1
|
Frara N, Jawawdeh K, Giaddui D, Tamas IP, Gares RP, McGonagle ER, Hilliard BA, Kolpakov MA, Bright-Rowe L, Braverman AS, Brown JM, Ruggieri MR, Barbe MF. Enhanced BDNF and ROS in Mucosa of Lower Motor Neuron Lesioned Dog Bladder Following Somatic Motor Nerve Transfer. Cells 2025; 14:406. [PMID: 40136655 PMCID: PMC11941061 DOI: 10.3390/cells14060406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Neurotrophic factors and reactive oxygen species (ROS) modulate neuronal plasticity. In a model of a lower motor neuron lesioned bladder, somatic nerve transfer was used as a reinnervation strategy. Levels of neurotrophins, ROS, and TNF-α in bladder mucosa and muscle layers collected from three groups of adult female dogs: (1) Decentralized, via bilateral transection of coccygeal and sacral spinal roots, lumbar 7 dorsal roots, and hypogastric nerves, then 6-21 mo recovery; (2) reinnervated (ObNT-Reinn), after similar decentralization for 12 mo, then bilateral obturator-to-vesical nerve transfer and 8-12 mo recovery; and (3) Controls. In mucosa, BDNF and ROS levels were highest in ObNT-Reinn bladders, GDNF and TNF-α levels were restored to Control levels in ObNT-Reinn bladders (lowest in Decentralized). NT-3 and ARTN were lower in ObNT-Reinn and Decentralized bladders versus Controls. In muscle, ROS was lower in ObNT-Reinn muscle versus Controls. BDNF mucosa levels correlated with bladder axonal density and detrusor layer thickness; and GDNF mucosal correlated with bladder contraction after vesical or transferred obturator nerve electrical stimulation, as did BDNF and GDNF muscle levels. The increased BDNF and GDNF in bladders that underwent somatic nerve transfer with subsequent recovery suggest that BDNF and GDNF may help promote the reestablishment of bladder innervation.
Collapse
Affiliation(s)
- Nagat Frara
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Kais Jawawdeh
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Dania Giaddui
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Istvan P. Tamas
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Ryan P. Gares
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Elizabeth R. McGonagle
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Brendan A. Hilliard
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Mikhail A. Kolpakov
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Lewis Bright-Rowe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Alan S. Braverman
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| | - Justin M. Brown
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Michael R. Ruggieri
- Center for Translational Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19040, USA; (K.J.); (D.G.); (R.P.G.); (M.R.R.S.)
| | - Mary F. Barbe
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (I.P.T.); (E.R.M.); (B.A.H.); (M.A.K.); (L.B.-R.); (A.S.B.)
| |
Collapse
|
2
|
Al KF, Parris J, Engelbrecht K, Reid G, Burton JP. Interconnected microbiomes-insights and innovations in female urogenital health. FEBS J 2025; 292:1378-1396. [PMID: 39080993 DOI: 10.1111/febs.17235] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 03/22/2025]
Abstract
The paradigm that the vaginal microbiota is a protective gateway for the urinary and reproductive systems has endured for more than a century and driven decades of probiotic research. Evidence robustly supports the notion that healthy urogenital microbiomes are predominantly colonized by lactobacilli, particularly Lactobacillus crispatus, which can acidify the local environment and protect against urogynecologic pathogen colonization. However, recent studies are beginning to delve deeper into the intricate mechanistic interactions connecting the microbiome, its diverse functional potential, host immunity, pathogens, and the development of urogenital diseases. Leveraging these emerging insights alongside past successes presents promising opportunities for future therapies aimed at enhancing the management of conditions such as bacterial vaginosis, urinary tract infections, bladder pain, urinary incontinence, and beyond.
Collapse
Affiliation(s)
- Kait F Al
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
- Lawson Health Research Institute, St. Joseph's Hospital, London, Canada
| | - Josh Parris
- Kimberly Clark Corporation, Global Research & Engineering, Roswell, GA, USA
| | | | - Gregor Reid
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
- Lawson Health Research Institute, St. Joseph's Hospital, London, Canada
- Division of Urology, Department of Surgery, The University of Western Ontario, London, Canada
| | - Jeremy P Burton
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
- Lawson Health Research Institute, St. Joseph's Hospital, London, Canada
- Division of Urology, Department of Surgery, The University of Western Ontario, London, Canada
| |
Collapse
|
3
|
Timm MR, Russell SK, Hultgren SJ. Urinary tract infections: pathogenesis, host susceptibility and emerging therapeutics. Nat Rev Microbiol 2025; 23:72-86. [PMID: 39251839 DOI: 10.1038/s41579-024-01092-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/11/2024]
Abstract
Urinary tract infections (UTIs), which include any infection of the urethra, bladder or kidneys, account for an estimated 400 million infections and billions of dollars in health-care spending per year. The most common bacterium implicated in UTI is uropathogenic Escherichia coli, but diverse pathogens including Klebsiella, Enterococcus, Pseudomonas, Staphylococcus and even yeast such as Candida species can also cause UTIs. UTIs occur in both women and men and in both healthy and immunocompromised patients. However, certain patient factors predispose to disease: for example, female sex, history of prior UTI, or the presence of a urinary catheter or other urinary tract abnormality. The current clinical paradigm for the treatment of UTIs involves the use of antibiotics. Unfortunately, the efficacy of this approach is dwindling as the prevalence of antimicrobial resistance rises among UTI isolates, and the immense quantity of antibiotics prescribed annually for these infections contributes to the emergence of resistant pathogens. Therefore, there is an urgent need for new antibiotics and non-antibiotic treatment and prevention strategies. In this Review, we discuss how recent studies of bacterial pathogenesis, recurrence, persistence, host-pathogen interactions and host susceptibility factors have elucidated new and promising targets for the treatment and prevention of UTIs.
