1
|
Germon P, Foucras G, Smith DGE, Rainard P. Invited review: Mastitis Escherichia coli strains-Mastitis-associated or mammo-pathogenic? J Dairy Sci 2025; 108:4485-4507. [PMID: 40139360 DOI: 10.3168/jds.2024-26109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Bovine mastitis remains a major concern for dairy farmers, mainly because of its effect on the economy of their activity and on animal welfare. Because Escherichia coli is considered a major mastitis pathogen, the diversity of E. coli strains isolated from mastitis cases has been studied for decades, with the aim to discover new ways to fight this infection. With the recent advances in whole-genome sequencing, a detailed view of the peculiarities of mastitis E. coli strains has emerged. This review aims to bring together the knowledge garnered over the years with the more recent results of whole-genome analyses. Whereas the concept of a mammary pathogenic E. coli has been proposed, because a common set of virulence genes cannot be identified among mastitis E. coli strains, we prefer the use of mastitis-associated E. coli (MAEC), with MAEC being more an "ecotype" rather than a "pathotype." Indeed, data available so far suggest that a common feature of MAEC would rather be an enrichment in fitness capabilities that makes them well-suited for survival and rapid adaptation to changing biotopes in the mammary gland, which we qualify as intramammary ecotopes.
Collapse
Affiliation(s)
- Pierre Germon
- INRAE, ISP UMR 1282, Université François Rabelais de Tours, 37380 Nouzilly, France.
| | - Gilles Foucras
- IHAP, Université de Toulouse, INRAE, ENVT, 31076 Toulouse, France
| | | | - Pascal Rainard
- INRAE, ISP UMR 1282, Université François Rabelais de Tours, 37380 Nouzilly, France
| |
Collapse
|
2
|
Gadhvi JG, Kenee PRM, Lutz KC, Khan F, Li Q, Zimmern PE, De Nisco NJ. Bladder-resident bacteria associated with increased risk of recurrence after electrofulguration in women with antibiotic-recalcitrant urinary tract infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.03.24309902. [PMID: 39006411 PMCID: PMC11245057 DOI: 10.1101/2024.07.03.24309902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Antibiotic-recalcitrant recurrent urinary tract infection (rUTI) is has become increasingly observed in postmenopausal women. Therefore, when standard antibiotic therapies have failed, some elect electrofulguration (EF) of areas of chronic cystitis when detected on office cystoscopy. EF is thought to remove tissue-resident bacteria that have been previously detected in the bladder walls of postmenopausal women with rUTI. We hypothesized that increased bladder bacterial burden may be associated with incomplete rUTI resolution following EF. Methods Following IRB approval, bladder biopsies were obtained from 34 consenting menopausal women electing EF for the advanced management of rUTI. 16S rRNA FISH was performed using both universal and Escherichia probes and tissue-resident bacterial load was quantified. Time to UTI relapse after EF was recorded during a six-month follow-up period and the association of bladder bacterial burden and clinical covariates with UTI relapse was assessed. Results We observed bladder-resident Escherichia in 52% of all participants and in 92% of participants with recent E. coli UTI. Time-to-relapse analysis revealed that women with high bladder bacterial burden as detected by the universal probe had a significantly ( p =0.035) higher risk of UTI within six months of EF (HR=3.15, 95% CI: 1.09-9.11). Interestingly, bladder-resident Escherichia was not significantly associated ( p =0.26) with a higher risk of UTI relapse (HR= 2.14, 95% CI: 0.58-7.90). Conclusions We observed that total bladder bacterial burden was associated with a 3.1x increased risk of rUTI relapse within six months. Continued analysis of the relationship between bladder bacterial burden and rUTI outcomes may provide insight into the management of these challenging patients.
Collapse
|
3
|
Marsh MC, Junquera GY, Stonebrook E, Spencer JD, Watson JR. Urinary Tract Infections in Children. Pediatr Rev 2024; 45:260-270. [PMID: 38689106 DOI: 10.1542/pir.2023-006017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Despite the American Academy of Pediatrics guidelines for the evaluation, treatment, and management of urinary tract infections (UTIs), UTI diagnosis and management remains challenging for clinicians. Challenges with acute UTI management stem from vague presenting signs and symptoms, diagnostic uncertainty, limitations in laboratory testing, and selecting appropriate antibiotic therapy in an era with increasing rates of antibiotic-resistant uropathogens. Recurrent UTI management remains difficult due to an incomplete understanding of the factors contributing to UTI, when to assess a child with repeated infections for kidney and urinary tract anomalies, and limited prevention strategies. To help reduce these uncertainties, this review provides a comprehensive overview of UTI epidemiology, risk factors, diagnosis, treatment, and prevention strategies that may help pediatricians overcome the challenges associated with acute and recurrent UTI management.
Collapse
Affiliation(s)
- Melanie C Marsh
- Division of Hospital Medicine, Department of Pediatrics, Advocate Aurora Atrium Health Systems, Chicago, IL
| | - Guillermo Yepes Junquera
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
- Division of Infectious Diseases
| | - Emily Stonebrook
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
- Division of Nephrology and Hypertension, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| | - John David Spencer
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
- Division of Nephrology and Hypertension, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| | - Joshua R Watson
- Center for Clinical Excellence, Nationwide Children's Hospital, Columbus, OH
- Division of Infectious Diseases
| |
Collapse
|
4
|
Naber KG, Alidjanov JF, Fünfstück R, Strohmaier WL, Kranz J, Cai T, Pilatz A, Wagenlehner FM. Therapeutic strategies for uncomplicated cystitis in women. GMS INFECTIOUS DISEASES 2024; 12:Doc01. [PMID: 38764941 PMCID: PMC11099625 DOI: 10.3205/id000086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Uncomplicated cystitis is affecting many women of all ages and has a great impact on the quality of life, especially in women suffering from recurrent, uncomplicated cystitis. By far the most frequent uropathogen, E. coli, may have acquired increasing resistance against a variety of oral antibiotics, which may differ between countries and regions. Therefore, local resistance data are important to be considered. On the other hand, non-antibiotic therapy has also become an option which should be discussed and offered to the patient. In patients suffering from recurrent uncomplicated cystitis, individual risk factors and possible behavioral changes should first be taken into account. Non-antimicrobial prophylactic strategies shown to be successful in well-designed clinical studies are the next options. Long term antibiotic prophylaxis, however, should only be considered as a last option. For some of those patients self-diagnosis and self-treatment may be suitable, e.g. by using a recognized questionnaire.
Collapse
Affiliation(s)
- Kurt G. Naber
- Department of Urology, Technical University of Munich, Germany
| | | | | | - Walter L. Strohmaier
- Medical School Regiomed, Coburg, Germany
- Julius Maximilian University, Wuerzburg, Germany
- University of Split, Croatia
| | - Jennifer Kranz
- Department of Urology and Pediatric Urology, University Medical Center RWTH Aachen, Germany
- Department of Urology and Kidney Transplantation, Martin Luther University, Halle (Saale), Germany
| | - Tommaso Cai
- Department of Urology, Santa Chiara Regional Hospital, Trento, Italy
| | - Adrian Pilatz
- Clinic for Urology, Pediatric Urology and Andrology, Justus Liebig University of Giessen, Germany
| | - Florian M. Wagenlehner
- Clinic for Urology, Pediatric Urology and Andrology, Justus Liebig University of Giessen, Germany
| |
Collapse
|
5
|
Li X, Zhou J, Liu X, Jin C, Liu L, Sun H, Wang Q, Wang Q, Liu R, Zheng X, Liu Y, Pang Y. Nucleoside-diphosphate kinase of uropathogenic Escherichia coli inhibits caspase-1-dependent pyroptosis facilitating urinary tract infection. Cell Rep 2024; 43:114051. [PMID: 38564334 DOI: 10.1016/j.celrep.2024.114051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/19/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the most common causative agent of urinary tract infection (UTI). UPEC invades bladder epithelial cells (BECs) via fusiform vesicles, escapes into the cytosol, and establishes biofilm-like intracellular bacterial communities (IBCs). Nucleoside-diphosphate kinase (NDK) is secreted by pathogenic bacteria to enhance virulence. However, whether NDK is involved in UPEC pathogenesis remains unclear. Here, we find that the lack of ndk impairs the colonization of UPEC CFT073 in mouse bladders and kidneys owing to the impaired ability of UPEC to form IBCs. Furthermore, we demonstrate that NDK inhibits caspase-1-dependent pyroptosis by consuming extracellular ATP, preventing superficial BEC exfoliation, and promoting IBC formation. UPEC utilizes the reactive oxygen species (ROS) sensor OxyR to indirectly activate the regulator integration host factor, which then directly activates ndk expression in response to intracellular ROS. Here, we reveal a signaling transduction pathway that UPEC employs to inhibit superficial BEC exfoliation, thus facilitating acute UTI.
Collapse
Affiliation(s)
- Xueping Li
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Jiarui Zhou
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Xingmei Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Chen Jin
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Le Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Hongmin Sun
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Qian Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Qiushi Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Ruiying Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Xiaoyu Zheng
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China
| | - Yutao Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China.
| | - Yu Pang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, P.R. China; The Key Laboratory of Molecular Microbiology and Technology, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Ministry of Education, Tianjin 300457, P.R. China.
| |
Collapse
|
6
|
Kwak Y, Kim HG, Seok J, Kim S, Kim EM, Kim A. The Critical Role of Intracellular Bacterial Communities in Uncomplicated Recurrent Urinary Cystitis: A Comprehensive Review of Detection Methods and Diagnostic Potential. Int Neurourol J 2024; 28:4-10. [PMID: 38569615 PMCID: PMC10990762 DOI: 10.5213/inj.2448066.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/06/2024] [Indexed: 04/05/2024] Open
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections worldwide and are particularly prevalent in women. Recurrent UTIs significantly diminish quality of life due to their symptoms and frequent relapses. Patients often experience immediate relapse following slightly strenuous activities or intense psychological stress. In this review, we explore why infections persist despite the advent of various treatments and suggest strategies to manage recurrent cystitis by targeting the mechanisms of adhesion and infection. Vitamin D levels and the expression of neutrophil gelatinase-associated lipocalin are linked to the recurrence of UTIs. During a UTI, bacteria employ adhesins to invade the urinary tract, adhere to urothelial cells, and then penetrate these cells, where they rapidly multiply to establish intracellular bacterial communities. Bacteria can also form quiescent intracellular reservoirs that escape immune responses and antibiotic treatments, leading to recurrence under certain conditions. The surface proteins of bacteria and D-mannose are crucial in the adhesion of bacteria to the urothelium. Understanding these processes provides valuable insights into potential therapeutic approaches that focus on preventing bacterial attachment and cluster formation. By disrupting the ability of bacteria to adhere to and form clusters on cells, we can better manage recurrent UTIs and improve patient outcomes.
Collapse
Affiliation(s)
- Yeonjoo Kwak
- Department of Stem Cell & Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Hyeong Gon Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Jaekwon Seok
- Department of Stem Cell & Regenerative Biotechnology, Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Sehwan Kim
- Department of Biomedical Engineering, School of Medicine, Dankook University, Cheonan, Korea
| | - Eun-Mee Kim
- Department of Paramedicine, Korea Nazarene University, Cheonan, Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, CA, USA
| |
Collapse
|
7
|
Pioch T, Fischer T, Schneider M. Aspherical, Nano-Structured Drug Delivery System with Tunable Release and Clearance for Pulmonary Applications. Pharmaceutics 2024; 16:232. [PMID: 38399290 PMCID: PMC10891959 DOI: 10.3390/pharmaceutics16020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/21/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Addressing the challenge of efficient drug delivery to the lungs, a nano-structured, microparticulate carrier system with defined and customizable dimensions has been developed. Utilizing a template-assisted approach and capillary forces, particles were rapidly loaded and stabilized. The system employs a biocompatible alginate gel as a stabilizing matrix, facilitating the breakdown of the carrier in body fluids with the subsequent release of its nano-load, while also mitigating long-term accumulation in the lung. Different gel strengths and stabilizing steps were applied, allowing us to tune the release kinetics, as evaluated by a quantitative method based on a flow-imaging system. The micro-cylinders demonstrated superior aerodynamic properties in Next Generation Impactor (NGI) experiments, such as a smaller median aerodynamic diameter (MMAD), while yielding a higher fine particle fraction (FPF) than spherical particles similar in critical dimensions. They exhibited negligible toxicity to a differentiated macrophage cell line (dTHP-1) for up to 24 h of incubation. The kinetics of the cellular uptake by dTHP-1 cells was assessed via fluorescence microscopy, revealing an uptake-rate dependence on the aspect ratio (AR = l/d); cylinders with high AR were phagocytosed more slowly than shorter rods and comparable spherical particles. This indicates that this novel drug delivery system can modulate macrophage uptake and clearance by adjusting its geometric parameters while maintaining optimal aerodynamic properties and featuring a biodegradable stabilizing matrix.
