128 results on '"Hall-Stoodley L"'
Search Results
2. 76 Surface-dependent inhibition of Mycobacterium abscessus by diverse Pseudomonas aeruginosa strains
- Author
-
Idosa, A., primary, Wozniak, D., additional, and Hall-Stoodley, L., additional
- Published
- 2023
- Full Text
- View/download PDF
3. ESCMID guideline for the diagnosis and treatment of biofilm infections 2014
- Author
-
Høiby, N., Bjarnsholt, T., Moser, C., Bassi, G.L., Coenye, T., Donelli, G., Hall-Stoodley, L., Holá, V., Imbert, C., Kirketerp-Møller, K., Lebeaux, D., Oliver, A., Ullmann, A.J., and Williams, C.
- Published
- 2015
- Full Text
- View/download PDF
4. On the mechanics of bacterial biofilms on non-dissolvable surgical sutures: A laser scanning confocal microscopy-based finite element study
- Author
-
Limbert, G., Bryan, R., Cotton, R., Young, P., Hall-Stoodley, L., Kathju, S., and Stoodley, P.
- Published
- 2013
- Full Text
- View/download PDF
5. G385 RATNO – Reducing Antibiotic Tolerance using Nitric Oxide in Cystic Fibrosis: report of a proof of concept clinical trial
- Author
-
Cathie, K, Howlin, R, Carroll, M, Clarke, S, Connett, G, Cornelius, V, Daniels, T, Duignan, C, Hall-Stoodley, L, Jefferies, J, Kelso, M, Kjelleberg, S, Legg, J, Pink, S, Rogers, G, Salib, R, Stoodley, P, Sukhtankar, P, Webb, J, and Faust, S
- Published
- 2014
- Full Text
- View/download PDF
6. The influence of fluid shear on the structure and material properties of sulphate-reducing bacterial biofilms
- Author
-
Dunsmore, B C, Jacobsen, A, Hall-Stoodley, L, Bass, C J, Lappin-Scott, H M, and Stoodley, P
- Published
- 2002
- Full Text
- View/download PDF
7. Mycobacterium fortuitum and Mycobacterium chelonae biofilm formation under high and low nutrient conditions
- Author
-
Hall-Stoodley, L., Keevil, C. W., and Lappin-Scott, H. M.
- Published
- 1999
8. Mycobacterium fortuitum and Mycobacterium chelonae biofilm formation under high and low nutrient conditions
- Author
-
Hall-Stoodley, L., Keevil, C. W., and Lappin-Scott, H. M.
- Published
- 1998
- Full Text
- View/download PDF
9. Low-dose nitric oxide as targeted anti-biofilm adjunctive therapy to treat chronic Pseudomonas aeruginosa infection in cystic fibrosis
- Author
-
Howlin, RP, Cathie, K, Hall-Stoodley, L, Cornelius, V, Duignan, C, Allan, RN, Fernandez, BO, Barraud, N, Bruce, KD, Jefferies, J, Kelso, M, Kjelleberg, S, Rice, SA, Rogers, GB, Pink, S, Smith, C, Sukhtankar, PS, Salib, R, Legg, J, Carroll, M, Daniels, T, Feelisch, M, Stoodley, P, Clarke, SC, Connett, G, Faust, SN, and Webb, JS
- Subjects
Adult ,BACTERIAL BIOFILMS ,Time Factors ,Cystic Fibrosis ,Adolescent ,Research & Experimental Medicine ,Nitric Oxide ,cystic fibrosis ,Young Adult ,nitric oxide ,SPUTUM ,10 Technology ,Humans ,ANTIBIOTIC SUSCEPTIBILITIES ,Pseudomonas Infections ,SILICA NANOPARTICLES ,Randomized Controlled Trials as Topic ,Genetics & Heredity ,Science & Technology ,Dose-Response Relationship, Drug ,Biofilm ,Sputum ,MIDDLE-EAR ,POLYMORPHONUCLEAR LEUKOCYTES ,CLINICAL-SAMPLES ,ADULTS ,11 Medical And Health Sciences ,Middle Aged ,IN-SITU HYBRIDIZATION ,06 Biological Sciences ,biochemical phenomena, metabolism, and nutrition ,Bacterial Load ,Anti-Bacterial Agents ,Biotechnology & Applied Microbiology ,Medicine, Research & Experimental ,Biofilms ,LUNG INFECTIONS ,Pseudomonas aeruginosa ,Original Article ,Life Sciences & Biomedicine ,Biotechnology - Abstract
Despite aggressive antibiotic therapy, bronchopulmonary colonization by Pseudomonas aeruginosa causes persistent morbidity and mortality in cystic fibrosis (CF). Chronic P. aeruginosa infection in the CF lung is associated with structured, antibiotic-tolerant bacterial aggregates known as biofilms. We have demonstrated the effects of non-bactericidal, low-dose nitric oxide (NO), a signaling molecule that induces biofilm dispersal, as a novel adjunctive therapy for P. aeruginosa biofilm infection in CF in an ex vivo model and a proof-of-concept double-blind clinical trial. Submicromolar NO concentrations alone caused disruption of biofilms within ex vivo CF sputum and a statistically significant decrease in ex vivo biofilm tolerance to tobramycin and tobramycin combined with ceftazidime. In the 12-patient randomized clinical trial, 10 ppm NO inhalation caused significant reduction in P. aeruginosa biofilm aggregates compared with placebo across 7 days of treatment. Our results suggest a benefit of using low-dose NO as adjunctive therapy to enhance the efficacy of antibiotics used to treat acute P. aeruginosa exacerbations in CF. Strategies to induce the disruption of biofilms have the potential to overcome biofilm-associated antibiotic tolerance in CF and other biofilm-related diseases., This paper reports the first example of targeted anti-biofilm therapy in human disease. We have demonstrated that using low-dose nitric oxide as a non-bactericidal signaling molecule to induce biofilm dispersal may be useful as a novel adjunctive therapy to treat chronic pseudomonal biofilm infection in cystic fibrosis.
- Published
- 2017
10. Cephalosporin-3’-diazeniumdiolate NO-donor prodrug PYRRO-C3D is active against Streptococcus pneumoniae biofilms despite the absence of -lactamases
- Author
-
Allan, Raymond N., Kelso, M.J., Rineh, A., Yepuri, N.R., Feelisch, M., Soren, O., Brito-Mutunayagam, S., Salib, R.J., Stoodley, P., Clarke, S.C., Webb, J.S., Hall-Stoodley, L., and Faust, S.N.
- Subjects
Streptococcus pneumoniae ,Biofilm ,Nitric oxide ,biochemical phenomena, metabolism, and nutrition - Abstract
The file attached to this record is the author's final peer reviewed version. The Publisher's final version can be found by following the DOI link. Bacterial biofilms show high tolerance towards antibiotics and are a significant problem in clinical settings where they are a primary cause of chronic infections. Novel therapeutic strategies are needed to improve anti-biofilm efficacy and support reduction in antibiotic use. Treatment with exogenous nitric oxide (NO) has been shown to modulate bacterial signaling and metabolic processes that render biofilms more susceptible to antibiotics.We previously reported on cephalosporin-3’-diazeniumdiolates (C3Ds) as NO-donor prodrugs designed to selectively deliver NO to bacterial infection sites following reaction with b-lactamases. With structures based on cephalosporins, C3Ds could, in principal, also be triggered to release NO following b-lactam cleavage mediated by transpeptidases/penicillin-binding proteins (PBPs), the antibacterial target of cephalosporin antibiotics. Transpeptidase-reactive C3Ds could potentially show both NO-mediated anti-biofilm properties and intrinsic (b-lactam-mediated) antibacterial effects. This dual-activity concept was explored using Streptococcus pneumoniae, a species that lacks b-lactamases but relies on transpeptidases for cell-wall synthesis. Treatment with PYRRO-C3D (a representative C3D containing the diazeniumdiolate NO donor PYRRO-NO) was found to significantly reduce viability of planktonic and biofilm pneumococci, demonstrating that C3Ds can elicit direct, cephalosporin-like antibacterial activity in the absence of b-lactamases. While NO release from PYRRO-C3D in the presence of pneumococci was confirmed, the anti-pneumococcal action of the compound was shown to arise exclusively from the b-lactam component and not through NO-mediated effects. The compound showed similar potency to amoxicillin against S. pneumoniae biofilms and greater efficacy than azithromycin, highlighting the potential of C3Ds as new agents for treating pneumococcal infections.