Collapse
Affiliation(s)
- Morgan R Timm
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Seongmi K Russell
- Department of Paediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Scott J Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Milosevic M, Magnutzki A, Braun T, Hussain S, Jakschitz T, Kragl M, Soeberdt M, Nausch B, Bonn GK, Huber LA, Valovka T. Anti-inflammatory and cytoprotective polypharmacology of Canephron N reveals targeting of the IKK-NF-κB and p38-MK2-RIPK1 axes. Biomed Pharmacother 2025; 182:117747. [PMID: 39671726 DOI: 10.1016/j.biopha.2024.117747] [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: 10/08/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024] Open
Abstract
Urinary tract infections are among the most frequently occurring forms of infection, and inflammation and tissue damage contribute significantly to symptoms, e.g., dysuria and urge. Canephron N is an orally bioavailable herbal medicine with anti-inflammatory, spasmolytic, anti-adhesive, and anti-nociceptive therapeutic effects that is approved for the treatment of uncomplicated urinary tract infections. Here, we used renal tubular epithelial HK-2 cells to study the anti-inflammatory and cytoprotective effects and molecular mechanisms of its active component, BNO 2103. BNO 2103 suppressed nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation by lipopolysaccharide (LPS) and tumor necrosis factor alpha (TNFα) and prevented inhibitory κB kinase (IKK)-dependent phosphorylation and degradation of inhibitor of nuclear factor kappa B alpha (IκBα). BNO 2103 also suppressed the inflammation-specific S536 phosphorylation of the NF-κB subunit p65 and the production of a specific set of inflammatory cytokines. Unlike other NF-κB inhibitors, BNO 2103 demonstrated cytoprotection against TNFα-induced cytotoxicity. Our data suggest that BNO 2103 acts primarily through the mitogen-activated protein kinase p38 (p38 MAPK)-MAPK-activated protein kinase 2 (MK2) axis by promoting receptor-interacting serine/threonine protein kinase 1 (RIPK1) phosphorylation at S320. Simultaneously, it suppresses S166 autophosphorylation and subsequent activation of RIPK1, which is required for apoptotic and necroptotic responses to TNFα. This study confirms Canephron N as an effective alternative to traditional anti-inflammatory drugs and provides initial evidence of its ability to inhibit apoptosis and necroptosis in the urogenital system. It also presents a detailed pathway investigation that identifies the specific targets of Canephron N within the NF-κB signaling cascade.
Collapse
Affiliation(s)
- Marija Milosevic
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Theodor Braun
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Shah Hussain
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | - Thomas Jakschitz
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria
| | | | | | | | - Günther K Bonn
- ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria.
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria; ADSI-Austrian Drug Screening Institute, Leopold-Franzens University of Innsbruck, Innsbruck 6020, Austria.
| | - Taras Valovka
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck 6020, Austria; Department of Pediatrics I, Medical University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
5
|
Kaleska B, Sluyter R, Chen Z, Mansfield KJ. Effect of pro-inflammatory cytokines on urothelial cell adenosine triphosphate release and breakdown. Bladder (San Franc) 2024; 11:e21200006. [PMID: 39539471 PMCID: PMC11555206 DOI: 10.14440/bladder.2024.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/25/2024] [Accepted: 08/20/2024] [Indexed: 11/16/2024] Open
Abstract
Objectives Urinary symptoms of urgency, frequency, and pain are thought to be the result of inflammation in several bladder pathologies although the cause of these symptoms remains uncertain. Extracellular adenosine triphosphate (ATP) released from the bladder urothelium during normal bladder stretch is believed to bind to purinergic receptors on afferent nerves to signal bladder sensation. This study examined pro-inflammatory cytokines in the urine of women with detrusor overactivity (DO) with or without urinary tract infection (UTI) compared to controls and then determined the effect of pro-inflammatory cytokines on ATP signaling (release and breakdown) from the urothelium. Methods The urinary concentrations of interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β) were determined in women with DO with or without UTI compared to female controls. The effect of pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-1β) on control and hypotonic-induced ATP release using human UROtsa urothelial cells was examined, as was the effect of these cytokines on nucleotide (ATP, adenosine diphosphate and adenosine monophosphate) breakdown. Results Urinary concentrations of IFN-γ, TNF-α, and IL-1β were increased in women with DO and UTI. Pre-treatment of urothelial cells with individual cytokines stimulated a decrease rather than an increase in ATP release whereas pre-treatment with a cocktail of all three cytokines stimulated a small but significant increase in hypotonic-induced ATP release. Pre-treatment of urothelial cells with cytokines significantly enhanced nucleotide breakdown. Conclusion Using a simple cell culture model we have demonstrated that the response of the urothelium to pro-inflammatory cytokines is complex, affecting both release and breakdown of ATP.
Collapse
Affiliation(s)
- Belinda Kaleska
- Graduate School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Biosciences, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Zhuoran Chen
- Department of Urogynaecology, St George Hospital, University of New South Wales, Kogarah, NSW 2522, Australia
| | - Kylie J Mansfield
- Graduate School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
6
|
Zou Z, Singh P, Pinkner JS, Obernuefemann CLP, Xu W, Nye TM, Dodson KW, Almqvist F, Hultgren SJ, Caparon MG. Dihydrothiazolo ring-fused 2-pyridone antimicrobial compounds treat Streptococcus pyogenes skin and soft tissue infection. SCIENCE ADVANCES 2024; 10:eadn7979. [PMID: 39093975 PMCID: PMC11296344 DOI: 10.1126/sciadv.adn7979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
We have developed GmPcides from a peptidomimetic dihydrothiazolo ring-fused 2-pyridone scaffold that has antimicrobial activities against a broad spectrum of Gram-positive pathogens. Here, we examine the treatment efficacy of GmPcides using skin and soft tissue infection (SSTI) and biofilm formation models by Streptococcus pyogenes. Screening our compound library for minimal inhibitory (MIC) and minimal bactericidal (MBC) concentrations identified GmPcide PS757 as highly active against S. pyogenes. Treatment of S. pyogenes biofilm with PS757 revealed robust efficacy against all phases of biofilm formation by preventing initial biofilm development, ceasing biofilm maturation and eradicating mature biofilm. In a murine model of S. pyogenes SSTI, subcutaneous delivery of PS757 resulted in reduced levels of tissue damage, decreased bacterial burdens, and accelerated rates of wound healing, which were associated with down-regulation of key virulence factors, including M protein and the SpeB cysteine protease. These data demonstrate that GmPcides show considerable promise for treating S. pyogenes infections.