Collapse
Affiliation(s)
| | | | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany; (T.P.); (T.F.)
| |
Collapse
|
8
|
Raatz M, de Azevedo-Lopes A, Drabik K, Traulsen A, Waclaw B. Pathogen non-planktonic phases within the urinary tract impact early infection and resistance evolution. THE ISME JOURNAL 2024; 18:wrae191. [PMID: 39325970 PMCID: PMC11499890 DOI: 10.1093/ismejo/wrae191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/12/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024]
Abstract
Treatment of urinary tract infections and the prevention of their recurrence is a pressing global health problem. In a urinary infection, pathogenic bacteria not only reside in the bladder lumen but also attach to and invade the bladder tissue. Planktonic, attached, and intracellular bacteria face different selection pressures from physiological processes such as micturition, immune response, and antibiotic treatment. Here, we use a mathematical model of the initial phase of infection to unravel the effects of these different selective pressures on the ecological and evolutionary dynamics of urinary infections. We explicitly model planktonic bacteria in the bladder lumen, bacteria attached to the bladder wall, and bacteria that have invaded the epithelial cells of the bladder. We find that the presence of non-planktonic bacteria substantially increases the risk of infection establishment and affects evolutionary trajectories leading to resistance during antibiotic treatment. We also show that competitive inoculation with a fast-growing non-pathogenic strain can reduce the pathogen load and increase the efficacy of an antibiotic, but only if the antibiotic is used in moderation. Our study shows that including different compartments is essential to create more realistic models of urinary infections, which may help guide new treatment strategies.
Collapse
Affiliation(s)
- Michael Raatz
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany
| | - Amanda de Azevedo-Lopes
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany
| | - Karolina Drabik
- Dioscuri Centre for Physics and Chemistry of Bacteria, Institute of Physical Chemistry (IChF), Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Arne Traulsen
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany
| | - Bartlomiej Waclaw
- Dioscuri Centre for Physics and Chemistry of Bacteria, Institute of Physical Chemistry (IChF), Polish Academy of Sciences, 01-224 Warsaw, Poland
- School of Physics and Astronomy, University of Edinburgh, EH9 3FD Edinburgh, United Kingdom
| |
Collapse
|
9
|
Boya BR, Lee JH, Lee J. Antimicrobial and antibiofilm activities of chromone derivatives against uropathogenic Escherichia coli. Microbiol Res 2024; 278:127537. [PMID: 37922697 DOI: 10.1016/j.micres.2023.127537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
Uropathogenic Escherichia coli (UPEC) is a urinary tract pathogen responsible for most nosocomial urinary tract infections and can cause severe conditions like acute cystitis of the bladder or pyelonephritis. UPEC harbors a host of virulence factors like curli, hemolysin, siderophore, and motility factors and can form biofilm-like communities and quiescent reservoirs that aid its survival. This study was performed to investigate the antibiofilm, antimicrobial, and antivirulence potentials of three chromone derivatives, namely, 6-bromo 3-formylchromone, 6-chloro 3-formylchromone, and 3-formyl 6-isopropylchromone. These chromones had MICs against UPEC of 20, 20, and 50 µg/ml, respectively, inhibited biofilm formation by 72-96% at 20 µg/ml, and inhibited UPEC-associated virulence factors, that is, hemolysis, motility, curli, siderophore production, indole production, quiescent colony formation, and cell surface hydrophobicity. Gene expression analysis indicated these three derivatives downregulated virulence genes associated with toxins, biofilm production, and stress regulation and suggested they might target two-component UvrY response regulator. 3D-QSAR analysis showed that substitutions at the third and sixth positions of the chromone scaffold favor antimicrobial activity against UPEC. Furthermore, ADME profiles and C. elegans cytotoxicity assays indicated that these chromone derivatives are potent, safe drug candidates.
Collapse
Affiliation(s)
- Bharath Reddy Boya
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
10
|
Gleicher S, Karram M, Wein AJ, Dmochowski RR. Recurrent and complicated urinary tract infections in women: Utility of advanced testing to enhance care. Neurourol Urodyn 2024; 43:161-166. [PMID: 37822027 DOI: 10.1002/nau.25280] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/03/2023] [Accepted: 08/28/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND The United States currently faces a public health crisis with regarding to antibiotic-resistant bacteria, and new urinary tract infection (UTI) diagnostics are needed. Women with recurrent UTI (rUTI) and complicated UTI (cUTI) are at particular risk given their complexity and the paucity of adequate testing modalities. The standard urine culture (SUC) is the cornerstone for diagnosis, but it has many shortcomings. These pitfalls lead to dissatisfaction and frustration among women afflicted with rUTI and cUTI, as well as overuse of antibiotics. One innovation is PCR UTI testing, which has been shown to outperform SUC among symptomatic women. AIMS This article discusses UTI PCR testing, as well as a possible role in clinical practice. MATERIALS AND METHODS Published literature was reviewed and summarized. RESULTS Management of rUTI and cUTI is complex, and providers should have all diagnostics available to facilitate providing optimal care. Urine PCR testing faces reimbursement issues despite fulfilling clinical indication parameters as described by insurance companies. DISCUSSION The role of UTI PCR testing remains unclear. Reimbursement issues have led to underuse and limited real-world outcomes reinforcing benefit. CONCLUSION This study proposes an algorithm for PCR testing among women with rUTI and cUTI.
Collapse
Affiliation(s)
| | - Mickey Karram
- The Christ Hospital Physicians, Cincinnati, Ohio, USA
| | - Alan J Wein
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Roger R Dmochowski
- Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Flores C, Ling J, Loh A, Maset RG, Aw A, White IJ, Fernando R, Rohn JL. A human urothelial microtissue model reveals shared colonization and survival strategies between uropathogens and commensals. SCIENCE ADVANCES 2023; 9:eadi9834. [PMID: 37939183 PMCID: PMC10631729 DOI: 10.1126/sciadv.adi9834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/05/2023] [Indexed: 11/10/2023]
Abstract
Urinary tract infection is among the most common infections worldwide, typically studied in animals and cell lines with limited uropathogenic strains. Here, we assessed diverse bacterial species in a human urothelial microtissue model exhibiting full stratification, differentiation, innate epithelial responses, and urine tolerance. Several uropathogens invaded intracellularly, but also commensal Escherichia coli, suggesting that invasion is a shared survival strategy, not solely a virulence hallmark. The E. coli adhesin FimH was required for intracellular bacterial community formation, but not for invasion. Other shared lifestyles included filamentation (Gram-negatives), chaining (Gram-positives), and hijacking of exfoliating cells, while biofilm-like aggregates were formed mainly with Pseudomonas and Proteus. Urothelial cells expelled invasive bacteria in Rab-/LC3-decorated structures, while highly cytotoxic/invasive uropathogens, but not commensals, disrupted host barrier function and strongly induced exfoliation and cytokine production. Overall, this work highlights diverse species-/strain-specific infection strategies and corresponding host responses in a human urothelial microenvironment, providing insights at the microtissue, cell, and molecular level.
Collapse
Affiliation(s)
- Carlos Flores
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Jefferson Ling
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Amanda Loh
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Ramón G. Maset
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Angeline Aw
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
| | - Ian J. White
- Laboratory for Molecular Cell Biology, University College London, WC1E 6BT London, UK
| | - Raymond Fernando
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
- Royal Free London NHS Foundation Trust & Anthony Nolan Laboratories, NW3 2QG London, UK
| | - Jennifer L. Rohn
- Centre for Urological Biology, Division of Medicine, University College London, WC1E 6BT London, UK
| |
Collapse
|
12
|
Kuhn HW, Hreha TN, Hunstad DA. Immune defenses in the urinary tract. Trends Immunol 2023; 44:701-711. [PMID: 37591712 PMCID: PMC10528756 DOI: 10.1016/j.it.2023.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 08/19/2023]
Abstract
Recent advances in preclinical modeling of urinary tract infections (UTIs) have enabled the identification of key facets of the host response that influence pathogen clearance and tissue damage. Here, we review new insights into the functions of neutrophils, macrophages, and antimicrobial peptides in innate control of uropathogens and in mammalian infection-related tissue injury and repair. We also discuss novel functions for renal epithelial cells in innate antimicrobial defense. In addition, epigenetic modifications during bacterial cystitis have been implicated in bladder remodeling, conveying susceptibility to recurrent UTI. In total, contemporary work in this arena has better defined host processes that shape UTI susceptibility and severity and might inform the development of novel preventive and therapeutic approaches for acute and recurrent UTI.
Collapse
Affiliation(s)
- Hunter W Kuhn
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Teri N Hreha
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Hunstad
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
13
|
Naskar M, Parekh VP, Abraham MA, Alibasic Z, Kim MJ, Suk G, Noh JH, Ko KY, Lee J, Kim C, Yoon H, Abraham SN, Choi HW. α-Hemolysin promotes uropathogenic E. coli persistence in Bladder epithelial cells Via abrogating bacteria-harboring lysosome acidification. PLoS Pathog 2023; 19:e1011388. [PMID: 37167325 DOI: 10.1371/journal.ppat.1011388] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 05/23/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023] Open
Abstract
There is a growing consensus that a significant proportion of recurrent urinary tract infections are linked to the persistence of uropathogens within the urinary tract and their re-emergence upon the conclusion of antibiotic treatment. Studies in mice and human have revealed that uropathogenic Escherichia coli (UPEC) can persist in bladder epithelial cells (BECs) even after the apparent resolution of the infection. Here, we found that, following the entry of UPEC into RAB27b+ fusiform vesicles in BECs, some bacteria escaped into the cytoplasmic compartment via a mechanism involving hemolysin A (HlyA). However, these UPEC were immediately recaptured within LC3A/B+ autophagosomes that matured into LAMP1+ autolysosomes. Thereafter, HlyA+ UPEC-containing lysosomes failed to acidify, which is an essential step for bacterial elimination. This lack of acidification was related to the inability of bacteria-harboring compartments to recruit V-ATPase proton pumps, which was attributed to the defragmentation of cytosolic microtubules by HlyA. The persistence of UPEC within LAMP1+ compartments in BECs appears to be directly linked to HlyA. Thus, through intravesicular instillation of microtubule stabilizer, this host defense response can be co-opted to reduce intracellular bacterial burden following UTIs in the bladder potentially preventing recurrence.
Collapse
Affiliation(s)
- Manisha Naskar
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Viraj P Parekh
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Mathew A Abraham
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Zehra Alibasic
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Min Jung Kim
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Gyeongseo Suk
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Joo Hwan Noh
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Kwan Young Ko
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Joonha Lee
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Chungho Kim
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Hana Yoon
- Department of Urology, Ewha Womans University, College of medicine, Seoul, South Korea
| | - Soman N Abraham
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- Molecular Genetics & Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Hae Woong Choi
- Division of Life Sciences, Korea University, Seoul, South Korea
| |
Collapse
|
14
|
Deng Y, Fu Y, Chua SL, Khoo BL. Biofilm Potentiates Cancer-Promoting Effects of Tumor-Associated Macrophages in a 3D Multi-Faceted Tumor Model. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205904. [PMID: 36748304 DOI: 10.1002/smll.202205904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/01/2023] [Indexed: 05/11/2023]
Abstract
Components of the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs), influence tumor progression. The specific polarization and phenotypic transition of TAMs in the tumor microenvironment lead to two-pronged impacts that can promote or hinder cancer development and treatment. Here, a novel microfluidic multi-faceted bladder tumor model (TAMPIEB ) is developed incorporating TAMs and cancer cells to evaluate the impact of bacterial distribution on immunomodulation within the tumor microenvironment in vivo. It is demonstrated for the first time that biofilm-induced inflammatory conditions within tumors promote the transition of macrophages from a pro-inflammatory M1-like to an anti-inflammatory/pro-tumor M2-like state. Consequently, multiple roles and mechanisms by which biofilms promote cancer by inducing pro-tumor phenotypic switch of TAMs are identified, including cancer hallmarks such as reducing susceptibility to apoptosis, enhancing cell viability, and promoting epithelial-mesenchymal transition and metastasis. Furthermore, biofilms formed by extratumoral bacteria can shield tumors from immune attack by TAMs, which can be visualized through various imaging assays in situ. The study sheds light on the underlying mechanism of biofilm-mediated inflammation on tumor progression and provides new insights into combined anti-biofilm therapy and immunotherapy strategies in clinical trials.
