1
|
Ambite I, Chao SM, Rosenblad T, Hopkins R, Storm P, Ng YH, Ganesan I, Lindén M, Haq F, Tran TH, Ahmadi S, Lee B, Chen SL, Godaly G, Brandström P, Connolly JE, Svanborg C. Molecular analysis of acute pyelonephritis-excessive innate and attenuated adaptive immunity. Life Sci Alliance 2025; 8:e202402926. [PMID: 40036168 PMCID: PMC11662066 DOI: 10.26508/lsa.202402926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 03/06/2025] Open
Abstract
This study investigated the molecular basis of disease severity in acute pyelonephritis (APN), a common and potentially life-threatening bacterial infection. Two cohorts of infants with febrile urinary tract infection were included. Renal involvement was defined by DMSA scans and molecular disease determinants by gene expression analysis and proteomic screens, at diagnosis and after 6 mo. Innate immune hyper-activation, systemically and locally in the urinary tract, was defined as a cytokine storm. Neutrophil degranulation and renal toxicity genes were strongly regulated, with overexpression in the APN group (first DMSA+). Adaptive immune attenuation in the APN group further supported the notion of an immune imbalance. DNA exome genotyping identified APN and febrile urinary tract infection as genetically distinct and scarring associated genes, but the activation of renal toxicity genes during acute infection was unrelated to the development of renal scarring. The results define APN as a hyper-inflammatory disorder with the characteristics of a cytokine storm combined with adaptive immune attenuation. The findings are consistent with innate immune dysfunctions and neutrophil disorders identified as determinants of APN susceptibility in genetic models.
Collapse
Affiliation(s)
- Ines Ambite
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sing Ming Chao
- Duke-National University of Singapore Academic Clinical Program, Pediatric Nephrology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Therese Rosenblad
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Pediatrics, Lund Children's Hospital, Lund, Sweden
| | - Richard Hopkins
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Petter Storm
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Yong Hong Ng
- Duke-National University of Singapore Academic Clinical Program, Pediatric Nephrology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Indra Ganesan
- Duke-National University of Singapore Academic Clinical Program, Pediatric Nephrology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Magnus Lindén
- Department of Pediatrics, Halland Hospital, Halmstad, Sweden
| | - Farhan Haq
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Shahram Ahmadi
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Bernett Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Swaine L Chen
- Laboratory of Bacterial Genomics, Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Infectious Diseases Translational Research Program, Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Gabriela Godaly
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Per Brandström
- Pediatric Uro-Nephrology Center, Queen Silvia's Children's Hospital, Gothenburg, Sweden
- University of Gothenburg, Gothenburg, Sweden
| | - John E Connolly
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Catharina Svanborg
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Ambite I, Wan MLY, Tran HT, Nazari A, Chaudhuri A, Krintel C, Gomes I, Sabari S, Ahmadi S, Carneiro ANBM, Ishac R, Haq F, Svanborg C. Multitarget mechanism of MYC inhibition by the bacterial lon protease in disease. Sci Rep 2025; 15:6778. [PMID: 40000737 PMCID: PMC11861601 DOI: 10.1038/s41598-025-88093-2] [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: 10/31/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Identifying specific inhibitors of the MYC oncogene has been challenging, due to off target effects associated with MYC inhibition. This study investigated how the recombinant Escherichia coli Lon protease (rLon), which targets MYC in human cells, inhibits MYC over-activation in models of infection and cancer. In silico predictions identified specific peptide domains of bacterial Lon that target MYC and the affinity of these peptides for MYC was investigated by surface plasmon resonance. The N-terminal domain of rLon was shown to interact with the C-terminal, leucine zipper domain of MYC and MAX and to prevent MYC/MAX dimerization. Furthermore, rLon targeted and degraded c-MYC in vitro and in cellular models, through the peptidase domain. In a model of kidney infection, rLon treatment prevented, c-MYC, N-MYC and L-MYC over-expression, MYC-dependent gene expression, specifically renal toxicity genes and pathology, suggesting that rLon recognizes and corrects MYC dysregulation in this disease. The findings describe a multitarget mechanism of MYC inhibition by rLon, and the combined effects achieved by the Lon domains, targeting different MYC epitopes and MYC-dependent functions, with no evidence of toxicity or detrimental effects on homeostatic MYC expression.
Collapse
Affiliation(s)
- Ines Ambite
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Murphy Lam Yim Wan
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Hien Thi Tran
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Atefeh Nazari
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Arunima Chaudhuri
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Christian Krintel
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Inês Gomes
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Samudra Sabari
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Shahram Ahmadi
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - António N B M Carneiro
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Rita Ishac
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Farhan Haq
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Catharina Svanborg
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden.
| |
Collapse
|
3
|
Yesilbas Aksel Y, Barut EN, Engin S. Fosaprepitant improves cyclophosphamide-induced bladder damage by alleviating inflammatory response in mice. Toxicol Appl Pharmacol 2024; 492:117120. [PMID: 39378958 DOI: 10.1016/j.taap.2024.117120] [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: 04/25/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
Inhibition of inflammatory process is a key therapeutic target for the treatment of interstitial cystitis (IC). Recent reports indicate that neurokinin 1 receptor (NK1R) antagonists have beneficial roles in inflammatory-based diseases. Herein, we investigate the protective effects of fosaprepitant (FOS), a NK1R antagonist, in cyclophosphamide (CP)-induced cystitis. The cystitis model was established multiple CP (80 mg/kg; i.p.) injection one day apart, and mice were treated with FOS (20 and 60 mg/kg/day; i.p.) for seven consecutive days. Detrusor contractility, vesical vascular permeability, myeloperoxidase (MPO) activity and protein expression levels of the TLR4 pathway were evaluated in mice bladder. Carbachol and electric field stimulation-evoked contractions of detrusor strips were significantly increased in CP-treated mice, which was significantly attenuated by FOS (60 mg/kg/day) treatment (p<0.001, p<0.05). Notably, vesical vascular permeability was markedly impaired in CP-induced cystitis, that was restored by FOS (60 mg/kg/day) treatment (p<0.01). MPO activity was significantly increased in cystitis group whereas FOS (20 and 60 mg/kg/day) treatment remarkably suppressed MPO activity in bladder tissue (p<0.001). Although TLR4 expression increased with cystitis, MyD88 and p-NFκBSer536/total NFκB did not change, FOS (20 and 60 mg/kg/day) treatment caused a dramatic decrease in TLR4 expression (p<0.001), indicating the anti-inflammatory effect of FOS. In conclusion, FOS improved detrusor overactivity and inflammatory response by inhibiting MPO activity and TLR4 expression, resulting in functional and histological recovery in CP-induced cystitis.
Collapse
Affiliation(s)
- Yaren Yesilbas Aksel
- Karadeniz Technical University, Graduate School of Health Sciences, Department of Pharmacology, Türkiye
| | - Elif Nur Barut
- Karadeniz Technical University, Faculty of Pharmacy, Department of Pharmacology, Trabzon, Türkiye.
| | - Seckin Engin
- Karadeniz Technical University, Faculty of Pharmacy, Department of Pharmacology, Trabzon, Türkiye
| |
Collapse
|
4
|
Yu J, Varella Pereira GM, Allen-Brady K, Cuffolo R, Siddharth A, Koch M, Chua JWF, Sorrentino F, Dytko O, Ng KY, Violette P, Khullar V, Wang ZT, Cartwright R. Genetic polymorphisms associated with urinary tract infection in children and adults: a systematic review and meta-analysis. Am J Obstet Gynecol 2024; 230:600-609.e3. [PMID: 38128862 DOI: 10.1016/j.ajog.2023.12.018] [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: 08/22/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
INTRODUCTION The lifetime risk of urinary tract infection is known from first-degree relative studies to be highly heritable. Associations have also been observed across the life course from pediatric urinary tract infection to recurrent urinary tract infection in adulthood, suggesting lifelong susceptibility factors. Candidate gene studies and genome-wide association studies have tested for genetic associations of urinary tract infection; however, no contemporary systematic synthesis of studies is available. OBJECTIVE We conducted a systematic review to identify all genetic polymorphisms tested for an association with urinary tract infection in children and adults; and to assess their strength, consistency, and risk of bias among reported associations. DATA SOURCES AND STUDY ELIGIBILITY CRITERIA PubMed, HuGE Navigator and Embase were searched from January 1, 2005 to November 16, 2023, using a combination of genetic and phenotype key words. STUDY APPRAISAL AND SYNTHESIS METHODS Fixed and random effects meta-analyses were conducted using codominant models of inheritance in metan. The interim Venice criteria were used to assess their credibility of pooled associations. RESULTS After removing 451 duplicates, 1821 studies reports were screened, with 106 selected for full-text review, 22 were included in the meta-analysis (7 adult studies and 15 pediatric studies). Our meta-analyses demonstrated significant pooled associations for pediatric urinary tract infection with variation in CXCR1, IL8, TGF, TLR4 and VDR; all of which have plausible roles in the pathogenesis of urinary tract infection. Our meta-analyses also demonstrated a significant pooled association for adult urinary tract infection with variation in CXCR1. All significant pooled associations were graded according to their epidemiological credibility, sample sizes, heterogeneity between studies, and risk of bias. CONCLUSION This systematic review provides a current synthesis of the known genetic architecture of urinary tract infection in childhood and adulthood; and should provide important information for researchers analysing future genetic association studies. Although, overall, the credibility of pooled associations was weak, the consistency of findings for rs2234671 single nucleotide polymorphisms of CXCR1 in both populations suggest a key role in the urinary tract infection pathogenesis.