Collapse
Affiliation(s)
- Zongsen Zou
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pardeep Singh
- Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Jerome S. Pinkner
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chloe L. P. Obernuefemann
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wei Xu
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Taylor M. Nye
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karen W. Dodson
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Scott J. Hultgren
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael G. Caparon
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Mohammad A, Laboulaye MA, Shenhar C, Dobberfuhl AD. Mechanisms of oxidative stress in interstitial cystitis/bladder pain syndrome. Nat Rev Urol 2024; 21:433-449. [PMID: 38326514 DOI: 10.1038/s41585-023-00850-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 02/09/2024]
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is characterized by bladder and/or pelvic pain, increased urinary urgency and frequency and nocturia. The pathophysiology of IC/BPS is poorly understood, and theories include chronic inflammation, autoimmune dysregulation, bacterial cystitis, urothelial dysfunction, deficiency of the glycosaminoglycan (GAG) barrier and urine cytotoxicity. Multiple treatment options exist, including behavioural interventions, oral medications, intravesical instillations and procedures such as hydrodistension; however, many clinical trials fail, and patients experience an unsatisfactory treatment response, likely owing to IC/BPS phenotype heterogeneity and the use of non-targeted interventions. Oxidative stress is implicated in the pathogenesis of IC/BPS as reactive oxygen species impair bladder function via their involvement in multiple molecular mechanisms. Kinase signalling pathways, nociceptive receptors, mast-cell activation, urothelial dysregulation and circadian rhythm disturbance have all been linked to reactive oxygen species and IC/BPS. However, further research is necessary to fully uncover the role of oxidative stress in the pathways driving IC/BPS pathogenesis. The development of new models in which these pathways can be manipulated will aid this research and enable further investigation of promising therapeutic targets.
Collapse
Affiliation(s)
- Ashu Mohammad
- Department of Urology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mallory A Laboulaye
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Chen Shenhar
- Department of Urology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Amy D Dobberfuhl
- Department of Urology, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
8
|
Henry S, Lewis SM, Cyrill SL, Callaway MK, Chatterjee D, Hanasoge Somasundara AV, Jones G, He XY, Caligiuri G, Ciccone MF, Diaz IA, Biswas AA, Hernandez E, Ha T, Wilkinson JE, Egeblad M, Tuveson DA, Dos Santos CO. Host response during unresolved urinary tract infection alters female mammary tissue homeostasis through collagen deposition and TIMP1. Nat Commun 2024; 15:3282. [PMID: 38627380 PMCID: PMC11021735 DOI: 10.1038/s41467-024-47462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/03/2024] [Indexed: 04/19/2024] Open
Abstract
Exposure to pathogens throughout a lifetime influences immunity and organ function. Here, we explore how the systemic host-response to bacterial urinary tract infection (UTI) induces tissue-specific alterations to the mammary gland. Utilizing a combination of histological tissue analysis, single cell transcriptomics, and flow cytometry, we identify that mammary tissue from UTI-bearing mice displays collagen deposition, enlarged ductal structures, ductal hyperplasia with atypical epithelial transcriptomes and altered immune composition. Bacterial cells are absent in the mammary tissue and blood of UTI-bearing mice, therefore, alterations to the distal mammary tissue are mediated by the systemic host response to local infection. Furthermore, broad spectrum antibiotic treatment resolves the infection and restores mammary cellular and tissue homeostasis. Systemically, unresolved UTI correlates with increased plasma levels of the metalloproteinase inhibitor, TIMP1, which controls extracellular matrix remodeling and neutrophil function. Treatment of nulliparous and post-lactation UTI-bearing female mice with a TIMP1 neutralizing antibody, restores mammary tissue normal homeostasis, thus providing evidence for a link between the systemic host response during UTI and mammary gland alterations.
Collapse
Affiliation(s)
- Samantha Henry
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Stony Brook University, Graduate Program in Genetics, Stony Brook, NY, USA
| | - Steven Macauley Lewis
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Stony Brook University, Graduate Program in Genetics, Stony Brook, NY, USA
| | | | | | | | | | - Gina Jones
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Xue-Yan He
- Department of Cell Biology and Physiology. School of Medicine in St. Louis. Washington University, St. Louis, MO, USA
| | | | | | | | - Amelia Aumalika Biswas
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- SUNY Downstate Health Sciences University, Neural and Behavior Science, Brooklyn, NY, USA
| | | | - Taehoon Ha
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - John Erby Wilkinson
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Mikala Egeblad
- Department of Cell Biology, Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
9
|
Ebrahimzadeh T, Basu U, Lutz KC, Gadhvi J, Komarovsky JV, Li Q, Zimmern PE, De Nisco NJ. Inflammatory markers for improved recurrent UTI diagnosis in postmenopausal women. Life Sci Alliance 2024; 7:e202302323. [PMID: 38331474 PMCID: PMC10853434 DOI: 10.26508/lsa.202302323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Recurrent urinary tract infection (rUTI) severely impacts postmenopausal women. The lack of rapid and accurate diagnostic tools is a major obstacle in rUTI management as current gold standard methods have >24-h diagnostic windows. Work in animal models and limited human cohorts have identified robust inflammatory responses activated during UTI. Consequently, urinary inflammatory cytokines secreted during UTI may function as diagnostic biomarkers. This study aimed to identify urinary cytokines that could accurately diagnose UTI in a controlled cohort of postmenopausal women. Women passing study exclusion criteria were classified into no UTI and active rUTI groups, and urinary cytokine levels were measured by immunoassay. Pro-inflammatory cytokines IL-8, IL-18, IL-1β, and monocyte chemoattractant protein-1 were significantly elevated in the active rUTI group, and anti-inflammatory cytokines IL-13 and IL-4 were elevated in women without UTI. We evaluated cytokine diagnostic performance and found that an IL-8, prostaglandin E2, and IL-13 multivariable model had the lowest misclassification rate and highest sensitivity. Our data identify urinary IL-8, prostaglandin E2, and IL-13 as candidate biomarkers that may be useful in the development of immunoassay-based UTI diagnostics.
Collapse
Affiliation(s)
| | - Ujjaini Basu
- Department of Biological Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Kevin C Lutz
- Department of Mathematics, University of Texas at Dallas, Dallas, TX, USA
| | - Jashkaran Gadhvi
- Department of Biological Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Jessica V Komarovsky
- Department of Biological Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Qiwei Li
- Department of Mathematics, University of Texas at Dallas, Dallas, TX, USA
| | - Philippe E Zimmern
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicole J De Nisco
- Department of Biological Sciences, University of Texas at Dallas, Dallas, TX, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
10
|
Molina JJ, Kohler KN, Gager C, Andersen MJ, Wongso E, Lucas ER, Paik A, Xu W, Donahue DL, Bergeron K, Klim A, Caparon MG, Hultgren SJ, Desai A, Ploplis VA, Flick MJ, Castellino FJ, Flores-Mireles AL. Fibrinolytic-deficiencies predispose hosts to septicemia from a catheter-associated UTI. Nat Commun 2024; 15:2704. [PMID: 38538626 PMCID: PMC10973455 DOI: 10.1038/s41467-024-46974-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Catheter-associated urinary tract infections (CAUTIs) are amongst the most common nosocomial infections worldwide and are difficult to treat partly due to development of multidrug-resistance from CAUTI-related pathogens. Importantly, CAUTI often leads to secondary bloodstream infections and death. A major challenge is to predict when patients will develop CAUTIs and which populations are at-risk for bloodstream infections. Catheter-induced inflammation promotes fibrinogen (Fg) and fibrin accumulation in the bladder which are exploited as a biofilm formation platform by CAUTI pathogens. Using our established mouse model of CAUTI, here we identified that host populations exhibiting either genetic or acquired fibrinolytic-deficiencies, inducing fibrin deposition in the catheterized bladder, are predisposed to severe CAUTI and septicemia by diverse uropathogens in mono- and poly-microbial infections. Furthermore, here we found that Enterococcus faecalis, a prevalent CAUTI pathogen, uses the secreted protease, SprE, to induce fibrin accumulation and create a niche ideal for growth, biofilm formation, and persistence during CAUTI.