- Published
- 2017
11. EPS1.05 CAT-5571 restores autophagy, a fundamental defect in cystic fibrosis, and is a potential new treatment for people with cystic fibrosis
- Author
-
Liu, F., primary, Krause, K., additional, Amer, A., additional, Hall-Stoodley, L., additional, Reilly, J.F., additional, and Nichols, A.J., additional
- Published
- 2018
- Full Text
- View/download PDF
12. An iron detection system determines bacterial swarming initiation and biofilm formation
- Author
-
Lin, Chuan-Sheng, Tsai, Yu-Huan, Chang, Chih-Jung, Tseng, Shun-Fu, Wu, Tsung-Ru, Lu, Chia-Chen, Wu, Ting-Shu, Lu, Jang-Jih, Horng, Jim-Tong, Martel, Jan, Ojcius, David M., Lai, Hsin-Chih, Young, John D., Andrews, S. C., Robinson, A. K., Rodriguez-Quinones, F., Touati, D., Yeom, J., Imlay, J. A., Park, W., Marx, J. J., Braun, V., Hantke, K., Cornelis, P., Wei, Q., Vinckx, T., Troxell, B., Hassan, H. M., Verstraeten, N., Lewis, K., Hall-Stoodley, L., Costerton, J. W., Stoodley, P., Kearns, D. B., Losick, R., Butler, M. T., Wang, Q., Harshey, R. M., Lai, S., Tremblay, J., Deziel, E., Overhage, J., Bains, M., Brazas, M. D., Hancock, R. E., Partridge, J. D., Kim, W., Surette, M. G., Givskov, M., Rather, P. N., Houdt, R. Van, Michiels, C. W., Mukherjee, S., Inoue, T., Frye, J. G., McClelland, M., McCarter, L., Silverman, M., Matilla, M. A., Wu, Y., Outten, F. W., Singh, P. K., Parsek, M. R., Greenberg, E. P., Welsh, M. J., Banin, E., Vasil, M. L., Wosten, M. M., Kox, L. F., Chamnongpol, S., Soncini, F. C., Groisman, E. A., Laub, M. T., Goulian, M., Krell, T., Lai, H. C., Lin, C. S., Soo, P. C., Tsai, Y. H., Wei, J. R., Wyckoff, E. E., Mey, A. R., Leimbach, A., Fisher, C. F., Payne, S. M., Livak, K. J., Schmittgen, T. D., Clarke, M. B., Hughes, D. T., Zhu, C., Boedeker, E. C., Sperandio, V., Stintzi, A., Clarke-Pearson, M. F., Brady, S. F., Drake, E. J., Gulick, A. M., Qaisar, U., Rowland, M. A., Deeds, E. J., Garcia, C. A., Alcaraz, E. S., Franco, M. A., Rossi, B. N. Passerini de, Mehi, O., Skaar, E. P., Visaggio, D., Nishino, K., Dietz, P., Gerlach, G., Beier, D., Bustin, S. A., Schwyn, B., Neilands, J. B., Sub algemeen U-talent, Dep Biologie, Sub Inorganic Chemistry and Catalysis, LS Infectiebiologie (Bacteriologie), Sub Condensed Matter and Interfaces, SGPL Stadsgeografie, and dI&I I&I-2
- Subjects
0301 basic medicine ,inorganic chemicals ,Iron ,030106 microbiology ,Swarming (honey bee) ,Microbial communities ,Flagellum ,Bacterial Physiological Phenomena ,Models, Biological ,Article ,Microbiology ,03 medical and health sciences ,Bacterial Proteins ,Models ,Coumarins ,Bacteriology ,Serratia marcescens ,Multidisciplinary ,biology ,Biofilm ,Bacterial ,Gene Expression Regulation, Bacterial ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Biological ,Cell biology ,Gene Expression Regulation ,Flagella ,Biofilms ,Signal transduction ,Bacteria ,Signal Transduction - Abstract
Iron availability affects swarming and biofilm formation in various bacterial species. However, how bacteria sense iron and coordinate swarming and biofilm formation remains unclear. Using Serratia marcescens as a model organism, we identify here a stage-specific iron-regulatory machinery comprising a two-component system (TCS) and the TCS-regulated iron chelator 2-isocyano-6,7-dihydroxycoumarin (ICDH-Coumarin) that directly senses and modulates environmental ferric iron (Fe3+) availability to determine swarming initiation and biofilm formation. We demonstrate that the two-component system RssA-RssB (RssAB) directly senses environmental ferric iron (Fe3+) and transcriptionally modulates biosynthesis of flagella and the iron chelator ICDH-Coumarin whose production requires the pvc cluster. Addition of Fe3+, or loss of ICDH-Coumarin due to pvc deletion results in prolonged RssAB signaling activation, leading to delayed swarming initiation and increased biofilm formation. We further show that ICDH-Coumarin is able to chelate Fe3+ to switch off RssAB signaling, triggering swarming initiation and biofilm reduction. Our findings reveal a novel cellular system that senses iron levels to regulate bacterial surface lifestyle.
- Published
- 2016
- Full Text
- View/download PDF
13. Helycobacter pilory Outer Membrane Vesicles (OMVs) as DNA 'shuttles' and structural components of biofilm EPS
- Author
-
Grande, R, Di Marcantonio, Mc, Robuffo, I, Pompilio, A, Primavera, Rosita, Di Francesco, M, Di Bartolomeo, E, Di Bonaventura, G, Stoodley, P, Hall Stoodley, L, and Mincione, G.
- Published
- 2015
14. An iron detection system determines bacterial swarming initiation and biofilm formation
- Author
-
Sub algemeen U-talent, Dep Biologie, Sub Inorganic Chemistry and Catalysis, LS Infectiebiologie (Bacteriologie), Sub Condensed Matter and Interfaces, SGPL Stadsgeografie, dI&I I&I-2, Lin, Chuan-Sheng, Tsai, Yu-Huan, Chang, Chih-Jung, Tseng, Shun-Fu, Wu, Tsung-Ru, Lu, Chia-Chen, Wu, Ting-Shu, Lu, Jang-Jih, Horng, Jim-Tong, Martel, Jan, Ojcius, David M., Lai, Hsin-Chih, Young, John D., Andrews, S. C., Robinson, A. K., Rodriguez-Quinones, F., Touati, D., Yeom, J., Imlay, J. A., Park, W., Marx, J. J., Braun, V., Hantke, K., Cornelis, P., Wei, Q., Vinckx, T., Troxell, B., Hassan, H. M., Verstraeten, N., Lewis, K., Hall-Stoodley, L., Costerton, J. W., Stoodley, P., Kearns, D. B., Losick, R., Butler, M. T., Wang, Q., Harshey, R. M., Lai, S., Tremblay, J., Deziel, E., Overhage, J., Bains, M., Brazas, M. D., Hancock, R. E., Partridge, J. D., Kim, W., Surette, M. G., Givskov, M., Rather, P. N., Houdt, R. Van, Michiels, C. W., Mukherjee, S., Inoue, T., Frye, J. G., McClelland, M., McCarter, L., Silverman, M., Matilla, M. A., Wu, Y., Outten, F. W., Singh, P. K., Parsek, M. R., Greenberg, E. P., Welsh, M. J., Banin, E., Vasil, M. L., Wosten, M. M., Kox, L. F., Chamnongpol, S., Soncini, F. C., Groisman, E. A., Laub, M. T., Goulian, M., Krell, T., Lai, H. C., Lin, C. S., Soo, P. C., Tsai, Y. H., Wei, J. R., Wyckoff, E. E., Mey, A. R., Leimbach, A., Fisher, C. F., Payne, S. M., Livak, K. J., Schmittgen, T. D., Clarke, M. B., Hughes, D. T., Zhu, C., Boedeker, E. C., Sperandio, V., Stintzi, A., Clarke-Pearson, M. F., Brady, S. F., Drake, E. J., Gulick, A. M., Qaisar, U., Rowland, M. A., Deeds, E. J., Garcia, C. A., Alcaraz, E. S., Franco, M. A., Rossi, B. N. Passerini de, Mehi, O., Skaar, E. P., Visaggio, D., Nishino, K., Dietz, P., Gerlach, G., Beier, D., Bustin, S. A., Schwyn, B., Neilands, J. B., Sub algemeen U-talent, Dep Biologie, Sub Inorganic Chemistry and Catalysis, LS Infectiebiologie (Bacteriologie), Sub Condensed Matter and Interfaces, SGPL Stadsgeografie, dI&I I&I-2, Lin, Chuan-Sheng, Tsai, Yu-Huan, Chang, Chih-Jung, Tseng, Shun-Fu, Wu, Tsung-Ru, Lu, Chia-Chen, Wu, Ting-Shu, Lu, Jang-Jih, Horng, Jim-Tong, Martel, Jan, Ojcius, David M., Lai, Hsin-Chih, Young, John D., Andrews, S. C., Robinson, A. K., Rodriguez-Quinones, F., Touati, D., Yeom, J., Imlay, J. A., Park, W., Marx, J. J., Braun, V., Hantke, K., Cornelis, P., Wei, Q., Vinckx, T., Troxell, B., Hassan, H. M., Verstraeten, N., Lewis, K., Hall-Stoodley, L., Costerton, J. W., Stoodley, P., Kearns, D. B., Losick, R., Butler, M. T., Wang, Q., Harshey, R. M., Lai, S., Tremblay, J., Deziel, E., Overhage, J., Bains, M., Brazas, M. D., Hancock, R. E., Partridge, J. D., Kim, W., Surette, M. G., Givskov, M., Rather, P. N., Houdt, R. Van, Michiels, C. W., Mukherjee, S., Inoue, T., Frye, J. G., McClelland, M., McCarter, L., Silverman, M., Matilla, M. A., Wu, Y., Outten, F. W., Singh, P. K., Parsek, M. R., Greenberg, E. P., Welsh, M. J., Banin, E., Vasil, M. L., Wosten, M. M., Kox, L. F., Chamnongpol, S., Soncini, F. C., Groisman, E. A., Laub, M. T., Goulian, M., Krell, T., Lai, H. C., Lin, C. S., Soo, P. C., Tsai, Y. H., Wei, J. R., Wyckoff, E. E., Mey, A. R., Leimbach, A., Fisher, C. F., Payne, S. M., Livak, K. J., Schmittgen, T. D., Clarke, M. B., Hughes, D. T., Zhu, C., Boedeker, E. C., Sperandio, V., Stintzi, A., Clarke-Pearson, M. F., Brady, S. F., Drake, E. J., Gulick, A. M., Qaisar, U., Rowland, M. A., Deeds, E. J., Garcia, C. A., Alcaraz, E. S., Franco, M. A., Rossi, B. N. Passerini de, Mehi, O., Skaar, E. P., Visaggio, D., Nishino, K., Dietz, P., Gerlach, G., Beier, D., Bustin, S. A., Schwyn, B., and Neilands, J. B.
- Published
- 2016
15. Medroxyprogesterone acetate and levonorgestrel increase genital mucosal permeability and enhance susceptibility to genital herpes simplex virus type 2 infection
- Author
-
Quispe Calla, N.E., primary, Vicetti Miguel, R.D., additional, Boyaka, P.N., additional, Hall-Stoodley, L., additional, Kaur, B., additional, Trout, W., additional, Pavelko, S.D., additional, and Cherpes, T.L., additional
- Published
- 2016
- Full Text
- View/download PDF
16. ESCMID guideline for the diagnosis and treatment of biofilm infections 2014
- Author
-
Høiby, N, Bjarnsholt, Thomas, Moser, C, Bassi, G L, Coenye, T, Donelli, G, Hall-Stoodley, L, Holá, V, Imbert, C, Kirketerp-Møller, K, Lebeaux, D, Oliver, A, Ullmann, A J, Williams, C, Høiby, N, Bjarnsholt, Thomas, Moser, C, Bassi, G L, Coenye, T, Donelli, G, Hall-Stoodley, L, Holá, V, Imbert, C, Kirketerp-Møller, K, Lebeaux, D, Oliver, A, Ullmann, A J, and Williams, C
- Abstract
Biofilms cause chronic infections in tissues or by developing on the surfaces of medical devices. Biofilm infections persist despite both antibiotic therapy and the innate and adaptive defence mechanisms of the patient. Biofilm infections are characterized by persisting and progressive pathology due primarily to the inflammatory response surrounding the biofilm. For this reason, many biofilm infections may be difficult to diagnose and treat efficiently. It is the purpose of the guideline to bring the current knowledge of biofilm diagnosis and therapy to the attention of clinical microbiologists and infectious disease specialists. Selected hallmark biofilm infections in tissues (e.g. cystic fibrosis with chronic lung infection, patients with chronic wound infections) or associated with devices (e.g. orthopaedic alloplastic devices, endotracheal tubes, intravenous catheters, indwelling urinary catheters, tissue fillers) are the main focus of the guideline, but experience gained from the biofilm infections included in the guideline may inspire similar work in other biofilm infections. The clinical and laboratory parameters for diagnosing biofilm infections are outlined based on the patient's history, signs and symptoms, microscopic findings, culture-based or culture-independent diagnostic techniques and specific immune responses to identify microorganisms known to cause biofilm infections. First, recommendations are given for the collection of appropriate clinical samples, for reliable methods to specifically detect biofilms, for the evaluation of antibody responses to biofilms, for antibiotic susceptibility testing and for improvement of laboratory reports of biofilm findings in the clinical microbiology laboratory. Second, recommendations are given for the prevention and treatment of biofilm infections and for monitoring treatment effectiveness. Finally, suggestions for future research are given to improve diagnosis and treatment of biofilm infections.