Collapse
Affiliation(s)
- Jiakun Yu
- University Hospitals Sussex NHS Foundation Trust, Brighton, United Kingdom.
| | - Glaucia Miranda Varella Pereira
- Department of Epidemiology and Biostatistics, Imperial College London, Norfolk Place, London, United Kingdom; Department of Urogynaecology, LNWH NHS Trust, London, United Kingdom; Department of Obstetrics and Gynaecology, School of Medical Sciences, University of Campinas, Brazil
| | - Kristina Allen-Brady
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Romana Cuffolo
- Department of Obstetrics and Gynaecology, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Aditi Siddharth
- John Radcliffe Hospital, Headley Way, Headington, Oxford, United Kingdom
| | - Marianne Koch
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - John W F Chua
- Frimley Health NHS Foundation Trust, Frimley, United Kingdom
| | - Felice Sorrentino
- Department of Medical and Surgical Sciences, Institute of Obstetrics and Gynecology, University of Foggia, Foggia, Italy
| | - Oskar Dytko
- Imperial College School of Medicine, Imperial College London, London, United Kingdom
| | - Kaa-Yung Ng
- Imperial College School of Medicine, Imperial College London, London, United Kingdom
| | - Philippe Violette
- Department of Health Research Methods, Evidence and Impact (HEI) and Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Vik Khullar
- Department of Urogynaecology, Imperial College London, United Kingdom
| | - Zhan Tao Wang
- Department of Surgery, Division of Urology, University of Western Ontario, London, Ontario, Canada
| | - Rufus Cartwright
- Department of Epidemiology and Biostatistics, Imperial College London, Norfolk Place, London, United Kingdom; Department of Urogynaecology, LNWH NHS Trust, London, United Kingdom; Department of Urogynaecology, Chelsea and Westminster NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
5
|
Mercado-Evans V, Chew C, Serchejian C, Saltzman A, Mejia ME, Zulk JJ, Cornax I, Nizet V, Patras KA. Tamm-Horsfall protein augments neutrophil NETosis during urinary tract infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578501. [PMID: 38370726 PMCID: PMC10871275 DOI: 10.1101/2024.02.01.578501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Urinary neutrophils are a hallmark of urinary tract infection (UTI), yet the mechanisms governing their activation, function, and efficacy in controlling infection remain incompletely understood. Tamm-Horsfall glycoprotein (THP), the most abundant protein in urine, uses terminal sialic acids to bind an inhibitory receptor and dampen neutrophil inflammatory responses. We hypothesized that neutrophil modulation is an integral part of THP-mediated host protection. In a UTI model, THP-deficient mice showed elevated urinary tract bacterial burdens, increased neutrophil recruitment, and more severe tissue histopathological changes compared to WT mice. Furthermore, THP-deficient mice displayed impaired urinary NETosis during UTI. To investigate the impact of THP on NETosis, we coupled in vitro fluorescence-based NET assays, proteomic analyses, and standard and imaging flow cytometry with peripheral human neutrophils. We found that THP increases proteins involved in respiratory chain, neutrophil granules, and chromatin remodeling pathways, enhances NETosis in an ROS-dependent manner, and drives NET-associated morphologic features including nuclear decondensation. These effects were observed only in the presence of a NETosis stimulus and could not be solely replicated with equivalent levels of sialic acid alone. We conclude that THP is a critical regulator of NETosis in the urinary tract, playing a key role in host defense against UTI.
Collapse
Affiliation(s)
- Vicki Mercado-Evans
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | - Claude Chew
- Cytometry and Cell Sorting Core, Baylor College of Medicine, Houston, Texas, USA
| | - Camille Serchejian
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Alexander Saltzman
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, Texas, USA
| | - Marlyd E. Mejia
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Ingrid Cornax
- Department of Pediatrics, UC San Diego, La Jolla, California, USA
| | - Victor Nizet
- Department of Pediatrics, UC San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, California, USA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Ambite I, Tran TH, Butler DSC, Cavalera M, Wan MLY, Ahmadi S, Svanborg C. Therapeutic Effects of IL-1RA against Acute Bacterial Infections, including Antibiotic-Resistant Strains. Pathogens 2023; 13:42. [PMID: 38251349 PMCID: PMC10820880 DOI: 10.3390/pathogens13010042] [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: 12/07/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Innate immunity is essential for the anti-microbial defense, but excessive immune activation may cause severe disease. In this study, immunotherapy was shown to prevent excessive innate immune activation and restore the anti-bacterial defense. E. coli-infected Asc-/- mice develop severe acute cystitis, defined by IL-1 hyper-activation, high bacterial counts, and extensive tissue pathology. Here, the interleukin-1 receptor antagonist (IL-1RA), which inhibits IL-1 hyper-activation in acute cystitis, was identified as a more potent inhibitor of inflammation and NK1R- and substance P-dependent pain than cefotaxime. Furthermore, IL-1RA treatment inhibited the excessive innate immune activation in the kidneys of infected Irf3-/- mice and restored tissue integrity. Unexpectedly, IL-1RA also accelerated bacterial clearance from infected bladders and kidneys, including antibiotic-resistant E. coli, where cefotaxime treatment was inefficient. The results suggest that by targeting the IL-1 response, control of the innate immune response to infection may be regained, with highly favorable treatment outcomes, including infections caused by antibiotic-resistant strains.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Catharina Svanborg
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, 221 84 Lund, Sweden; (I.A.); (T.H.T.); (D.S.C.B.); (M.C.); (M.L.Y.W.); (S.A.)
| |
Collapse
|
7
|
Schwartz L, de Dios Ruiz-Rosado J, Stonebrook E, Becknell B, Spencer JD. Uropathogen and host responses in pyelonephritis. Nat Rev Nephrol 2023; 19:658-671. [PMID: 37479904 PMCID: PMC10913074 DOI: 10.1038/s41581-023-00737-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections seen in clinical practice. The ascent of UTI-causing pathogens to the kidneys results in pyelonephritis, which can trigger kidney injury, scarring and ultimately impair kidney function. Despite sizable efforts to understand how infections develop or are cleared in the bladder, our appreciation of the mechanisms by which infections develop, progress or are eradicated in the kidney is limited. The identification of virulence factors that are produced by uropathogenic Escherichia coli to promote pyelonephritis have begun to fill this knowledge gap, as have insights into the mechanisms by which kidney tubular epithelial cells oppose uropathogenic E. coli infection to prevent or eradicate UTIs. Emerging data also illustrate how specific cellular immune responses eradicate infection whereas other immune cell populations promote kidney injury. Insights into the mechanisms by which uropathogenic E. coli circumvent host immune defences or antibiotic therapy to cause pyelonephritis is paramount to the development of new prevention and treatment strategies to mitigate pyelonephritis and its associated complications.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Juan de Dios Ruiz-Rosado
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Stonebrook
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
8
|
Butler D, Ambite I, Wan MLY, Tran TH, Wullt B, Svanborg C. Immunomodulation therapy offers new molecular strategies to treat UTI. Nat Rev Urol 2022; 19:419-437. [PMID: 35732832 PMCID: PMC9214477 DOI: 10.1038/s41585-022-00602-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
Abstract
Innovative solutions are needed for the treatment of bacterial infections, and a range of antibacterial molecules have been explored as alternatives to antibiotics. A different approach is to investigate the immune system of the host for new ways of making the antibacterial defence more efficient. However, the immune system has a dual role as protector and cause of disease: in addition to being protective, increasing evidence shows that innate immune responses can become excessive and cause acute symptoms and tissue pathology during infection. This role of innate immunity in disease suggests that the immune system should be targeted therapeutically, to inhibit over-reactivity. The ultimate goal is to develop therapies that selectively attenuate destructive immune response cascades, while augmenting the protective antimicrobial defence but such treatment options have remained underexplored, owing to the molecular proximity of the protective and destructive effects of the immune response. The concept of innate immunomodulation therapy has been developed successfully in urinary tract infections, based on detailed studies of innate immune activation and disease pathogenesis. Effective, disease-specific, immunomodulatory strategies have been developed by targeting specific immune response regulators including key transcription factors. In acute pyelonephritis, targeting interferon regulatory factor 7 using small interfering RNA or treatment with antimicrobial peptide cathelicidin was protective and, in acute cystitis, targeting overactive effector molecules such as IL-1β, MMP7, COX2, cAMP and the pain-sensing receptor NK1R has been successful in vivo. Furthermore, other UTI treatment strategies, such as inhibiting bacterial adhesion and vaccination, have also shown promise. Hyperactivation of innate immunity is a disease determinant in urinary tract infections (UTIs). Modulation of innate immunity has promise as a therapy for UTIs. In this Review, the authors discuss potential mechanisms and immunomodulatory therapeutic strategies in UTIs. Excessive innate immune responses to infection cause symptoms and pathology in acute pyelonephritis and acute cystitis. Innate immunomodulation therapy is, therefore, a realistic option for treating these conditions. Targeting excessive innate immune responses at the level of transcription has been successful in animal models. Innate immunomodulation therapy reduces excessive inflammation and tissue pathology and accelerates bacterial clearance from infected kidneys and bladders in mice. Innate immunomodulation therapy also accelerates the clearance of antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
- Daniel Butler
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ines Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Björn Wullt
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
Ambite I, Filenko NA, Zaldastanishvili E, Butler DS, Tran TH, Chaudhuri A, Esmaeili P, Ahmadi S, Paul S, Wullt B, Putze J, Chen SL, Dobrindt U, Svanborg C. Active bacterial modification of the host environment through RNA polymerase II inhibition. J Clin Invest 2021; 131:140333. [PMID: 33320835 DOI: 10.1172/jci140333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 12/09/2020] [Indexed: 01/17/2023] Open
Abstract
Unlike pathogens, which attack the host, commensal bacteria create a state of friendly coexistence. Here, we identified a mechanism of bacterial adaptation to the host niche, where they reside. Asymptomatic carrier strains were shown to inhibit RNA polymerase II (Pol II) in host cells by targeting Ser2 phosphorylation, a step required for productive mRNA elongation. Assisted by a rare, spontaneous loss-of-function mutant from a human carrier, the bacterial NlpD protein was identified as a Pol II inhibitor. After internalization by host cells, NlpD was shown to target constituents of the Pol II phosphorylation complex (RPB1 and PAF1C), attenuating host gene expression. Therapeutic efficacy of a recombinant NlpD protein was demonstrated in a urinary tract infection model, by reduced tissue pathology, accelerated bacterial clearance, and attenuated Pol II-dependent gene expression. The findings suggest an intriguing, evolutionarily conserved mechanism for bacterial modulation of host gene expression, with a remarkable therapeutic potential.