Collapse
Affiliation(s)
- Jonathan J Molina
- Integrated Biomedical Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Kurt N Kohler
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Christopher Gager
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Marissa J Andersen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Ellsa Wongso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Elizabeth R Lucas
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Andrew Paik
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Wei Xu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Deborah L Donahue
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Karla Bergeron
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aleksandra Klim
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael G Caparon
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Scott J Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alana Desai
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Urology, University of Washington Medical Center, Seattle, WA, 98133-9733, USA
| | - Victoria A Ploplis
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Matthew J Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- UNC Blood Research Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Francis J Castellino
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Ana L Flores-Mireles
- Integrated Biomedical Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN, 46556, USA.
| |
Collapse
|
11
|
Konesan J, Wang J, Moore KH, Mansfield KJ, Liu L. Cranberry, but not D-mannose and ibuprofen, prevents against uropathogenic Escherichia coli-induced cell damage and cell death in MDCK cells. Front Microbiol 2023; 14:1319785. [PMID: 38098676 PMCID: PMC10719950 DOI: 10.3389/fmicb.2023.1319785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/17/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction The main function of the urinary tract is to form an impermeable barrier against urinary solutes and bacteria. However, this barrier can be compromised by urinary tract infections, most commonly caused by uropathogenic Escherichia coli (UPEC). This can result in damage to the epithelial barrier, leading to decreased epithelial thickness, loss of tight junctions, loss of epithelial integrity, and apoptosis. Due to the rise in antimicrobial resistance, there is worldwide interest in exploring non-antibiotic agents as alternative therapy. Methods Using the Madin-Darby canine kidney (MDCK) cell line, a widely accepted epithelial cell model for the urinary tract, and the UPEC strain UTI89, this paper aimed to investigate the impact of UPEC on cell integrity, permeability, and barrier functions, and determine whether cranberry, D-mannose and ibuprofen could counteract the effects induced by UPEC. Furthermore, the study examined the protective potential of these agents against UPEC-induced increase in reactive oxygen species (ROS) production and programmed death-ligand 1 (PD-L1) expression. Results The results demonstrated that UTI89 caused a marked reduction in cell viability and monolayer integrity. Cranberry (3 mg/mL) was protective against these changes. In addition, cranberry exhibited protective effects against UPEC-induced damage to cell barrier integrity, escalation of oxidative stress, and UPEC/TNFα-triggered PD-L1 expression. However, no effect was observed for D-mannose and ibuprofen in alleviating UPEC-induced cell damage and changes in ROS and PD-L1 levels. Conclusion Overall, cranberry, but not D-mannose or ibuprofen, has a protective influence against UPEC associated damage in urinary epithelial cells.
Collapse
Affiliation(s)
- Jenane Konesan
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Jenny Wang
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Kate H. Moore
- St George Hospital, UNSW Sydney, Sydney, NSW, Australia
| | - Kylie J. Mansfield
- Graduate School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Lu Liu
- School of Biomedical Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
12
|
Hawas S, Vagenas D, Haque A, Totsika M. Bladder-draining lymph nodes support germinal center B cell responses during urinary tract infection in mice. Infect Immun 2023; 91:e0031723. [PMID: 37882531 PMCID: PMC10652902 DOI: 10.1128/iai.00317-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
Bacterial urinary tract infections (UTIs) are both common and exhibit high recurrence rates in women. UTI healthcare costs are increasing due to the rise of multidrug-resistant (MDR) bacteria, necessitating alternative approaches for infection control. Here, we directly observed host adaptive immune responses in acute UTI. We employed a mouse model in which wild-type C57BL/6J mice were transurethrally inoculated with a clinically relevant MDR UTI strain of uropathogenic Escherichia coli (UPEC). Firstly, we noted that rag1-/- C57BL/6J mice harbored larger bacterial burdens than wild-type counterparts, consistent with a role for adaptive immunity in UTI control. Consistent with this, UTI triggered in the bladders of wild-type mice early increases of myeloid cells, including CD11chi conventional dendritic cells, suggesting possible involvement of these professional antigen-presenting cells. Importantly, germinal center B cell responses developed by 4 weeks post-infection in bladder-draining lymph nodes of wild-type mice and, although modest in magnitude and transient in nature, could not be boosted with a second UTI. Thus, our data reveal for the first time in a mouse model that UPEC UTI induces local B cell immune responses in bladder-draining lymph nodes, which could potentially serve to control infection.
Collapse
Affiliation(s)
- Sophia Hawas
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Dimitrios Vagenas
- Research Methods Group, School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ashraful Haque
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
13
|
Russell SK, Harrison JK, Olson BS, Lee HJ, O'Brien VP, Xing X, Livny J, Yu L, Roberson EDO, Bomjan R, Fan C, Sha M, Estfanous S, Amer AO, Colonna M, Stappenbeck TS, Wang T, Hannan TJ, Hultgren SJ. Uropathogenic Escherichia coli infection-induced epithelial trained immunity impacts urinary tract disease outcome. Nat Microbiol 2023; 8:875-888. [PMID: 37037942 PMCID: PMC10159856 DOI: 10.1038/s41564-023-01346-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/20/2023] [Indexed: 04/12/2023]
Abstract
Previous urinary tract infections (UTIs) can predispose one to future infections; however, the underlying mechanisms affecting recurrence are poorly understood. We previously found that UTIs in mice cause differential bladder epithelial (urothelial) remodelling, depending on disease outcome, that impacts susceptibility to recurrent UTI. Here we compared urothelial stem cell (USC) lines isolated from mice with a history of either resolved or chronic uropathogenic Escherichia coli (UPEC) infection, elucidating evidence of molecular imprinting that involved epigenetic changes, including differences in chromatin accessibility, DNA methylation and histone modification. Epigenetic marks in USCs from chronically infected mice enhanced caspase-1-mediated cell death upon UPEC infection, promoting bacterial clearance. Increased Ptgs2os2 expression also occurred, potentially contributing to sustained cyclooxygenase-2 expression, bladder inflammation and mucosal wounding-responses associated with severe recurrent cystitis. Thus, UPEC infection acts as an epi-mutagen reprogramming the urothelial epigenome, leading to urothelial-intrinsic remodelling and training of the innate response to subsequent infection.
