombe Research and Development Centre, Agriculture and Agri-Food Canada, Lacombe, Alberta, Canada, 3. Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, Alberta, Canada. Beef cattle in Canada frequently receive an antibiotic at feedlot placement to manage bronchopneumonia. The impact of these antibiotics on the bovine microbiome is largely unknown. Therefore, the objective of this study was to evaluate the longitudinal effects of two commonly administered veterinary antibiotics on the fecal and nasopharyngeal (NP) microbiota of beef cattle that were transported to a feedlot. Nasopharyngeal swabs and feces were collected from calves (N = 12 per treatment) at a farm (d minus 5). Subsequently, calves were transported to a feedlot and injected with oxytetracycline or tulathromycin, while a control group received no antimicrobials. Calves were sampled immediately before injection (d 0), and again 2, 5, 12, 19, and 34 days after treatment. DNA was extracted from samples and the 16S rRNA gene was sequenced to evaluate bacteria. Oxytetracycline and tulathromycin perturbation of the fecal and NP microbiota was greatest at d 2 and 5. Although the NP microbiota of the tulathromycin-treated cattle recovered by d 12, the NP microbiota of oxytetracycline-treated cattle remained altered through d 34. Members of the bacterial Microbacteriaceae family were most affected by antibiotic administration in the NP microbiota. Both antibiotics protected against Pasteurella spp. in the nasopharynx at d 2 and 5, whereas oxytetracycline enriched Mycoplasma at d 34. Overall, the NP microbiota appeared to be more sensitive to antibiotic treatment than the fecal microbiota. This study showed that the bovine microbiota is altered by antimicrobial treatment and that recovery can vary depending on the class of antimicrobial administered.

21

Page 22: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P22. Mechanisms underlying Pseudomonas aeruginosa susceptibility to antimicrobials in clinically relevant conditions. Corrie Belanger, Amy Lee, Daniel Pletzer and Robert E.W. Hancock. University of British Columbia, 235-2259 Lower Mall Research Station, Vancouver, BC Conventional antibiotic assays do not reflect the biochemical composition found in human infections, and thus are not very predictive of clinical antibacterial activity. For this reason, our research assessed the ability and mechanism of antibiotics and antimicrobial peptides to treat the opportunistic pathogen P. aeruginosa in physiologically relevant conditions compared to standard laboratory conditions. Testing susceptibility in media relevant to the lungs of cystic fibrosis patients or the blood of sepsis patients, five antibiotics showed differential inhibitory and antibiotic:peptide synergistic effects against P. aeruginosa when compared to standard laboratory media. To unravel potential causes for this altered susceptibility, RNA-Seq was used to explore changes in gene expression of P. aeruginosa grown or treated with azithromycin (AZM) in laboratory and clinically relevant conditions. More than 60 known resistome genes were dysregulated in media representing the host with and without AZM treatment, including important surface modification genes. Dysregulation of these genes may disrupt Lipopolysaccharide modification and prevent Pseudomonas from blocking antibiotic uptake in host conditions. Additionally, genes encoding universal stress proteins and genes encoding diguanlyate cyclases were down and upregulated, respectively in P. aeruginosa grown in conditions relevant to the host compared to synthetic media. Mutants for stringent response genes relA and spoT showed increased susceptibility to AZM and significant synergy was seen in vivo between AZM and host defense peptides known to target stringent response. These results indicate that AZM may downregulate stress responses relying on synthesis of the signaling molecule c-di-GMP, and may be synergizing with peptides due to the ability of the peptides to simultaneously target the alternative overlapping RelA SpoT stringent response triggered by P. aeruginosa in physiologically relevant conditions. This research is important for understanding the mechanisms that underlie bacterial susceptibility to antimicrobials in clinically relevant conditions and could help to change standard approaches to drug testing and lead to more predictive drug discovery routes.

P21. PAAR-dependent effector delivery of the type VI secretion system. Brianne J. Burkinshaw1, Xiaoye Liang1, Megan Wong1, Alexander N. H. Le1, Linh Lam1 and Tao G. Dong1*. 1Ecosystem and Public Health, Faculty of Veterinary Medicine; Biochemistry and Molecular Biology, Cumming School of Medicine; Snyder Institute for Chronic Diseases, University of Calgary, 3330 Hospital Dr. NW, Calgary, AB, T2N4Z6, Canada. The type VI secretion system (T6SS) is a needle-like protein delivery machine that is widely distributed among Gram-negative bacteria and plays a key role in interbacterial and host-microbe interaction. The T6SS injects its inner needle Hcp tube, sharpening tip complex consisting of VgrG and PAAR, and toxic effectors into neighboring cells. Dedicated effectors are delivered through direct binding to the inner tube or the VgrG (Valine-Glycine-Repeat protein G) proteins of the tip complex. Here, we report a new mechanism of effector delivery that requires direct interaction with a component of the tip complex, PAAR (proline-alanine-alanine-arginine) repeat protein, and effector-specific chaperone and co-chaperone proteins. The Pseudomonas aeruginosa PAO1 TOX-REase-5 domain-containing effector TseT directly interacts with PAAR4, a small dedicated PAAR protein, and a chaperone TecT. TecT forms a complex with its co-chaperone, co-TecT, which is disrupted by the C-terminal tail of PAAR4. Delivery of TseT via the H2-T6SS requires VgrG4b and VgrG6 and is greatly stimulated in the vgrG2a and vgrG2b mutants. Finally, we predicted a large family of PAAR-dependent TseT effectors in Gram-negative bacteria and validated the antibacterial role of a TseT homolog in Burkholderia. Our results demonstrate yet another way of T6SS effector secretion via PAAR, chaperone and co-chaperone proteins and highlight a multilayered competition process that dictates effector delivery.