Collapse
Affiliation(s)
- Inès Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Nina A Filenko
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Daniel Sc Butler
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Arunima Chaudhuri
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Parisa Esmaeili
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Shahram Ahmadi
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sanchari Paul
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Björn Wullt
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Johannes Putze
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Swaine L Chen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Infectious Diseases Group, Genome Institute Singapore, A*STAR, Singapore
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
10
|
McLellan LK, McAllaster MR, Kim AS, Tóthová Ľ, Olson PD, Pinkner JS, Daugherty AL, Hreha TN, Janetka JW, Fremont DH, Hultgren SJ, Virgin HW, Hunstad DA. A host receptor enables type 1 pilus-mediated pathogenesis of Escherichia coli pyelonephritis. PLoS Pathog 2021; 17:e1009314. [PMID: 33513212 PMCID: PMC7875428 DOI: 10.1371/journal.ppat.1009314] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 02/10/2021] [Accepted: 01/13/2021] [Indexed: 12/26/2022] Open
Abstract
Type 1 pili have long been considered the major virulence factor enabling colonization of the urinary bladder by uropathogenic Escherichia coli (UPEC). The molecular pathogenesis of pyelonephritis is less well characterized, due to previous limitations in preclinical modeling of kidney infection. Here, we demonstrate in a recently developed mouse model that beyond bladder infection, type 1 pili also are critical for establishment of ascending pyelonephritis. Bacterial mutants lacking the type 1 pilus adhesin (FimH) were unable to establish kidney infection in male C3H/HeN mice. We developed an in vitro model of FimH-dependent UPEC binding to renal collecting duct cells, and performed a CRISPR screen in these cells, identifying desmoglein-2 as a primary renal epithelial receptor for FimH. The mannosylated extracellular domain of human DSG2 bound directly to the lectin domain of FimH in vitro, and introduction of a mutation in the FimH mannose-binding pocket abolished binding to DSG2. In infected C3H/HeN mice, type 1-piliated UPEC and Dsg2 were co-localized within collecting ducts, and administration of mannoside FIM1033, a potent small-molecule inhibitor of FimH, significantly attenuated bacterial loads in pyelonephritis. Our results broaden the biological importance of FimH, specify the first renal FimH receptor, and indicate that FimH-targeted therapeutics will also have application in pyelonephritis. Urinary tract infections (UTIs) are among the most common bacterial infections in humans. While much has been discovered about how E. coli cause bladder infections, less is known about the host-pathogen interactions that underlie kidney infection (pyelonephritis). We employed recently developed mouse models to show that bacterial surface fibers called type 1 pili, which bear the adhesive protein FimH and are known to mediate E. coli binding to bladder epithelium, are also required for ascending kidney infection. We developed a cell-culture model of bacterial binding to renal collecting duct, then performed a screen using the gene-editing tool CRISPR to identify the first known FimH receptor in the kidney. This epithelial cell-surface protein, desmoglein-2, was shown to directly bind FimH, and we localized this binding to specific extracellular domains of DSG2. Further, we showed that mannosides, small-molecule FimH inhibitors currently in development to treat bladder infection, are also effective in experimental kidney infection. Our study reveals a novel host-pathogen interaction during pyelonephritis and demonstrates how this interaction may be therapeutically targeted.
Collapse
Affiliation(s)
- Lisa K. McLellan
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael R. McAllaster
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Arthur S. Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ľubomíra Tóthová
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Patrick D. Olson
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jerome S. Pinkner
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Allyssa L. Daugherty
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Teri N. Hreha
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - James W. Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Scott J. Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Herbert W. Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David A. Hunstad
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
11
|
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
|
12
|
Ching C, Schwartz L, Spencer JD, Becknell B. Innate immunity and urinary tract infection. Pediatr Nephrol 2020; 35:1183-1192. [PMID: 31197473 PMCID: PMC6908784 DOI: 10.1007/s00467-019-04269-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/17/2019] [Accepted: 04/25/2019] [Indexed: 01/31/2023]
Abstract
Urinary tract infections are a severe public health problem. The emergence and spread of antimicrobial resistance among uropathogens threaten to further compromise the quality of life and health of people who develop acute and recurrent upper and lower urinary tract infections. The host defense mechanisms that prevent invasive bacterial infection are not entirely delineated. However, recent evidence suggests that versatile innate immune defenses play a key role in shielding the urinary tract from invading uropathogens. Over the last decade, considerable advances have been made in defining the innate mechanisms that maintain immune homeostasis in the kidney and urinary tract. When these innate defenses are compromised or dysregulated, pathogen susceptibility increases. The objective of this review is to provide an overview of how basic science discoveries are elucidating essential innate host defenses in the kidney and urinary tract. In doing so, we highlight how these findings may ultimately translate into the clinic as new biomarkers or therapies for urinary tract infection.
Collapse
Affiliation(s)
- Christina Ching
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Urology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Laura Schwartz
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - John David Spencer
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Pediatric Nephrology, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Brian Becknell
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA.
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
- Division of Pediatric Nephrology, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| |
Collapse
|
13
|
Hingane S, Joshi N, Surjit M, Ranjith-Kumar CT. Hepatitis E Virus ORF2 Inhibits RIG-I Mediated Interferon Response. Front Microbiol 2020; 11:656. [PMID: 32351484 PMCID: PMC7174656 DOI: 10.3389/fmicb.2020.00656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/23/2020] [Indexed: 01/18/2023] Open
Abstract
Understanding the dynamics of host innate immune responses against a pathogen marks the first step toward developing intervention strategies against the pathogen. The cytosolic pattern recognition receptor retinoic acid-inducible gene I (RIG-I) has been shown to be the major innate immune sensor for hepatitis E virus (HEV). Here, we show that HEV capsid protein (ORF2), a 660 amino acid long protein, interferes with the RIG-I signaling. Interestingly, only the full length ORF2 protein but not the 112-608 ORF2 protein inhibited RIG-I dependent interferon response. Both synthetic agonist and virus induced RIG-I activation was modulated by ORF2. Interference of interferon response was confirmed by reporter assays involving different interferon inducible promoters, qRT PCR, ELISA, and immunofluorescence microscopy. Neither glycosylation nor dimerization of the ORF2 protein had any effect on the observed inhibition. Further analyses revealed that the ORF2 protein antagonized Toll-like receptor (TLR) pathways as well. ORF2 inhibited signaling by RIG-I and TLR adapters, IPS-1, MyD88, and TRIF but was unable to inhibit activation by ectopically expressed IRF3 suggesting that it may be acting at a site upstream of IRF3 and downstream of adapter proteins. Our data uncover a new mechanism by which HEV may interfere with the host antiviral signaling.
Collapse
Affiliation(s)
- Smita Hingane
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India
| | - Nishant Joshi
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India
| | - Milan Surjit
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India
| | - C T Ranjith-Kumar
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India.,University School of Biotechnology, Guru Gobind Singh Indraprastha University, Dwarka, India
| |
Collapse
|
14
|
Chung KW, Dhillon P, Huang S, Sheng X, Shrestha R, Qiu C, Kaufman BA, Park J, Pei L, Baur J, Palmer M, Susztak K. Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. Cell Metab 2019; 30:784-799.e5. [PMID: 31474566 PMCID: PMC7054893 DOI: 10.1016/j.cmet.2019.08.003] [Citation(s) in RCA: 409] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/18/2019] [Accepted: 08/02/2019] [Indexed: 12/24/2022]
Abstract
Fibrosis is the final common pathway leading to end-stage renal failure. By analyzing the kidneys of patients and animal models with fibrosis, we observed a significant mitochondrial defect, including the loss of the mitochondrial transcription factor A (TFAM) in kidney tubule cells. Here, we generated mice with tubule-specific deletion of TFAM (Ksp-Cre/Tfamflox/flox). While these mice developed severe mitochondrial loss and energetic deficit by 6 weeks of age, kidney fibrosis, immune cell infiltration, and progressive azotemia causing death were only observed around 12 weeks of age. In renal cells of TFAM KO (knockout) mice, aberrant packaging of the mitochondrial DNA (mtDNA) resulted in its cytosolic translocation, activation of the cytosolic cGAS-stimulator of interferon genes (STING) DNA sensing pathway, and thus cytokine expression and immune cell recruitment. Ablation of STING ameliorated kidney fibrosis in mouse models of chronic kidney disease, demonstrating how TFAM sequesters mtDNA to limit the inflammation leading to fibrosis.