Collapse
Affiliation(s)
- Seongmi K Russell
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Jessica K Harrison
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Benjamin S Olson
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Hyung Joo Lee
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Valerie P O'Brien
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
- Fred Hutchinson Cancer Center, Human Biology Division, Seattle, WA, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Jonathan Livny
- Infectious Disease and Microbiome Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Lu Yu
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Elisha D O Roberson
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St Louis, MO, USA
| | - Rajdeep Bomjan
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Changxu Fan
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Marina Sha
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, Ohio State University, Columbus, OH, USA
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy Helwan University, Cairo, Egypt
| | - Amal O Amer
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, Ohio State University, Columbus, OH, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA.
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA.
| | - Thomas J Hannan
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| | - Scott J Hultgren
- Department of Molecular Microbiology and Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
14
|
Peelaerts W, Mercado G, George S, Villumsen M, Kasen A, Aguileta M, Linstow C, Sutter AB, Kuhn E, Stetzik L, Sheridan R, Bergkvist L, Meyerdirk L, Lindqvist A, Gavis MLE, Van den Haute C, Hultgren SJ, Baekelandt V, Pospisilik JA, Brudek T, Aznar S, Steiner JA, Henderson MX, Brundin L, Ivanova MI, Hannan TJ, Brundin P. Urinary tract infections trigger synucleinopathy via the innate immune response. Acta Neuropathol 2023; 145:541-559. [PMID: 36991261 PMCID: PMC10119259 DOI: 10.1007/s00401-023-02562-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 03/31/2023]
Abstract
Symptoms in the urogenital organs are common in multiple system atrophy (MSA), also in the years preceding the MSA diagnosis. It is unknown how MSA is triggered and these observations in prodromal MSA led us to hypothesize that synucleinopathy could be triggered by infection of the genitourinary tract causing ɑ-synuclein (ɑSyn) to aggregate in peripheral nerves innervating these organs. As a first proof that peripheral infections could act as a trigger in MSA, this study focused on lower urinary tract infections (UTIs), given the relevance and high frequency of UTIs in prodromal MSA, although other types of infection might also be important triggers of MSA. We performed an epidemiological nested-case control study in the Danish population showing that UTIs are associated with future diagnosis of MSA several years after infection and that it impacts risk in both men and women. Bacterial infection of the urinary bladder triggers synucleinopathy in mice and we propose a novel role of ɑSyn in the innate immune system response to bacteria. Urinary tract infection with uropathogenic E. coli results in the de novo aggregation of ɑSyn during neutrophil infiltration. During the infection, ɑSyn is released extracellularly from neutrophils as part of their extracellular traps. Injection of MSA aggregates into the urinary bladder leads to motor deficits and propagation of ɑSyn pathology to the central nervous system in mice overexpressing oligodendroglial ɑSyn. Repeated UTIs lead to progressive development of synucleinopathy with oligodendroglial involvement in vivo. Our results link bacterial infections with synucleinopathy and show that a host response to environmental triggers can result in ɑSyn pathology that bears semblance to MSA.
Collapse
Affiliation(s)
- Wouter Peelaerts
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Louvain, Belgium
- Laboratory for Virology and Gene Therapy, Department of Pharmacy and Pharmaceutical Sciences, KU Leuven, Louvain, Belgium
| | - Gabriela Mercado
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Sonia George
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Marie Villumsen
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Alysa Kasen
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Miguel Aguileta
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Christian Linstow
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Alexandra B Sutter
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily Kuhn
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Lucas Stetzik
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Rachel Sheridan
- Flow Cytometry Core Facility, Van Andel Institute, Grand Rapids, MI, USA
| | - Liza Bergkvist
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Lindsay Meyerdirk
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Allison Lindqvist
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Martha L Escobar Gavis
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Louvain, Belgium
- Leuven Viral Vector Core, Department of Neurosciences, KU Leuven, Louvain, Belgium
| | - Scott J Hultgren
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Louvain, Belgium
- Leuven Viral Vector Core, Department of Neurosciences, KU Leuven, Louvain, Belgium
| | | | - Tomasz Brudek
- Centre for Neuroscience and Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Susana Aznar
- Centre for Neuroscience and Stereology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Jennifer A Steiner
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Michael X Henderson
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Lena Brundin
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA
| | - Magdalena I Ivanova
- Neuroscience Graduate Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Biophysics Program, University of Michigan, Ann Arbor, MI, USA
| | - Tom J Hannan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Patrik Brundin
- Department of Neurodegenerative Science, Parkinson's Disease Center, Van Andel Institute, Grand Rapids, MI, USA.
- Pharma Research and Early Development (pRED), F. Hoffmann-La Roche, Basel, Switzerland.
| |
Collapse
|
15
|
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
|
16
|
Abstract
The bladder is a major component of the urinary tract, an organ system that expels metabolic waste and excess water, which necessitates proximity to the external environment and its pathogens. It also houses a commensal microbiome. Therefore, its tissue immunity must resist pathogen invasion while maintaining tolerance to commensals. Bacterial infection of the bladder is common, with half of women globally experiencing one or more episodes of cystitis in their lifetime. Despite this, our knowledge of bladder immunity, particularly in humans, is incomplete. Here we consider the current view of tissue immunity in the bladder, with a focus on defense against infection. The urothelium has robust immune functionality, and its defensive capabilities are supported by resident immune cells, including macrophages, dendritic cells, natural killer cells, and γδ T cells. We discuss each in turn and consider why adaptive immune responses are often ineffective in preventing recurrent infection, as well as areas of priority for future research.