22

Page 23: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P23. Lactobacilli-mediated Rel/NF-κB activation and ROS-generation mitigates IMI-induced susceptibility to infection and microbial dysbiosis. Brendan A. Daisley1,2*, John E. Chmiel1,2, Mark Trinder1,2, Tim W. McDowell3, Bethany Rogers1,2, Hylke Welle1,2,4, Josh S. Dube2, Sohrab N. Ali2,5, Hon S. Leong2,6, Mark W. Sumarah3, Jeremy P. Burton1,2,6, and Gregor Reid1,2,6. 1Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON. 2Department of Microbiology and Immunology, The University of Western Ontario, London, ON. 3London Research and Development Center, Agriculture and Agri-Food Canada, London, ON. 4Vrije Universiteit Amsterdam, Faculty Earth and Life Sciences, Institute of Molecular Cell Biology, Amsterdam, Netherlands. 5Department of Surgery, Division of Urology, University of Ottawa, Ottawa, ON. 6Department of Surgery, The University of Western Ontario, London, ON. Background: Neonicotinoids are neuro-active acetylcholine (ACh) mimics that are used as insecticides due to their selective nicotinic ACh receptor (nAChR) inhibiting properties. Imidacloprid (IMI) is the most widely used neonicotinoids and has been detected 52-66% of all fruits and vegetables worldwide, demonstrating its ubiquity as a dietary staple in the global food supply. Recent evidence suggests chronic nAChR activation in non-neuronal cells can reduce Rel/NF-κB activation and reactive oxygen species (ROS)-generation, both of which are highly conserved divisions of innate immunity that respond to bacterial pathogens and regulate the intestinal microbiota. Hypothesis: Innate immune stimulation by Lactobacillus plantarum Lp39 could mitigate IMI-induced susceptibility towards bacterial infection and microbial dysbiosis in Drosophila melanogaster. Results: IMD (upstream of Rel/NF-κB) deficient D. melanogaster were significantly more susceptible to IMI toxicity, as demonstrated by reduced survival in Rel-/-, Upd1-/-, Upd2/3-/-, and Hop2-/- mutants. However, overexpressing Rel/NF-κB (via constitutive expression of the active N-terminal subunit, Rel-68) was unable to rescue the phenotype. Interestingly, orally supplementing Lp39 led to an increase in Diptericin (Rel/NF-κB-downstream antimicrobial peptide gene) expression and effectively mitigated sublethal IMI-induced susceptibility of D. melanogaster to Serratia marcescens septic infection. Furthermore, prophylactically supplementing Lp39 for 48-hours prior to sublethal IMI exposure was sufficient in ameliorating IMI-induced increases in intestinal bacterial loads of Acetobacter and Lactobacillus species, an effect that coincided with Lp39-induced expression of ROS-generating transmembrane proteins, NOX and DUOX. Conclusion: Immunomodulatory lactobacilli can mitigate the biologically-relevant effects of sublethal IMI exposure on host innate immunity and subsequent microbial regulation.

P24. Functional Characterization of the Group A Streptococci Bacteriocins SpbJK and SpbMN. Lana Estafanos1, Brent D. Armstrong1, John K. McCormick1,2. 1Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON. 2Lawson Health Research Institute, London, ON. Group A Streptococcus (GAS) is a globally prominent bacterial pathogen with a propensity to colonize the skin and upper respiratory tract of humans. Infections are mediated through mucosal colonization, yet the mechanisms by which GAS competes for ecological stability are poorly understood. Our lab has identified two novel Class IIb bacteriocin systems – Streptococcus pyogenes bacteriocin (Spb) JK and MN – which may contribute to nasopharyngeal colonization. Using bioactivity assays, we identified galactose- and CO2-rich environments as distinct regulatory elements which induce antimicrobial activity. Under these conditions, bacteriocin-producing strains have the ability to inhibit growth of closely related bacterial species in vitro. Notably, we show that nasal co-infection of mice with the parental S. pyogenes M18 serotype and a bacteriocin-deficient strain results in a wild-type dominant population, suggesting that bacteriocin-deficient strains have reduced survival fitness in a polymicrobial environment. We intend to further evaluate the changes in microbial diversity by 16S ribosomal sequencing. Taken together, these data highlight the need to further explore the roles of these novel bacteriocin systems. These studies will determine how GAS competes with other microorganisms in its biological niche, with the goal of understanding of how GAS facilitates colonization and persistence in the human host.

23

Page 24: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P 25. Engineering a broadly cross-protective vaccine against Haemophilus influenzae using transferrin binding proteins. Nikolas Ewasechko, Anthony Schryvers. Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary. Haemophilus influenzae is a Gram-negative bacterium that causes pneumonia, otitis media, and invasive infections such as meningitis and bacteremia. A vaccine that effectively protects against infection by H. influenzae serotype b currently exists, but infections caused by other serotypes and non-typeable strains are still prevalent. The aim of our project is therefore to engineer a broadly cross-protective vaccine that will protect against colonization and infection by all H. influenzae strains. Our strategy for developing such a vaccine is to target iron acquisition receptors known as transferrin binding proteins. As previous studies have demonstrated that binding-defective transferrin binding protein B (TbpB) variants elicit a superior protective immune response compared to the native protein, our strategy also involves the design and production of TbpB mutants that are unable to bind to human transferrin (Tf). To prevent vaccine escape, we have extensively analyzed the sequence diversity among TbpB variants. Based on this analysis, we identified several different clades and selected TbpB variants representing each of these clades as candidates for site-directed mutagenesis. Each of these representative variants was then expressed and tested by solid-phase binding assays to determine the protein’s Tf-binding ability. We are now in the process of determining the structure of both native and mutant H. influenzae TbpB variants using protein X-ray crystallography. This will help us understand whether substantial changes in binding affinity can be induced without changing the overall structural conformation of the protein, thereby disrupting Tf binding while preserving the conformational epitopes displayed on the native TbpB.

P26. A novel, non-immunity-mediated resistance to type-six secretion system attack. Steven J. Hersch, Mavis Lee, and Tao G. Dong. The type six secretion system (T6SS) is employed by many gram negative bacteria to compete with neighbouring microbes both in the environment and in the context of infection. Resembling a molecular spear, the T6SS machinery contracts to thrust its tip into neighbouring cells, thereby delivering attached effector proteins. These toxins can target a variety of different cellular processes in both eukaryotes and prokaryotes and T6SS+ bacteria protect themselves against self-killing by expressing immunity genes to each of their secreted effectors. Vibrio cholerae, the causative agent of cholera, employs its T6SS to deliver five known effector proteins. Here we characterise a cryptic effector of V. cholerae, describing its mechanism of attachment onto the T6SS for delivery and furthermore demonstrating that it selectively kills a relatively narrower spectrum of species in comparison with the other V. cholerae effectors. To elucidate this difference and the underlying mechanism of selectivity, we employed genome comparisons as well as the E. coli Keio collection of specific gene knockouts. We identified a concerted effort of multiple stress response pathways and a variety of downstream genes that are critical to E. coli’s survival. Indeed, deletion of individual components of this response results in E. coli becoming susceptible to killing, suggesting that every piece of armour must be in place for a successful defense. In summary, this work characterizes a cryptic T6SS effector and reveals its killing activity in a T6SS-delivery setting. Moreover, we demonstrate a novel, non-immunity-mediated resistance to T6SS attack.