Collapse
Affiliation(s)
- Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong
| | - Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Kowloon, 999077, Hong Kong
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, Kowloon, 999077, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong SAR, Kowloon, 999077, China
- Shenzhen Key Laboratory of Food Biological Safety Control, Kowloon, 999077, Hong Kong
- Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Hong Kong SAR, Kowloon, 999077, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, 999077, Hong Kong
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Kowloon, 999077, Hong Kong
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen-Futian Research Institute, Shenzhen, 518057, China
| |
Collapse
|
15
|
Ullah H, Minno AD, Filippis AD, Sommella E, Buccato DG, Lellis LFD, El-Seedi HR, Khalifa SAM, Piccinocchi R, Galdiero M, Campiglia P, Daglia M. In Vitro Antimicrobial and Antibiofilm Properties and Bioaccessibility after Oral Digestion of Chemically Characterized Extracts Obtained from Cistus × incanus L., Scutellaria lateriflora L., and Their Combination. Foods 2023; 12:1826. [PMID: 37174364 PMCID: PMC10178160 DOI: 10.3390/foods12091826] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Periodontal diseases are oral inflammatory diseases ranging from gingivitis to chronic periodontitis. Porphyromonas gingivalis is one of the major pathogens responsible for severe and chronic periodontitis. Plant extracts with antimicrobial activity could be considered possible alternatives to chlorhexidine, an antiseptic substance used in oral hygiene thatcan cause bacteria resistance. Here, two commercial extracts obtained from Cistus × incanus L. and Scutellaria lateriflora L. were chemically characterized usingUltra-High-Performance Liquid Chromatography (UHPLC) coupled with a Q-Exactive Hybrid Quadrupole Orbitrap Mass Spectrometer. The extracts were studied for their bioaccessibility after simulated in vitro oral digestion, their antimicrobial activity against P. gingivalis, their protective effects against cellular invasion by P. gingivalis, and their antibiofilm activity. The extracts were found to contain very complex mixtures of polyphenols, which were quite stable after in vitro simulated oral digestion and demonstrated mild, dose-dependent inhibitory activity against P. gingivalis growth. This activity increased with the combination of the two extracts. Moreover, the combination of the extracts induced a reduction in P. gingivalis HaCaT invasiveness, and the reduction in biofilm came to around 80%. In conclusion, a combination of C. incanus and S. lateriflora showed promising effects useful in the treatment of gingivitis.
Collapse
Affiliation(s)
- Hammad Ullah
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, NA, Italy; (H.U.); (A.D.M.); (D.G.B.); (L.F.D.L.)
| | - Alessandro Di Minno
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, NA, Italy; (H.U.); (A.D.M.); (D.G.B.); (L.F.D.L.)
- CEINGE-BiotecnologieAvanzate, Via Gaetano Salvatore 486, 80145 Naples, NA, Italy
| | - Anna De Filippis
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, University of Campania “L. Vanvitelli”, Via De Crecchio, 7, 80138 Naples, NA, Italy; (A.D.F.); (M.G.)
| | - Eduardo Sommella
- Department of Pharmacy, University of Salerno, 84084 Fisciano, SA, Italy; (E.S.); (P.C.)
| | - Daniele Giuseppe Buccato
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, NA, Italy; (H.U.); (A.D.M.); (D.G.B.); (L.F.D.L.)
| | - Lorenza Francesca De Lellis
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, NA, Italy; (H.U.); (A.D.M.); (D.G.B.); (L.F.D.L.)
| | - Hesham R. El-Seedi
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, SE-751 24 Uppsala, Sweden;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Shaden A. M. Khalifa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91 Stockholm, Sweden;
| | - Roberto Piccinocchi
- Level 1 Medical Director Anaesthesia and Resuscitation A. U. O. Luigi Vanvitelli, Via Santa Maria di Costantinopoli, 80138 Naples, NA, Italy;
| | - Massimiliano Galdiero
- Department of Experimental Medicine, Section of Microbiology and Clinical Microbiology, University of Campania “L. Vanvitelli”, Via De Crecchio, 7, 80138 Naples, NA, Italy; (A.D.F.); (M.G.)
- UOC of Virology and Microbiology, University Hospital of Campania “Luigi Vanvitelli”, 80138 Naples, NA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, SA, Italy; (E.S.); (P.C.)
- European Biomedical Research Institute of Salerno, Via De Renzi 50, 84125 Salerno, SA, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, NA, Italy; (H.U.); (A.D.M.); (D.G.B.); (L.F.D.L.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
16
|
Morris CJ, Rohn JL, Glickman S, Mansfield KJ. Effective Treatments of UTI—Is Intravesical Therapy the Future? Pathogens 2023; 12:pathogens12030417. [PMID: 36986339 PMCID: PMC10058863 DOI: 10.3390/pathogens12030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Urinary tract infection (UTI) afflicts millions of patients globally each year. While the majority of UTIs are successfully treated with orally administered antibiotics, the impact of oral antibiotics on the host microbiota is under close research scrutiny and the potential for dysbiosis is a cause for concern. Optimal treatment of UTI relies upon the selection of an agent which displays appropriate pharmacokinetic-pharmacodynamic (PK-PD) properties that will deliver appropriately high concentrations in the urinary tract after oral administration. Alternatively, high local concentrations of antibiotic at the urothelial surface can be achieved by direct instillation into the urinary tract. For antibiotics with the appropriate physicochemical properties, this can be of critical importance in cases for which an intracellular urothelial bacterial reservoir is suspected. In this review, we summarise the underpinning biopharmaceutical barriers to effective treatment of UTI and provide an overview of the evidence for the deployment of the intravesical administration route for antibiotics.
Collapse
Affiliation(s)
- Chris J. Morris
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Jennifer L. Rohn
- Division of Medicine, University College London, Royal Free Hospital Campus, Rowland Hill Street, London NW3 2PF, UK
| | | | - Kylie J. Mansfield
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
- Correspondence: ; Tel.: +61-242-215-851
| |
Collapse
|
17
|
Chieng CCY, Kong Q, Liou NSY, Khasriya R, Horsley H. The clinical implications of bacterial pathogenesis and mucosal immunity in chronic urinary tract infection. Mucosal Immunol 2023; 16:61-71. [PMID: 36642381 DOI: 10.1016/j.mucimm.2022.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023]
Abstract
Urinary tract infections (UTIs) exert a significant health and economic cost globally. Approximately one in four people with a previous history of UTI continue to develop recurrent or chronic infections. Research on UTI has primarily concentrated on pathogen behavior, with the focus gradually shifting to encompass the host immune response. However, these are centered on mouse models of Escherichia coli infection, which may not fully recapitulate the infective etiology and immune responses seen in humans. The emerging field of the urobiome also inadvertently confounds the discrimination of true UTI-causing pathogens from commensals. This review aims to present a novel perspective on chronic UTI by linking microbiology with immunology, which is commonly divergent in this field of research. It also describes the challenges in understanding chronic UTI pathogenesis and the human bladder immune response, largely conjectured from murine studies. Lastly, it outlines the shortcomings of current diagnostic methods in identifying individuals with chronic UTI and consequently treating them, potentially aggravating their disease due to mismanagement of prior episodes. This discourse highlights the need to consider these knowledge gaps and encourages more relevant studies of UTIs in humans.
Collapse
Affiliation(s)
| | - Qingyang Kong
- Department of Microbial Diseases, Eastman Dental Institute, University College London, London, United Kingdom
| | - Natasha S Y Liou
- Department of Renal Medicine, University College London, London, United Kingdom; EGA Institute for Women's Health, University College London, London, United Kingdom
| | - Rajvinder Khasriya
- Department of Microbial Diseases, Eastman Dental Institute, University College London, London, United Kingdom
| | - Harry Horsley
- Department of Renal Medicine, University College London, London, United Kingdom.
| |
Collapse
|
18
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
19
|
A Model of Intracellular Persistence of Pseudomonas aeruginosa in Airway Epithelial Cells. Cell Microbiol 2022. [DOI: 10.1155/2022/5431666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pseudomonas aeruginosa (P.a.) is a major human pathogen capable of causing chronic infections in hosts with weakened barrier functions and host defenses, most notably airway infections commonly observed in individuals with the genetic disorder cystic fibrosis (CF). While mainly described as an extracellular pathogen, previous in vitro studies have described the molecular events leading to P.a. internalization in diverse epithelial cell types. However, the long-term fate of intracellular P.a. remains largely unknown. Here, we developed a model allowing for a better understanding of long-term (up to 120 h) intracellular bacterial survival in the airway epithelial cell line BEAS-2B. Using a tobramycin protection assay, we characterized the internalization, long-term intracellular survival, and cytotoxicity of the lab strain PAO1, as well as clinical CF isolates, and conducted analyses at the single-cell level using confocal microscopy and flow cytometry techniques. We observed that infection at low multiplicity of infection allows for intracellular survival up to 120 h post-infection without causing significant host cytotoxicity. Finally, infection with clinical isolates revealed significant strain-to-strain heterogeneity in intracellular survival, including a high persistence phenotype associated with bacterial replication within host cells. Future studies using this model will further elucidate the host and bacterial mechanisms that promote P. aeruginosa intracellular persistence in airway epithelial cells, a potentially unrecognized bacterial reservoir during chronic infections.
Collapse
|
20
|
Tomasek K, Leithner A, Glatzova I, Lukesch MS, Guet CC, Sixt M. Type 1 piliated uropathogenic Escherichia coli hijack the host immune response by binding to CD14. eLife 2022; 11:e78995. [PMID: 35881547 PMCID: PMC9359703 DOI: 10.7554/elife.78995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
A key attribute of persistent or recurring bacterial infections is the ability of the pathogen to evade the host's immune response. Many Enterobacteriaceae express type 1 pili, a pre-adapted virulence trait, to invade host epithelial cells and establish persistent infections. However, the molecular mechanisms and strategies by which bacteria actively circumvent the immune response of the host remain poorly understood. Here, we identified CD14, the major co-receptor for lipopolysaccharide detection, on mouse dendritic cells (DCs) as a binding partner of FimH, the protein located at the tip of the type 1 pilus of Escherichia coli. The FimH amino acids involved in CD14 binding are highly conserved across pathogenic and non-pathogenic strains. Binding of the pathogenic strain CFT073 to CD14 reduced DC migration by overactivation of integrins and blunted expression of co-stimulatory molecules by overactivating the NFAT (nuclear factor of activated T-cells) pathway, both rate-limiting factors of T cell activation. This response was binary at the single-cell level, but averaged in larger populations exposed to both piliated and non-piliated pathogens, presumably via the exchange of immunomodulatory cytokines. While defining an active molecular mechanism of immune evasion by pathogens, the interaction between FimH and CD14 represents a potential target to interfere with persistent and recurrent infections, such as urinary tract infections or Crohn's disease.
Collapse
Affiliation(s)
- Kathrin Tomasek
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | | | - Ivana Glatzova
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | | | - Calin C Guet
- Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Michael Sixt
- Institute of Science and Technology AustriaKlosterneuburgAustria
| |
Collapse
|
21
|
Fleming BA, Blango MG, Rousek AA, Kincannon WM, Tran A, Lewis A, Russell C, Zhou Q, Baird LM, Barber A, Brannon JR, Beebout C, Bandarian V, Hadjifrangiskou M, Howard M, Mulvey M. A tRNA modifying enzyme as a tunable regulatory nexus for bacterial stress responses and virulence. Nucleic Acids Res 2022; 50:7570-7590. [PMID: 35212379 PMCID: PMC9303304 DOI: 10.1093/nar/gkac116] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Post-transcriptional modifications can impact the stability and functionality of many different classes of RNA molecules and are an especially important aspect of tRNA regulation. It is hypothesized that cells can orchestrate rapid responses to changing environmental conditions by adjusting the specific types and levels of tRNA modifications. We uncovered strong evidence in support of this tRNA global regulation hypothesis by examining effects of the well-conserved tRNA modifying enzyme MiaA in extraintestinal pathogenic Escherichia coli (ExPEC), a major cause of urinary tract and bloodstream infections. MiaA mediates the prenylation of adenosine-37 within tRNAs that decode UNN codons, and we found it to be crucial to the fitness and virulence of ExPEC. MiaA levels shifted in response to stress via a post-transcriptional mechanism, resulting in marked changes in the amounts of fully modified MiaA substrates. Both ablation and forced overproduction of MiaA stimulated translational frameshifting and profoundly altered the ExPEC proteome, with variable effects attributable to UNN content, changes in the catalytic activity of MiaA, or availability of metabolic precursors. Cumulatively, these data indicate that balanced input from MiaA is critical for optimizing cellular responses, with MiaA acting much like a rheostat that can be used to realign global protein expression patterns.