Collapse
Affiliation(s)
- Georgina S Bowyer
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom;
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Kevin W Loudon
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom;
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Ondrej Suchanek
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom;
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom;
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
17
|
O’Brien VP, Lewis AL, Gilbert NM. Bladder Exposure to Gardnerella Activates Host Pathways Necessary for Escherichia coli Recurrent UTI. Front Cell Infect Microbiol 2021; 11:788229. [PMID: 34938672 PMCID: PMC8685330 DOI: 10.3389/fcimb.2021.788229] [Citation(s) in RCA: 5] [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] [Received: 10/01/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022] Open
Abstract
Recurrent urinary tract infections (rUTI) are a costly clinical problem affecting millions of women worldwide each year. The majority of rUTI cases are caused by uropathogenic Escherichia coli (UPEC). Data from humans and mouse models indicate that some instances of rUTI are caused by UPEC emerging from latent reservoirs in the bladder. Women with vaginal dysbiosis, typically characterized by high levels of Gardnerella and other anaerobes, are at increased risk of UTI. Multiple studies have detected Gardnerella in urine collected by transurethral catheterization (to limit vaginal contamination), suggesting that some women experience routine urinary tract exposures. We recently reported that inoculation of Gardnerella into the bladder triggers rUTI from UPEC bladder reservoirs in a mouse model. Here we performed whole bladder RNA-seq to identify host pathways involved in Gardnerella-induced rUTI. We identified a variety host pathways differentially expressed in whole bladders following Gardnerella exposure, such as pathways involved in inflammation/immunity and epithelial turnover. At the gene level, we identified upregulation of Immediate Early (IE) genes, which are induced in various cell types shortly following stimuli like infection and inflammation. One such upregulated IE gene was the orphan nuclear receptor Nur77 (aka Nr4a1). Pilot experiments in Nur77-/- mice suggest that Nur77 is necessary for Gardnerella exposure to trigger rUTI from UPEC reservoirs. These findings demonstrate that bladder gene expression can be impacted by short-lived exposures to urogenital bacteria and warrant future examination of responses in distinct cell types, such as with single cell transcriptomic technologies. The biological validation studies in Nur77-/- mice lay the groundwork for future studies investigating Nur77 and the Immediate Early response in rUTI.
Collapse
Affiliation(s)
- Valerie P. O’Brien
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Amanda L. Lewis
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, CA, United States
| | - Nicole M. Gilbert
- Department of Pediatrics, Division of Infectious Diseases, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
18
|
Sharma K, Thacker VV, Dhar N, Clapés Cabrer M, Dubois A, Signorino-Gelo F, Mullenders J, Knott GW, Clevers H, McKinney JD. Early invasion of the bladder wall by solitary bacteria protects UPEC from antibiotics and neutrophil swarms in an organoid model. Cell Rep 2021; 36:109351. [PMID: 34289360 DOI: 10.1016/j.celrep.2021.109351] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/26/2021] [Accepted: 06/15/2021] [Indexed: 01/24/2023] Open
Abstract
Recurrence of uropathogenic Escherichia coli (UPEC) infections has been attributed to reactivation of quiescent intracellular reservoirs (QIRs) in deep layers of the bladder wall. QIRs are thought to arise late during infection following dispersal of bacteria from intracellular bacterial communities (IBCs) in superficial umbrella cells. Here, we track the formation of QIR-like bacteria in a bladder organoid model that recapitulates the stratified uroepithelium within a volume suitable for high-resolution live-cell imaging. Bacteria injected into the organoid lumen enter umbrella-like cells and proliferate to form IBC-like bodies. In parallel, single bacteria penetrate deeper layers of the organoid wall, where they localize within or between uroepithelial cells. These "solitary" bacteria evade killing by antibiotics and neutrophils and are morphologically distinct from bacteria in IBCs. We conclude that bacteria with QIR-like properties may arise at early stages of infection, independent of IBC formation and rupture.
Collapse
Affiliation(s)
- Kunal Sharma
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Vivek V Thacker
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Neeraj Dhar
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Maria Clapés Cabrer
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Anaëlle Dubois
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - François Signorino-Gelo
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Jasper Mullenders
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, the Netherlands
| | - Graham W Knott
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Utrecht, the Netherlands
| | - John D McKinney
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
19
|
Ebrahimzadeh T, Kuprasertkul A, Neugent ML, Lutz KC, Fuentes JL, Gadhvi J, Khan F, Zhang C, Sharon BM, Orth K, Li Q, Zimmern PE, De Nisco NJ. Urinary prostaglandin E2 as a biomarker for recurrent UTI in postmenopausal women. Life Sci Alliance 2021; 4:4/7/e202000948. [PMID: 33958485 PMCID: PMC8200289 DOI: 10.26508/lsa.202000948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 01/11/2023] Open
Abstract
This work uses controlled human cohorts to investigate urinary prostaglandin E2, the product of cyclooxygenase-2, as both a diagnostic and prognostic biomarker of recurrent UTI postmenopausal women. Urinary tract infection (UTI) is one of the most common adult bacterial infections and exhibits high recurrence rates, especially in postmenopausal women. Studies in mouse models suggest that cyclooxygenase-2 (COX-2)–mediated inflammation sensitizes the bladder to recurrent UTI (rUTI). However, COX-2–mediated inflammation has not been robustly studied in human rUTI. We used human cohorts to assess urothelial COX-2 production and evaluate its product, PGE2, as a biomarker for rUTI in postmenopausal women. We found that the percentage of COX-2–positive cells was elevated in inflamed versus uninflamed bladder regions. We analyzed the performance of urinary PGE2 as a biomarker for rUTI in a controlled cohort of 92 postmenopausal women and PGE2 consistently outperformed all other tested clinical variables as a predictor of rUTI status. Furthermore, time-to-relapse analysis indicated that the risk of rUTI relapse was 3.6 times higher in women with above median urinary PGE2 levels than with below median levels. Taken together, these data suggest that urinary PGE2 may be a clinically useful diagnostic and prognostic biomarker for rUTI in postmenopausal women.
Collapse
Affiliation(s)
- Tahmineh Ebrahimzadeh
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Amy Kuprasertkul
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael L Neugent
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Kevin C Lutz
- Depatment of Mathematics, University of Texas at Dallas, Richardson, TX, USA
| | - Jorge L Fuentes
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jashkaran Gadhvi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Fatima Khan
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Cong Zhang
- Depatment of Mathematics, University of Texas at Dallas, Richardson, TX, USA
| | - Belle M Sharon
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qiwei Li
- Depatment of Mathematics, University of Texas at Dallas, Richardson, TX, USA
| | - Philippe E Zimmern
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicole J De Nisco
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
20
|
Zhang Z, Wang M, Zhang Y, Zhang Y, Bartkuhn M, Markmann M, Hossain H, Chakraborty T, Hake SB, Jia Z, Meinhardt A, Bhushan S. Uropathogenic Escherichia coli Virulence Factor α-Hemolysin Reduces Histone Acetylation to Inhibit Expression of Proinflammatory Cytokine Genes. J Infect Dis 2021; 223:1040-1051. [PMID: 33453118 DOI: 10.1093/infdis/jiab018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Urinary tract infections are common and costly diseases affecting millions of people. Uropathogenic Escherichia coli (UPEC) is a primary cause of these infections and has developed multiple strategies to avoid the host immune response. Here, we dissected the molecular mechanisms underpinning UPEC inhibition of inflammatory cytokine in vitro and in vivo. We found that UPEC infection simulates nuclear factor-κB activation but does not result in transcription of cytokine genes. Instead, UPEC-mediated suppression of the metabolic enzyme ATP citrate lyase results in decreased acetyl-CoA levels, leading to reduced H3K9 histone acetylation in the promotor region of CXCL8. These effects were dependent on the UPEC virulence factor α-hemolysin and were reversed by exogenous acetate. In a murine cystitis model, prior acetate supplementation rapidly resolved UPEC-elicited immune responses and improved tissue recovery. Thus, upon infection, UPEC rearranges host cell metabolism to induce chromatin remodeling processes that subvert expression of host innate immune response genes.