24

Page 25: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P 27. Vasoactive Intestinal Peptide (VIP) Promotes Intestinal Host Defense by Modulating Group 3 Innate Lymphoid Cells. Hong Bing Yu1,#, Hyungjun Yang1, Caixia Ma1, Gregor S. Reid2, James A. Waschek3, Lisa C. Osborne4, Bruce A. Vallance1, Kevan Jacobson1,#. 1Division of Gastroenterology, Hepatology and Nutrition, 2Division of Oncology, Department of Pediatrics, BC Children’s Hospital Research Institute and the University of British Columbia, Vancouver, Canada, 3The Semel Institute and Department of Psychiatry, the David Geffen School of Medicine, University of California, Los Angeles, CA, 4Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada. The mammalian gastrointestinal (GI) tract is a highly innervated organ system, with enteric nerves and neuropeptides, including vasoactive intestinal peptide (VIP), controlling many physiologic responses that promote normal gut function. Herein, we found a significant increase in the intestinal secretion of VIP in mice infected with Citrobacter rodentium, a mouse pathogen widely used as a surrogate organism to study the pathogenesis of human pathogens: enteropathogenic E. coli (EPEC) and enterohemorrhagic E. coli (EHEC). This increased VIP secretion appears to be a protective host response, as mice deficient in VIP (Vip-/-) showed increased morbidity and mortality compared to wild type mice during infection. Intestinal tissues from Vip-/- mice secreted significantly less interleukin (IL)-22, a cytokine known to promote antimicrobial defense and epithelial repair during C. rodentium infection. Consistent with this observation, the expansion of group 3 innate lymphoid cells (ILC3s), a large cellular source of IL-22, was drastically reduced in the intestines of infected Vip-/- mice. Intestinal ILC3s from Vip-/- mice also produced reduced levels of IL-22 in response to IL-23 stimulation. Notably, administration of exogenous IL-22 or adoptive transfer of ILC3s into Vip-/- mice was sufficient to increase the resistance of Vip-/- mice to infection. Moreover, exogenous treatment of Vip-/- mice with recombinant VIP ameliorated pathogen-induced colitis, accompanied by partial restoration of IL-22 production. Collectively, these data identify a novel neuroimmune axis whereby the neuropeptide VIP controls the expansion and function of ILC3s in the GI tract, thereby playing a key role in optimizing mucosal defense against intestinal pathogens. #Corresponding authors: [email protected] and [email protected]

P28. Reduction of Zika virus Infectious Titer After Treatment with Anti-Inflammatory Compound, Triptolide. Hossaena Ayele2,4,5, Kathleen Glover2,3, Dr. Adam Burgener2,4,5,6, Dr. Kevin Coombs1,2, 3 1Department of Microbiology, Faculty of Science, University of Manitoba, Winnipeg, MB, 2Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB, 3Manitoba Centre for Proteomics and Systems Biology, 4National HIV and Retrovirology Labs, JC Wilt Center for Infectious Diseases, 5Public Health Agency of Canada, Winnipeg, Canada, 6Unit of Infectious Diseases, Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden. Abstract: In 2015, the Zika Virus (ZIKV) outbreak in the Americas was deemed a public health emergency by the World Health Organization (WHO) after several cases of microcephaly, Guillain Barré syndrome and other fetal abnormalities were observed. The discovery of anti-viral compounds and other therapeutic agents against ZIKV, is thus imperative for further prevention of ZIKV related complications, especially in pregnant women. Here we evaluated the efficacy of Triptolide used for the treatment of immune-inflammatory disorders due to its immunosuppression and anti-inflammatory capacities, to reduce infectious ZIKV titer. This was accomplished by use of U-251 human glioblastoma cells, which underwent treatment with Triptolide at various dilutions where cell viability was >80%, with subsequent ZIKV infection. Standard plaque assay with African Green Monkey epithelial (Vero) cells was then utilized for determination of infectious ZIKV presence at each concentration of Triptolide. Further analysis of viral proteins by western blot exhibited the absence of ZIKV Envelope (E) protein in Triptolide treated Vero cells. The compilation of lack of observable plaques and ZIKV E protein with Triptolide treatment underlines the ability of Triptolide to reduce infectious ZIKV virus titer, and may suggest its use as an anti-viral, either alone or in combination with other pharmacologic agents.

25

Page 26: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

26

P29. The effect of exogenous sugar alcohol (erythritol, sorbitol and mannitol) on Group B streptococcus virulence. Maram Hulbah and Gregory J. Tyrrell. Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta and ProvLab, Edmonton, Alberta. Group B streptococci (GBS) are bacterial pathogens of neonates. A cofactor thought associated with GBS virulence in neonates are polyols. Polyols (erythritol, sorbitol and mannitol) are sugar alcohols found in amniotic fluid of humans. The objective was to investigate the effect of polyols on GBS virulence. Methods: GBS were grown in 0%, 1%, 2%, 4% erythritol, sorbitol or mannitol overnight. Bacteria were harvested and virulence phenotypes assayed. Assays included measuring the expression of phosphoglycerate kinase (PGK – a plasminogen binding protein on GBS). Measuring antiphagocytic activity of GBS using fresh human blood and the effect of polyols on the invasion by GBS of HeLa cells. Results. GBS showed no significant difference in growth in concentrations of polyols used. GBS growth in presence of 1%, 2%, 4% erythritol enhanced expression of GBS-PGK compared to 0% erythritol. There was no significant difference in PGK surface expression in presence of 1%, 2%, 4% of sorbitol or mannitol. The antiphagocytic activity of GBS in 1%, 2%, 4% erythritol or sorbitol was significantly increased compared to 0%. In the presence of 1% mannitol, the antiphagocytic activity of GBS was not significantly increased in comparison with 2% and 4% mannitol which was significantly increased compared to 0%. GBS invaded HeLa cells at a higher rate (164%) in 1% erythritol compared to 0% erythritol. Invasion was not affected by sorbitol or mannitol. In summary, the presence of erythritol in culture media enhanced GBS virulence suggesting this polyol may play a role in GBS pathogenesis during neonatal infection.