- Published
- 2015
17. Adenoid Reservoir for Pathogenic Biofilm Bacteria
- Author
-
Nistico, L., Kreft, R., Gieseke, A., Coticchia, J., Burrows, A., Khampang, P., Liu, Y., Kerschner, J., Post, J., Lonergan, S., Sampath, R., Hu, F., Ehrlich, G., Stoodley, P., and Hall-Stoodley, L.
- Abstract
Biofilms of pathogenic bacteria are present on the middle ear mucosa of children with chronic otitis media (COM) and may contribute to the persistence of pathogens and the recalcitrance of COM to antibiotic treatment. Controlled studies indicate that adenoidectomy is effective in the treatment of COM, suggesting that the adenoids may act as a reservoir for COM pathogens. To investigate the bacterial community in the adenoid, samples were obtained from 35 children undergoing adenoidectomy for chronic OM or obstructive sleep apnea. We used a novel, culture-independent molecular diagnostic methodology, followed by confocal microscopy, to investigate the in situ distribution and organization of pathogens in the adenoids to determine whether pathogenic bacteria exhibited criteria characteristic of biofilms. The Ibis T5000 Universal Biosensor System was used to interrogate the extent of the microbial diversity within adenoid biopsy specimens. Using a suite of 16 broad-range bacterial primers, we demonstrated that adenoids from both diagnostic groups were colonized with polymicrobial biofilms. Haemophilus influenzae was present in more adenoids from the COM group (P = 0.005), but there was no significant difference between the two patient groups for Streptococcus pneumoniae or Staphylococcus aureus. Fluorescence in situ hybridization, lectin binding, and the use of antibodies specific for host epithelial cells demonstrated that pathogens were aggregated, surrounded by a carbohydrate matrix, and localized on and within the epithelial cell surface, which is consistent with criteria for bacterial biofilms.
- Published
- 2011
18. The role of bacterial biofilms in infections of catheters and shunts
- Author
-
Thomsen, Trine Rolighed, Hall-Stoodley, L, Moser, Claus Ernst, Stoodley, P, Thomsen, Trine Rolighed, Hall-Stoodley, L, Moser, Claus Ernst, and Stoodley, P
- Published
- 2010
19. Nitric oxide-mediated dispersal and enhanced antibiotic sensitivity in pseudomonas aeruginosa biofilms from the cystic fibrosis lung
- Author
-
Howlin, R., primary, Cathie, K., additional, Hall-Stoodley, L., additional, Niehaus, L., additional, Connett, G., additional, Legg, J., additional, Daniels, T., additional, Carroll, M., additional, Jefferies, J., additional, Clarke, S. C., additional, Stoodley, P., additional, Webb, J., additional, and Faust, S. N., additional
- Published
- 2011
- Full Text
- View/download PDF
20. Adenoid Reservoir for Pathogenic Biofilm Bacteria
- Author
-
Nistico, L., primary, Kreft, R., additional, Gieseke, A., additional, Coticchia, J. M., additional, Burrows, A., additional, Khampang, P., additional, Liu, Y., additional, Kerschner, J. E., additional, Post, J. C., additional, Lonergan, S., additional, Sampath, R., additional, Hu, F. Z., additional, Ehrlich, G. D., additional, Stoodley, P., additional, and Hall-Stoodley, L., additional
- Published
- 2011
- Full Text
- View/download PDF
21. Laser disruption and killing of methicillin-resistant Staphylococcus aureus biofilms.
- Author
-
Krespi YP, Kizhner V, Nistico L, Hall-Stoodley L, and Stoodley P
- Published
- 2011
- Full Text
- View/download PDF
22. Characterization of biofilm matrix, degradation by DNase treatment and evidence of capsule downregulation in Streptococcus pneumoniae clinical isolates
- Author
-
Johnson Candice, Mershon William J, Nguyen Duc, Dice Bethany, Sambanthamoorthy Karthik, Nistico Laura, Hall-Stoodley Luanne, Ze Hu Fen, Stoodley Paul, Ehrlich Garth D, and Post J Christopher
- Subjects
Microbiology ,QR1-502 - Abstract
Abstract Background Streptococcus pneumoniae is a common respiratory pathogen and a major causative agent of respiratory infections, including otitis media (OM). Pneumococcal biofilms have been demonstrated on biopsies of the middle ear mucosa in children receiving tympanostomy tubes, supporting the hypothesis that chronic OM may involve biofilm development by pathogenic bacteria as part of the infectious process. To better understand pneumococcal biofilm formation six low-passage encapsulated nasopharyngeal isolates of S. pneumoniae were assessed over a six-eight day period in vitro. Results Multiparametric analysis divided the strains into two groups. Those with a high biofilm forming index (BFI) were structurally complex, exhibited greater lectin colocalization and were more resistant to azithromycin. Those with a low BFI developed less extensive biofilms and were more susceptible to azithromycin. dsDNA was present in the S. pneumoniae biofilm matrix in all strains and treatment with DNase I significantly reduced biofilm biomass. Since capsule expression has been hypothesized to be associated with decreased biofilm development, we also examined expression of cpsA, the first gene in the pneumococcal capsule operon. Interestingly, cpsA was downregulated in biofilms in both high and low BFI strains. Conclusion All pneumococcal strains developed biofilms that exhibited extracellular dsDNA in the biofilm matrix, however strains with a high BFI correlated with greater carbohydrate-associated structural complexity and antibiotic resistance. Furthermore, all strains of S. pneumoniae showed downregulation of the cpsA gene during biofilm growth compared to planktonic culture, regardless of BFI ranking, suggesting downregulation of capsule expression occurs generally during adherent growth.
- Published
- 2008
- Full Text
- View/download PDF
23. Disruption of nontuberculous mycobacteria biofilms induces a highly vulnerable to antibiotic killing phenotype.
- Author
-
Kurbatfinski N, Hill PJ, Tobin N, Kramer CN, Wickham J, Goodman SD, Hall-Stoodley L, and Bakaletz LO
- Abstract
Objectives: Structural or mucus hypersecretory pulmonary diseases such as cystic fibrosis (CF), wherein viscous mucus accumulates and clearance functions are impaired, predispose people to lung infection by inhaled bacteria that form biofilm aggregates. Nontuberculous mycobacteria (NTM), primarily Mycobacterium abscessus and Mycobacterium avium, are the growing cause of these lung infections and are extremely challenging to treat due to antibiotic recalcitrance. Better therapeutic approaches are urgently needed. We developed a humanized monoclonal antibody (HuTipMab) directed against a biofilm structural linchpin, the bacterial DNABII proteins, that rapidly disrupts biofilms and generates highly vulnerable newly released bacteria (NRel)., Methods: HuTipMab's ability to recognize HupB, NTM's DNABII homologue was determined by ELISA. Relative ability of HuTipMab to disrupt biofilms formed by lab-passaged and clinical isolates of NTM was assessed by CLSM. Relative sensitivity of NTM NRel to antibiotic killing compared to when grown planktonically was evaluated by plate count., Results: HuTipMab recognized HupB and significantly disrupted NTM biofilms in a time- and dose-dependent manner. Importantly, NTM NRel of lab-passaged and clinical isolates were now highly sensitive to killing by amikacin and azithromycin., Conclusions: If successful, this combinatorial treatment strategy would empower existing antibiotics to more effectively kill NTM newly released from a biofilm by HuTipMab and thereby both improve clinical outcomes and perhaps decrease length of antibiotic treatment for people that are NTM culture-positive., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Lauren O. Bakaletz reports financial support was provided by 10.13039/100000897Cystic Fibrosis Foundation. Steven D Goodman reports financial support was provided by 10.13039/100000897Cystic Fibrosis Foundation. Luanne Hall-Stoodley reports financial support was provided by 10.13039/100000897Cystic Fibrosis Foundation. L.O.B. and S.D.G. are inventors of technology related to the DNABII-directed approach, rights to which have been licensed to Clarametyx Biosciences, Inc., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
24. The Many Hosts of Mycobacteria 9 (MHM9): A conference report.
- Author
-
Klever AM, Alexander KA, Almeida D, Anderson MZ, Ball RL, Beamer G, Boggiatto P, Buikstra JE, Chandler B, Claeys TA, Concha AE, Converse PJ, Derbyshire KM, Dobos KM, Dupnik KM, Endsley JJ, Endsley MA, Fennelly K, Franco-Paredes C, Hagge DA, Hall-Stoodley L, Hayes D Jr, Hirschfeld K, Hofman CA, Honda JR, Hull NM, Kramnik I, Lacourciere K, Lahiri R, Lamont EA, Larsen MH, Lemaire T, Lesellier S, Lee NR, Lowry CA, Mahfooz NS, McMichael TM, Merling MR, Miller MA, Nagajyothi JF, Nelson E, Nuermberger EL, Pena MT, Perea C, Podell BK, Pyle CJ, Quinn FD, Rajaram MVS, Mejia OR, Rothoff M, Sago SA, Salvador LCM, Simonson AW, Spencer JS, Sreevatsan S, Subbian S, Sunstrum J, Tobin DM, Vijayan KKV, Wright CTO, and Robinson RT
- Subjects
- Animals, Cattle, Humans, Nontuberculous Mycobacteria, Coinfection, Mycobacterium Infections, Nontuberculous microbiology, Mycobacterium tuberculosis, Tuberculosis, Bovine
- Abstract
The Many Hosts of Mycobacteria (MHM) meeting series brings together basic scientists, clinicians and veterinarians to promote robust discussion and dissemination of recent advances in our knowledge of numerous mycobacterial diseases, including human and bovine tuberculosis (TB), nontuberculous mycobacteria (NTM) infection, Hansen's disease (leprosy), Buruli ulcer and Johne's disease. The 9th MHM conference (MHM9) was held in July 2022 at The Ohio State University (OSU) and centered around the theme of "Confounders of Mycobacterial Disease." Confounders can and often do drive the transmission of mycobacterial diseases, as well as impact surveillance and treatment outcomes. Various confounders were presented and discussed at MHM9 including those that originate from the host (comorbidities and coinfections) as well as those arising from the environment (e.g., zoonotic exposures), economic inequality (e.g. healthcare disparities), stigma (a confounder of leprosy and TB for millennia), and historical neglect (a confounder in Native American Nations). This conference report summarizes select talks given at MHM9 highlighting recent research advances, as well as talks regarding the historic and ongoing impact of TB and other infectious diseases on Native American Nations, including those in Southwestern Alaska where the regional TB incidence rate is among the highest in the Western hemisphere., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