Collapse
Affiliation(s)
- Brittany A Fleming
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Matthew G Blango
- Junior Research Group RNA Biology of Fungal Infections, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute (Leibniz-HKI), 07745 Jena, Germany
| | - Alexis A Rousek
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | - Alexander Tran
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Adam J Lewis
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Colin W Russell
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Qin Zhou
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Lisa M Baird
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Amelia E Barber
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - John R Brannon
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Connor J Beebout
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Vahe Bandarian
- Department of Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Maria Hadjifrangiskou
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael T Howard
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Matthew A Mulvey
- Division of Microbiology and Immunology, Pathology Department, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
22
|
Krawczyk B, Wysocka M, Michalik M, Gołębiewska J. Urinary Tract Infections Caused by K. pneumoniae in Kidney Transplant Recipients – Epidemiology, Virulence and Antibiotic Resistance. Front Cell Infect Microbiol 2022; 12:861374. [PMID: 35531341 PMCID: PMC9068989 DOI: 10.3389/fcimb.2022.861374] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Abstract
Urinary tract infections are the most common complication in kidney transplant recipients, possibly resulting in the deterioration of a long-term kidney allograft function and an increased risk of recipient’s death. K. pneumoniae has emerged as one of the most prevalent etiologic agents in the context of recurrent urinary tract infections, especially with multidrug resistant strains. This paper discusses the epidemiology and risk factors associated with urinary tract infections in kidney transplant recipients, multi-drug resistance of K. pneumoniae (ESBL, KPC, NDM), treatment and pathogenesis of K. pneumoniae infections, and possible causes of recurrent UTIs. It also addresses the issue of colonization/becoming a carrier of K. pneumoniae in the gastrointestinal tract and asymptomatic bacteriuria in relation to a symptomatic UTI development and epidemiology.
Collapse
Affiliation(s)
- Beata Krawczyk
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
- *Correspondence: Beata Krawczyk,
| | - Magdalena Wysocka
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | | | - Justyna Gołębiewska
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
23
|
Robinson CK, Saenkham-Huntsinger P, Hanson BS, Adams LG, Subashchandrabose S. Vaginal Inoculation of Uropathogenic Escherichia coli during Estrus Leads to Genital and Renal Colonization. Infect Immun 2022; 90:e0053221. [PMID: 35357220 PMCID: PMC9022555 DOI: 10.1128/iai.00532-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Urinary tract infection (UTI) is one of the most prevalent bacterial infections, particularly in women, children, and the elderly. Uropathogenic Escherichia coli (UPEC) is the predominant etiological agent of UTI. Uropathogens are directly instilled in the urinary bladder, bypassing the lower urogenital tract, in the widely used murine model of UTI. We assessed whether vaginal inoculation of UPEC led to UTI and how stages of the estrous cycle would impact bacterial colonization in mice. Mice in proestrus, estrus, metestrus, and diestrus were identified by vaginal cytology and inoculated with UPEC in the vaginal tract. Mice were euthanized 1 day after infection, and bacterial loads in the urogenital tract, liver, and spleen were enumerated. Mice in estrus exhibited the highest and most consistent UPEC burdens in all organs, except the bladder. Vaginal inoculation resulted in bladder colonization in a UPEC strain-specific manner. In contrast, transurethral inoculation of UPEC led to bladder colonization. Importantly, inoculation by both routes led to vaginal and uterine colonization and concomitant systemic dissemination to the spleen and liver. The kinetics of bacterial colonization over 2 weeks following vaginal inoculation was comparable in the urogenital tract. Tissue sections revealed the induction of vaginitis and cystitis upon the vaginal instillation of UPEC. In summary, vaginal inoculation of UPEC in mice during estrus represents a novel approach to investigate infection of the kidneys and genital tract and systemic dissemination from the urogenital tract. Our findings suggest that estrogen primes the urogenital tract to create a conducive milieu for UPEC colonization.
Collapse
Affiliation(s)
- Christen K. Robinson
- Comparative Medicine Program, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Panatda Saenkham-Huntsinger
- Comparative Medicine Program, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Braden S. Hanson
- Comparative Medicine Program, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - L. Garry Adams
- Comparative Medicine Program, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Sargurunathan Subashchandrabose
- Comparative Medicine Program, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
24
|
Non-antibiotic Approaches to Preventing Pediatric UTIs: a Role for D-Mannose, Cranberry, and Probiotics? Curr Urol Rep 2022; 23:113-127. [PMID: 35441976 DOI: 10.1007/s11934-022-01094-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW While antibiotics have been a staple in the management and even prevention of urinary tract infections (UTIs), it is not without significant consequences due to intolerance and development of antibiotic resistant bacteria. These concerns necessitate alternatives to antibiotic use in the management of pediatric UTIs. This review seeks to evaluate non-antibiotic means of preventing UTI in the pediatric population. RECENT FINDINGS The search for preventative alternatives to antibiotics has included D-mannose, cranberry, and probiotics. These products similarly work through competitive inhibition of uropathogens in the urinary tract. Pediatric studies exist highlighting the use of cranberry extract/juice and probiotics in UTI prevention, although significant heterogeneity amongst studies have limited overarching recommendations for their use. Data of D-mannose use is extrapolated from adult literature. More studies are required in the utility of each treatment, with some emphasis on larger sample sizes and clarifications regarding dosing and formulation.
Collapse
|
25
|
Bladder epithelial cell phosphate transporter inhibition protects mice against uropathogenic Escherichia coli infection. Cell Rep 2022; 39:110698. [PMID: 35443182 DOI: 10.1016/j.celrep.2022.110698] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/12/2022] [Accepted: 03/28/2022] [Indexed: 12/20/2022] Open
Abstract
Urinary tract infections are predominantly caused by uropathogenic Escherichia coli (UPEC). UPEC infects bladder epithelial cells (BECs) via fusiform vesicles, escapes into the cytosol to evade exocytosis, and establishes intracellular bacterial communities (IBCs) for the next round of infection. The UPEC vesicle escape mechanism remains unclear. Here we show that UPEC senses host immune responses and initiates escape by upregulating a key phospholipase. The UPEC phospholipase PldA disrupts the vesicle membrane, and pldA expression is activated by phosphate reduction in vesicles. The host phosphate transporter PIT1 is located on the fusiform vesicle membrane, transporting phosphate into the cytosol. UPEC infection upregulates PIT1 via nuclear factor κB (NF-κB), resulting in phosphate reduction. Silencing PIT1 blocks UPEC vesicle escape in BECs, inhibits IBC formation in mouse bladders, and protects mice from UPEC infection. Our results shed light on pathogenic bacteria responding to intracellular phosphate shortage and tackling host defense and provide insights for development of new therapeutic agents to treat UPEC infection.
Collapse
|
26
|
Kaushik V, Sharma S, Tiwari M, Tiwari V. Anti-persister strategies against stress induced bacterial persistence. Microb Pathog 2022; 164:105423. [PMID: 35092834 DOI: 10.1016/j.micpath.2022.105423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 01/22/2023]
Abstract
The increase in antibiotic non-responsive bacteria is the leading concern in current research-oriented to eliminate pathogens. Nowadays, the excess use of antibiotics without specifically understanding the potentiality of killing pathogens and bacterial survival patterns has helped bacteria emerge indefatigably. Bacteria use various mechanisms such as resistance, persistence, and tolerance to ensure survival. Among these, persistence is a mechanism by which bacteria reside in their dormant state, bypassing the effects of treatments, making it crucial for bacterial survival. Persistent bacterial cells arise from the normal bacterial population as a slow-growing subset of bacteria with no metabolic flux. This behavior renders it to survive for a longer duration and at higher concentrations of antibiotics. They are one of the underlying causes of recurrence of bacterial infections. The present article explains the detailed molecular mechanisms and strategies of bacterial persistence, including the toxin-antitoxin modules, DNA damage, the formation of inactive ribosomal complexes, (p)ppGpp network, antibiotic-induced persistence, which are triggered by drug-induced stress. The article also comprehensively covers the epigenetic memory of persistence in bacteria, and anti-persistent therapeutics like antimicrobial molecules, synthetic peptides, acyldepsipeptide antibiotics, and endolysin therapy to reduce persister cell formation and control their frequency. These strategies could be utilized in combating the pathogenic bacteria undergoing persistence.
Collapse
Affiliation(s)
- Vaishali Kaushik
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Saroj Sharma
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Bandarsindri, Ajmer, 305817, India.
| |
Collapse
|
27
|
Montalbetti N, Dalghi MG, Bastacky SI, Clayton DR, Ruiz WG, Apodaca G, Carattino MD. Bladder infection with uropathogenic Escherichia coli increases the excitability of afferent neurons. Am J Physiol Renal Physiol 2022; 322:F1-F13. [PMID: 34779263 PMCID: PMC8698541 DOI: 10.1152/ajprenal.00167.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/05/2021] [Indexed: 01/03/2023] Open
Abstract
Urinary tract infections (UTIs) cause bladder hyperactivity and pelvic pain, but the underlying causes of these symptoms remain unknown. We investigated whether afferent sensitization contributes to the bladder overactivity and pain observed in mice suffering from experimentally induced bacterial cystitis. Inoculation of mouse bladders with the uropathogenic Escherichia coli strain UTI89 caused pelvic allodynia, increased voiding frequency, and prompted an acute inflammatory process marked by leukocytic infiltration and edema of the mucosa. Compared with controls, isolated bladder sensory neurons from UTI-treated mice exhibited a depolarized resting membrane potential, lower action potential threshold and rheobase, and increased firing in response to suprathreshold stimulation. To determine whether bacterial virulence factors can contribute to the sensitization of bladder afferents, neurons isolated from naïve mice were incubated with supernatants collected from bacterial cultures with or depleted of lipopolysaccharide (LPS). Supernatants containing LPS prompted the sensitization of bladder sensory neurons with both tetrodotoxin (TTX)-resistant and TTX-sensitive action potentials. However, bladder sensory neurons with TTX-sensitive action potentials were not affected by bacterial supernatants depleted of LPS. Unexpectedly, ultrapure LPS increased the excitability only of bladder sensory neurons with TTX-resistant action potentials, but the supplementation of supernatants depleted of LPS with ultrapure LPS resulted in the sensitization of both population of bladder sensory neurons. In summary, the results of our study indicate that multiple virulence factors released from UTI89 act on bladder sensory neurons to prompt their sensitization. These sensitized bladder sensory neurons mediate, at least in part, the bladder hyperactivity and pelvic pain seen in mice inoculated with UTI89.NEW & NOTEWORTHY Urinary tract infection (UTI) produced by uropathogenic Escherichia coli (UPEC) promotes sensitization of bladder afferent sensory neurons with tetrodotoxin-resistant and tetrodotoxin-sensitive action potentials. Lipopolysaccharide and other virulence factors produced by UPEC contribute to the sensitization of bladder afferents in UTI. In conclusion, sensitized afferents contribute to the voiding symptoms and pelvic pain present in mice bladder inoculated with UPEC.
Collapse
Affiliation(s)
- Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marianela G Dalghi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dennis R Clayton
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wily G Ruiz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Kell DB. The Transporter-Mediated Cellular Uptake and Efflux of Pharmaceutical Drugs and Biotechnology Products: How and Why Phospholipid Bilayer Transport Is Negligible in Real Biomembranes. Molecules 2021; 26:5629. [PMID: 34577099 PMCID: PMC8470029 DOI: 10.3390/molecules26185629] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Over the years, my colleagues and I have come to realise that the likelihood of pharmaceutical drugs being able to diffuse through whatever unhindered phospholipid bilayer may exist in intact biological membranes in vivo is vanishingly low. This is because (i) most real biomembranes are mostly protein, not lipid, (ii) unlike purely lipid bilayers that can form transient aqueous channels, the high concentrations of proteins serve to stop such activity, (iii) natural evolution long ago selected against transport methods that just let any undesirable products enter a cell, (iv) transporters have now been identified for all kinds of molecules (even water) that were once thought not to require them, (v) many experiments show a massive variation in the uptake of drugs between different cells, tissues, and organisms, that cannot be explained if lipid bilayer transport is significant or if efflux were the only differentiator, and (vi) many experiments that manipulate the expression level of individual transporters as an independent variable demonstrate their role in drug and nutrient uptake (including in cytotoxicity or adverse drug reactions). This makes such transporters valuable both as a means of targeting drugs (not least anti-infectives) to selected cells or tissues and also as drug targets. The same considerations apply to the exploitation of substrate uptake and product efflux transporters in biotechnology. We are also beginning to recognise that transporters are more promiscuous, and antiporter activity is much more widespread, than had been realised, and that such processes are adaptive (i.e., were selected by natural evolution). The purpose of the present review is to summarise the above, and to rehearse and update readers on recent developments. These developments lead us to retain and indeed to strengthen our contention that for transmembrane pharmaceutical drug transport "phospholipid bilayer transport is negligible".