P30. The Role of Streptococcus pyogenes Virulence Factors in Colonization and Rheumatic Heart Disease. Jacklyn R. Hurst1 and John K. McCormick1,2. 1Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, London, ON, Canada, 2 Lawson Health Research Institute, Western University, London, ON, Canada Rheumatic heart disease (RHD) is the leading cause acquired heart disease by an infectious agent worldwide, emerging as a severe autoimmune consequence of repetitive and untreated Group A Streptococcus (GAS) pharyngeal infections. The M protein, hyaluronic acid (HA) capsule and N-acetylglucosamine (GlcNAc) immunodominant epitope expressed on streptococcal surfaces are postulated to play a role during early GAS colonization and stimulate autoimmune responses in RHD. Isogenic mutant strains for each virulence factor in the parental M18 serotype strain were inoculated into whole human blood and analyzed for immune evasion capabilities. The survival of the gacI mutant strain did not significantly differ from the wild-type strain, however, deletion of the M18 protein and HA capsule drastically impeded growth and showed increased susceptibility to killing by whole human blood. The influence of each virulence factor on streptococcal colonization was explored through nasal inoculation of mutant strains into transgenic mice expressing human MHC class II molecules. Results suggest the GlcNAc and M18 protein do not offer important survival advantages for initial stages of infection as there was similar bacterial recovery between wild-type, gacI and emm18 mutant strains from isolated murine nasal turbinates. Notably, deletion of the HA capsule significantly hindered bacterial recovery and is therefore considered an essential virulence factor for GAS nasopharyngeal colonization. To understand the autoantigenic role of the M protein HA capsule, and GlcNAc, future experiments will involve the analysis of cardiac function by echocardiography following repetitive live GAS nasal infections to simulate repeated pharyngitis attacks that progress to RHD.

Page 27: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P31. Title: Exploring the function of a novel porin in Helicobacter pylori. K. Yogendirarajah1 & C. Creuzenet1. 1The University of Western Ontario, Canada. Introduction Helicobacter pylori (HP) colonizes over 50% of the world’s population and is the causative agent of both gastric ulcers and cancer. Due to high antibiotic failure rates, the WHO has listed HP as a high priority pathogen to promote research/development of new antibiotics. We focus on OmpX, currently known as a major nonspecific porin, and aim to investigate its role in antibiotic resistance, intraspecies interplay, and host-pathogen interactions. Hypothesis & Objectives We propose that OmpX may play a role in antibiotic susceptibility, and that the large-surface exposed loops of OmpX may function as an adhesin. Our objectives are: optimize the detection of OmpX using new anti-OmpX antibodies to distinguish between wild-type (WT) and ompX mutant; create a ompX/waaL double KO to remove the lipopolysaccharide O-antigen ligase WaaL functionality, allowing better characterization of OmpX’s function; and assess the ability of the WT and KO strains to mediate interactions with the host and other HP cells. Methods Western blotting was performed on OMPs extracted from the WT and ompX mutant, using anti-OmpX for detection. The ompX/waaL double KO was created by chromosomal integration of the ompX KO construct into the waaL KO strain. Antibiotic sensitivity was determined via spot plating, Etest strips, and disk diffusion method. Bacterial adhesion was tested via fibronectin/laminin binding, biofilm formation and auto-agglutination assays. Results We have successfully detected OmpX and confirmed creation of the ompX/waaL double KO. Other experiments are still in progress; however, all assays have been optimized and final results will be acquired shortly.

27

P32. A novel regulatory mechanism of the Vibrio cholerae Type 6 Secretion System. Kevin Manera and Tao Dong. Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary. The Type 6 Secretion System (T6SS) is a bacterial nano-machine capable of secreting toxic effectors into other bacteria or eukaryotes. T6SS delivers effectors to these cells using a contractile sheath which propels a sharp needle carrying cargo effectors through neighbouring cell membranes. In recent years, significant advances have been made in understanding the structure and mechanism of T6SS as well as the activities of its effectors. However, despite the large energy commitment required by the cell to create a functional T6SS, knowledge about regulation of its expression is lacking. We used RNA sequencing (RNA-Seq) to observe T6SS expression patterns in Vibrio cholerae. This data was confirmed with both qPCR and reporter gene assays and suggests the presence of a novel regulatory mechanism. To characterize this system, we performed Transposon Sequencing (Tn-Seq) to identify genes that are essential for regulation. We made gene knockouts of the top hits identified in the Tn-Seq screen and used qPCR and gene reporters to confirm their disruption interfered with normal regulation. Having identified genes that are putatively involved, we are now examining protein-protein and protein-DNA interactions of the gene products to elucidate the mechanism of regulation. By gaining an understanding of this novel T6SS control in the pathogen V. cholerae, we can start to better understand the role T6SS plays in V. cholerae infections.

Page 28: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P33. Using High-Throughput Proteomics to Characterize the Largest Arsenal of Bacterial Effector Proteins. Harley O'Connor Mount, Malene L. Urbanus, Atina G. Cote, Frederick P. Roth, Alex W. Ensminger. The intracellular bacterial pathogen Legionella pneumophila infects a broad range of eukaryotic hosts separated by more than a billion years of evolution(Parfrey et al., 2011). The generalist pathogen lifestyle has imposed selective pressure to expand the arsenal of encoded virulence proteins. As a result L. pneumophila harbors the largest bacterial effector repertoire sequenced to date(Ensminger, 2016). Currently only about 10-20% of this effector arsenal has an assigned function(Qiu and Luo, 2017), and host target information is known for even fewer. As an intracellular bacterial pathogen Legionella must maintain a fine control over its virulence program to prevent premature host killing. Recent work from our lab(Urbanus et al., 2016) and others(Jeong et al., 2015; Kubori et al., 2010; Neunuebel et al., 2011;Tan and Luo, 2011) has highlighted a role for effector proteins regulating the activity of each other both directly, and indirectly. These direct regulators, dubbed ‘metaeffectors’, likely provide enhanced spatiotemporal regulation to effector proteins during infection(Urbanus et al., 2016). To probe this regulatory complexity, we decided to assay the entire L. pneumophila effector proteome for physical interactions in all pairwise combinations using inducible barcode fusion genetic-yeast two-hybrid (iBFG-Y2H)(Yachie et al., 2016). This approach promises to reveal novel mechanisms of effector-mediated regulation, and effector complexes utilized by L. pneumophila during pathogenesis. In the future I will expand this approach to begin investigating metaeffectors across a range of intracellular bacterial pathogens.