25. Interferon-induced transmembrane protein 3 (IFITM3) limits lethality of SARS-CoV-2 in mice.
- Author
-
Kenney AD, Zani A, Kawahara J, Eddy AC, Wang XL, Mahesh KC, Lu M, Thomas J, Kohlmeier JE, Suthar MS, Hemann EA, Li J, Peeples ME, Hall-Stoodley L, Forero A, Cai C, Ma J, and Yount JS
- Subjects
- Animals, Mice, Interferons genetics, Lung, Mice, Knockout, COVID-19 genetics, SARS-CoV-2
- Abstract
Interferon-induced transmembrane protein 3 (IFITM3) is an antiviral protein that alters cell membranes to block fusion of viruses. Conflicting reports identified opposing effects of IFITM3 on SARS-CoV-2 infection of cells, and its impact on viral pathogenesis in vivo remains unclear. Here, we show that IFITM3 knockout (KO) mice infected with SARS-CoV-2 experience extreme weight loss and lethality compared to mild infection in wild-type (WT) mice. KO mice have higher lung viral titers and increases in inflammatory cytokine levels, immune cell infiltration, and histopathology. Mechanistically, we observe disseminated viral antigen staining throughout the lung and pulmonary vasculature in KO mice, as well as increased heart infection, indicating that IFITM3 constrains dissemination of SARS-CoV-2. Global transcriptomic analysis of infected lungs shows upregulation of gene signatures associated with interferons, inflammation, and angiogenesis in KO versus WT animals, highlighting changes in lung gene expression programs that precede severe lung pathology and fatality. Our results establish IFITM3 KO mice as a new animal model for studying severe SARS-CoV-2 infection and overall demonstrate that IFITM3 is protective in SARS-CoV-2 infections in vivo., (© 2023 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
26. Cystic fibrosis macrophage function and clinical outcomes after elexacaftor/tezacaftor/ivacaftor.
- Author
-
Zhang S, Shrestha CL, Robledo-Avila F, Jaganathan D, Wisniewski BL, Brown N, Pham H, Carey K, Amer AO, Hall-Stoodley L, McCoy KS, Bai S, Partida-Sanchez S, and Kopp BT
- Subjects
- Humans, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Chlorides metabolism, Chloride Channel Agonists therapeutic use, Mutation, Macrophages metabolism, Cystic Fibrosis drug therapy
- Abstract
Background: Abnormal macrophage function caused by dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) is a critical contributor to chronic airway infections and inflammation in people with cystic fibrosis (PWCF). Elexacaftor/tezacaftor/ivacaftor (ETI) is a new CFTR modulator therapy for PWCF. Host-pathogen and clinical responses to CFTR modulators are poorly described. We sought to determine how ETI impacts macrophage CFTR function, resulting effector functions and relationships to clinical outcome changes., Methods: Clinical information and/or biospecimens were obtained at ETI initiation and 3, 6, 9 and 12 months post-ETI in 56 PWCF and compared with non-CF controls. Peripheral blood monocyte-derived macrophages (MDMs) were isolated and functional assays performed., Results: ETI treatment was associated with increased CF MDM CFTR expression, function and localisation to the plasma membrane. CF MDM phagocytosis, intracellular killing of CF pathogens and efferocytosis of apoptotic neutrophils were partially restored by ETI, but inflammatory cytokine production remained unchanged. Clinical outcomes including increased forced expiratory volume in 1 s (+10%) and body mass index (+1.0 kg·m
-2 ) showed fluctuations over time and were highly individualised. Significant correlations between post-ETI MDM CFTR function and sweat chloride levels were observed. However, MDM CFTR function correlated with clinical outcomes better than sweat chloride., Conclusion: ETI is associated with unique changes in innate immune function and clinical outcomes., Competing Interests: Conflict of interest: The authors declare that no conflicts of interest exists., (Copyright ©The authors 2023. For reproduction rights and permissions contact permissions@ersnet.org.)- Published
- 2023
- Full Text
- View/download PDF
27. Surface Dependent Inhibition of Mycobacterium abscessus by Diverse Pseudomonas aeruginosa Strains.
- Author
-
Idosa AW, Wozniak DJ, and Hall-Stoodley L
- Subjects
- Humans, Pseudomonas aeruginosa genetics, Anti-Bacterial Agents pharmacology, Quorum Sensing, Lung microbiology, Biofilms, Mycobacterium abscessus, Cystic Fibrosis microbiology, Pseudomonas Infections microbiology
- Abstract
Both Pseudomonas aeruginosa and Mycobacterium abscessus are bacteria that cause pulmonary infection in people with inflammatory lung disease, including individuals with cystic fibrosis (CF). These bacterial species inhabit the same environmental reservoirs (soil and water) and can be coisolated in the lungs of people with CF. We investigated the interaction of these bacteria and found an antagonistic interaction favoring P. aeruginosa that was observed in biofilms but not in planktonic cultures. This antagonism extended to multiple P. aeruginosa strains and against Mycobacterium smegmatis. We tested known P. aeruginosa mutants for genes that can play roles in interbacterial contact-dependent (type III and type VI secretion systems) and contact-independent (quorum sensing, type II secretion) antagonism pathways to interrogate the mechanism of action. Our results indicate that well-known mechanisms of interbacterial competition are not responsible for the antagonism of P. aeruginosa toward M. abscessus, suggesting a novel antibacterial strategy. IMPORTANCE The biofilm lifestyle is favored by many organisms, and understanding interbacterial interactions that occur between coisolated bacterial species can provide new information regarding bacterial defense mechanisms and antibacterial targets. This may also provide insights into possible interbacterial interactions impacting host immunity during coinfection. Here, we investigate an antagonistic interaction favoring P. aeruginosa over M. abscessus exclusively in dual-species biofilms and not in liquid coculture.
- Published
- 2022
- Full Text
- View/download PDF
28. The biofilm life cycle: expanding the conceptual model of biofilm formation.
- Author
-
Sauer K, Stoodley P, Goeres DM, Hall-Stoodley L, Burmølle M, Stewart PS, and Bjarnsholt T
- Subjects
- Animals, Life Cycle Stages, Biofilms, Pseudomonas aeruginosa physiology
- Abstract
Bacterial biofilms are often defined as communities of surface-attached bacteria and are typically depicted with a classic mushroom-shaped structure characteristic of Pseudomonas aeruginosa. However, it has become evident that this is not how all biofilms develop, especially in vivo, in clinical and industrial settings, and in the environment, where biofilms often are observed as non-surface-attached aggregates. In this Review, we describe the origin of the current five-step biofilm development model and why it fails to capture many aspects of bacterial biofilm physiology. We aim to present a simplistic developmental model for biofilm formation that is flexible enough to include all the diverse scenarios and microenvironments where biofilms are formed. With this new expanded, inclusive model, we hereby introduce a common platform for developing an understanding of biofilms and anti-biofilm strategies that can be tailored to the microenvironment under investigation., (© 2022. Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
29. Biofilm aggregates and the host airway-microbial interface.
- Author
-
Hall-Stoodley L and McCoy KS
- Subjects
- Bacteria, Biofilms, Haemophilus influenzae physiology, Humans, Pseudomonas aeruginosa physiology, Respiratory System pathology, Cystic Fibrosis microbiology, Pseudomonas Infections
- Abstract
Biofilms are multicellular microbial aggregates that can be associated with host mucosal epithelia in the airway, gut, and genitourinary tract. The host environment plays a critical role in the establishment of these microbial communities in both health and disease. These host mucosal microenvironments however are distinct histologically, functionally, and regarding nutrient availability. This review discusses the specific mucosal epithelial microenvironments lining the airway, focusing on: i) biofilms in the human respiratory tract and the unique airway microenvironments that make it exquisitely suited to defend against infection, and ii) how airway pathophysiology and dysfunctional barrier/clearance mechanisms due to genetic mutations, damage, and inflammation contribute to biofilm infections. The host cellular responses to infection that contribute to resolution or exacerbation, and insights about evaluating and therapeutically targeting airway-associated biofilm infections are briefly discussed. Since so many studies have focused on Pseudomonas aeruginosa in the context of cystic fibrosis (CF) or on Haemophilus influenzae in the context of upper and lower respiratory diseases, these bacteria are used as examples. However, there are notable differences in diseased airway microenvironments and the unique pathophysiology specific to the bacterial pathogens themselves., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Hall-Stoodley and McCoy.)
- Published
- 2022
- Full Text
- View/download PDF
30. Tissue-localized immune responses in people with cystic fibrosis and respiratory nontuberculous mycobacteria infection.
- Author
-
Hayes D Jr, Shukla RK, Cheng Y, Gecili E, Merling MR, Szczesniak RD, Ziady AG, Woods JC, Hall-Stoodley L, Liyanage NP, and Robinson RT
- Subjects
- Adult, Humans, Immunity, Nontuberculous Mycobacteria, Cystic Fibrosis complications, Mycobacterium Infections, Nontuberculous complications, Mycobacterium Infections, Nontuberculous microbiology
- Abstract
Nontuberculous mycobacteria (NTM) are an increasingly common cause of respiratory infection in people with cystic fibrosis (PwCF). Relative to those with no history of NTM infection (CF-NTMNEG), PwCF and a history of NTM infection (CF-NTMPOS) are more likely to develop severe lung disease and experience complications over the course of treatment. In other mycobacterial infections (e.g., tuberculosis), an overexuberant immune response causes pathology and compromises organ function; however, since the immune profiles of CF-NTMPOS and CF-NTMNEG airways are largely unexplored, it is unknown which, if any, immune responses distinguish these cohorts or concentrate in damaged tissues. Here, we evaluated lung lobe-specific immune profiles of 3 cohorts (CF-NTMPOS, CF-NTMNEG, and non-CF adults) and found that CF-NTMPOS airways are distinguished by a hyperinflammatory cytokine profile. Importantly, the CF-NTMPOS airway immune profile was dominated by B cells, classical macrophages, and the cytokines that support their accumulation. These and other immunological differences between cohorts, including the near absence of NK cells and complement pathway members, were enriched in the most damaged lung lobes. The implications of these findings for our understanding of lung disease in PwCF are discussed, as are how they may inform the development of host-directed therapies to improve NTM disease treatment.