Collapse
Affiliation(s)
- Zhengguo Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Ming Wang
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Marek Bartkuhn
- Institute for Genetics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Melanie Markmann
- Institute of Medical Microbiology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Hamid Hossain
- Klinikum St Marien Amberg, Amberg and Kliniken Nordoberpfalz AG, Weiden, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sandra B Hake
- Institute for Genetics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Zhankui Jia
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andreas Meinhardt
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sudhanshu Bhushan
- Institute of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
21
|
Ambite I, Butler D, Wan MLY, Rosenblad T, Tran TH, Chao SM, Svanborg C. Molecular determinants of disease severity in urinary tract infection. Nat Rev Urol 2021; 18:468-486. [PMID: 34131331 PMCID: PMC8204302 DOI: 10.1038/s41585-021-00477-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
The most common and lethal bacterial pathogens have co-evolved with the host. Pathogens are the aggressors, and the host immune system is responsible for the defence. However, immune responses can also become destructive, and excessive innate immune activation is a major cause of infection-associated morbidity, exemplified by symptomatic urinary tract infections (UTIs), which are caused, in part, by excessive innate immune activation. Severe kidney infections (acute pyelonephritis) are a major cause of morbidity and mortality, and painful infections of the urinary bladder (acute cystitis) can become debilitating in susceptible patients. Disease severity is controlled at specific innate immune checkpoints, and a detailed understanding of their functions is crucial for strategies to counter microbial aggression with novel treatment and prevention measures. One approach is the use of bacterial molecules that reprogramme the innate immune system, accelerating or inhibiting disease processes. A very different outcome is asymptomatic bacteriuria, defined by low host immune responsiveness to bacteria with attenuated virulence. This observation provides the rationale for immunomodulation as a new therapeutic tool to deliberately modify host susceptibility, control the host response and avoid severe disease. The power of innate immunity as an arbitrator of health and disease is also highly relevant for emerging pathogens, including the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Ines Ambite
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Daniel Butler
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Therese Rosenblad
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sing Ming Chao
- Nephrology Service, Department of Paediatrics, KK Hospital, Singapore, Singapore
| | - Catharina Svanborg
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Ligon MM, Wang C, DeJong EN, Schulz C, Bowdish DME, Mysorekar IU. Single cell and tissue-transcriptomic analysis of murine bladders reveals age- and TNFα-dependent but microbiota-independent tertiary lymphoid tissue formation. Mucosal Immunol 2020; 13:908-918. [PMID: 32366865 PMCID: PMC7572484 DOI: 10.1038/s41385-020-0290-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/02/2020] [Accepted: 03/31/2020] [Indexed: 02/04/2023]
Abstract
Aging has multifaceted effects on the immune system, but how aging affects tissue-specific immunity is not well-defined. Bladder diseases characterized by chronic inflammation are highly prevalent in older women, but mechanisms by which aging promotes these pathologies remain unknown. Tissue transcriptomics of unperturbed, young and aged bladders identified a highly altered immune landscape as a fundamental feature of the aging female bladder. Detailed mapping of immune cells using single cell RNA-sequencing revealed novel subsets of macrophages and dendritic cells and unique changes to the immune repertoire in the aged bladder. B and T cells are highly enriched in aged bladders and spontaneously form organized bladder tertiary lymphoid tissues (bTLTs). Naïve, activated, and germinal center B cells and IgA+ plasma cells are found within bTLT and associated with increased urinary IgA concentrations. bTLTs form with increasing age and their formation is dependent on TNFα. Microbiota are not required to form bTLT, as aged germfree mice also harbor them. Thus, bTLTs require age-dependent TNFα but are independent of the microbiota. Our results indicate that chronic, age-associated inflammation underlies a fundamental alteration to the bladder and establishes a resource for further investigation of the bladder immune system in homeostasis, aging, and disease.
Collapse
Affiliation(s)
- Marianne M. Ligon
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Caihong Wang
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Erica N. DeJong
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Christian Schulz
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Dawn M. E. Bowdish
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Indira U. Mysorekar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,To whom correspondence should be addressed: Indira U. Mysorekar, Ph.D., Washington University School of Medicine, Depts. of Obstetrics and Gynecology & Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, Phone: 314-747-1329, Fax: 314-747-1350,
| |
Collapse
|
23
|
Chamoun MN, Sullivan MJ, Goh KGK, Acharya D, Ipe DS, Katupitiya L, Gosling D, Peters KM, Sweet MJ, Sester DP, Schembri MA, Ulett GC. Restriction of chronic Escherichia coli urinary tract infection depends upon T cell-derived interleukin-17, a deficiency of which predisposes to flagella-driven bacterial persistence. FASEB J 2020; 34:14572-14587. [PMID: 32901999 DOI: 10.1096/fj.202000760r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 01/04/2023]
Abstract
Urinary tract infections (UTI) frequently progress to chronicity in infected individuals but the mechanisms of pathogenesis underlying chronic UTI are not well understood. We examined the role of interleukin (IL)-17A in UTI because this cytokine promotes innate defense against uropathogenic Escherichia coli (UPEC). Analysis of UPEC persistence and pyelonephritis in mice deficient in IL-17A revealed that UPEC CFT073 caused infection at a rate higher than the multidrug resistant strain EC958. Il17a-/- mice exhibited pyelonephritis with kidney bacterial burdens higher than those of wild-type (WT) mice. Synthesis of IL-17A in the bladder reflected a combination of γδ-T and TH 17 cell responses. Analysis of circulating inflammatory mediators at 24h postinoculation identified predictors of progression to chronicity, including IL-6 and monocyte chemoattractant protein-1 (MCP-1). Histological analysis identified infiltrating populations of neutrophils, NK cells, and γδ T cells in the bladder, whereas neutrophils predominated in the kidney. Analysis of the contribution of flagella to chronicity using hyper-flagellated and fliC-deficient UPEC in WT and Il17a-/- mice revealed that, in a host that is deficient for the production of IL-17A, flagella contribute to bacterial persistence. These findings show a role for IL-17A in defense against chronic UTI and a contribution of flagella to the pathogenesis of infection.