P34. Making antibiotics work against high density infections by multidrug resistant ESKAPE pathogens. Daniel Pletzer, Sarah C. Mansour, Heidi Wolfmeier, Robert E.W. Hancock. University of British Columbia, 235-2259 Lower Mall Research Station, Vancouver, BC. There has been enormous publicity about the inexorable rise of resistance and the dearth of new therapies. However less attention has been placed on adaptively multidrug-resistant high density bacterial infections for which antibiotics are highly used but no effective therapies currently exist. Here we have provided new hope for this previously intractable class of infections as typified by abscess infections that are responsible for 3.2 million annual emergency room visits in the US alone. We show how to enhance the activity of antibiotics to treat multidrug-resistant Gram-positive and Gram-negative bacteria, using synthetic peptides that target the bacterial stress response, persister-based resistance and the outer membrane permeability barrier. In particular we have employed a new bacterial subcutaneous abscess mouse model to demonstrate that: (a) 7 of the society’s most recalcitrant pathogens formed cutaneous abscesses (including all ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter cloacae) pathogens and Escherichia coli) and even when antibiotics were directly delivered into abscess tissues, they showed poor efficacy; (b) By combining antibiotics with the local administration of anti-biofilm peptides that target cellular (stringent) stress responses, we could pharmacologically treat the infection and reduce the severity of cutaneous abscesses; (c) This synergy was due to increased outer membrane permeability as well as the disruption of the conserved stringent stress response that controls virulence and antibiotic resistance, particularly due to so-called persisters. These peptides have therefore the potential to broaden our limited antibiotic arsenal for a group of extremely difficult to treat infections.

28

Page 29: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P35. Investigating the Mechanism and Impact of Neutrophil Cell Death Following Intra-phagosomal Infection with Leishmania major. Adam J. Ranson, Vivian Martins, and Nathan C. Peters. Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary. Following in-vivo dermal infection with the intra-phagosomal pathogen Leishmania major (L. major), neutrophils are rapidly recruited to the skin and represent the majority of infected cells. Comparison of infected and uninfected neutrophils from the same site of dermal infection has revealed that Leishmania infection of neutrophils modulates neutrophil cell death. However, the mechanism of this modulation, and the effects of modulated neutrophil cell death on neighbouring cells of the innate immune system and the dermal inflammatory environment have not been well characterized. In this study, an in-vitro model was established using purified, murine, bone-marrow-derived neutrophils infected with L. major in order to investigate mechanisms of neutrophil cell death, the modulation of the cell death program by Leishmania, and subsequent extracellular communication with surrounding bystander phagocytes. Using flow cytometry, confocal microscopy, and western blot analysis, we have determined the optimal ratio of parasites to neutrophils for use in our studies, established the importance of opsonization of the parasite on the infection outcome of neutrophils, investigated the effects of neutrophil derived reactive oxygen species (ROS) on parasite survival and neutrophil cell death, detected the release of an extra-cellular alarmin, HMGB1, from neutrophils infected with L. major, and begun to characterize the molecular mechanism of cell death in neutrophils infected with L. major, including the role of canonical and non-canonical inflammasome activation. This study establishes the groundwork for future investigation into the mechanism of pathogen-mediated modulation of neutrophil cell death and the impact of this modulation on neighbouring bystander phagocytes and the inflammatory environment at sites of infection.

P36. Bat cells persistently infected with Middle East respiratory syndrome (MERS) coronavirus: a molecular model to study spillover. Arinjay Banerjee1, Sonu Subudhi1, Noreen Rapin1, Darryl Falzarano2 and Vikram Misra1. 1 Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Canada, 2 Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO/InterVac), Saskatoon, Canada. Middle-East respiratory syndrome coronavirus (MERS-CoV) is an emerging pathogen that causes disease in humans with a mortality of thirty-five percent. Bats are believed to be the original hosts for coronaviruses parasitizing many other mammalian species. However, little is known about how these viruses are retained in bat populations or the factors that lead to their spread. We have demonstrated that North American little brown bats are persistently infected with a coronavirus related to MERS-CoV, and that stress of secondary fungal infection suppresses innate antiviral responses potentially leading to increase in viral replication and spillover. To examine the molecular mechanisms that may regulate the coronavirus-bat relationship during viral persistence we tested the hypothesis that big brown bat cells can be persistently infected with MERS-CoV and disrupting the antiviral pathways will lead to an increase in virus propagation in these cells. We infected bat kidney cells with MERS-CoV at a multiplicity of infection of 0.01 and cultured the surviving cells for over three months. We characterized the persistently infected bat cells by demonstrating the presence of infectious virus by TCID50, viral proteins by confocal microscopy and immune blots, viral RNA by in-situ hybridization and virus particles by electron microscopy. Our data suggest that bat kidney cells can be persistently infected with MERS-CoV. Future studies will explore innate antiviral pathways in these persistently infected cells to identify molecules that control virus propagation. Knock-down/out studies will allow us to dissect the molecular mechanisms that lead to virus spillover from this intriguing viral reservoir.

29

Page 30: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P37. Identification of microsporidia host-exposed proteins reveals a repertoire of rapidly evolving proteins. Aaron Reinke1, Eric Bennett2 and Emily Troemel2.

1Department of Molecular Genetics, University of Toronto, 2Division of Biological Sciences, University of California, San Diego. Although it is clear that intracellular pathogens use a variety of secreted and surface proteins to interact with and manipulate their hosts, a systematic approach for identifying such proteins has been lacking. Because of this, the identity of these host-exposed proteins is often unknown in many pathogens. Additionally, little is known about how conserved repertoires of host-exposed proteins are between related species. Microsporidia are a large phylum of eukaryotic obligate intracellular parasites that can specifically infect a variety of different animal species. We identified host-exposed proteins from microsporidia using spatially restricted enzymatic tagging followed by mass spectrometry on Caenorhabditis elegans infected with two related species of Nematocida microsporidia. Using this approach, we identified 82 microsporidia proteins that are exposed inside of host intestinal cells, including several in the nucleus. These proteins are enriched in targeting signals, lack conservation with other microsporidia species, are rapidly evolving, and lack domains with known function. Almost half of the identified proteins belong to large, Nematocida-specific gene families that are undergoing species-specific radiations. We also find that large, species-specific families with targeting signals are common throughout microsporidia species. Our data suggest that the use of a large number of rapidly evolving species-specific proteins represent a common strategy for these intracellular pathogens to interact with their hosts.