Collapse
Affiliation(s)
- Ki Wung Chung
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Poonam Dhillon
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shizheng Huang
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Xin Sheng
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rojesh Shrestha
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengxiang Qiu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Brett A Kaufman
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Jihwan Park
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Liming Pei
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joseph Baur
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Matthew Palmer
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Ambite I, Butler DSC, Stork C, Grönberg-Hernández J, Köves B, Zdziarski J, Pinkner J, Hultgren SJ, Dobrindt U, Wullt B, Svanborg C. Fimbriae reprogram host gene expression - Divergent effects of P and type 1 fimbriae. PLoS Pathog 2019; 15:e1007671. [PMID: 31181116 PMCID: PMC6557620 DOI: 10.1371/journal.ppat.1007671] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/01/2019] [Indexed: 01/03/2023] Open
Abstract
Pathogens rely on a complex virulence gene repertoire to successfully attack their hosts. We were therefore surprised to find that a single fimbrial gene reconstitution can return the virulence-attenuated commensal strain Escherichia coli 83972 to virulence, defined by a disease phenotype in human hosts. E. coli 83972pap stably reprogrammed host gene expression, by activating an acute pyelonephritis-associated, IRF7-dependent gene network. The PapG protein was internalized by human kidney cells and served as a transcriptional agonist of IRF-7, IFN-β and MYC, suggesting direct involvement of the fimbrial adhesin in this process. IRF-7 was further identified as a potent upstream regulator (-log (p-value) = 61), consistent with the effects in inoculated patients. In contrast, E. coli 83972fim transiently attenuated overall gene expression in human hosts, enhancing the effects of E. coli 83972. The inhibition of RNA processing and ribosomal assembly indicated a homeostatic rather than a pathogenic end-point. In parallel, the expression of specific ion channels and neuropeptide gene networks was transiently enhanced, in a FimH-dependent manner. The studies were performed to establish protective asymptomatic bacteriuria in human hosts and the reconstituted E. coli 83972 variants were developed to improve bacterial fitness for the human urinary tract. Unexpectedly, P fimbriae were able to drive a disease response, suggesting that like oncogene addiction in cancer, pathogens may be addicted to single super-virulence factors. Urinary tract infections affect millions of individuals annually, and many patients suffer from recurring infections several times a year. Antibiotic resistance is increasing rapidly and new strategies are needed to treat even these common bacterial infections. One approach is to use the protective power of asymptomatic bacterial carriage, which has been shown to protect the host against symptomatic urinary tract infection. Instilling “nice” bacteria in the urinary bladder is therefore a promising alternative approach to antibiotic therapy. In an effort to increase the therapeutic use of asymptomatic bacteriuria, we reintroduced bacterial adhesion molecules into the therapeutic Escherichia coli strain 83972 and inoculated patients who are in need of alternative therapy. To our great surprise, the P fimbriated variant caused symptoms, despite lacking other virulence factors commonly thought to be necessary to cause disease. In contrast, type 1 fimbriae, did not provoke symptoms but enhanced the beneficial properties of the wild-type strain. This is explained by a divergent effect of these fimbrial types on host gene expression, where P fimbriae activate the IRF-7 transcription factor that regulates pathology in infected kidneys, suggesting that a single, potent virulence gene may be sufficient to create virulence in human hosts.
Collapse
Affiliation(s)
- Ines Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Klinikgatan, Lund, Sweden
| | - Daniel S. C. Butler
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Klinikgatan, Lund, Sweden
| | - Christoph Stork
- Institute of Hygiene, University of Münster, Mendelstr, Münster, Germany
| | - Jenny Grönberg-Hernández
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Klinikgatan, Lund, Sweden
| | - Bela Köves
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Klinikgatan, Lund, Sweden
| | - Jaroslaw Zdziarski
- Institute for Molecular Biology of Infectious Diseases, University of Würzburg, Würzburg, Germany
| | - Jerome Pinkner
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Center for Women's Infectious Disease Research (CWIDR), Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Scott J. Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Center for Women's Infectious Disease Research (CWIDR), Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Mendelstr, Münster, Germany
- Institute for Molecular Biology of Infectious Diseases, University of Würzburg, Würzburg, Germany
| | - Björn Wullt
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Klinikgatan, Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Klinikgatan, Lund, Sweden
- * E-mail:
| |
Collapse
|
16
|
Saxena V, Fitch J, Ketz J, White P, Wetzel A, Chanley MA, Spencer JD, Becknell B, Pierce KR, Arregui SW, Nelson RD, Schwartz GJ, Velazquez V, Walker LA, Chen X, Yan P, Hains DS, Schwaderer AL. Whole Transcriptome Analysis of Renal Intercalated Cells Predicts Lipopolysaccharide Mediated Inhibition of Retinoid X Receptor alpha Function. Sci Rep 2019; 9:545. [PMID: 30679625 PMCID: PMC6345901 DOI: 10.1038/s41598-018-36921-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 11/23/2018] [Indexed: 01/14/2023] Open
Abstract
The renal collecting duct consists of intercalated cells (ICs) and principal cells (PCs). We have previously demonstrated that collecting ducts have a role in the innate immune defense of the kidney. Transcriptomics is an important tool used to enhance systems-level understanding of cell biology. However, transcriptomics performed on whole kidneys provides limited insight of collecting duct cell gene expression, because these cells comprise a small fraction of total kidney cells. Recently we generated reporter mouse models to enrich collecting duct specific PC and ICs and reported targeted gene expression of anti-microbial peptide genes. Here we report transcriptomics on enriched ICs and PCs and performed a pilot study sequencing four single ICs. We identified 3,645 genes with increased relative expression in ICs compared to non-ICs. In comparison to non-PCs, 2,088 genes had higher relative expression in PCs. IC associated genes included the innate interleukin 1 receptor, type 1 and the antimicrobial peptide(AMP) adrenomedullin. The top predicted canonical pathway for enriched ICs was lipopolysaccharide/Interleukin 1 mediated inhibition of Retinoid X Receptor alpha function and decreased Retinoid X Receptor expression was confirmed to occur 1-hour post experimental murine UTI in ICs but not in non-ICs.
Collapse
Affiliation(s)
- Vijay Saxena
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| | - James Fitch
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - John Ketz
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
| | - Peter White
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Amy Wetzel
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Melinda A Chanley
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
| | - John D Spencer
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Brian Becknell
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Keith R Pierce
- Innate Immunity Translational Research Center, Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, Tennessee, United States
| | - Sam W Arregui
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States
| | - Raoul D Nelson
- Division of Nephrology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - George J Schwartz
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, United States
| | - Victoria Velazquez
- Research Institute at Nationwide Children's Hospital Flow Cytometry Core Laboratory, Columbus, Ohio, United States
| | - Logan A Walker
- Department of Physics, College of Arts and Sciences, The Ohio State University, Columbus, Ohio, United States
| | - Xi Chen
- Genomics Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Pearlly Yan
- Genomics Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - David S Hains
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| | - Andrew L Schwaderer
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| |
Collapse
|
17
|
Yu L, Zhang L, Yang H, Gui G, Liu Y, Xiao Y. Identification and characterization of the myeloid differentiation factor 88 gene in yellow catfish. 3 Biotech 2018; 8:430. [PMID: 30305999 PMCID: PMC6163110 DOI: 10.1007/s13205-018-1448-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/24/2018] [Indexed: 10/28/2022] Open
Abstract
Myeloid differentiation factor 88 (MyD88) is an important adapter protein of the innate immune system, but it has never before been reported in yellow catfish (Pelteobagrus fulvidraco). In this study, we cloned and characterized the yellow catfish MyD88 gene. The gene was 1230 bp in length and contained an 876-bp open reading frame which encodes a polypeptide of 291 amino acid residues. The theoretical molecular mass and isoelectric point of this polypeptide were 33.4341 kDa and 5.17, respectively. Furthermore, bioinformatic and phylogenetic analyses grouped yellow catfish MyD88 with MyD88 of other fish. We found that the deduced amino acid sequence showed that the conserved N-terminal death domain and the C-terminal typical Toll/interleukin-1 receptor domain were very similar to those of other fish. Moreover, reverse transcription PCR showed that yellow catfish MyD88 is ubiquitously expressed in all tissues examined, with highest expression levels observed in the spleen and lowest levels in the intestine. Importantly, MyD88 was shown to be significantly up-regulated in the intestines after 30-day dietary supplement of Clostridium butyricum. Collectively, these results indicate that yellow catfish MyD88 has a conserved structure and is probably an important component of innate immunity in yellow catfish. This study is the first to identify and characterize MyD88 in yellow catfish, thereby providing a reference for further research into the yellow catfish innate immune system.