- Published
- 2022
- Full Text
- View/download PDF
31. Caspase-4/11 exacerbates disease severity in SARS-CoV-2 infection by promoting inflammation and immunothrombosis.
- Author
-
Eltobgy MM, Zani A, Kenney AD, Estfanous S, Kim E, Badr A, Carafice C, Daily K, Whitham O, Pietrzak M, Webb A, Kawahara J, Eddy AC, Denz P, Lu M, Kc M, Peeples ME, Li J, Zhu J, Que J, Robinson R, Rosas Mejia O, Rayner RE, Hall-Stoodley L, Seveau S, Gavrilin MA, Zhang X, Thomas J, Kohlmeier JE, Suthar MS, Oltz E, Tedeschi A, Robledo-Avila FH, Partida-Sanchez S, Hemann EA, Abdelrazik E, Forero A, Nimjee SM, Boyaka PN, Cormet-Boyaka E, Yount JS, and Amer AO
- Subjects
- Animals, Caspases, Initiator genetics, Disease Progression, Humans, Lung pathology, Mice, Mice, Knockout, Severity of Illness Index, COVID-19 enzymology, COVID-19 pathology, Caspases, Initiator metabolism, SARS-CoV-2, Thromboinflammation enzymology, Thromboinflammation genetics
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS–CoV-2) is a worldwide health concern, and new treatment strategies are needed. Targeting inflammatory innate immunity pathways holds therapeutic promise, but effective molecular targets remain elusive. Here, we show that human caspase-4 (CASP4) and its mouse homolog, caspase-11 (CASP11), are up-regulated in SARS–CoV-2 infections and that CASP4 expression correlates with severity of SARS–CoV-2 infection in humans. SARS–CoV-2–infected Casp11−/− mice were protected from severe weight loss and lung pathology, including blood vessel damage, compared to wild-type (WT) mice and mice lacking the caspase downstream effector gasdermin-D (Gsdmd−/−). Notably, viral titers were similar regardless of CASP11 knockout. Global transcriptomics of SARS–CoV-2–infected WT, Casp11−/−, and Gsdmd−/− lungs identified restrained expression of inflammatory molecules and altered neutrophil gene signatures in Casp11−/− mice. We confirmed that protein levels of inflammatory mediators interleukin (IL)-1β, IL-6, and CXCL1, as well as neutrophil functions, were reduced in Casp11−/− lungs. Additionally, Casp11−/− lungs accumulated less von Willebrand factor, a marker for endothelial damage, but expressed more Kruppel-Like Factor 2, a transcription factor that maintains vascular integrity. Overall, our results demonstrate that CASP4/11 promotes detrimental SARS–CoV-2–induced inflammation and coagulopathy, largely independently of GSDMD, identifying CASP4/11 as a promising drug target for treatment and prevention of severe COVID-19.
- Published
- 2022
- Full Text
- View/download PDF
32. Interferon-induced transmembrane protein 3 (IFITM3) limits lethality of SARS-CoV-2 in mice.
- Author
-
Zani A, Kenney AD, Kawahara J, Eddy AC, Wang XL, Kc M, Lu M, Hemann EA, Li J, Peeples ME, Hall-Stoodley L, Forero A, Cai C, Ma J, and Yount JS
- Abstract
Interferon-induced transmembrane protein 3 (IFITM3) is a host antiviral protein that alters cell membranes to block fusion of viruses. Published reports have identified conflicting pro- and antiviral effects of IFITM3 on SARS-CoV-2 in cultured cells, and its impact on viral pathogenesis in vivo remains unclear. Here, we show that IFITM3 knockout (KO) mice infected with mouse-adapted SARS-CoV-2 experienced extreme weight loss and lethality, while wild type (WT) mice lost minimal weight and recovered. KO mice had higher lung viral titers and increases in lung inflammatory cytokine levels, CD45-positive immune cell infiltration, and histopathology, compared to WT mice. Mechanistically, we observed disseminated viral antigen staining throughout the lung tissue and pulmonary vasculature in KO mice, while staining was observed in confined regions in WT lungs. Global transcriptomic analysis of infected lungs identified upregulation of gene signatures associated with interferons, inflammation, and angiogenesis in KO versus WT animals, highlighting changes in lung gene expression programs that precede severe lung pathology and fatality. Corroborating the protective effect of IFITM3 in vivo , K18-hACE2/IFITM3 KO mice infected with non-adapted SARS-CoV-2 showed enhanced, rapid weight loss and early death compared to control mice. Increased heart infection was observed in both mouse models in the absence of IFITM3, indicating that IFITM3 constrains extrapulmonary dissemination of SARS-CoV-2. Our results establish IFITM3 KO mice as a new animal model for studying severe SARS-CoV-2 infection of the lung and cardiovascular system, and overall demonstrate that IFITM3 is protective in SARS-CoV-2 infections of mice.
- Published
- 2021
- Full Text
- View/download PDF
33. Mycobacterium abscessus biofilms have viscoelastic properties which may contribute to their recalcitrance in chronic pulmonary infections.
- Author
-
Gloag ES, Wozniak DJ, Stoodley P, and Hall-Stoodley L
- Subjects
- Cell Wall ultrastructure, Elasticity, Glycopeptides chemistry, Glycopeptides isolation & purification, Humans, Lipopeptides chemistry, Lipopeptides isolation & purification, Mycobacterium abscessus ultrastructure, Pseudomonas aeruginosa chemistry, Pseudomonas aeruginosa ultrastructure, Rheology, Shear Strength, Viscosity, Biofilms growth & development, Biomechanical Phenomena physiology, Cell Wall chemistry, Mycobacterium abscessus chemistry
- Abstract
Mycobacterium abscessus is emerging as a cause of recalcitrant chronic pulmonary infections, particularly in people with cystic fibrosis (CF). Biofilm formation has been implicated in the pathology of this organism, however the role of biofilm formation in infection is unclear. Two colony-variants of M. abscessus are routinely isolated from CF samples, smooth (Ma
Sm ) and rough (MaRg ). These two variants display distinct colony morphologies due to the presence (MaSm ) or absence (MaRg ) of cell wall glycopeptidolipids (GPLs). We hypothesized that MaSm and MaRg variant biofilms might have different mechanical properties. To test this hypothesis, we performed uniaxial mechanical indentation, and shear rheometry on MaSm and MaRg colony-biofilms. We identified that MaRg biofilms were significantly stiffer than MaSm under a normal force, while MaSm biofilms were more pliant compared to MaRg , under both normal and shear forces. Furthermore, using theoretical indices of mucociliary and cough clearance, we identified that M. abscessus biofilms may be more resistant to mechanical forms of clearance from the lung, compared to another common pulmonary pathogen, Pseudomonas aeruginosa. Thus, the mechanical properties of M. abscessus biofilms may contribute to the persistent nature of pulmonary infections caused by this organism.- Published
- 2021
- Full Text
- View/download PDF
34. Opposing activities of IFITM proteins in SARS-CoV-2 infection.
- Author
-
Shi G, Kenney AD, Kudryashova E, Zani A, Zhang L, Lai KK, Hall-Stoodley L, Robinson RT, Kudryashov DS, Compton AA, and Yount JS
- Subjects
- Angiotensin-Converting Enzyme 2 genetics, Animals, Cell Line, Chlorocebus aethiops, Humans, Mice, Mutation, Serine Endopeptidases, Virus Internalization, Antigens, Differentiation genetics, COVID-19 genetics, Membrane Proteins genetics, RNA-Binding Proteins genetics, SARS-CoV-2 physiology
- Abstract
Interferon-induced transmembrane proteins (IFITMs) restrict infections by many viruses, but a subset of IFITMs enhance infections by specific coronaviruses through currently unknown mechanisms. We show that SARS-CoV-2 Spike-pseudotyped virus and genuine SARS-CoV-2 infections are generally restricted by human and mouse IFITM1, IFITM2, and IFITM3, using gain- and loss-of-function approaches. Mechanistically, SARS-CoV-2 restriction occurred independently of IFITM3 S-palmitoylation, indicating a restrictive capacity distinct from reported inhibition of other viruses. In contrast, the IFITM3 amphipathic helix and its amphipathic properties were required for virus restriction. Mutation of residues within the IFITM3 endocytosis-promoting YxxФ motif converted human IFITM3 into an enhancer of SARS-CoV-2 infection, and cell-to-cell fusion assays confirmed the ability of endocytic mutants to enhance Spike-mediated fusion with the plasma membrane. Overexpression of TMPRSS2, which increases plasma membrane fusion versus endosome fusion of SARS-CoV-2, attenuated IFITM3 restriction and converted amphipathic helix mutants into infection enhancers. In sum, we uncover new pro- and anti-viral mechanisms of IFITM3, with clear distinctions drawn between enhancement of viral infection at the plasma membrane and amphipathicity-based mechanisms used for endosomal SARS-CoV-2 restriction., (© 2020 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
35. Neutralizing antibody against SARS-CoV-2 spike in COVID-19 patients, health care workers, and convalescent plasma donors.
- Author
-
Zeng C, Evans JP, Pearson R, Qu P, Zheng YM, Robinson RT, Hall-Stoodley L, Yount J, Pannu S, Mallampalli RK, Saif L, Oltz E, Lozanski G, and Liu SL
- Subjects
- COVID-19 virology, Cohort Studies, Humans, Immunization, Passive, SARS-CoV-2 immunology, COVID-19 Serotherapy, Antibodies, Neutralizing immunology, COVID-19 immunology, COVID-19 therapy, SARS-CoV-2 isolation & purification, Spike Glycoprotein, Coronavirus immunology
- Abstract
Rapid and specific antibody testing is crucial for improved understanding, control, and treatment of COVID-19 pathogenesis. Herein, we describe and apply a rapid, sensitive, and accurate virus neutralization assay for SARS-CoV-2 antibodies. The assay is based on an HIV-1 lentiviral vector that contains a secreted intron Gaussia luciferase (Gluc) or secreted nano-luciferase reporter cassette, pseudotyped with the SARS-CoV-2 spike (S) glycoprotein, and is validated with a plaque-reduction assay using an authentic, infectious SARS-CoV-2 strain. The assay was used to evaluate SARS-CoV-2 antibodies in serum from individuals with a broad range of COVID-19 symptoms; patients included those in the intensive care unit (ICU), health care workers (HCWs), and convalescent plasma donors. The highest neutralizing antibody titers were observed among ICU patients, followed by general hospitalized patients, HCWs, and convalescent plasma donors. Our study highlights a wide phenotypic variation in human antibody responses against SARS-CoV-2 and demonstrates the efficacy of a potentially novel lentivirus pseudotype assay for high-throughput serological surveys of neutralizing antibody titers in large cohorts.