P38. Characterization of a pre-mobilized MCR-1 ortholog: crystal structure of the intrinsic colistin resistance enzyme from Moraxella catarrhalis. Peter J. Stogios1, 2,*, Georgina Cox3, Haley L. Zubyk3, Elena Evdokimova1, 2, Zdzislaw Wawrzak4, Gerard D. Wright3, Alexei Savchenko1,2,5. 1Department of Chemical Engineering and Applied Chemistry, University of Toronto, [email protected] 2Center for Structural Genomics of Infectious Diseases (CSGID), 3Department of Biochemistry and Biomedical Sciences, McMaster University, 4LS-CAT, Synchrotron Research Center, Northwestern University, Argonne, Illinois 60439, 5Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Health Research Innovation Centre, Calgary, Alberta, T2N 4N1 *Research Associate Lipid A phosphoethanolamine (PEtN) transferases render bacteria resistant to the last resort antibiotic colistin. The recent discoveries of pathogenic bacteria harboring plasmid-borne PEtN transferases (mcr) genes have illustrated the serious potential for wide dissemination of these resistance elements. The origin of mcr-1 is traced to Moraxella species co-occupying environmental niches with Enterobacteriaceae. Here, we describe the crystal structure of the catalytic domain of the chromosomally-encoded colistin resistance PEtN transferase, ICRMc (for intrinsic colistin resistance) of Moraxella catarrhalis. The ICRMc structure in complex with PEtN reveals key molecular details including specific residues involved in catalysis and PEtN binding. It also demonstrates that ICRMc catalytic domain dimerization is required for substrate binding. Our structure-guided phylogenetic analysis provides sequence signatures defining potentially colistin-active representatives in this enzyme family. Combined, these results advance the molecular and mechanistic understanding of PEtN transferases and illuminate their origins.

30

Page 31: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P39. Stressors in persistently infected bats may influence the severity of the disease and/or increase virus shedding. Sonu Subudhi1, Neha Paranjape1, Noreen Rapin1, Christina M. Davy2,3, Michael E. Donaldson2, Lisa Warnecke4, James M. Turner4, Trent K. Bollinger5, Christopher J. Kyle6, Craig K.R. Willis4, and Vikram Misra1. 1Department of Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 2Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, 3Ontario Ministry of Natural Resources and Forestry, Wildlife Research and Monitoring Section, Trent University, Peterborough, ON, 4Department of Biology, University of Winnipeg, Manitoba, 5Canadian Wildlife Health Cooperative and Department of Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 6Forensic Science Department, Trent University, Peterborough, ON. In recent years, several viruses that cause no obvious disease in their natural bat hosts have spilled over to other species resulting in frequently fatal disease. These spillovers may be promoted by factors that disrupt the balanced relationship between viruses and the reservoir bat they have co-evolved with, leading to increased virus replication and shedding. We have detected and characterized corona and herpesviruses that infect two North American bat species – Myotis lucifugus and Eptesicus fuscus. Many (~30%) M. lucifugus are persistently infected with a coronavirus (Myl-CoV) and most (>70%) E. fuscus are latently infected with a gammaherpesvirus (EfHV). In order to examine this bat-virus relationship, we are testing the hypothesis that stressors such as secondary infections will alter the bat-virus relationship, leading to increased Myl-CoV replication and reactivation of latent EfHV. We show that the intestines of bats infected with Pseudogymnoascus destructans (Pd), a fungus that causes fatal White Nose Syndrome (WNS), contain on average 60-fold more Myl-CoV RNA than singly-infected bats and that the level of viral RNA correlates with the severity of WNS-related pathology. The intestines of bats with WNS also contain lower levels of transcripts for immune-modulatory genes. Our results suggest that the systemic effects of WNS down-regulate anti-viral responses in bats persistently infected with Myl-CoV, thereby increasing the potential for virus shedding. Further, we are artificially altering cellular stress-response pathways, namely unfolded-protein response, heat shock reponse, and hypoxia induced response to determine the molecular basis for the effect of stressors on viral replication and reactivation in-vitro.

31

P40. Developing new therapies to treat bloodstream infections caused by Klebsiella pneumoniae. Brent S. Weber1 and Eric. D. Brown1,2. 1 Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON 2 Contact: [email protected]. Hospital-acquired infections caused by opportunistic bacterial pathogens are a significant contributor to patient death and a major healthcare burden. Coupled with an alarming increase in antibiotic resistance, and few new antibiotics in development, drug-resistant bacterial pathogens have quickly established themselves as a global threat to healthcare security. Bloodstream infections (BSI) caused by antibiotic resistant Gram-negative bacteria are a leading cause of mortality and can be challenging to treat. The bloodstream is a hostile environment for bacteria, containing a multitude of factors that can kill bacteria. Blood serum contains soluble components, such as complement, that can efficiently lyse susceptible bacteria, and bacteria must adapt to varied levels of nutrients and sequestered micronutrients in serum by synthesizing important metabolites de novo or evolving systems to acquire them from the host. We hypothesized that the mechanisms used by bacteria to survive in this challenging environment represent novel targets for the development of new antibacterial drugs to treat BSI. We utilized a sequence-defined transposon library of Klebsiella pneumoniae, an important cause of BSI, to identify the factors required by this organism to grow in human serum. Using this data as a target list, we undertook a high-throughput screening campaign aimed at finding chemical inhibitors of these essential factors. This screen yielded several compounds that show enhanced activity in human serum, and therefore could be beneficial for developing new therapeutics against bacterial bloodstream infections.

Page 32: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P41. Visualizing the Role of Alpha Toxin in Real Time During Staphylococcus aureus Sepsis.

Bas Surewaard PhD. Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta. Sepsis is a clinical manifestation of the host’s inflammatory reaction to bloodstream infections causing microvascular perfusion problems leading to multiple organ dysfunction. Clinically, one of the biggest problems during sepsis is the formation of large platelet aggregates in the vasculature of poorly understood etiology leading to thrombotic microangiopathy, vascular occlusion and multi-organ failure. Insight into Staphylococcus aureus infection-mediated vascular damage and multi-organ dysfunction is limited. Therefore, using intravital spinning-disk microscopy we visualized rapid dynamic intravascular events in the microcirculation following Staphylococcal a-toxin (AT a-hemolysin, Hla) intoxication or infection. Intravenous AT injection induced rapid platelet aggregation, forming dynamic micro-thrombi in all organs examined. While the aggregates were washed away from the lung and skin vasculature, they were retained in the liver sinusoids and glomeruli of the kidney causing multi-organ dysfunction. Platelet aggregation and subsequent tissue damage in the liver were prevented with the AT neutralizing antibody MEDI4893*. Acute S. aureus infection resulted in sequestration of most bacteria by liver macrophages. Platelets were recruited to these cells early in the infection in a complement-dependent manner, but with time, an aberrant and damaging AT-dependent thrombosis predominated in the liver. Similarly, prophylaxis with MEDI4893* prior to S. aureus infection did not reduce early beneficial platelet recruitment, but did significantly reduce hepatic platelet deposition and thrombocytopenia resulting in reduced liver damage. These results indicate that AT elicits a damaging platelet response that can be prevented with MEDI14893*. This may represent a promising molecule to prevent staphylococcal induced intravascular coagulation, poor tissue perfusion and organ dysfunction.