Collapse
Affiliation(s)
- Michelle N Chamoun
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Matthew J Sullivan
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Kelvin G K Goh
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Dhruba Acharya
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Deepak S Ipe
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Lahiru Katupitiya
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Dean Gosling
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| | - Kate M Peters
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - David P Sester
- TRI Flow Cytometry Suite (TRI.fcs), Translational Research Institute, Wooloongabba, QLD, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Glen C Ulett
- School of Medical Sciences, And Menzies Health Institute Queensland, Griffith University, Parklands, QLD, Australia
| |
Collapse
|
24
|
Moldoveanu AL, Rycroft JA, Helaine S. Impact of bacterial persisters on their host. Curr Opin Microbiol 2020; 59:65-71. [PMID: 32866708 DOI: 10.1016/j.mib.2020.07.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023]
Abstract
The rise of antibiotic failure poses a severe threat to global health. There is growing concern that this failure is not solely driven by stable antibiotic resistance but also by a subpopulation of transiently non-growing, antibiotic tolerant bacteria. These 'persisters' have been proposed to seed relapsing infections, an important clinical outcome of treatment failure - although definitive evidence for this direct link remains elusive. Recent advances in the field have revealed the complex nature of intra-host persisters which drive their high adaptability through biosynthetic activity. These features of persisters contribute to evolution of antimicrobial resistance and modulation of host immune responses, despite clinically efficacious treatment.
Collapse
Affiliation(s)
- Ana L Moldoveanu
- Centre for Molecular Bacteriology and Infection, Imperial College London, South Kensington, London SW7 2AZ, UK
| | - Julian A Rycroft
- Department of Microbiology, Harvard Medical School, 77 Ave Pasteur, Boston, MA 02115, USA
| | - Sophie Helaine
- Department of Microbiology, Harvard Medical School, 77 Ave Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Lacerda Mariano L, Ingersoll MA. The immune response to infection in the bladder. Nat Rev Urol 2020; 17:439-458. [PMID: 32661333 DOI: 10.1038/s41585-020-0350-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2020] [Indexed: 12/22/2022]
Abstract
The bladder is continuously protected by passive defences such as a mucus layer, antimicrobial peptides and secretory immunoglobulins; however, these defences are occasionally overcome by invading bacteria that can induce a strong host inflammatory response in the bladder. The urothelium and resident immune cells produce additional defence molecules, cytokines and chemokines, which recruit inflammatory cells to the infected tissue. Resident and recruited immune cells act together to eradicate bacteria from the bladder and to develop lasting immune memory against infection. However, urinary tract infection (UTI) is commonly recurrent, suggesting that the induction of a memory response in the bladder is inadequate to prevent reinfection. Additionally, infection seems to induce long-lasting changes in the urothelium, which can render the tissue more susceptible to future infection. The innate immune response is well-studied in the field of UTI, but considerably less is known about how adaptive immunity develops and how repair mechanisms restore bladder homeostasis following infection. Furthermore, data demonstrate that sex-based differences in immunity affect resolution and infection can lead to tissue remodelling in the bladder following resolution of UTI. To combat the rise in antimicrobial resistance, innovative therapeutic approaches to bladder infection are currently in development. Improving our understanding of how the bladder responds to infection will support the development of improved treatments for UTI, particularly for those at risk of recurrent infection.
Collapse
Affiliation(s)
- Livia Lacerda Mariano
- Department of Immunology, Institut Pasteur, Paris, France.,Inserm, U1223, Paris, France
| | - Molly A Ingersoll
- Department of Immunology, Institut Pasteur, Paris, France. .,Inserm, U1223, Paris, France.
| |
Collapse
|
26
|
Advances in Understanding the Human Urinary Microbiome and Its Potential Role in Urinary Tract Infection. mBio 2020; 11:mBio.00218-20. [PMID: 32345639 PMCID: PMC7188990 DOI: 10.1128/mbio.00218-20] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent advances in the analysis of microbial communities colonizing the human body have identified a resident microbial community in the human urinary tract (UT). Compared to many other microbial niches, the human UT harbors a relatively low biomass. Studies have identified many genera and species that may constitute a core urinary microbiome. However, the contribution of the UT microbiome to urinary tract infection (UTI) and recurrent UTI (rUTI) pathobiology is not yet clearly understood. Evidence suggests that commensal species within the UT and urogenital tract (UGT) microbiomes, such as Lactobacillus crispatus, may act to protect against colonization with uropathogens. However, the mechanisms and fundamental biology of the urinary microbiome-host relationship are not understood. The ability to measure and characterize the urinary microbiome has been enabled through the development of next-generation sequencing and bioinformatic platforms that allow for the unbiased detection of resident microbial DNA. Translating technological advances into clinical insight will require further study of the microbial and genomic ecology of the urinary microbiome in both health and disease. Future diagnostic, prognostic, and therapeutic options for the management of UTI may soon incorporate efforts to measure, restore, and/or preserve the native, healthy ecology of the urinary microbiomes.
Collapse
|
27
|
Urinary tract infections: microbial pathogenesis, host-pathogen interactions and new treatment strategies. Nat Rev Microbiol 2020; 18:211-226. [PMID: 32071440 DOI: 10.1038/s41579-020-0324-0] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2020] [Indexed: 12/14/2022]
Abstract
Urinary tract infections (UTIs) are common, recurrent infections that can be mild to life-threatening. The continued emergence of antibiotic resistance, together with our increasing understanding of the detrimental effects conferred by broad-spectrum antibiotic use on the health of the beneficial microbiota of the host, has underscored the weaknesses in our current treatment paradigm for UTIs. In this Review, we discuss how recent microbiological, structural, genetic and immunological studies have expanded our understanding of host-pathogen interactions during UTI pathogenesis. These basic scientific findings have the potential to shift the strategy for UTI treatment away from broad-spectrum antibiotics targeting conserved aspects of bacterial replication towards pathogen-specific antibiotic-sparing therapeutics that target core determinants of bacterial virulence at the host-pathogen interface.
Collapse
|