- Published
- 2020
- Full Text
- View/download PDF
36. Opposing activities of IFITM proteins in SARS-CoV-2 infection.
- Author
-
Shi G, Kenney AD, Kudryashova E, Zhang L, Hall-Stoodley L, Robinson RT, Kudryashov DS, Compton AA, and Yount JS
- Abstract
Interferon-induced transmembrane proteins (IFITMs) restrict infections by many viruses, but a subset of IFITMs enhance infections by specific coronaviruses through currently unknown mechanisms. Here we show that SARS-CoV-2 Spike-pseudotyped virus and genuine SARS-CoV-2 infections are generally restricted by expression of human IFITM1, IFITM2, and IFITM3, using both gain- and loss-of-function approaches. Mechanistically, restriction of SARS-CoV-2 occurred independently of IFITM3 S -palmitoylation sites, indicating a restrictive capacity that is distinct from reported inhibition of other viruses. In contrast, the IFITM3 amphipathic helix and its amphipathic properties were required for virus restriction. Mutation of residues within the human IFITM3 endocytosis-promoting YxxΦ motif converted human IFITM3 into an enhancer of SARS-CoV-2 infection, and cell-to-cell fusion assays confirmed the ability of endocytic mutants to enhance Spike-mediated fusion with the plasma membrane. Overexpression of TMPRSS2, which reportedly increases plasma membrane fusion versus endosome fusion of SARS-CoV-2, attenuated IFITM3 restriction and converted amphipathic helix mutants into strong enhancers of infection. In sum, these data uncover new pro- and anti-viral mechanisms of IFITM3, with clear distinctions drawn between enhancement of viral infection at the plasma membrane and amphipathicity-based mechanisms used for endosomal virus restriction. Indeed, the net effect of IFITM3 on SARS-CoV-2 infections may be a result of these opposing activities, suggesting that shifts in the balance of these activities could be coopted by viruses to escape this important first line innate defense mechanism.
- Published
- 2020
- Full Text
- View/download PDF
37. Neutralizing antibody against SARS-CoV-2 spike in COVID-19 patients, health care workers and convalescent plasma donors: a cohort study using a rapid and sensitive high-throughput neutralization assay.
- Author
-
Zeng C, Evans JP, Pearson R, Qu P, Zheng YM, Robinson RT, Hall-Stoodley L, Yount J, Pannu S, Mallampalli RK, Saif L, Oltz E, Lozanski G, and Liu SL
- Abstract
Rapid and specific antibody testing is crucial for improved understanding, control, and treatment of COVID-19 pathogenesis. Herein, we describe and apply a rapid, sensitive, and accurate virus neutralization assay for SARS-CoV-2 antibodies. The new assay is based on an HIV-1 lentiviral vector that contains a secreted intron Gaussia luciferase or secreted Nano-luciferase reporter cassette, pseudotyped with the SARS-CoV-2 spike (S) glycoprotein, and is validated with a plaque reduction assay using an authentic, infectious SARS-CoV-2 strain. The new assay was used to evaluate SARS-CoV-2 antibodies in serum from individuals with a broad range of COVID-19 symptoms, including intensive care unit (ICU) patients, health care workers (HCWs), and convalescent plasma donors. The highest neutralizing antibody titers were observed among ICU patients, followed by general hospitalized patients, HCWs and convalescent plasma donors. Our study highlights a wide phenotypic variation in human antibody responses against SARS-CoV-2, and demonstrates the efficacy of a novel lentivirus pseudotype assay for high-throughput serological surveys of neutralizing antibody titers in large cohorts.
- Published
- 2020
- Full Text
- View/download PDF
38. The Many Hosts of Mycobacteria 8 (MHM8): A conference report.
- Author
-
Larsen MH, Lacourciere K, Parker TM, Kraigsley A, Achkar JM, Adams LB, Dupnik KM, Hall-Stoodley L, Hartman T, Kanipe C, Kurtz SL, Miller MA, Salvador LCM, Spencer JS, and Robinson RT
- Subjects
- Animals, Congresses as Topic, Diffusion of Innovation, Disease Models, Animal, Host-Pathogen Interactions, Humans, Mycobacterium genetics, Mycobacterium Infections, Nontuberculous diagnosis, Mycobacterium Infections, Nontuberculous epidemiology, Tuberculosis diagnosis, Tuberculosis epidemiology, Bacteriology, Biomedical Research, Infectious Disease Medicine, Mycobacterium pathogenicity, Mycobacterium Infections, Nontuberculous microbiology, Tuberculosis microbiology
- Abstract
Mycobacteria are important causes of disease in human and animal hosts. Diseases caused by mycobacteria include leprosy, tuberculosis (TB), nontuberculous mycobacteria (NTM) infections and Buruli Ulcer. To better understand and treat mycobacterial disease, clinicians, veterinarians and scientists use a range of discipline-specific approaches to conduct basic and applied research, including conducting epidemiological surveys, patient studies, wildlife sampling, animal models, genetic studies and computational simulations. To foster the exchange of knowledge and collaboration across disciplines, the Many Hosts of Mycobacteria (MHM) conference series brings together clinical, veterinary and basic scientists who are dedicated to advancing mycobacterial disease research. Started in 2007, the MHM series recently held its 8th conference at the Albert Einstein College of Medicine (Bronx, NY). Here, we review the diseases discussed at MHM8 and summarize the presentations on research advances in leprosy, NTM and Buruli Ulcer, human and animal TB, mycobacterial disease comorbidities, mycobacterial genetics and 'omics, and animal models. A mouse models workshop, which was held immediately after MHM8, is also summarized. In addition to being a resource for those who were unable to attend MHM8, we anticipate this review will provide a benchmark to gauge the progress of future research concerning mycobacteria and their many hosts., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. The expression of Mirc1/Mir17-92 cluster in sputum samples correlates with pulmonary exacerbations in cystic fibrosis patients.
- Author
-
Krause K, Kopp BT, Tazi MF, Caution K, Hamilton K, Badr A, Shrestha C, Tumin D, Hayes D Jr, Robledo-Avila F, Hall-Stoodley L, Klamer BG, Zhang X, Partida-Sanchez S, Parinandi NL, Kirkby SE, Dakhlallah D, McCoy KS, Cormet-Boyaka E, and Amer AO
- Subjects
- Adolescent, Adult, Biomarkers metabolism, Chloride Channel Agonists administration & dosage, Correlation of Data, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Disease Progression, Drug Combinations, Drug Monitoring methods, Female, Gene Expression Profiling, Humans, Male, RNA, Long Noncoding, Respiratory Function Tests methods, Sputum metabolism, Aminophenols administration & dosage, Aminopyridines administration & dosage, Benzodioxoles administration & dosage, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Cystic Fibrosis metabolism, Cystic Fibrosis physiopathology, MicroRNAs metabolism, Nerve Tissue Proteins metabolism, Quinolones administration & dosage, Respiratory System drug effects, Respiratory System metabolism, Respiratory System physiopathology
- Abstract
Introduction: Cystic fibrosis (CF) is a multi-organ disorder characterized by chronic sino-pulmonary infections and inflammation. Many patients with CF suffer from repeated pulmonary exacerbations that are predictors of worsened long-term morbidity and mortality. There are no reliable markers that associate with the onset or progression of an exacerbation or pulmonary deterioration. Previously, we found that the Mirc1/Mir17-92a cluster which is comprised of 6 microRNAs (Mirs) is highly expressed in CF mice and negatively regulates autophagy which in turn improves CF transmembrane conductance regulator (CFTR) function. Therefore, here we sought to examine the expression of individual Mirs within the Mirc1/Mir17-92 cluster in human cells and biological fluids and determine their role as biomarkers of pulmonary exacerbations and response to treatment., Methods: Mirc1/Mir17-92 cluster expression was measured in human CF and non-CF plasma, blood-derived neutrophils, and sputum samples. Values were correlated with pulmonary function, exacerbations and use of CFTR modulators., Results: Mirc1/Mir17-92 cluster expression was not significantly elevated in CF neutrophils nor plasma when compared to the non-CF cohort. Cluster expression in CF sputum was significantly higher than its expression in plasma. Elevated CF sputum Mirc1/Mir17-92 cluster expression positively correlated with pulmonary exacerbations and negatively correlated with lung function. Patients with CF undergoing treatment with the CFTR modulator Ivacaftor/Lumacaftor did not demonstrate significant change in the expression Mirc1/Mir17-92 cluster after six months of treatment., Conclusions: Mirc1/Mir17-92 cluster expression is a promising biomarker of respiratory status in patients with CF including pulmonary exacerbation., (Published by Elsevier B.V.)
- Published
- 2018
- Full Text
- View/download PDF
40. Mycobacterium abscessus Smooth and Rough Morphotypes Form Antimicrobial-Tolerant Biofilm Phenotypes but Are Killed by Acetic Acid.