P42. Defining the function of VipF effector conserved across Legionella species. Lukas Syriste1, Peter J. Stogios2, Andy Quaile2, Dylan Valleau2,3, Zdzislaw Wawrzak5, Rosa Di Leo2, Tatiana Skarina2, Joanna Sandoval2, Alexander Ensminger3,4, Alexei Savchenko1,2. 1 Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, AB, 2 Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, 3 Department of Molecular Genetics, University of Toronto, Toronto, ON, 4 Department of Biochemistry, University of Toronto, Toronto, ON, 5 Life Sciences Collaborative Access Team, Northwestern University, Argonne, IL, USA. The pathogenic strategy of severe pneumonia-causing Legionella pneumophila is based on delivery of over 300 pathogenic factors called effectors inside the host cell via the Dot/Icm secretion system. The genomic analysis of 38 Legionella strains demonstrated that only seven “core” effectors are conserved across this genus. This included the uncharacterised VipF effector encoded by lpg0103 in L. pneumophila. We have determined the crystal structure of L. pneumophila VipF, which revealed features typical of Gcn5-related N-acetyltransferase (GNAT) superfamily of acetyltransferases. In vitro acetylation assay using Ellman’s thiol reagent confirmed the acetyltransferase activity of VipF. In order to identify the possible substrate of this enzyme in host cells we purified the VipF homologues from L. pneumophila, L. hackeliae, L. longbeachae, L. micdadei, and L. fallonii in fusion with streptavidin-binding peptide (SBP). These VipF homologues were used as bait in affinity pull-down assays from human U937 macrophage cell lysates. Co-precipitated human proteins were analyzed using nano-scale liquid chromatography electrospray-ionization tandem mass spectrometry (nLC-MS/MS). This analysis suggests that VipF is targeting a specific human protein typically localized to nucleus. Given the conservation of VipF effector among Legionella species, the elucidation of its molecular function holds potential for illuminating fundamental mechanisms of host manipulation by this intracellular pathogen.

32

Page 33: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

P43. Vascular dissemination of Borrelia burgdorferi. Xi Tan1,2, Priyanka Mukherjee1,2,3, Jennifer A. Caine4,5,*, Jenifer Coburn4,5,6 and George Chaconas1,2,3. 1Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, 2Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, 3Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Alberta, 4Graduate Program in Microbiology, Immunology, and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA, 5Center for Infectious Disease Research, Medical College of Wisconsin, Milwaukee, WI, USA, 6Division of Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA. *Current address: Department of Microbiology and Immunology, Medical College of Wisconsin. Lyme disease, caused by various members of the genus Borrelia, is the most prevalent tick-borne illness in the northern hemisphere. B. burgdorferi hematogenous dissemination is a multistep process and consists of several successive stages: tethering and dragging interactions, followed by stationary adhesion and extravasation. However, the mechanism of vascular transmigration by Lyme disease spirochetes is largely uncharacterized. To study the spirochete transmigration pathway, we are using intravital microscopy. Using this technique we can detect Borrelia extravasation in the peripheral knee vasculature in living Cd1d-/- mice, where iNKT cells are absent and do not disrupt the process. B. burgdorferi produces adhesins involved in binding to the vascular endothelium and the extracellular matrix. Whether these proteins act at different stages of infection or in colonization of specific tissues is not yet known. We have previously shown by intravital microscopy that the Fn/GAG binding protein BBK32 is not directly involved in vascular transmigration, but that the integrin binding porin, P66, is required though its integrin binding region. We are currently assessing the role of the B. burgdorferi adhesins VlsE, OspC and DbpA,B in vascular transmigration and adhesion using GFP-expressing B. burgdorferi strains carrying mutations in these genes. To date we have accumulated data suggesting that the outer membrane protein OspC is required for vascular transmigration into the mouse knee joint. Experiments are also in progress to distinguish between transcellular and paracellular transmigration pathways to initiate mechanistic studies and elucidate the molecular details of the process.

33

P44. Motility Kills: A mucus induced lifestyle that promotes antibiotic resistance. Evelyn Sun, Erin E. Gill, Reza Falsafi, Amy Yeung, Nicole Liu, and Robert E.W. Hancock, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC. Pseudomonas aeruginosa is a gram-negative opportunistic pathogen associated with a high incidence of lung infections in hospitalized and cystic fibrosis patients. P. aeruginosa is highly adaptable and exhibits diverse lifestyle adaptations depending on its surrounding conditions. Changes in viscosity, nitrogen sources, and other factors can induce major changes in adaptations ranging from swarming motility to biofilm formation. Here we show how the presence of mucin, a glycoprotein that is found in large abundance in the cystic fibrosis lung, induced a novel form of motile surface lifestyle known as surfing and how surfing is associated with broad-spectrum antibiotic resistance. Using disk diffusion assays, we investigated the adaptive antibiotic resistance of P. aeruginosa under conditions in which surfing motility occurs compared to cells undergoing swimming motility. P. aeruginosa surfing cells were significantly more resistant to several classes of antibiotics including aminoglycosides, carbapenems, polymyxins, and fluroquinolones. To investigate the basis of resistance we performed RNA-Seq to look for dysregulation in genes known to be involved in resistance when dysregulated, namely resistome genes. Screening transposon mutants of resistome genes that were dysregulated under surfing conditions revealed that several of these mutants, including mutants in armR, recG, atpB, clpS, nuoB, exhibited surfing-dependent changes in susceptibility to one or more antibiotics, consistent with a contribution to the observed adaptive resistance. Antibiotic incorporation assays confirmed a concentration-dependent inhibition of surfing motility that occurred at concentrations much higher than those needed to inhibit swimming. Therefore, we propose that surfing adaption led to high multidrug adaptive resistance as a result of the collective dysregulation of diverse resistome genes.