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, UK;
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs Lyngby, Denmark
- Mellizyme Biotechnology Ltd., IC1, Liverpool Science Park, Mount Pleasant, Liverpool L3 5TF, UK
| |
Collapse
|
29
|
Sharma K, Dhar N, Thacker VV, Simonet TM, Signorino-Gelo F, Knott GW, McKinney JD. Dynamic persistence of UPEC intracellular bacterial communities in a human bladder-chip model of urinary tract infection. eLife 2021; 10:66481. [PMID: 34219648 PMCID: PMC8354636 DOI: 10.7554/elife.66481] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 07/04/2021] [Indexed: 12/23/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) proliferate within superficial bladder umbrella cells to form intracellular bacterial communities (IBCs) during early stages of urinary tract infections. However, the dynamic responses of IBCs to host stresses and antibiotic therapy are difficult to assess in situ. We develop a human bladder-chip model wherein umbrella cells and bladder microvascular endothelial cells are co-cultured under flow in urine and nutritive media respectively, and bladder filling and voiding mimicked mechanically by application and release of linear strain. Using time-lapse microscopy, we show that rapid recruitment of neutrophils from the vascular channel to sites of infection leads to swarm and neutrophil extracellular trap formation but does not prevent IBC formation. Subsequently, we tracked bacterial growth dynamics in individual IBCs through two cycles of antibiotic administration interspersed with recovery periods which revealed that the elimination of bacteria within IBCs by the antibiotic was delayed, and in some instances, did not occur at all. During the recovery period, rapid proliferation in a significant fraction of IBCs reseeded new foci of infection through bacterial shedding and host cell exfoliation. These insights reinforce a dynamic role for IBCs as harbors of bacterial persistence, with significant consequences for non-compliance with antibiotic regimens. Urinary tract infections are one of the most common reasons people need antibiotics. These bacterial infections are typically caused by uropathogenic Escherichia coli (also known as UPEC), which either float freely in the urine and wash away when the bladder empties, or form communities inside cells that the bladder struggles to clear. It is possible that the bacteria living within cells are also more protected from the immune system and antibiotics. But this is hard to study in animal models. To overcome this, Sharma et al. built a ‘bladder-chip’ which mimics the interface between the blood vessels and the tissue layers of the human bladder. Similar chip devices have also been made for other organs. However, until now, no such model had been developed for the bladder. On the chip created by Sharma et al. is a layer of bladder cells which sit at the bottom of a channel filled with diluted human urine. These cells were infected with UPEC, and then imaged over time to see how the bacteria moved, interacted with the bladder cells, and aggregated together. Immune cells from human blood were then added to a vascular channel underneath the bladder tissue, which is coated with endothelial cells that normally line blood vessels. The immune cells rapidly crossed the endothelial barrier and entered the bladder tissue, and swarmed around sites of infection. In some instances, they released the contents of their cells to form net-like traps to catch the bacteria. But these traps failed to remove the bacteria living inside bladder cells. Antibiotics were then added to the urine flowing over the bladder cells as well as the vascular channel, similar to how drugs would be delivered in live human tissue. Sharma et al. discovered that the antibiotics killed bacteria residing in bladder cells slower than bacteria floating freely in the urine. Furthermore, they found that bacteria living in tightly packed communities within bladder cells were more likely to survive treatment and go on to re-infect other parts of the tissue. Antibiotic resistance is a pressing global challenge, and recurrent urinary tract infections are a significant contributor. The bladder-chip presented here could further our understanding of how these bacterial infections develop in vivo and how good antibiotics are at removing them. This could help researchers identify the best dosing and treatment strategies, as well as provide a platform for rapidly testing new antibiotic drugs and other therapies.
Collapse
Affiliation(s)
- Kunal Sharma
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Neeraj Dhar
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Vivek V Thacker
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Thomas M Simonet
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Francois Signorino-Gelo
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Graham W Knott
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - John D McKinney
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
30
|
Murray BO, Flores C, Williams C, Flusberg DA, Marr EE, Kwiatkowska KM, Charest JL, Isenberg BC, Rohn JL. Recurrent Urinary Tract Infection: A Mystery in Search of Better Model Systems. Front Cell Infect Microbiol 2021; 11:691210. [PMID: 34123879 PMCID: PMC8188986 DOI: 10.3389/fcimb.2021.691210] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Urinary tract infections (UTIs) are among the most common infectious diseases worldwide but are significantly understudied. Uropathogenic E. coli (UPEC) accounts for a significant proportion of UTI, but a large number of other species can infect the urinary tract, each of which will have unique host-pathogen interactions with the bladder environment. Given the substantial economic burden of UTI and its increasing antibiotic resistance, there is an urgent need to better understand UTI pathophysiology - especially its tendency to relapse and recur. Most models developed to date use murine infection; few human-relevant models exist. Of these, the majority of in vitro UTI models have utilized cells in static culture, but UTI needs to be studied in the context of the unique aspects of the bladder's biophysical environment (e.g., tissue architecture, urine, fluid flow, and stretch). In this review, we summarize the complexities of recurrent UTI, critically assess current infection models and discuss potential improvements. More advanced human cell-based in vitro models have the potential to enable a better understanding of the etiology of UTI disease and to provide a complementary platform alongside animals for drug screening and the search for better treatments.
Collapse
Affiliation(s)
- Benjamin O. Murray
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| | - Carlos Flores
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| | - Corin Williams
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Deborah A. Flusberg
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Elizabeth E. Marr
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Karolina M. Kwiatkowska
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| | - Joseph L. Charest
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Brett C. Isenberg
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Jennifer L. Rohn
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| |
Collapse
|
31
|
Othman MA, Ezzat HM, Rizk DEE, Kamal AH, Al-Mahameed AE, Marwani AM, Bindyna KM, Salvatore S. Induction of bacterial cystitis in female rabbits by uropathogenic Escherichia coli and the differences between the bladder dome and trigone. Ultrastruct Pathol 2021; 45:159-166. [PMID: 34030600 DOI: 10.1080/01913123.2021.1920653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This study is designed to evaluate the histological effects of uropathogenic Escherichia coli (UPEC) infection in the urinary bladder of female rabbits and compare the differences between the dome and trigone. Bacterial cystitis was induced in 13 female rabbits by transurethral inoculation of UPEC into the urinary bladder. Eight animals served as controls. Urine samples were collected by catheterization and cultured for bacterial growth after 12 and 24 hours then every 48 hours. Infection was defined as ≥(1X105) colony-forming unit/ml of UPEC in the first two urine samples. Bladder dome and trigonal specimens were examined by light and scanning electron microscopy eight days after infection. There was a sustained increase in bacterial count, with urethral bleeding and rabbit weakness suggesting bladder colonization in the 10/13 study group (77%). Infection was not demonstrated in two animals and was spontaneously cleared in the third after 48 hours. No control animals developed an infection. In infected rabbits (n = 10), the dome showed inflammatory changes including the epithelial loss or thinning, inflammatory cell infiltration, and congested blood vessels compared to controls. The trigone showed a more pronounced inflammatory response than the dome. The presence of urinary bacterial growth, infection manifestations, and inflammatory changes that were more severe in the trigone than in the dome indicate successful bacterial inoculation and induction of cystitis. This animal model can be used for clinical trials on female cystitis. Our histological findings support a possible role of trigone in the pathogenesis of urinary tract infection.
Collapse
Affiliation(s)
- Manal A Othman
- Department of Anatomy, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain.,Department of Histology, Faculty of Medicine, Assiut University, Assuit, Egypt
| | - Hicham M Ezzat
- Department of Microbiology, Immunology & Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain.,Department of Microbiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Diaa E E Rizk
- Department of Obstetrics and Gynecology, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Amer H Kamal
- Department of Physiology, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Ali E Al-Mahameed
- Department of Microbiology, Immunology & Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Ammar M Marwani
- Animal Facility Laboratory Manager, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Khalid M Bindyna
- Department of Microbiology, Immunology & Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Stefano Salvatore
- Urogynecology Unit, Department of Obstetrics and Gynecology, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
32
|
Penaranda C, Chumbler NM, Hung DT. Dual transcriptional analysis reveals adaptation of host and pathogen to intracellular survival of Pseudomonas aeruginosa associated with urinary tract infection. PLoS Pathog 2021; 17:e1009534. [PMID: 33901267 PMCID: PMC8102004 DOI: 10.1371/journal.ppat.1009534] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/06/2021] [Accepted: 04/06/2021] [Indexed: 11/21/2022] Open
Abstract
Long-term survival of bacterial pathogens during persistent bacterial infections can be associated with antibiotic treatment failure and poses a serious public health problem. Infections caused by the Gram-negative pathogen Pseudomonas aeruginosa, which can cause both acute and chronic infections, are particularly challenging due to its high intrinsic resistance to antibiotics. The ineffectiveness of antibiotics is exacerbated when bacteria reside intracellularly within host cells where they can adopt a drug tolerant state. While the early steps of adherence and entry of P. aeruginosa into mammalian cells have been described, the subsequent fate of internalized bacteria, as well as host and bacterial molecular pathways facilitating bacterial long-term survival, are not well defined. In particular, long-term survival within bladder epithelial cells has not been demonstrated and this may have important implications for the understanding and treatment of UTIs caused by P. aeruginosa. Here, we demonstrate and characterize the intracellular survival of wild type (WT) P. aeruginosa inside bladder epithelial cells and a mutant with a disruption in the bacterial two-component regulator AlgR that is unable to survive intracellularly. Using simultaneous dual RNA-seq transcriptional profiling, we define the transcriptional response of intracellular bacteria and their corresponding invaded host cells. The bacterial transcriptional response demonstrates that WT bacteria rapidly adapt to the stress encountered in the intracellular environment in contrast to ΔalgR bacteria. Analysis of the host transcriptional response to invasion suggests that the NF-κB signaling pathway, previously shown to be required for extracellular bacterial clearance, is paradoxically also required for intracellular bacterial survival. Lastly, we demonstrate that intracellular survival is important for pathogenesis of P. aeruginosa in vivo using a model of murine urinary tract infection. We propose that the unappreciated ability of P. aeruginosa to survive intracellularly may play an important role in contributing to the chronicity and recurrence of P. aeruginosa in urinary tract infections. Chronic persistent bacterial infections are a serious and growing public health problem worsened by the rise in antibiotic resistance, yet new approaches for treating these infections are lacking. These long-term infections can occur when bacteria invade and survive inside host cells where they can hide from the immune system and become less susceptible to killing by antibiotics. Pseudomonas aeruginosa, a bacterium conventionally considered an extracellular pathogen, can cause chronic infections of many organ systems, including the urinary tract. Here, we show that P. aeruginosa can in fact survive inside bladder epithelial cells and becomes tolerant to antibiotic treatment. Using gene expression analysis, we show that bacteria quickly adapt to the intracellular environment while the corresponding host cells upregulate the NF-κB signaling pathway. We demonstrate that this response, which had previously been shown to be required for clearance of extracellular bacteria, is paradoxically also required for survival of intracellular bacteria. We propose that the ability of P. aeruginosa to survive intracellularly plays an important role in contributing to the chronicity and recurrence of P. aeruginosa infections and that targeting host pathways, such as NF-κB signaling, could transform our ability to manage chronic and/or recurrent infections.