Collapse
Affiliation(s)
- Lintian Yu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130 China
- Guangxi Agricultural Vocational College, Nanning, 530007 Guangxi China
| | - Long Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130 China
- Institute of Ecology, China West Normal University, Nanchong, 637009 China
| | - Hua Yang
- Institute of Quality Standards for Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021 China
| | - Guohong Gui
- Institute of Quality Standards for Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021 China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130 China
| | - Yingping Xiao
- Institute of Quality Standards for Agro-Products, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021 China
| |
Collapse
|
18
|
Anigilaje EA. A Putative Role of Apolipoprotein L1 Polymorphism in Renal Parenchymal Scarring Following Febrile Urinary Tract Infection in Nigerian Under-Five Children: Proposal for a Case-Control Association Study. JMIR Res Protoc 2018; 7:e156. [PMID: 29903699 PMCID: PMC6024104 DOI: 10.2196/resprot.9514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/23/2018] [Accepted: 04/10/2018] [Indexed: 01/13/2023] Open
Abstract
Background Although urinary tract infection (UTI) resolves with prompt treatment in a majority of children, some children, especially those aged less than 5 years, also develop renal parenchymal scarring (RPS). RPS causes high blood pressure that may lead to severe chronic kidney disease and end-stage renal disease (ESRD). Although the risk of UTI is higher in white children than in black children, it is unknown whether RPS is more common in white children than in black children as data are scarce in this regard. A common genetic predisposition to kidney disease in African Americans and the sub-Saharan African blacks is the possession of apolipoprotein L1 (APOL1). APOL1 risk variants regulate the production of APOL1. APOL1 circulates in the blood, and it is also found in the kidney tissue. While circulating, APOL1 kills the trypanosome parasites; an increased APOL1 in kidney tissues, under the right environmental conditions, can also result in the death of kidney tissue (vascular endothelium, the podocytes, proximal tubules, and arterial cells), which, ultimately, is replaced by fibrous tissue. APOL1 may influence the development of RPS, as evidence affirms that its expression is increased in kidney tissue following UTI caused by bacteria. Thus, UTI may be a putative environmental risk factor responsible for APOL1-induced kidney injury. Objective The aim of this proposal was to outline a study that seeks to determine if the possession of two copies of either G1 or G2 APOL1 variant increases the risk of having RPS, 6 months following a febrile UTI among Nigerian under-five children. Methods This case-control association study seeks to determine whether the risk of RPS from febrile UTI is conditional on having 2 APOL1 risk alleles (either G1 or G2). Cases will be children with a confirmed RPS following a febrile UTI. Controls will be age-, gender-, and ethnic-matched children with a febrile UTI but without RPS. Children with vesicoureteral reflux and other congenital anomalies of the urinary tract are to be excluded. Association between predictor variables (ethnicity, APOL1 G1 or G2, and others) and RPS will be tested at bivariate logistic regression analyses. Predictors that attained significance at a P value of ˂.05 will be considered for multiple logistic regressions. Likelihood-based tests will be used for hypothesis testing. Estimation will be done for the effect size for each of the APOL1 haplotypes using a generalized linear model. Results The study is expected to last for 3 years. Conclusions The study is contingent on having a platform for undergoing a research-based PhD program in any willing university in Europe or elsewhere. The findings of this study will be used to improve the care of African children who may develop RPS following febrile UTI. Registered Report Identifier RR1-10.2196/9514
Collapse
Affiliation(s)
- Emmanuel Ademola Anigilaje
- Nephrology Unit, Department of Paediatrics, College of Health Sciences, University of Abuja, Abuja, Nigeria
| |
Collapse
|
19
|
Collectins in urinary tract and kidney diseases. Int Urol Nephrol 2017; 50:695-703. [PMID: 29071557 DOI: 10.1007/s11255-017-1728-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/16/2017] [Indexed: 10/18/2022]
Abstract
The innate immune system serves as the frontline defense against invading pathogens and initiates an inflammatory response to microorganisms. Collectins are C-type lectins that are structurally characterized by a collagen-like sequence and a carbohydrate recognition domain. Moreover, they are widely expressed throughout the body and are involved in the innate immunity against a variety of pathogens, regulating inflammation, and protecting the lungs from pathogens. Recently, two classical collectins, surfactant protein A (SP-A) and surfactant protein D (SP-D), as well as novel collectin 11, were found present in urinary tract tissues. They are increasingly recognized as key players in activating the humoral arm of innate immunity and host defense in urinary tract and kidney diseases, although their biological features, functions, and mechanisms in this regard remain largely unclear. In this review, we aim to integrate results reported by ourselves and others to summarize and gain a better understanding of the functions of collectins (SP-A, SP-D, and collectin 11) in urinary tract and kidney diseases.
Collapse
|
20
|
Susceptibility to Urinary Tract Infection: Benefits and Hazards of the Antibacterial Host Response. Microbiol Spectr 2017; 4. [PMID: 27337480 DOI: 10.1128/microbiolspec.uti-0019-2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A paradigm shift is needed to improve and personalize the diagnosis of infectious disease and to select appropriate therapies. For many years, only the most severe and complicated bacterial infections received more detailed diagnostic and therapeutic attention as the efficiency of antibiotic therapy has guaranteed efficient treatment of patients suffering from the most common infections. Indeed, treatability almost became a rationale not to analyze bacterial and host parameters in these larger patient groups. Due to the rapid spread of antibiotic resistance, common infections like respiratory tract- or urinary-tract infections (UTIs) now pose new and significant therapeutic challenges. It is fortunate and timely that infectious disease research can offer such a wealth of new molecular information that is ready to use for the identification of susceptible patients and design of new suitable therapies. Paradoxically, the threat of antibiotic resistance may become a window of opportunity, by encouraging the implementation of new diagnostic and therapeutic approaches. The frequency of antibiotic resistance is rising rapidly in uropathogenic organisms and the molecular and genetic understanding of UTI susceptibility is quite advanced. More bold translation of the new molecular diagnostic and therapeutic tools would not just be possible but of great potential benefit in this patient group. This chapter reviews the molecular basis for susceptibility to UTI, including recent advances in genetics, and discusses the consequences for diagnosis and therapy. By dissecting the increasingly well-defined molecular interactions between bacteria and host and the molecular features of excessive bacterial virulence or host-response malfunction, it is becoming possible to isolate the defensive from the damaging aspects of the host response. Distinguishing "good" from "bad" inflammation has been a long-term quest of biomedical science and in UTI, patients need the "good" aspects of the inflammatory response to resist infection while avoiding the "bad" aspects, causing chronicity and tissue damage.
Collapse
|
21
|
Piao Z, Yuan H, Wang C, Shi L. IRF3 Inhibits Neutrophil Recruitment in Mice Infected with Pseudomonas aeruginosa. Inflammation 2017; 40:735-744. [PMID: 28181039 DOI: 10.1007/s10753-017-0517-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Pseudomonas aeruginosa is the major cause of morbidity and mortality in patients with ventilator-associated pneumonia. Interferon regulatory factor 3 (IRF3) is a transcription factor that plays an important role in the immune response to viral infection via the IRF3/IFN-β signaling pathway. Controversial data exist regarding the role of IRF3 in immune cell recruitment during bacterial infections. IRF3 has been shown to promote neutrophil recruitment and bacterial clearance in mice infected with P. aeruginosa by inducing the production of specific chemokines and cytokines. In contrast, our study showed that IRF3 knockout (KO) mice infected with P. aeruginosa exhibited greater survival rates, demonstrated enhanced bacterial clearance, and showed significantly increased neutrophil recruitment to the lungs, when compared with the wild-type (WT) mice. The peritoneal lavage fluid collected from IRF3 KO mice 4 h after intraperitoneal injection with P. aeruginosa or 3% thioglycolate contained a significantly increased number of neutrophils. Furthermore, neutrophils from the bone marrow (BM) of IRF3 KO mice showed greater adhesiveness to the extracellular matrix when compared with those of WT mice, post-P. aeruginosa infection. In addition, IRF3 induced the expression of target genes in WT neutrophils infected with P. aeruginosa. These findings indicate that IRF3 exacerbates P. aeruginosa-induced mortality in mice by inhibiting neutrophil adhesion and recruitment to the lungs. Together, these data indicate that the inhibition of IRF3 might provide a possible mechanism for controlling P. aeruginosa infections.
Collapse
Affiliation(s)
- Zhenghao Piao
- Department of Basic Medical Science, School of Medicine, Hangzhou Normal University, Xuelin Street 16#, Hangzhou, 310036, China.
| | - Haiying Yuan
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cuili Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liyun Shi
- Department of Immunology, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, 210023, China.
| |
Collapse
|
22
|
Forster CS, Johnson K, Patel V, Wax R, Rodig N, Barasch J, Bachur R, Lee RS. Urinary NGAL deficiency in recurrent urinary tract infections. Pediatr Nephrol 2017; 32:1077-1080. [PMID: 28210838 PMCID: PMC5417074 DOI: 10.1007/s00467-017-3607-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/04/2016] [Accepted: 01/19/2017] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Children with recurrent urinary tract infections (rUTI) often show no identifiable cause of their infections. Neutrophil gelatinase-associated lipocalin (NGAL) is known to be upregulated within the uroepithelium and kidney of patients with UTI and exhibits a localized bacteriostatic effect through iron chelation. We hypothesize that some patients with rUTI without an identifiable cause of their recurrent infections have locally deficient NGAL production. We therefore explored whether a lack of NGAL production may be a factor in the pathogenesis of rUTI. MATERIALS AND METHODS Patients seen in the urology clinic for rUTI who were <21 years of age were enrolled. Patients were excluded if they had UTI at the time of enrollment, evidence of renal disease, decreased renal function, known anatomic abnormality of the genitourinary tract, or other reasons that predispose to UTI, such as neurogenic bladder, the need for intermittent catheterization, or unrepaired posterior urethral valves. Control patients were healthy children enrolled from the emergency department with no history of UTI or renal dysfunction, normal urinalysis at the time of enrollment, and presenting no diagnosis associated with increased NGAL levels, such as acute kidney injury or infection. NGAL was measured by immunoblot. RESULTS Fifteen cases and controls were enrolled. Median urinary NGAL levels were significantly decreased in rUTI patients compared with controls [15 (14-29) ng/ml vs 30 (27-61) ng/ml; p = 0.002)] Although comparatively diminished, measurable NGAL levels were present in all patients with rUTI. CONCLUSIONS Urinary NGAL is significantly decreased in patients with compared with patients without rUTI. These data suggest that some patients with rUTI may be predisposed to UTI because of a relative local deficiency in urinary NGAL production.