Page 34: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

34

P45. Membrane damage-responsive biosensors for the discovery of antimicrobials from Lenzites betulina and Haploporus odorus. Steve Shideler1,2,3, Shauna Reckseidler-Zenteno1,2, Roland Treu1, Azka Qasim3, and Shawn Lewenza1,2 1 Athabasca University, Faculty of Science and Technology, Athabasca, AB, 2 University of Calgary, Dept. Microbiology, Immunology and Infectious Diseases, Calgary AB, 3 University of Calgary, Dept Biological Sciences, Calgary AB. Abstract: Antibiotic resistance of pathogens is a growing concern in medical treatment of infections. Conventional methods for antimicrobial screening rely on detecting direct antimicrobial activity. However, this method is not sensitive enough to detect sub-lethal activities. We have developed biosensors in Pseudomonas aeruginosa that produce light from transcriptional lux fusions in response to compounds that damage the outer membrane. Sub-lethal exposure to antimicrobial peptides, the antibiotic cycloserine, and cation chelators induces the expression of arnC::lux (PA3553) and speE2::lux (PA4774). These genes encode outer surface modifications that ultimately protect the P. aeruginosa outer membrane from exposure to sub-lethal concentrations of membrane-active compounds. We used high throughput screening in 96-well microplate format to test the cell-free, culture supernatants from a panel of fungal species isolated in Alberta. Preliminary results showed a significant induction of both strains of lux biosensors in 23 of the 29 fungal species screened. Due to the significant biosensor response by the supernatants from Lenzites betulina, Haploporus odorus, Ganoderma applanation, and Lentinula edodes, an 8-week time course of experiments was performed to observe optimal induction of the biosensor strains when challenged with cell free fungal supernatant. Protease digestion of the supernatants also provided evidence that some of the supernatants contained active protein compounds, whereas other supernatants showed no change in activity, indicating a non-protein compound with membrane activity. Large batch cultures were grown and supernatant was harvested at optimal time points. Proteins were then concentrated and fractionated using fast protein liquid chromatography (FPLC) on Superdex™ 200 GL 10/300 and Superdex™ peptide 10/300 columns. The fractions were then retested for biosensor activity. Preliminary mass spectrometry has identified the presence of histones and glycoside hydrolases, proteins known to possess antimicrobial activity, in active fractions. Synthetic peptides derived from a fungal glycoside hydrolase are being tested for antimicrobial activity. Our efforts directed at the purification and isolation of the active components found in the fungal cell free supernatants have met with preliminary success and further identification is currently underway. In summary, we developed a rapid and sensitive approach to detect antimicrobials from fungi that target the Gram-negative outer membrane.

Page 35: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

35

P47. Pseudomonas aeruginosa Swarming Motility: Mechanisms of Antibiotic Resistance. Coleman, S.R. and Hancock, R.E.W. Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC.

Abstract: In infectious diseases, bacterial pathogens are proficient at acquiring drug resistance over time due to a number of different factors. First, their short generation time enables resistant subpopulations to rapidly dominate. Second, bacteria possess genetic plasticity allowing for the uptake and exchange of foreign DNA. Third, pathogenic bacteria are extremely versatile and can respond to environmental stress with massive changes in gene expression leading to adaptive resistance. This is particularly true of the opportunistic and nosocomial pathogen Pseudomonas aeruginosa, which has a genome that encodes approximately 10% transcriptional regulators. Under conditions relevant to the human lung (semisolid with amino acids as a nitrogen source), P. aeruginosa undergoes swarming motility, a multicellular phenomenon where rafts of cells propel themselves across a surface, resulting in rapid colonization. Here we show that P. aeruginosa is resistant to multiple antibiotic classes under swarming conditions as compared to control swimming and spread plate bacteria. This phenomenon was further investigated by examining the transcriptome of swarm cells for resistance determinants. Mutants in candidate genes were tested for altered antibiotic susceptibility under swarming conditions. Results indicate that PrtN, a transcriptional regulator of pyocin, influences resistance to tobramycin and trimethoprim; and WbpW, involved in LPS biosynthesis, contributes to tobramycin susceptibility. These determinants may be relevant for drug resistance in infectious human diseases such as pneumonia and cystic fibrosis.

Page 37: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

37

Participants’ E-mail list

Karin Aistleitner [email protected]

Sara Andres Lasheras [email protected]

Hossaena Ayele [email protected]

Sahar Bagheri [email protected]

Arinjay Banerjee [email protected]

Corrie Belanger [email protected]

Amit Bhavsar [email protected]

Agostina Carestia [email protected]

Somshukla Chaudhuri [email protected]

Deirdre Church [email protected]

Shannon Coleman [email protected]

John Conly [email protected]

Carole Creuzenet [email protected]

Brendan Daisley [email protected]

Tao Dong [email protected]

Zinaida Eltsova [email protected]

Alex Ensminger [email protected]

Lana Estafanos [email protected]

Nikolas Ewasechko [email protected]

Caleigh Gerow [email protected]

Mariya Goncheva [email protected]

Steven Hersch [email protected]

Maram Hulbah [email protected]

Jacklyn Hurst [email protected]

Fatima Kamal [email protected]

Ayesha Kashif [email protected]

Sung Soo Kim [email protected]

Linh Lam [email protected]

Ian Lewis [email protected]

Kevin Manera [email protected]

Claudia Fabiola Martinez de la Pena [email protected]

John McCormick [email protected]

Vikram Misra [email protected]

Dion Neame [email protected]

Phu Hai NGUYEN [email protected]

Sobia Nizami [email protected]

Harley O'Connnor Mount [email protected]

Neha Paranjape [email protected]

Page 38: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

38

Nathan Peters [email protected]

Daniel Pletzer [email protected]

Adam Ranson [email protected]

Noreen Rapin [email protected]

Aaron Reinke [email protected]

Jamie Rothenburger [email protected]

Anthony Schryvers [email protected]

Steve Shideler [email protected]

Olivia Steele-Mortimer [email protected]

Maria Silvina Stietz [email protected]

Peter Stogios [email protected]

DOUG STOREY [email protected]

Sonu Subudhi [email protected]

Evelyn Sun [email protected]

Bas Surewaard [email protected]

Lukas Syriste [email protected]

Xi Tan [email protected]

Le Tang [email protected]

Ankita Tulangekar [email protected]

Nienke Van Houten [email protected]

Gehua Wang [email protected]

Brent Weber [email protected]

Heidi Wolfmeier [email protected]

Keertika Yogendirarajah [email protected]

HONG BING YU [email protected]

Amanda Zucoloto [email protected]

Page 39: PowerPoint Presentation · 1961. He then joined the Laboratory of Angus Graham at the Wistar Institute in Philadelphia, where he made his most important early contribution to science,

BCID 2016 - Notes

39