Collapse
Affiliation(s)
- Cristina Penaranda
- Infectious Disease and Microbiome Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nicole M. Chumbler
- Infectious Disease and Microbiome Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Deborah T. Hung
- Infectious Disease and Microbiome Program, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
33
|
Gill K, Horsley H, Swamy S, Khasriya R, Malone-Lee J. A prospective observational study of urinary cytokines and inflammatory response in patients with Overactive Bladder Syndrome. BMC Urol 2021; 21:39. [PMID: 33740940 PMCID: PMC7980577 DOI: 10.1186/s12894-021-00809-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/03/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Contemporary studies have discredited the methods used to exclude urinary tract infection (UTI) when treating overactive bladder (OAB). Thus we must revisit the OAB phenotype to check that UTI has not been overlooked. AIMS To examine the differences in urinary cytokines IL6 and lactoferrin in OAB patients compared to controls, with references to microscopy of urine and enhanced quantitative urine culture. METHODS A blinded, prospective cohort study with normal controls using six repeated measures, achieved two-monthly, over 12 months. RESULTS The differences between patients and controls in urine IL6 (F = 49.0, p < .001) and lactoferrin (F = 228.5, p < .001) were significant and of a magnitude to have clinical implications. These differences were for lactoferrin correlated to symptoms (9.3, p = .003); for both to pyuria (IL6 F = 66.2, p < .001, Lactoferrin F = 73.9, p < .001); and for IL6 microbial abundance (F = 5.1, p = .024). The pathological markers had been missed by urinary dipsticks and routine MSU culture. CONCLUSION The OAB phenotype may encompass patients with UTI that is being overlooked because of the failure of standard screening methods.
Collapse
Affiliation(s)
- Kiren Gill
- Women’s Health, Whittington Health NHS Trust, Magdala Avenue, London, N19 5FN UK
| | - Harry Horsley
- Bladder Infection and Immunity Group (BIIG), Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF UK
| | - Sheela Swamy
- Women’s Health, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH UK
| | - Rajvinder Khasriya
- Women’s Health, Whittington Health NHS Trust, Magdala Avenue, London, N19 5FN UK
| | - James Malone-Lee
- Bladder Infection and Immunity Group (BIIG), Department of Renal Medicine, Division of Medicine, University College London, 10 Harley Street, London, W1G 9PF UK
| |
Collapse
|
34
|
Gomes AÉI, Pacheco T, Dos Santos CDS, Pereira JA, Ribeiro ML, Darrieux M, Ferraz LFC. Functional Insights From KpfR, a New Transcriptional Regulator of Fimbrial Expression That Is Crucial for Klebsiella pneumoniae Pathogenicity. Front Microbiol 2021; 11:601921. [PMID: 33552015 PMCID: PMC7861041 DOI: 10.3389/fmicb.2020.601921] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022] Open
Abstract
Although originally known as an opportunistic pathogen, Klebsiella pneumoniae has been considered a worldwide health threat nowadays due to the emergence of hypervirulent and antibiotic-resistant strains capable of causing severe infections not only on immunocompromised patients but also on healthy individuals. Fimbriae is an essential virulence factor for K. pneumoniae, especially in urinary tract infections (UTIs), because it allows the pathogen to adhere and invade urothelial cells and to form biofilms on biotic and abiotic surfaces. The importance of fimbriae for K. pneumoniae pathogenicity is highlighted by the large number of fimbrial gene clusters on the bacterium genome, which requires a coordinated and finely adjusted system to control the synthesis of these structures. In this work, we describe KpfR as a new transcriptional repressor of fimbrial expression in K. pneumoniae and discuss its role in the bacterium pathogenicity. K. pneumoniae with disrupted kpfR gene exhibited a hyperfimbriated phenotype with enhanced biofilm formation and greater adhesion to and replication within epithelial host cells. Nonetheless, the mutant strain was attenuated for colonization of the bladder in a murine model of urinary tract infection. These results indicate that KpfR is an important transcriptional repressor that, by negatively controlling the expression of fimbriae, prevents K. pneumoniae from having a hyperfimbriated phenotype and from being recognized and eliminated by the host immune system.
Collapse
Affiliation(s)
- Ana Érika Inácio Gomes
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Thaisy Pacheco
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | | | - José Aires Pereira
- Laboratório de Biologia Molecular e Celular de Tumores, Universidade São Francisco, Bragança Paulista, Brazil
| | - Marcelo Lima Ribeiro
- Laboratório de Imunofarmacologia e Biologia Molecular, Universidade São Francisco, Bragança Paulista, Brazil
| | - Michelle Darrieux
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lúcio Fábio Caldas Ferraz
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| |
Collapse
|
35
|
Huang WC, Hashimoto M, Shih YL, Wu CC, Lee MF, Chen YL, Wu JJ, Wang MC, Lin WH, Hong MY, Teng CH. Peptidoglycan Endopeptidase Spr of Uropathogenic Escherichia coli Contributes to Kidney Infections and Competitive Fitness During Bladder Colonization. Front Microbiol 2021; 11:586214. [PMID: 33391204 PMCID: PMC7774453 DOI: 10.3389/fmicb.2020.586214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/25/2020] [Indexed: 11/27/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the most common pathogen of urinary tract infections (UTIs). Antibiotic therapy is the conventional measure to manage such infections. However, the rapid emergence of antibiotic resistance has reduced the efficacy of antibiotic treatment. Given that the bacterial factors required for the full virulence of the pathogens are potential therapeutic targets, identifying such factors may facilitate the development of novel therapeutic strategies against UPEC UTIs. The peptidoglycan (PG) endopeptidase Spr (also named MepS) is required for PG biogenesis in E. coli. In the present study, we found that Spr deficiency attenuated the ability of UPEC to infect kidneys and induced a fitness defect during bladder colonization in a mouse model of UTI. Based on the liquid chromatography (LC)/mass spectrometry (MS)/MS analysis of the bacterial envelope, spr deletion changed the levels of some envelope-associated proteins, suggesting that Spr deficiency interfere with the components of the bacterial structure. Among the proteins, FliC was significantly downregulated in the spr mutant, which is resulted in reduced motility. Lack of Spr might hinder the function of the flagellar transcriptional factor FlhDC to decrease FliC expression. The motility downregulation contributed to the reduced fitness in urinary tract colonization. Additionally, spr deletion compromised the ability of UPEC to evade complement-mediated attack and to resist intracellular killing of phagocytes, consequently decreasing UPEC bloodstream survival. Spr deficiency also interfered with the UPEC morphological switch from bacillary to filamentous shapes during UTI. It is known that bacterial filamentation protects UPEC from phagocytosis by phagocytes. In conclusion, Spr deficiency was shown to compromise multiple virulence properties of UPEC, leading to attenuation of the pathogen in urinary tract colonization and bloodstream survival. These findings indicate that Spr is a potential antimicrobial target for further studies attempting to develop novel strategies in managing UPEC UTIs.
Collapse
Affiliation(s)
- Wen-Chun Huang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Masayuki Hashimoto
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ling Shih
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chia-Ching Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Mei-Feng Lee
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - Ya-Lei Chen
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - Jiunn-Jong Wu
- Department of Biotechnology and Laboratory Science in Medicine, School of Biomedical Science and Engineering, National Yang Ming University, Taipei, Taiwan
| | - Ming-Cheng Wang
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Hung Lin
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Yuan Hong
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Hao Teng
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
36
|
Lau WK, Dharmasena D, Horsley H, Jafari NV, Malone-Lee J, Stride E, Edirisinghe M, Rohn JL. Novel antibiotic-loaded particles conferring eradication of deep tissue bacterial reservoirs for the treatment of chronic urinary tract infection. J Control Release 2020; 328:490-502. [PMID: 32882271 DOI: 10.1016/j.jconrel.2020.08.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/09/2020] [Accepted: 08/24/2020] [Indexed: 11/24/2022]
Abstract
A significant proportion of urinary tract infection (UTI) patients experience recurrent episodes, due to deep tissue infection and treatment-resistant bacterial reservoirs. Direct bladder instillation of antibiotics has proved disappointing in treating UTI, likely due to the failure of infused antibiotics to penetrate the bladder epithelium and accumulate to high enough levels to kill intracellular bacteria. This work investigates the use of nitrofurantoin loaded poly(lactic-co-glycolic acid) (PLGA) particles to improve delivery to intracellular targets for the treatment of chronic UTI. Using electrohydrodynamic atomisation, we produced particles with an average diameter of 2.8 μm. In broth culture experiments, the biodegradable particles were effective against a number of UTI-relevant bacterial strains. Dye-loaded particles demonstrated that intracellular delivery was achieved in all cells in 2D cultures of a human bladder epithelial progenitor cell line in a dose-dependent manner, achieving far higher efficiency and concentration than equivalent quantities of free drug. Time-lapse video microscopy confirmed that delivery occurred within 30 min of administration, to 100% of cells. Moreover, the particles were able to deliver the drug to cells through multiple layers of a 3D human bladder organoid model causing minimal cell toxicity, displaying superior killing of bacterial reservoirs harboured within bladder cells compared with unencapsulated drug. The particles were also able to kill bacterial biofilms more effectively than the free drug. These results illustrate the potential for using antibiotic-loaded microparticles to effectively treat chronic UTIs. Such a delivery method could be extrapolated to other clinical indications where robust intracellular delivery is required, such as oncology and gene therapy.
Collapse
Affiliation(s)
- Wai K Lau
- Department of Mechanical Engineering, University College London, London, UK
| | - Dhanuson Dharmasena
- Department of Renal Medicine, Division of Medicine, University College London, London, UK
| | - Harry Horsley
- Department of Renal Medicine, Division of Medicine, University College London, London, UK
| | - Nazila V Jafari
- Department of Renal Medicine, Division of Medicine, University College London, London, UK
| | - James Malone-Lee
- Department of Renal Medicine, Division of Medicine, University College London, London, UK
| | - Eleanor Stride
- The Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London, London, UK
| | - Jennifer L Rohn
- Department of Renal Medicine, Division of Medicine, University College London, London, UK.
| |
Collapse
|
37
|
Insights into the Oxidative Stress Response of Salmonella enterica serovar Enteritidis Revealed by the Next Generation Sequencing Approach. Antioxidants (Basel) 2020; 9:antiox9090849. [PMID: 32927804 PMCID: PMC7555449 DOI: 10.3390/antiox9090849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/27/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022] Open
Abstract
As a facultative intracellular pathogen, Salmonella Enteritidis must develop an effective oxidative stress response to survive exposure to reactive oxygen species within the host. To study this defense mechanism, we carried out a series of oxidative stress assays in parallel with a comparative transcriptome analyses using a next generation sequencing approach. It was shown that the expression of 45% of the genome was significantly altered upon exposure to H2O2. Quantitatively the most significant (≥100 fold) gene expression alterations were observed among genes encoding the sulfur utilization factor of Fe-S cluster formation and iron homeostasis. Our data point out the multifaceted nature of the oxidative stress response. It includes not only numerous mechanisms of DNA and protein repair and redox homeostasis, but also the key genes associated with osmotic stress, multidrug efflux, stringent stress, decrease influx of small molecules, manganese and phosphate starvation stress responses. Importantly, this study revealed that oxidatively stressed S. Enteritidis cells simultaneously repressed key motility encoding genes and induced a wide range of adhesin- and salmonellae-essential virulence-encoding genes, that are critical for the biofilm formation and intracellular survival, respectively. This finding indicates a potential intrinsic link between oxidative stress and pathogenicity in non-typhoidal Salmonella that needs to be empirically evaluated.
Collapse
|
38
|
Ernst CM, Braxton JR, Rodriguez-Osorio CA, Zagieboylo AP, Li L, Pironti A, Manson AL, Nair AV, Benson M, Cummins K, Clatworthy AE, Earl AM, Cosimi LA, Hung DT. Adaptive evolution of virulence and persistence in carbapenem-resistant Klebsiella pneumoniae. Nat Med 2020; 26:705-711. [PMID: 32284589 PMCID: PMC9194776 DOI: 10.1038/s41591-020-0825-4] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022]
Abstract
Among the most urgent public health threats is the worldwide emergence of carbapenem-resistant Enterobacteriaceae1-4, which are resistant to the antibiotic class of 'last resort'. In the United States and Europe, carbapenem-resistant strains of the Klebsiella pneumoniae ST258 (ref. 5) sequence type are dominant, endemic6-8 and associated with high mortality6,9,10. We report the global evolution of pathogenicity in carbapenem-resistant K. pneumoniae, resulting in the repeated convergence of virulence and carbapenem resistance in the United States and Europe, dating back to as early as 2009. We demonstrate that K. pneumoniae can enhance its pathogenicity by adopting two opposing infection programs through easily acquired gain- and loss-of-function mutations. Single-nucleotide polymorphisms in the capsule biosynthesis gene wzc lead to hypercapsule production, which confers phagocytosis resistance, enhanced dissemination and increased mortality in animal models. In contrast, mutations disrupting capsule biosynthesis genes impair capsule production, which enhances epithelial cell invasion, in vitro biofilm formation and persistence in urinary tract infections. These two types of capsule mutants have emerged repeatedly and independently in Europe and the United States, with hypercapsule mutants associated with bloodstream infections and capsule-deficient mutants associated with urinary tract infections. In the latter case, drug-tolerant K. pneumoniae can persist to yield potentially untreatable, persistent infection.