Collapse
Affiliation(s)
- Catherine S. Forster
- Harvard Medical School and Boston Children’s Hospital, Department of Medicine, Boston MA
| | - Kathryn Johnson
- Harvard Medical School and Boston Children’s Hospital, Department of Urology, Boston MA
| | - Viral Patel
- Harvard Medical School and Boston Children’s Hospital, Department of Urology, Boston MA
| | - Rebecca Wax
- Columbia University, Department of Medicine, New York, NY
| | - Nancy Rodig
- Harvard Medical School and Boston Children’s Hospital, Department of Medicine, Boston MA
| | | | - Richard Bachur
- Harvard Medical School and Boston Children’s Hospital, Department of Medicine, Boston MA
| | - Richard S. Lee
- Harvard Medical School and Boston Children’s Hospital, Department of Urology, Boston MA
| |
Collapse
|
23
|
Li B, Haridas B, Jackson AR, Cortado H, Mayne N, Kohnken R, Bolon B, McHugh KM, Schwaderer AL, Spencer JD, Ching CB, Hains DS, Justice SS, Partida-Sanchez S, Becknell B. Inflammation drives renal scarring in experimental pyelonephritis. Am J Physiol Renal Physiol 2017; 312:F43-F53. [PMID: 27760770 PMCID: PMC5283888 DOI: 10.1152/ajprenal.00471.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/13/2016] [Indexed: 12/30/2022] Open
Abstract
Acquired renal scarring occurs in a subset of patients following febrile urinary tract infections and is associated with hypertension, proteinuria, and chronic kidney disease. Limited knowledge of histopathology, immune cell recruitment, and gene expression changes during pyelonephritis restricts the development of therapies to limit renal scarring. Here, we address this knowledge gap using immunocompetent mice with vesicoureteral reflux. Transurethral inoculation of uropathogenic Escherichia coli in C3H/HeOuJ mice leads to renal mucosal injury, tubulointerstitial nephritis, and cortical fibrosis. The extent of fibrosis correlates most significantly with inflammation at 7 and 28 days postinfection. The recruitment of neutrophils and inflammatory macrophages to infected kidneys is proportional to renal bacterial burden. Transcriptome analysis reveals molecular signatures associated with renal ischemia-reperfusion injury, immune cell chemotaxis, and leukocyte activation. This murine model recapitulates the cardinal histopathological features observed in humans with acquired renal scarring following pyelonephritis. The integration of histopathology, quantification of cellular immune influx, and unbiased transcriptional profiling begins to define potential mechanisms of tissue injury during pyelonephritis in the context of an intact immune response. The clear relationship between inflammatory cell recruitment and fibrosis supports the hypothesis that acquired renal scarring arises as a consequence of excessive host inflammation and suggests that immunomodulatory therapies should be investigated to reduce renal scarring in patients with pyelonephritis.
Collapse
Affiliation(s)
- Birong Li
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Babitha Haridas
- Department of Neurology, State University of New York at Buffalo, Buffalo, New York
| | - Ashley R Jackson
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Hanna Cortado
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Nicholas Mayne
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Rebecca Kohnken
- College of Veterinary Medicine and Comparative Pathology and Mouse Phenotyping Shared Resource, The Ohio State University, Columbus, Ohio
| | | | - Kirk M McHugh
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Anatomy, The Ohio State University College of Allied Health Sciences, Columbus, Ohio
| | - Andrew L Schwaderer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Division of Nephrology, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - John David Spencer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Division of Nephrology, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Christina B Ching
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Division of Urology, Department of Surgery, The Ohio State University, Columbus, Ohio
| | - David S Hains
- Children's Research Foundation Institute, Le Bonheur Children's Hospital, Memphis, Tennessee; and
| | - Sheryl S Justice
- Division of Urology, Department of Surgery, The Ohio State University, Columbus, Ohio
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Brian Becknell
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio;
- Division of Nephrology, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
24
|
Ambite I, Rydstrom G, Schwaderer AL, Hains DS. The Genetics of Urinary Tract Infections and the Innate Defense of the Kidney and Urinary tract. J Pediatr Genet 2016; 5:25-32. [PMID: 27617139 DOI: 10.1055/s-0035-1557110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 04/02/2015] [Indexed: 12/31/2022]
Abstract
The urinary tract is a sterile organ system. Urinary tract infections (UTIs) are common and often serious infections. Research has focused on uropathogen, environment, and host factors leading to UTI pathogenesis. A growing body of evidence exists implicating genetic factors that can contribute to UTI risks. In this review, we highlight genetic variations in aspects of the innate immune system critical to the host response to uropathogens. This overview includes genetic variations in pattern recognition receptor molecules, chemokines/cytokines, and neutrophil activation. We also comprehensively cover murine knockout models of UTI, genetic variations involved in renal scarring as a result of ascending UTIs, and asymptomatic bacteriuria.
Collapse
Affiliation(s)
- Ines Ambite
- Section of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Gustav Rydstrom
- Section of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Andrew L Schwaderer
- Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, United States
| | - David S Hains
- Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, Tennessee, United States; Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
25
|
Richter E, Mostertz J, Hochgräfe F. Proteomic discovery of host kinase signaling in bacterial infections. Proteomics Clin Appl 2016; 10:994-1010. [PMID: 27440122 PMCID: PMC5096009 DOI: 10.1002/prca.201600035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/08/2016] [Accepted: 07/18/2016] [Indexed: 12/15/2022]
Abstract
Protein phosphorylation catalyzed by protein kinases acts as a reversible molecular switch in signal transduction, providing a mechanism for the control of protein function in cellular processes. During microbial infection, cellular signaling essentially contributes to immune control to restrict the dissemination of invading pathogens within the host organism. However, pathogenic microbes compete for the control of host signaling to create a beneficial environment for successful invasion and infection. Although efforts to achieve a better understanding of the host–pathogen interaction and its molecular consequences have been made, there is urgent need for a comprehensive characterization of infection‐related host signaling processes. System‐wide and hypothesis‐free analysis of phosphorylation‐mediated host signaling during host–microbe interactions by mass spectrometry (MS)‐based methods is not only promising in view of a greater understanding of the pathogenesis of the infection but also may result in the identification of novel host targets for preventive or therapeutic intervention. Here, we review state‐of‐the‐art MS‐based techniques for the system‐wide identification and quantitation of protein phosphorylation and compare them to array‐based phosphoprotein analyses. We also provide an overview of how phosphoproteomics and kinomics have contributed to our understanding of protein kinase‐driven phosphorylation networks that operate during host–microbe interactions.
Collapse
Affiliation(s)
- Erik Richter
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, Greifswald, Germany
| | - Jörg Mostertz
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, Greifswald, Germany
| | - Falko Hochgräfe
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, Greifswald, Germany.
| |
Collapse
|
26
|
Wullt B, Svanborg C. Deliberate Establishment of Asymptomatic Bacteriuria-A Novel Strategy to Prevent Recurrent UTI. Pathogens 2016; 5:E52. [PMID: 27483325 PMCID: PMC5039432 DOI: 10.3390/pathogens5030052] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 01/06/2023] Open
Abstract
We have established a novel strategy to reduce the risk for recurrent urinary tract infection (UTI), where rapidly increasing antibiotic resistance poses a major threat. Epidemiologic studies have demonstrated that asymptomatic bacteriuria (ABU) protects the host against symptomatic infections with more virulent strains. To mimic this protective effect, we deliberately establish ABU in UTI-prone patients, who are refractory to conventional therapy. The patients are inoculated with Escherichia coli (E. coli) 83972, now widely used as a prototype ABU strain. Therapeutic efficacy has been demonstrated in a placebo-controlled trial, supporting the feasibility of using E. coli 83972 as a tool to prevent recurrent UTI and, potentially, to outcompete antibiotic-resistant strains from the human urinary tract. In addition, the human inoculation protocol offers unique opportunities to study host-parasite interaction in vivo in the human urinary tract. Here, we review the clinical evidence for protection using this approach as well as some molecular insights into the pathogenesis of UTI that have been gained during these studies.
Collapse
Affiliation(s)
- Björn Wullt
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, 221 00 Lund, Sweden.