- Author
-
Clary G, Sasindran SJ, Nesbitt N, Mason L, Cole S, Azad A, McCoy K, Schlesinger LS, and Hall-Stoodley L
- Subjects
- Azithromycin pharmacology, Acetic Acid pharmacology, Anti-Bacterial Agents pharmacology, Biofilms drug effects, Mycobacterium abscessus drug effects
- Abstract
Mycobacterium abscessus has emerged as an important pathogen in people with chronic inflammatory lung diseases such as cystic fibrosis, and recent reports suggest that it may be transmissible by fomites. M. abscessus exhibits two major colony morphology variants: a smooth morphotype ( Ma
Sm ) and a rough morphotype ( MaRg ). Biofilm formation, prolonged intracellular survival, and colony variant diversity can each contribute to the persistence of M. abscessus and other bacterial pathogens in chronic pulmonary diseases. A prevailing paradigm of chronic M. abscessus infection is that MaSm is a noninvasive, biofilm-forming, persistent phenotype and MaRg an invasive phenotype that is unable to form biofilms. We show that MaRg is hyperaggregative and forms biofilm-like aggregates, which, like MaSm biofilm aggregates, are significantly more tolerant than planktonic variants to acidic pHs, hydrogen peroxide (H2 O2 ), and treatment with amikacin or azithromycin. We further show that both variants are recalcitrant to antibiotic treatment inside human macrophage-like cells and that MaRg is more refractory than MaSm to azithromycin. Our results indicate that biofilm-like aggregation and protracted intracellular survival may each contribute to the persistence of this problematic pathogen in the face of antimicrobial agents regardless of morphotype. Biofilms of each M. abscessus variant are rapidly killed, however, by acetic acid, which may help to prevent local fomite transmission., (Copyright © 2018 Clary et al.)- Published
- 2018
- Full Text
- View/download PDF
41. Checks and Balances between Autophagy and Inflammasomes during Infection.
- Author
-
Seveau S, Turner J, Gavrilin MA, Torrelles JB, Hall-Stoodley L, Yount JS, and Amer AO
- Subjects
- Animals, Bacteria immunology, Caspases immunology, Humans, Interleukin-18 immunology, Interleukin-1beta immunology, Viruses immunology, Autophagy, Bacterial Infections immunology, Inflammasomes immunology, Virus Diseases immunology
- Abstract
Autophagy and inflammasome complex assembly are physiological processes that control homeostasis, inflammation, and immunity. Autophagy is a ubiquitous pathway that degrades cytosolic macromolecules or organelles, as well as intracellular pathogens. Inflammasomes are multi-protein complexes that assemble in the cytosol of cells upon detection of pathogen- or danger-associated molecular patterns. A critical outcome of inflammasome assembly is the activation of the cysteine protease caspase-1, which activates the pro-inflammatory cytokine precursors pro-IL-1β and pro-IL-18. Studies on chronic inflammatory diseases, heart diseases, Alzheimer's disease, and multiple sclerosis revealed that autophagy and inflammasomes intersect and regulate each other. In the context of infectious diseases, however, less is known about the interplay between autophagy and inflammasome assembly, although it is becoming evident that pathogens have evolved multiple strategies to inhibit and/or subvert these pathways and to take advantage of their intricate crosstalk. An improved appreciation of these pathways and their subversion by diverse pathogens is expected to help in the design of anti-infective therapeutic interventions., (Copyright © 2017. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
42. CASP4/caspase-11 promotes autophagosome formation in response to bacterial infection.
- Author
-
Krause K, Caution K, Badr A, Hamilton K, Saleh A, Patel K, Seveau S, Hall-Stoodley L, Hegazi R, Zhang X, Gavrilin MA, and Amer AO
- Subjects
- Animals, Autophagosomes microbiology, Autophagy immunology, Bacterial Infections genetics, Bacterial Infections metabolism, Burkholderia Infections genetics, Burkholderia Infections immunology, Burkholderia Infections metabolism, Burkholderia cenocepacia metabolism, Caspases genetics, Caspases, Initiator, Cells, Cultured, Escherichia coli immunology, Escherichia coli metabolism, Inflammasomes genetics, Inflammasomes metabolism, Macrophages immunology, Macrophages metabolism, Macrophages microbiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Phagosomes genetics, Phagosomes metabolism, Phagosomes microbiology, Phagosomes pathology, Autophagosomes metabolism, Autophagy genetics, Bacterial Infections immunology, Burkholderia cenocepacia immunology, Caspases physiology
- Abstract
CASP4/caspase-11-dependent inflammasome activation is important for the clearance of various Gram-negative bacteria entering the host cytosol. Additionally, CASP4 modulates the actin cytoskeleton to promote the maturation of phagosomes harboring intracellular pathogens such as Legionella pneumophila but not those enclosing nonpathogenic bacteria. Nevertheless, this non-inflammatory role of CASP4 regarding the trafficking of vacuolar bacteria remains poorly understood. Macroautophagy/autophagy, a catabolic process within eukaryotic cells, is also implicated in the elimination of intracellular pathogens such as Burkholderia cenocepacia. Here we show that CASP4-deficient macrophages exhibit a defect in autophagosome formation in response to B. cenocepacia infection. The absence of CASP4 causes an accumulation of the small GTPase RAB7, reduced colocalization of B. cenocepacia with LC3 and acidic compartments accompanied by increased bacterial replication in vitro and in vivo. Together, our data reveal a novel role of CASP4 in regulating autophagy in response to B. cenocepacia infection.
- Published
- 2018
- Full Text
- View/download PDF
43. Targeting microbial biofilms: current and prospective therapeutic strategies.
- Author
-
Koo H, Allan RN, Howlin RP, Stoodley P, and Hall-Stoodley L
- Subjects
- Anti-Bacterial Agents pharmacology, Bacteria classification, Bacterial Physiological Phenomena, Biofilms growth & development, Drug Resistance, Bacterial
- Abstract
Biofilm formation is a key virulence factor for a wide range of microorganisms that cause chronic infections. The multifactorial nature of biofilm development and drug tolerance imposes great challenges for the use of conventional antimicrobials and indicates the need for multi-targeted or combinatorial therapies. In this Review, we focus on current therapeutic strategies and those under development that target vital structural and functional traits of microbial biofilms and drug tolerance mechanisms, including the extracellular matrix and dormant cells. We emphasize strategies that are supported by in vivo or ex vivo studies, highlight emerging biofilm-targeting technologies and provide a rationale for multi-targeted therapies aimed at disrupting the complex biofilm microenvironment.
- Published
- 2017
- Full Text
- View/download PDF
44. Primary ciliary dyskinesia ciliated airway cells show increased susceptibility to Haemophilus influenzae biofilm formation.
- Author
-
Walker WT, Jackson CL, Allan RN, Collins SA, Kelso MJ, Rineh A, Yepuri NR, Nicholas B, Lau L, Johnston D, Lackie P, Faust SN, Lucas JSA, and Hall-Stoodley L
- Subjects
- Adolescent, Adult, Anti-Bacterial Agents pharmacology, Bacterial Adhesion, Bacterial Proteins metabolism, Biofilms growth & development, Case-Control Studies, Child, Child, Preschool, Cytokines metabolism, Female, Haemophilus influenzae pathogenicity, Haemophilus influenzae physiology, Humans, Kartagener Syndrome physiopathology, Male, Middle Aged, Primary Cell Culture, Young Adult, Epithelial Cells microbiology, Haemophilus Infections physiopathology, Kartagener Syndrome microbiology, Nitric Oxide pharmacology
- Abstract
Non-typeable Haemophilus influenzae (NTHi) is the most common pathogen in primary ciliary dyskinesia (PCD) patients. We hypothesised that abnormal ciliary motility and low airway nitric oxide (NO) levels on airway epithelial cells from PCD patients might be permissive for NTHi colonisation and biofilm development.We used a primary epithelial cell co-culture model to investigate NTHi infection. Primary airway epithelial cells from PCD and non-PCD patients were differentiated to ciliation using an air-liquid interface culture and then co-cultured with NTHi.NTHi adherence was greater on PCD epithelial cells compared to non-PCD cells (p<0.05) and the distribution of NTHi on PCD epithelium showed more aggregated NTHi in biofilms (p<0.001). Apart from defective ciliary motility, PCD cells did not significantly differ from non-PCD epithelial cells in the degree of ciliation and epithelial integrity or in cytokine, LL-37 and NO production. Treatment of PCD epithelia using exogenous NO and antibiotic significantly reduced NTHi viability in biofilms compared with antibiotic treatment alone.Impaired ciliary function was the primary defect in PCD airway epithelium underlying susceptibility to NTHi biofilm development compared with non-PCD epithelium. Although NO responses were similar, use of targeted NO with antibiotics enhanced killing of NTHi in biofilms, suggesting a novel therapeutic approach., Competing Interests: Conflict of interest: Disclosures can be found alongside this article at erj.ersjournals.com, (Copyright ©ERS 2017.)
- Published
- 2017
- Full Text
- View/download PDF
45. IL-4-secreting eosinophils promote endometrial stromal cell proliferation and prevent Chlamydia -induced upper genital tract damage.
- Author
-
Vicetti Miguel RD, Quispe Calla NE, Dixon D, Foster RA, Gambotto A, Pavelko SD, Hall-Stoodley L, and Cherpes TL
- Subjects
- Animals, Chlamydia Infections genetics, Chlamydia Infections microbiology, Chlamydia trachomatis physiology, Endometrium cytology, Eosinophils metabolism, Female, Genitalia, Female metabolism, Genitalia, Female microbiology, Immunity, Innate genetics, Immunity, Innate immunology, Interleukin-4 genetics, Interleukin-4 metabolism, Leukocyte Count, Mice, Inbred BALB C, Mice, Knockout, Stromal Cells metabolism, Cell Proliferation, Chlamydia Infections immunology, Chlamydia trachomatis immunology, Eosinophils immunology, Genitalia, Female immunology, Interleukin-4 immunology, Stromal Cells immunology
- Abstract
Genital Chlamydia trachomatis infections in women typically are asymptomatic and do not cause permanent upper genital tract (UGT) damage. Consistent with this presentation, type 2 innate and T
H 2 adaptive immune responses associated with dampened inflammation and tissue repair are elicited in the UGT of Chlamydia -infected women. Primary C. trachomatis infection of mice also causes no genital pathology, but unlike women, does not generate Chlamydia -specific TH 2 immunity. Herein, we explored the significance of type 2 innate immunity for restricting UGT tissue damage in Chlamydia -infected mice, and in initial studies intravaginally infected wild-type, IL-10-/- , IL-4-/- , and IL-4Rα-/- mice with low-dose C. trachomatis inoculums. Whereas Chlamydia was comparably cleared in all groups, IL-4-/- and IL-4Rα-/- mice displayed endometrial damage not seen in wild-type or IL-10-/- mice. Congruent with the aberrant tissue repair in mice with deficient IL-4 signaling, we found that IL-4Rα and STAT6 signaling mediated IL-4-induced endometrial stromal cell (ESC) proliferation ex vivo, and that genital administration of an IL-4-expressing adenoviral vector greatly increased in vivo ESC proliferation. Studies with IL-4-IRES-eGFP (4get) reporter mice showed eosinophils were the main IL-4-producing endometrial leukocyte (constitutively and during Chlamydia infection), whereas studies with eosinophil-deficient mice identified this innate immune cell as essential for endometrial repair during Chlamydia infection. Together, our studies reveal IL-4-producing eosinophils stimulate ESC proliferation and prevent Chlamydia -induced endometrial damage. Based on these results, it seems possible that the robust type 2 immunity elicited by Chlamydia infection of human genital tissue may analogously promote repair processes that reduce phenotypic disease expression., Competing Interests: The authors declare no conflict of interest.- Published
- 2017
- Full Text
- View/download PDF
46. D-methionine interferes with non-typeable Haemophilus influenzae peptidoglycan synthesis during growth and biofilm formation.