Collapse
Affiliation(s)
- Christoph M Ernst
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Julian R Braxton
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Carlos A Rodriguez-Osorio
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Anna P Zagieboylo
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Li Li
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | | | - Anil V Nair
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Maura Benson
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Kaelyn Cummins
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Anne E Clatworthy
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Ashlee M Earl
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lisa A Cosimi
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Deborah T Hung
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Collins L, Sathiananthamoorthy S, Rohn J, Malone-Lee J. A revalidation and critique of assumptions about urinary sample collection methods, specimen quality and contamination. Int Urogynecol J 2020; 31:1255-1262. [PMID: 32140752 PMCID: PMC7270983 DOI: 10.1007/s00192-020-04272-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/18/2020] [Indexed: 01/21/2023]
Abstract
Introduction and hypothesis Midstream urine (MSU) is key in assessing lower urinary tract syndrome (LUTS), but contingent on some assumptions. The aim of this study was to compare the occurrence of contamination and the quality of substrates obtained from four different collections: MSU, catheter specimen urine (CSU), a commercial MSU collecting device (Peezy) and a natural void. Contamination was quantified by differential, uroplakin-positive, urothelial cell counts. Methods This was a single blind, crossover study conducted in two phases. First, we compared the MSU with CSU using urine culture, pyuria counts and differential counting of epithelial cells after immunofluorescence staining for uroplakin III (UP3). Second, we compared the three non-invasive (MSU, Peezy MSU™, natural void) methods using UP3 antibody staining only. Results The natural void was best at collecting bladder urinary sediment, with the majority of epithelial cells present derived from the urinary tract. CSU sampling missed much of the urinary sediment and showed sparse culture results. Finally, the MSU collection methods did not capture much of the bladder sediment. Conclusion We found little evidence for contamination with the four methods. Natural void was the best method for harvesting shed urothelial cells and white blood cells. It provides a richer sample of the inflammatory exudate, including parasitised urothelial cells and the microbial substrate. However, if the midstream sample is believed to be important, the MSU collection device is advantageous.
Collapse
Affiliation(s)
- Linda Collins
- School of Nursing, Kingston University, Frank Lampl Building, Kingston Hill Campus, London, UK.
| | | | - Jennifer Rohn
- Department of Renal Medicine, Division of Medicine, University College London, London, UK
| | - James Malone-Lee
- Department of Renal Medicine, Division of Medicine, University College London, London, UK
| |
Collapse
|
40
|
Adamus-Białek W, Wawszczak M, Arabski M, Majchrzak M, Gulba M, Jarych D, Parniewski P, Głuszek S. Ciprofloxacin, amoxicillin, and aminoglycosides stimulate genetic and phenotypic changes in uropathogenic Escherichia coli strains. Virulence 2020; 10:260-276. [PMID: 30938219 PMCID: PMC6527016 DOI: 10.1080/21505594.2019.1596507] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antibiotic therapy and its consequences in bacterial and human aspects are widely investigated. Despite this, the emergence of new multidrug resistant bacteria is still a current problem. The scope of our work included the observation of changes among uropathogenic Escherichia coli strains after the treatment with a subinhibitory concentration of different antibiotics. The sensitive strains with or without virulence factors were incubated with amoxicillin, ciprofloxacin, gentamycin, or tobramycin. After each passage, the E. coli derivatives were compared to their wild types based on their susceptibility profiles, virulence genes, biofilm formations and the fingerprint profiles of PCR products amplified with using the (N)(6)(CGG)(4) primer. It turned out that antibiotics caused significant changes in the repertoire of bacterial virulence and biofilm formation, corresponding to acquired cross-resistance. The genomic changes among the studied bacteria were reflected in the changed profiles of the CGG-PCR products. In conclusion, the inappropriate application of antibiotics may cause a rapid rise of Multidrug Resistant (MDR) strains and give bacteria a chance to modulate their own pathogenicity. This phenomenon has been easily observed among uropathogenic E. coli strains and it is one of the main reasons for recurrent infections of the urinary tract.
Collapse
Affiliation(s)
- Wioletta Adamus-Białek
- a Department of Surgery and Surgical Nursery with Laboratory of Genetics, Faculty of Medicine and Health Sciences , Jan Kochanowski University , Kielce , Poland
| | - Monika Wawszczak
- a Department of Surgery and Surgical Nursery with Laboratory of Genetics, Faculty of Medicine and Health Sciences , Jan Kochanowski University , Kielce , Poland
| | - Michał Arabski
- b Department of Biochemistry & Genetics , Jan Kochanowski University , Kielce , Poland
| | - Michał Majchrzak
- a Department of Surgery and Surgical Nursery with Laboratory of Genetics, Faculty of Medicine and Health Sciences , Jan Kochanowski University , Kielce , Poland
| | - Martyna Gulba
- a Department of Surgery and Surgical Nursery with Laboratory of Genetics, Faculty of Medicine and Health Sciences , Jan Kochanowski University , Kielce , Poland
| | - Dariusz Jarych
- c Institute of Medical Biology, Polish Academy of Sciences , Łódź , Poland
| | - Paweł Parniewski
- c Institute of Medical Biology, Polish Academy of Sciences , Łódź , Poland
| | - Stanisław Głuszek
- a Department of Surgery and Surgical Nursery with Laboratory of Genetics, Faculty of Medicine and Health Sciences , Jan Kochanowski University , Kielce , Poland
| |
Collapse
|
41
|
Zhu M, Wang S, Zhu Y, Wang Z, Zhao M, Chen D, Zhou C. Behavioral and dietary risk factors of recurrent urinary tract infection in Chinese postmenopausal women: a case-control study. J Int Med Res 2020; 48:300060519889448. [PMID: 31840544 PMCID: PMC7783248 DOI: 10.1177/0300060519889448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/29/2019] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE The present study aimed to examine the behavioral and dietary risk factors of recurrent urinary tract infection (RUTI) in postmenopausal patients in China. METHODS We performed a population-based case-control study with 193 postmenopausal women with RUTI and 193 age-matched healthy female controls with no history of RUTI. The study was conducted between January 2016 and June 2018 in Changzhou, China. Data were collected using an interviewer-based questionnaire, including information on demographics, lifestyle behavior, and habitual diet. Conditional logistic regression analysis was conducted to examine the risk factors associated with RUTI. RESULTS Wiping from back to front after toilet use, sedentary behavior >6 hours/day, delayed voiding, and chronic constipation were associated with an increased risk of RUTI. Drinking more than three cups of green tea per month showed an inverse association with RUTI. However, there was no evidence of dose dependency for overall consumption. Additionally, the three-cup association involved a small proportion of cases and may reflect statistical artifact. CONCLUSIONS Wiping from back to front after toilet use, sedentary behavior, delayed voiding, and chronic constipation are associated with an increased risk of RUTI in postmenopausal women.
Collapse
Affiliation(s)
- Meifeng Zhu
- Department of Nephrology, Changzhou Affiliated Hospital of
Nanjing University of Chinese Medicine, Changzhou, Jiangsu, China
- Department of Febrile Disease, Basic Medicine College, Nanjing
University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shenju Wang
- Department of Nephrology, Changzhou Affiliated Hospital of
Nanjing University of Chinese Medicine, Changzhou, Jiangsu, China
| | - Ying Zhu
- Department of Nephrology, Changshu Hospital Affiliated to
Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| | - Zhixia Wang
- Department of Obstetrics and Gynecology, The Affiliated BenQ
Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min Zhao
- Department of Nephrology, Changzhou Affiliated Hospital of
Nanjing University of Chinese Medicine, Changzhou, Jiangsu, China
| | - Dai Chen
- Department of Nephrology, Changzhou Affiliated Hospital of
Nanjing University of Chinese Medicine, Changzhou, Jiangsu, China
| | - Chunxiang Zhou
- Department of Febrile Disease, Basic Medicine College, Nanjing
University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
42
|
Sun J, Deering RW, Peng Z, Najia L, Khoo C, Cohen PS, Seeram NP, Rowley DC. Pectic Oligosaccharides from Cranberry Prevent Quiescence and Persistence in the Uropathogenic Escherichia coli CFT073. Sci Rep 2019; 9:19590. [PMID: 31862919 PMCID: PMC6925298 DOI: 10.1038/s41598-019-56005-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 12/04/2019] [Indexed: 11/26/2022] Open
Abstract
Urinary tract infections (UTIs) caused by Escherichia coli create a large burden on healthcare and frequently lead to recurrent infections. Part of the success of E. coli as an uropathogenic bacterium can be attributed to its ability to form quiescent intracellular reservoirs in bladder cells and its persistence after antibiotic treatment. Cranberry juice and related products have been used for the prevention of UTIs with varying degrees of success. In this study, a group of cranberry pectic oligosaccharides (cPOS) were found to both inhibit quiescence and reduce the population of persister cells formed by the uropathogenic strain, CFT073. This is the first report detailing constituents of cranberry with the ability to modulate these important physiological aspects of uropathogenic E. coli. Further studies investigating cranberry should be keen to include oligosaccharides as part of the ‘active’ cocktail of chemical compounds.
Collapse
Affiliation(s)
- Jiadong Sun
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA.,Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Robert W Deering
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Zhiyuan Peng
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Laila Najia
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Christina Khoo
- Ocean Spray Cranberries, Inc., One Ocean Spray Drive, Lakeville-Middleboro, MA, 02349, USA
| | - Paul S Cohen
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI, 02881, USA
| | - Navindra P Seeram
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - David C Rowley
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA.
| |
Collapse
|
43
|
Dixon M, Sha S, Stefil M, McDonald M. Is it Time to Say Goodbye to Culture and Sensitivity? The Case for Culture-independent Urology. Urology 2019; 136:112-118. [PMID: 31786305 DOI: 10.1016/j.urology.2019.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/28/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
Next-generation sequencing has highlighted the limitations of conventional culture methods in the role of urology while discovering the intricate details of the role of microbiota in urologic health and disease. This review article explores: the utility and limitations of conventional culture methods; how culture-independent technologies are revolutionizing medicine; and how the implementation of these technologies may lead to improved patient outcomes. Finally, this article discusses the barriers to widespread adoption of culture-independent technologies, with suggestions for how these hurdles may be overcome.
Collapse
Affiliation(s)
- Matthew Dixon
- Norwich Medical School, University of East Anglia, Norwich, UK.
| | - Sybil Sha
- Geisel School of Medicine, Dartmouth College, Hanover, NH
| | - Maria Stefil
- Norwich Medical School, University of East Anglia, Norwich, UK
| | | |
Collapse
|
44
|
Compounds targeting YadC of uropathogenic Escherichia coli and its host receptor annexin A2 decrease bacterial colonization in bladder. EBioMedicine 2019; 50:23-33. [PMID: 31757778 PMCID: PMC6921372 DOI: 10.1016/j.ebiom.2019.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Background Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs), and fimbrial tip adhesins, play important roles in UPEC colonization. Few fimbrial tip adhesins and their receptors on host cells, which have the potential to be the therapeutic targets, have been identified. Methods the UPEC wild-type strain CFT073, ΔyadC and the complemented strain were used to perform assays in vitro and in vivo. The effects of D-xylose targeting YadC on UPEC colonization were evaluated. A YadC receptor was identified by far-western blotting, LC-MS/MS and co-immunoprecipitation. The effects of compounds targeting the receptor on UPEC colonization were tested. Findings YadC was investigated for its mediation of UPEC adhesion and invasion to bladder epithelial cells in vitro; and its promotion of UPEC colonization in bladder in vivo. D-xylose, targeting YadC, showed prophylactic and therapeutic effects on UPEC colonization. Annexin A2 (ANXA2) was identified as a YadC receptor, involved in UPEC infection. ANXA2 inhibitors attenuated UPEC infections. The yadC gene was widely present in UPEC clinical isolates and phylogenetic analysis of yadC was performed. Interpretation YadC and its receptor ANXA2 play important roles in UPEC colonization in bladder, leading to novel treatment strategies targeting YadC or ANXA2 for acute UTIs. Fund This study was supported by grants from the National Natural Science Foundation of China (NSFC) Programs (31670071 and 31970133), the National Key Technologies R&D Program, Intergovernmental international innovation cooperation (2018YFE0102000), Tianjin Science and Technology Commissioner Project (18JCZDJC36000), the Science & Technology Development Fund of Tianjin Education Commission for Higher Education (2017ZD12). The Science Foundation of Tianjin Medical University (2016KY2M08).