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, 221 00 Lund, Sweden.
| |
Collapse
|
27
|
Bacterial Suppression of RNA Polymerase II-Dependent Host Gene Expression. Pathogens 2016; 5:pathogens5030049. [PMID: 27420101 PMCID: PMC5039429 DOI: 10.3390/pathogens5030049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 05/17/2016] [Accepted: 07/04/2016] [Indexed: 12/21/2022] Open
Abstract
Asymptomatic bacteriuria (ABU) is a bacterial carrier state in the urinary tract that resembles commensalism at other mucosal sites. ABU strains often lack the virulence factors that characterize uropathogenic Escherichia coli (E. coli) strains and therefore elicit weak innate immune responses in the urinary tract. In addition, ABU strains are active modifiers of the host environment, which they influence by suppressing RNA polymerase II (Pol II)-dependent host gene expression. In patients inoculated with the ABU strain E. coli 83972, gene expression was markedly reduced after 24 h (>60% of all regulated genes). Specific repressors and activators of Pol II-dependent transcription were modified, and Pol II Serine 2 phosphorylation was significantly inhibited, indicating reduced activity of the polymerase. This active inhibition included disease–associated innate immune response pathways, defined by TLR4, IRF-3 and IRF-7, suggesting that ABU strains persist in human hosts by active suppression of the antibacterial defense. In a search for the mechanism of inhibition, we compared the whole genome sequences of E. coli 83972 and the uropathogenic strain E. coli CFT073. In addition to the known loss of virulence genes, we observed that the ABU strain has acquired several phages and identified the lytic Prophage 3 as a candidate Pol II inhibitor. Intact phage particles were released by ABU during in vitro growth in human urine. To address if Prophage 3 affects Pol II activity, we constructed a Prophage 3 negative deletion mutant in E. coli 83972 and compared the effect on Pol II phosphorylation between the mutant and the E. coli 83972 wild type (WT) strains. No difference was detected, suggesting that the Pol II inhibitor is not encoded by the phage. The review summarizes the evidence that the ABU strain E. coli 83972 modifies host gene expression by inhibition of Pol II phosphorylation, and discusses the ability of ABU strains to actively create an environment that enhances their persistence.
Collapse
|
28
|
|
29
|
Puthia M, Ambite I, Cafaro C, Butler D, Huang Y, Lutay N, Rydström G, Gullstrand B, Swaminathan B, Nadeem A, Nilsson B, Svanborg C. IRF7inhibition prevents destructive innate immunity—A target for nonantibiotic therapy of bacterial infections. Sci Transl Med 2016; 8:336ra59. [DOI: 10.1126/scitranslmed.aaf1156] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/08/2016] [Indexed: 12/16/2022]
|
30
|
Urinary Tract Infection Molecular Mechanisms and Clinical Translation. Pathogens 2016; 5:pathogens5010024. [PMID: 26927188 PMCID: PMC4810145 DOI: 10.3390/pathogens5010024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 11/17/2022] Open
Abstract
Rapid developments in infection biology create new and exciting options for individualized diagnostics and therapy. Such new practices are needed to improve patient survival and reduce morbidity. Molecular determinants of host resistance to infection are being characterized, making it possible to identify susceptible individuals and to predict their risk for future morbidity. Immunotherapy is emerging as a new strategy to treat infections worldwide and controlled boosting of the host immune defense represents an important therapeutic alternative to antibiotics. In proof of concept studies, we have demonstrated that this approach is feasible. The long-term goal is not just to remove the pathogens but to also develop technologies that restore resistance to infection in disease-prone patients and devise personalized therapeutic interventions. Here, we discuss some approaches to reaching these goals, in patients with urinary tract infection (UTI). We describe critical host signaling pathways that define symptoms and pathology and the genetic control of innate immune responses that balance protection against tissue damage. For some of these genes, human relevance has been documented in clinical studies, identifying them as potential targets for immune-modulatory therapies, as a complement to antibiotics.
Collapse
|
31
|
Asymtomatic Bacteriuria as a Model to Study the Coevolution of Hosts and Bacteria. Pathogens 2016; 5:pathogens5010021. [PMID: 26891332 PMCID: PMC4810142 DOI: 10.3390/pathogens5010021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/28/2016] [Accepted: 02/03/2016] [Indexed: 01/21/2023] Open
Abstract
During asymptomatic bacteriuria (ABU), bacteria colonize the urinary tract for extended periods of time without causing symptoms of urinary tract infection. Previous studies indicate that many Escherichia coli (E. coli) strains that cause ABU have evolved from uropathogenic E. coli (UPEC) by reductive evolution and loss of the ability to express functional virulence factors. For instance, the prototype ABU strain 83972 has a smaller genome than UPEC strains with deletions or point mutations in several virulence genes. To understand the mechanisms of bacterial adaptation and to find out whether the bacteria adapt in a host-specific manner, we compared the complete genome sequences of consecutive reisolates of ABU strain 83972 from different inoculated individuals and compared them with the genome of the parent strain. Reisolates from different hosts exhibited individual patterns of genomic alterations. Non-synonymous SNPs predominantly occurred in coding regions and often affected the amino acid sequence of proteins with global or pleiotropic regulatory function. These gene products are involved in different bacterial stress protection strategies, and metabolic and signaling pathways. Our data indicate that adaptation of E. coli 83972 to prolonged growth in the urinary tract involves responses to specific growth conditions and stresses present in the individual hosts. Accordingly, modulation of gene expression required for survival and growth under stress conditions seems to be most critical for long-term growth of E. coli 83972 in the urinary tract.
Collapse
|
32
|
Ipe DS, Horton E, Ulett GC. The Basics of Bacteriuria: Strategies of Microbes for Persistence in Urine. Front Cell Infect Microbiol 2016; 6:14. [PMID: 26904513 PMCID: PMC4744864 DOI: 10.3389/fcimb.2016.00014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/22/2016] [Indexed: 01/09/2023] Open
Abstract
Bacteriuria, the presence of bacteria in urine, is associated with asymptomatic, as well as symptomatic, urinary tract infection (UTI). Bacteriuria underpins some of the dynamics of microbial colonization of the urinary tract, and probably impacts the progression and persistence of infection in some individuals. Recent molecular discoveries in vitro have elucidated how some key bacterial traits can enable organisms to survive and grow in human urine as a means of microbial fitness adaptation for UTI. Several microbial characteristics that confer bacteruric potential have been identified including de novo synthesis of guanine, relative resistance to D-serine, and catabolism of malic acid. Microbial characteristics such as these are increasingly being defined through the use of synthetic human urine (SHU) in vitro as a model to mimic the in vivo environment that bacteria encounter in the bladder. There is considerable variation in the SHU model systems that have been used to study bacteriuria to date, and this influences the utility of these models. In this review, we discuss recent advances in our understanding of bacteruric potential with a focus on the specific mechanisms underlying traits that promote the growth of bacteria in urine. We also review the application of SHU in research studies modeling UTI and discuss the chemical makeup, and benefits and limitations that are encountered in utilizing SHU to study bacterial growth in urine in vitro.
Collapse
Affiliation(s)
| | | | - Glen C. Ulett
- School of Medical Science, Menzies Health Institute Queensland, Griffith UniversityGold Coast, QLD, Australia
| |
Collapse
|
33
|
Association of O-Antigen Serotype with the Magnitude of Initial Systemic Cytokine Responses and Persistence in the Urinary Tract. J Bacteriol 2016; 198:964-72. [PMID: 26755631 DOI: 10.1128/jb.00664-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/05/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Urinary tract infection (UTI) is one of the most common ailments requiring both short-term and prophylactic antibiotic therapies. Progression of infection from the bladder to the kidney is associated with more severe clinical symptoms (e.g., fever and vomiting) as well as with dangerous disease sequelae (e.g., renal scaring and sepsis). Host-pathogen interactions that promote bacterial ascent to the kidney are not completely understood. Prior studies indicate that the magnitude of proinflammatory cytokine elicitation in vitro by clinical isolates of uropathogenic Escherichia coli (UPEC) inversely correlates with the severity of clinical disease. Therefore, we hypothesize that the magnitude of initial proinflammatory responses during infection defines the course and severity of disease. Clinical UPEC isolates obtained from patients with a nonfebrile UTI elicited high systemic proinflammatory responses early during experimental UTI in a murine model and were attenuated in bladder and kidney persistence. Conversely, UPEC isolates obtained from patients with febrile UTI elicited low systemic proinflammatory responses early during experimental UTI and exhibited prolonged persistence in the bladder and kidney. Soluble factors in the supernatant from saturated cultures as well as the lipopolysaccharide (LPS) serotype correlated with the magnitude of proinflammatory responses in vitro. Our data suggest that the structure of the O-antigen sugar moiety of the LPS may determine the strength of cytokine induction by epithelial cells. Moreover, the course and severity of disease appear to be the consequence of the magnitude of initial cytokines produced by the bladder epithelium during infection. IMPORTANCE The specific host-pathogen interactions that determine the extent and course of disease are not completely understood. Our studies demonstrate that modest changes in the magnitude of cytokine production observed using in vitro models of infection translate into significant ramifications for bacterial persistence and disease severity. While many studies have demonstrated that modifications of the LPS lipid A moiety modulate the extent of Toll-like receptor 4 (TLR4) activation, our studies implicate the O-antigen sugar moiety as another potential rheostat for the modulation of proinflammatory cytokine production.
Collapse
|
34
|
Hu F, Ding G, Zhang Z, Gatto LA, Hawgood S, Poulain FR, Cooney RN, Wang G. Innate immunity of surfactant proteins A and D in urinary tract infection with uropathogenic Escherichia coli. Innate Immun 2015; 22:9-20. [PMID: 26511057 DOI: 10.1177/1753425915609973] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/14/2015] [Indexed: 12/20/2022] Open
Abstract
To investigate the effects of surfactant proteins A and D (SP-A and SP-D, respectively) in urinary tract infection (UTI), SP-A and SP-D double knockout (SP-A/D KO) and wild type (WT) C57BL/6 female mice were infected with uropathogenic Escherichia coli by intravesical inoculation. Compared with WT mice SP-A/D KO mice showed increased susceptibility to UTI, as evidenced by higher bacterial CFU, more infiltrating neutrophils and severe pathological changes. Keratinocyte-derived chemokine increased in the kidney of WT mice but not in SP-A/D KO mice 24 h post-infection. Compared with control, the level of IL-17 was elevated in the kidney of infected WT and SP-A/D KO mice and the level of IL-17 was higher in the infected SP-A/D KO mice than in infected WT mice 24 and 48 h post-infection. The basal level of p38 MAPK phosphorylation in SP-A/D KO mice was higher than in WT mice. The phosphorylated p38 level was elevated in the kidney of WT mice post infection but not in SP-A/D KO mice. Furthermore, in vitro growth of uropathogenic E. coli was inhibited by SP-A and SP-D. We conclude that SP-A and SP-D function as mediators of innate immunity by inhibiting bacterial growth and modulating renal inflammation in part by regulating p38 MAPK-related pathway in murine UTI.