- Author
-
Dawe H, Berger E, Sihlbom C, Angus EM, Howlin RP, Laver JR, Tebruegge M, Hall-Stoodley L, Stoodley P, Faust SN, and Allan RN
- Subjects
- Anti-Bacterial Agents pharmacology, Bacterial Proteins genetics, Bacterial Proteins metabolism, Gene Expression Regulation, Bacterial, Haemophilus influenzae drug effects, Haemophilus influenzae genetics, Humans, Biofilms, Haemophilus Infections microbiology, Haemophilus influenzae growth & development, Haemophilus influenzae metabolism, Methionine metabolism, Peptidoglycan biosynthesis
- Abstract
Non-typeable Haemophilus influenzae (NTHi) is an opportunistic pathogen that plays a major role in a number of respiratory tract infections, including otitis media, cystic fibrosis and chronic obstructive pulmonary disease. Biofilm formation has been implicated in both NTHi colonization and disease, and is responsible for the increased tolerance of this pathogen towards antibiotic treatment. Targeting metabolic pathways that are important in NTHi biofilm formation represents a potential strategy to combat this antibiotic recalcitrance. A previous investigation demonstrated increased expression of a putative d-methionine uptake protein following exposure of NTHi biofilms to the ubiquitous signalling molecule, nitric oxide. We therefore hypothesized that treatment with exogenous d-methionine would impact on NTHi biofilm formation and increase antibiotic sensitivity. Treatment of NTHi during the process of biofilm formation resulted in a reduction in biofilm viability, increased biomass, changes in the overall biofilm architecture and the adoption of an amorphous cellular morphology. Quantitative proteomic analyses identified 124 proteins that were differentially expressed following d-methionine treatment, of which 51 (41 %) were involved in metabolic and transport processes. Nine proteins involved in peptidoglycan synthesis and cell division showed significantly increased expression. Furthermore, d-methionine treatment augmented the efficacy of azithromycin treatment and highlighted the potential of d-methionine as an adjunctive therapeutic approach for NTHi biofilm-associated infections.
- Published
- 2017
- Full Text
- View/download PDF
47. Detection and Physicochemical Characterization of Membrane Vesicles (MVs) of Lactobacillus reuteri DSM 17938.
- Author
-
Grande R, Celia C, Mincione G, Stringaro A, Di Marzio L, Colone M, Di Marcantonio MC, Savino L, Puca V, Santoliquido R, Locatelli M, Muraro R, Hall-Stoodley L, and Stoodley P
- Abstract
Membrane vesicles (MVs) are bilayer structures which bleb from bacteria, and are important in trafficking biomolecules to other bacteria or host cells. There are few data about MVs produced by the Gram-positive commensal-derived probiotic Lactobacillus reuteri ; however, MVs from this species may have potential therapeutic benefit. The aim of this study was to detect and characterize MVs produced from biofilm (bMVs), and planktonic (pMVs) phenotypes of L. reuteri DSM 17938. MVs were analyzed for structure and physicochemical characterization by Scanning Electron Microscope (SEM) and Dynamic Light Scattering (DLS). Their composition was interrogated using various digestive enzyme treatments and subsequent Transmission Electron Microscopy (TEM) analysis. eDNA (extracellular DNA) was detected and quantified using PicoGreen. We found that planktonic and biofilm of L. reuteri cultures generated MVs with a broad size distribution. Our data also showed that eDNA was associated with pMVs and bMVs (e
MVs DNA). DNase I treatment demonstrated no modifications of MVs, suggesting that an eDNA-MVs complex protected the eMVs DNA. Proteinase K and Phospholipase C treatments modified the structure of MVs, showing that lipids and proteins are important structural components of L. reuteri MVs. The biological composition and the physicochemical characterization of MVs generated by the probiotic L. reuteri may represent a starting point for future applications in the development of vesicles-based therapeutic systems.- Published
- 2017
- Full Text
- View/download PDF
48. Cephalosporin-NO-donor prodrug PYRRO-C3D shows β-lactam-mediated activity against Streptococcus pneumoniae biofilms.
- Author
-
Allan RN, Kelso MJ, Rineh A, Yepuri NR, Feelisch M, Soren O, Brito-Mutunayagam S, Salib RJ, Stoodley P, Clarke SC, Webb JS, Hall-Stoodley L, and Faust SN
- Subjects
- Amoxicillin pharmacology, Anti-Inflammatory Agents, Non-Steroidal chemistry, Azithromycin pharmacology, Azo Compounds chemistry, Cephalosporins chemistry, Nitric Oxide analysis, Nitric Oxide Donors chemistry, Penicillinase chemistry, Plankton microbiology, Prodrugs chemistry, Anti-Inflammatory Agents, Non-Steroidal pharmacology, Azo Compounds pharmacology, Biofilms drug effects, Cephalosporins pharmacology, Nitric Oxide Donors pharmacology, Prodrugs pharmacology, Streptococcus pneumoniae drug effects
- Abstract
Bacterial biofilms show high tolerance towards antibiotics and are a significant problem in clinical settings where they are a primary cause of chronic infections. Novel therapeutic strategies are needed to improve anti-biofilm efficacy and support reduction in antibiotic use. Treatment with exogenous nitric oxide (NO) has been shown to modulate bacterial signaling and metabolic processes that render biofilms more susceptible to antibiotics. We previously reported on cephalosporin-3'-diazeniumdiolates (C3Ds) as NO-donor prodrugs designed to selectively deliver NO to bacterial infection sites following reaction with β-lactamases. With structures based on cephalosporins, C3Ds could, in principal, also be triggered to release NO following β-lactam cleavage mediated by transpeptidases/penicillin-binding proteins (PBPs), the antibacterial target of cephalosporin antibiotics. Transpeptidase-reactive C3Ds could potentially show both NO-mediated anti-biofilm properties and intrinsic (β-lactam-mediated) antibacterial effects. This dual-activity concept was explored using Streptococcus pneumoniae, a species that lacks β-lactamases but relies on transpeptidases for cell-wall synthesis. Treatment with PYRRO-C3D (a representative C3D containing the diazeniumdiolate NO donor PYRRO-NO) was found to significantly reduce viability of planktonic and biofilm pneumococci, demonstrating that C3Ds can elicit direct, cephalosporin-like antibacterial activity in the absence of β-lactamases. While NO release from PYRRO-C3D in the presence of pneumococci was confirmed, the anti-pneumococcal action of the compound was shown to arise exclusively from the β-lactam component and not through NO-mediated effects. The compound showed similar potency to amoxicillin against S. pneumoniae biofilms and greater efficacy than azithromycin, highlighting the potential of C3Ds as new agents for treating pneumococcal infections., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
49. Microbiological diagnosis of device-related biofilm infections.
- Author
-
Xu Y, Larsen LH, Lorenzen J, Hall-Stoodley L, Kikhney J, Moter A, and Thomsen TR
- Subjects
- Animals, Bacteria genetics, Bacteria growth & development, Bacterial Infections therapy, Humans, Prosthesis-Related Infections therapy, Bacterial Infections diagnosis, Bacterial Infections microbiology, Bacterial Physiological Phenomena, Biofilms, Prosthesis-Related Infections diagnosis, Prosthesis-Related Infections microbiology
- Abstract
Medical device-related infections cause undue patient distress, increased morbidity and mortality and pose a huge financial burden on healthcare services. The pathogens are frequently distributed heterogeneously in biofilms, which can persist without being effectively cleared by host immune defenses and antibiotic therapy. At present, there is no 'gold standard' available to reveal the presence of device-related biofilm infections. However, adequate sample collection and logistics, standardised diagnostic methods, and interpretation of results by experienced personnel are important steps in efficient diagnosis and treatment of these infections. The focus of this mini review is on prosthethic joint and cardiovascular implantable device infections, which exemplify permanent devices that are placed in a sterile body site. These device-related infections represent some of the most challenging in terms of both diagnosis and treatment., (© 2017 APMIS. Published by John Wiley & Sons Ltd.)
- Published
- 2017
- Full Text
- View/download PDF
50. Cephalosporin-3'-Diazeniumdiolate NO Donor Prodrug PYRRO-C3D Enhances Azithromycin Susceptibility of Nontypeable Haemophilus influenzae Biofilms.
- Author
-
Collins SA, Kelso MJ, Rineh A, Yepuri NR, Coles J, Jackson CL, Halladay GD, Walker WT, Webb JS, Hall-Stoodley L, Connett GJ, Feelisch M, Faust SN, Lucas JS, and Allan RN
- Subjects
- Anti-Bacterial Agents pharmacology, Azithromycin pharmacology, Chromatography, Liquid, Drug Resistance, Bacterial, Mass Spectrometry, Microbial Sensitivity Tests, Nitrogen Oxides metabolism, Proteomics, beta-Lactamases metabolism, Azo Compounds pharmacology, Biofilms drug effects, Cephalosporins pharmacology, Haemophilus influenzae drug effects, Nitric Oxide Donors pharmacology, Prodrugs pharmacology
- Abstract
PYRRO-C3D is a cephalosporin-3-diazeniumdiolate nitric oxide (NO) donor prodrug designed to selectively deliver NO to bacterial infection sites. The objective of this study was to assess the activity of PYRRO-C3D against nontypeable Haemophilus influenzae (NTHi) biofilms and examine the role of NO in reducing biofilm-associated antibiotic tolerance. The activity of PYRRO-C3D on in vitro NTHi biofilms was assessed through CFU enumeration and confocal microscopy. NO release measurements were performed using an ISO-NO probe. NTHi biofilms grown on primary ciliated respiratory epithelia at an air-liquid interface were used to investigate the effects of PYRRO-C3D in the presence of host tissue. Label-free liquid chromatography-mass spectrometry (LC/MS) proteomic analyses were performed to identify differentially expressed proteins following NO treatment. PYRRO-C3D specifically released NO in the presence of NTHi, while no evidence of spontaneous NO release was observed when the compound was exposed to primary epithelial cells. NTHi lacking β-lactamase activity failed to trigger NO release. Treatment significantly increased the susceptibility of in vitro NTHi biofilms to azithromycin, causing a log fold reduction (10-fold reduction or 1-log-unit reduction) in viability (P < 0.05) relative to azithromycin alone. The response was more pronounced for biofilms grown on primary respiratory epithelia, where a 2-log-unit reduction was observed (P < 0.01). Label-free proteomics showed that NO increased expression of 16 proteins involved in metabolic and transcriptional/translational functions. NO release from PYRRO-C3D enhances the efficacy of azithromycin against NTHi biofilms, putatively via modulation of NTHi metabolic activity. Adjunctive therapy with NO mediated through PYRRO-C3D represents a promising approach for reducing biofilm-associated antibiotic tolerance., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.