Collapse
|
45
|
Orthosipon stamineus extract exerts inhibition of bacterial adhesion and chaperon-usher system of uropathogenic Escherichia coli—a transcriptomic study. Appl Microbiol Biotechnol 2019; 103:8571-8584. [DOI: 10.1007/s00253-019-10120-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/12/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022]
|
46
|
Horsley H, Owen J, Browning R, Carugo D, Malone-Lee J, Stride E, Rohn JL. Ultrasound-activated microbubbles as a novel intracellular drug delivery system for urinary tract infection. J Control Release 2019; 301:166-175. [PMID: 30904501 DOI: 10.1016/j.jconrel.2019.03.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/27/2022]
Abstract
The development of new modalities for high-efficiency intracellular drug delivery is a priority for a number of disease areas. One such area is urinary tract infection (UTI), which is one of the most common infectious diseases globally and which imposes an immense economic and healthcare burden. Common uropathogenic bacteria have been shown to invade the urothelial wall during acute UTI, forming latent intracellular reservoirs that can evade antimicrobials and the immune response. This behaviour likely facilitates the high recurrence rates after oral antibiotic treatments, which are not able to penetrate the bladder wall and accumulate to an effective concentration. Meanwhile, oral antibiotics may also exacerbate antimicrobial resistance and cause systemic side effects. Using a human urothelial organoid model, we tested the ability of novel ultrasound-activated lipid microbubbles to deliver drugs into the cytoplasm of apical cells. The gas-filled lipid microbubbles were decorated with liposomes containing the non-cell-permeant antibiotic gentamicin and a fluorescent marker. The microbubble suspension was added to buffer at the apical surface of the bladder model before being exposed to ultrasound (1.1 MHz, 2.5 Mpa, 5500 cycles at 20 ms pulse duration) for 20 s. Our results show that ultrasound-activated intracellular delivery using microbubbles was over 16 times greater than the control group and twice that achieved by liposomes that were not associated with microbubbles. Moreover, no cell damage was detected. Together, our data show that ultrasound-activated microbubbles can safely deliver high concentrations of drugs into urothelial cells, and have the potential to be a more efficacious alternative to traditional oral antibiotic regimes for UTI. This modality of intracellular drug delivery may prove useful in other clinical indications, such as cancer and gene therapy, where such penetration would aid in treatment.
Collapse
Affiliation(s)
- H Horsley
- Department of Renal Medicine, Division of Medicine, University College, London, UK
| | - J Owen
- Institute of Biomedical Engineering, University of Oxford, UK
| | - R Browning
- Institute of Biomedical Engineering, University of Oxford, UK
| | - D Carugo
- Faculty of Physical Sciences and Engineering, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | - J Malone-Lee
- Department of Renal Medicine, Division of Medicine, University College, London, UK
| | - E Stride
- Institute of Biomedical Engineering, University of Oxford, UK
| | - J L Rohn
- Department of Renal Medicine, Division of Medicine, University College, London, UK.
| |
Collapse
|
47
|
Uropathogenic Escherichia coli employs both evasion and resistance to subvert innate immune-mediated zinc toxicity for dissemination. Proc Natl Acad Sci U S A 2019; 116:6341-6350. [PMID: 30846555 PMCID: PMC6442554 DOI: 10.1073/pnas.1820870116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is responsible for most urinary tract infections and is also a frequent cause of sepsis, thus necessitating an understanding of UPEC-mediated subversion of innate immunity. The role of zinc in the innate immune response against UPEC infection, and whether this pathogen counters this response, has not been examined. Here we demonstrate, both in vitro and in vivo, that UPEC both evades and resists innate immune-mediated zinc toxicity to persist and disseminate within the host. Moreover, we have defined the set of UPEC genes conferring zinc resistance and have developed highly selective E. coli reporter systems to track zinc toxicity. These innovative approaches substantially enhance our understanding of immune-mediated metal ion toxicity and bacterial pathogenesis. Toll-like receptor (TLR)-inducible zinc toxicity is a recently described macrophage antimicrobial response used against bacterial pathogens. Here we investigated deployment of this pathway against uropathogenic Escherichia coli (UPEC), the major cause of urinary tract infections. Primary human macrophages subjected EC958, a representative strain of the globally disseminated multidrug-resistant UPEC ST131 clone, to zinc stress. We therefore used transposon-directed insertion site sequencing to identify the complete set of UPEC genes conferring protection against zinc toxicity. Surprisingly, zinc-susceptible EC958 mutants were not compromised for intramacrophage survival, whereas corresponding mutants in the nonpathogenic E. coli K-12 strain MG1655 displayed significantly reduced intracellular bacterial loads within human macrophages. To investigate whether the intramacrophage zinc stress response of EC958 reflected the response of only a subpopulation of bacteria, we generated and validated reporter systems as highly specific sensors of zinc stress. Using these tools we show that, in contrast to MG1655, the majority of intramacrophage EC958 evades the zinc toxicity response, enabling survival within these cells. In addition, EC958 has a higher tolerance to zinc than MG1655, with this likely being important for survival of the minor subset of UPEC cells exposed to innate immune-mediated zinc stress. Indeed, analysis of zinc stress reporter strains and zinc-sensitive mutants in an intraperitoneal challenge model in mice revealed that EC958 employs both evasion and resistance against zinc toxicity, enabling its dissemination to the liver and spleen. We thus demonstrate that a pathogen of global significance uses multiple mechanisms to effectively subvert innate immune-mediated zinc poisoning for systemic spread.
Collapse
|
48
|
Reassessment of Routine Midstream Culture in Diagnosis of Urinary Tract Infection. J Clin Microbiol 2019; 57:JCM.01452-18. [PMID: 30541935 PMCID: PMC6425166 DOI: 10.1128/jcm.01452-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022] Open
Abstract
Midstream urine (MSU) culture remains the gold standard diagnostic test for confirming urinary tract infection (UTI). We previously showed that patients with chronic lower urinary tract symptoms (LUTS) below the diagnostic cutoff on MSU culture may still harbor bacterial infection and that their antibiotic treatment was associated with symptom resolution. Here, we evaluated the results of the United Kingdom's MSU culture in symptomatic patients and controls. Next, we compared the bacterial enrichment capabilities of the MSU culture with those of a 50-µl uncentrifuged culture, a 30-ml centrifuged sediment culture, and 16S rRNA gene sequencing. This study was conducted on urine specimens from 33 LUTS patients attending their first clinical appointment (mean age, 48.7 years; standard deviation [SD], 16.5 years), 30 LUTS patients on treatment (mean age, 47.8 years; SD, 16.5 years) whose symptoms had relapsed, and 29 asymptomatic controls (mean age, 40.7 years, SD, 15.7 years). We showed that the routine MSU culture, adopting the UK interpretation criteria tailored to acute UTI, failed to detect a variety of bacterial species, including recognized uropathogens. Moreover, the diagnostic MSU culture was unable to discriminate between patients and controls. In contrast, genomic analysis of urine enriched by centrifugation discriminated between the groups, generating a more accurate understanding of species richness. In conclusion, the United Kingdom's MSU protocol misses a significant proportion of bacteria, which include recognized uropathogens, and may be unsuitable for excluding UTI in patients with LUTS.
Collapse
|
49
|
Adamus-Białek W, Vollmerhausen TL, Janik K. Hydrogen peroxide stimulates uropathogenic Escherichia coli strains to cellulose production. Microb Pathog 2018; 126:287-291. [PMID: 30447422 DOI: 10.1016/j.micpath.2018.11.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/23/2022]
Abstract
Reactive oxygen intermediates, such as hydrogen peroxide, are toxic molecules produced by immune cells in response to bacterial invasion into the host. Bacteria try to protect themselves against the immune system through specific properties such as biofilm formation. This phenomenon occurs also during urinary tract infections. Cellulose is an important factor of Escherichia coli biofilm and contributes to building a protective shield around bacterial cells upon the host immune response. In this study, we aimed to analyze the effect of hydrogen peroxide on the production of this biofilm component. To achieve this goal, 25 clinical E. coli strains isolated from patients with urinary tract infections were used. These bacterial strains were characterized based on their growth characteristics, their ability to form biofilm and their capacity to produce cellulose upon exposure to sub-lethal concentrations of hydrogen peroxide growth, and the biofilm formation of these strains was analyzed. Our results revealed that the analyzed uropathogenic E. coli strains slightly, but significantly, reduced growth and biofilm production upon hydrogen peroxide treatment. However, when separating these strains regarding their ability to produce cellulose, we found that general biofilm production was reduced but cellulose expression was induced upon peroxide treatment. This finding contributes to a better understanding of how bacterial biofilm formation is triggered and provides interesting insights into how uropathogenic E. coli protect themselves in an inhospitable environment.
Collapse
Affiliation(s)
- Wioletta Adamus-Białek
- Jan Kochanowski University, Institute of Medical Sciences, Kielce, Poland; Department of Microbiology, Tumor and Cell Biology, Karolinska University Hospital & Karolinska Institutet, Stockholm, Sweden.
| | - Tara L Vollmerhausen
- Department of Microbiology, Tumor and Cell Biology, Karolinska University Hospital & Karolinska Institutet, Stockholm, Sweden
| | - Katrin Janik
- Department of Microbiology, Tumor and Cell Biology, Karolinska University Hospital & Karolinska Institutet, Stockholm, Sweden; Functional Genomics, Laimburg Research Centre, Laimburg, Italy
| |
Collapse
|
50
|
Hardison RL, Heimlich DR, Harrison A, Beatty WL, Rains S, Moseley MA, Thompson JW, Justice SS, Mason KM. Transient Nutrient Deprivation Promotes Macropinocytosis-Dependent Intracellular Bacterial Community Development. mSphere 2018; 3:3/5/e00286-18. [PMID: 30209128 PMCID: PMC6135960 DOI: 10.1128/msphere.00286-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nutrient limitation restricts bacterial growth in privileged sites such as the middle ear. Transient heme-iron restriction of nontypeable Haemophilus influenzae (NTHI), the major causative agent of chronic and recurrent otitis media (OM), promotes new and diverse phenotypes that can influence planktonic, biofilm, and intracellular lifestyles of NTHI. However, the bacterial responses to nutrient restriction that impact intracellular fate and survival of NTHI are unknown. In this work, we provide evidence for the role of transient heme-iron restriction in promoting the formation of intracellular bacterial communities (IBCs) of NTHI both in vitro and in vivo in a preclinical model of OM. We show that transient heme-iron restriction of NTHI results in significantly increased invasion and intracellular populations that escape or evade the endolysosomal pathway for increased intracellular survival. In contrast, NTHI continuously exposed to heme-iron traffics through the endolysosomal pathway for degradation. The use of pharmacological inhibitors revealed that prior heme-iron status does not appear to influence NTHI internalization through endocytic pathways. However, inhibition of macropinocytosis altered the intracellular fate of transiently restricted NTHI for degradation in the endolysosomal pathway. Furthermore, prevention of macropinocytosis significantly reduced the number of IBCs in cultured middle ear epithelial cells, providing evidence for the feasibility of this approach to reduce OM persistence. These results reveal that microenvironmental cues can influence the intracellular fate of NTHI, leading to new mechanisms for survival during disease progression.IMPORTANCE Otitis media is the most common bacterial infection in childhood. Current therapies are limited in the prevention of chronic or recurrent otitis media which leads to increased antibiotic exposure and represents a significant socioeconomic burden. In this study, we delineate the effect of nutritional limitation on the intracellular trafficking pathways used by nontypeable Haemophilus influenzae (NTHI). Moreover, transient limitation of heme-iron led to the development of intracellular bacterial communities that are known to contribute to persistence and recurrence in other diseases. New approaches for therapeutic interventions that reduce the production of intracellular bacterial communities and promote trafficking through the endolysosomal pathway were revealed through the use of pharmacological inhibition of macropinocytosis. This work demonstrates the importance of an intracellular niche for NTHI and provides new approaches for intervention for acute, chronic, and recurring episodes of otitis media.
Collapse
Affiliation(s)
- Rachael L Hardison
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Derek R Heimlich
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Alistair Harrison
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Wandy L Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sarah Rains
- Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
| | - M Arthur Moseley
- Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
| | - J Will Thompson
- Duke Proteomics and Metabolomics Core Facility, Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
| | - Sheryl S Justice
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Kevin M Mason
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|