Collapse
Affiliation(s)
- Fengqi Hu
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiyong Zhang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Louis A Gatto
- Department of Biological Sciences, SUNY Cortland, Cortland, NY, USA
| | - Samuel Hawgood
- Department of Pediatrics and the Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Francis R Poulain
- Division of Neonatology, Department of Pediatrics, Children's Hospital, University of California Davis Medical Center, Davis, CA, USA
| | - Robert N Cooney
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
35
|
Abstract
Purpose of review Urinary tract infections (UTIs) are common, dangerous and interesting. Susceptible individuals experience multiple, often clustered episodes, and in a subset of patients, infections progress to acute pyelonephritis (APN), sometimes accompanied by uro-sepsis. Others develop asymptomatic bacteriuria (ABU). Here, we review the molecular basis for these differences, with the intention to distinguish exaggerated host responses that drive disease from attenuated responses that favour protection and to highlight the genetic basis for these extremes, based on knock-out mice and clinical studies. Recent findings The susceptibility to UTI is controlled by specific innate immune signalling and by promoter polymorphisms and transcription factors that modulate the expression of genes controlling these pathways. Gene deletions that disturb innate immune activation either favour asymptomatic bacteriuria or create acute morbidity and disease. Promoter polymorphisms and transcription factor variants affecting those genes are associated with susceptibility in UTI-prone patients. Summary It is time to start using genetics in UTI-prone patients, to improve diagnosis and to assess the risk for chronic sequels such as renal malfunction, hypertension, spontaneous abortions, dialysis and transplantation. Furthermore, the majority of UTI patients do not need follow-up, but for lack of molecular markers, they are unnecessarily investigated.
Collapse
|
36
|
Abstract
Both urinary tract infection (UTI) and asymptomatic bacteriuria (ASB) are common problems among elderly adults and represent a significant health care burden. Despite their frequency, differentiating between ASB and true UTI remains controversial among health care providers. Several challenges exist in the evaluation of urinary symptoms in the elderly patient. Symptoms of UTI are variable; problems are encountered in the collection, testing, and interpretation of urine specimens; and results of urinalysis are often misinterpreted and mishandled. Multiple studies have shown no morbidity or mortality benefit to antibiotic therapy in either community or long-term care facility residents with ASB.
Collapse
Affiliation(s)
- Keri Detweiler
- Touro University College of Osteopathic Medicine - California, 1310 Club Drive, Vallejo, CA 94592, USA
| | - Daniel Mayers
- Touro University College of Osteopathic Medicine - California, 1310 Club Drive, Vallejo, CA 94592, USA
| | - Sophie G Fletcher
- Kaiser Permanente Northern California, 401 Bicentennial Way, Santa Rosa, CA 95403, USA.
| |
Collapse
|
37
|
Carey AJ, Tan CK, Ipe DS, Sullivan MJ, Cripps AW, Schembri MA, Ulett GC. Urinary tract infection of mice to model human disease: Practicalities, implications and limitations. Crit Rev Microbiol 2015; 42:780-99. [PMID: 26006172 DOI: 10.3109/1040841x.2015.1028885] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections in humans. Murine models of human UTI are vital experimental tools that have helped to elucidate UTI pathogenesis and advance knowledge of potential treatment and infection prevention strategies. Fundamentally, several variables are inherent in different murine models, and understanding the limitations of these variables provides an opportunity to understand how models may be best applied to research aimed at mimicking human disease. In this review, we discuss variables inherent in murine UTI model studies and how these affect model usage, data analysis and data interpretation. We examine recent studies that have elucidated UTI host-pathogen interactions from the perspective of gene expression, and review new studies of biofilm and UTI preventative approaches. We also consider potential standards for variables inherent in murine UTI models and discuss how these might expand the utility of models for mimicking human disease and uncovering new aspects of pathogenesis.
Collapse
Affiliation(s)
- Alison J Carey
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Chee K Tan
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Deepak S Ipe
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Matthew J Sullivan
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| | - Allan W Cripps
- b Menzies Health Institute Queensland, Griffith University , Gold Coast , Australia , and
| | - Mark A Schembri
- c School of Chemistry and Molecular Biosciences, University of Queensland , Brisbane , Australia
| | - Glen C Ulett
- a Menzies Health Institute Queensland & School of Medical Sciences, Griffith University , Gold Coast , Australia
| |
Collapse
|
38
|
Simultaneous assessment of NF-κB/p65 phosphorylation and nuclear localization using imaging flow cytometry. J Immunol Methods 2015; 423:3-11. [PMID: 25862606 DOI: 10.1016/j.jim.2015.03.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 03/20/2015] [Accepted: 03/30/2015] [Indexed: 01/12/2023]
Abstract
Aberrant activity of Nuclear Factor-kappaB (NF-κB) is associated with many diseases and is therapeutically targeted. Post-translational modifications, particularly phosphorylation of the RELA/p65 sub-unit, are essential for cytoplasmic to nuclear localization of NF-κB/p65 and initiation of transcription of downstream target genes. Immunoblot and phospho-flow cytometry have been used to study the relationship between phosphorylation motifs and NF-κB activation and microscopic analysis of nuclear localization of p65 is also used as a parameter for activation. The labor intensive nature of these approaches commonly limits the number of sampling points or replicates. Recent insights into the relationship between p65 phosphorylation motifs and their nuclear localization indicate that these parameters have different significances and should not be used interchangeably. In this study, we demonstrate feasibility and reproducibility of studying the relationship between p65 phosphorylation and nuclear translocation using imaging flow cytometry (IFC). TNFα- or PMA/Ionomycin-induced phosphorylation of p65 at serine 529 in cell line models and healthy donor lymphocytes served as the experimental model. IFC analysis demonstrated that expression of phosphorylated serine 529 (P-p65(s529)) increased rapidly following stimulation and that nuclear localization of P-p65(s529) followed the nuclear localization pattern of total p65. However, in the presence of tacrolimus, P-p65(s529) expression was inhibited without affecting nuclear localization of total p65. The data demonstrate the application of IFC to simultaneously assess phosphorylation of p65 and its cellular localization and the results obtained by this analysis corroborate current insights regarding the specific effect of tacrolimus on serine 529 phosphorylation.
Collapse
|
39
|
|
40
|
The Humoral Pattern Recognition Molecule PTX3 Is a Key Component of Innate Immunity against Urinary Tract Infection. Immunity 2014; 40:621-32. [DOI: 10.1016/j.immuni.2014.02.015] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 02/14/2014] [Indexed: 11/17/2022]
|
41
|
Ingersoll MA, Albert ML. From infection to immunotherapy: host immune responses to bacteria at the bladder mucosa. Mucosal Immunol 2013; 6:1041-53. [PMID: 24064671 DOI: 10.1038/mi.2013.72] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/20/2013] [Indexed: 02/04/2023]
Abstract
The pathogenesis of urinary tract infection and mechanisms of the protective effect of Bacillus Calmette-Guerin (BCG) therapy for bladder cancer highlight the importance of studying the bladder as a unique mucosal surface. Innate responses to bacteria are reviewed, and although our collective knowledge remains incomplete, we discuss how adaptive immunity may be generated following bacterial challenge in the bladder microenvironment. Interestingly, the widely held belief that the bladder is sterile has been challenged recently, indicating the need for further study of the impact of commensal microorganisms on the immune response to uropathogen infection or intentional instillation of BCG. This review addresses the aspects of bladder biology that have been well explored and defines what still must be discovered about the immunobiology of this understudied organ.
Collapse
Affiliation(s)
- M A Ingersoll
- 1] Unité d'Immunobiologie des Cellules Dendritiques, Department of Immunology, Institut Pasteur, Paris, France [2] INSERM U818, Department of Immunology, Institut Pasteur, Paris, France [3] Université Paris Descartes, Paris, France
| | | |
Collapse
|
42
|
Lutay N, Ambite I, Grönberg Hernandez J, Rydström G, Ragnarsdóttir B, Puthia M, Nadeem A, Zhang J, Storm P, Dobrindt U, Wullt B, Svanborg C. Bacterial control of host gene expression through RNA polymerase II. J Clin Invest 2013; 123:2366-79. [PMID: 23728172 DOI: 10.1172/jci66451] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 02/27/2013] [Indexed: 01/25/2023] Open
Abstract
The normal flora furnishes the host with ecological barriers that prevent pathogen attack while maintaining tissue homeostasis. Urinary tract infections (UTIs) constitute a highly relevant model of microbial adaptation in which some patients infected with Escherichia coli develop acute pyelonephritis, while other patients with bacteriuria exhibit an asymptomatic carrier state similar to bacterial commensalism. It remains unclear if the lack of destructive inflammation merely reflects low virulence or if carrier strains actively inhibit disease-associated responses in the host. Here, we identify a new mechanism of bacterial adaptation through broad suppression of RNA polymerase II–dependent (Pol II–dependent) host gene expression. Over 60% of all genes were suppressed 24 hours after human inoculation with the prototype asymptomatic bacteriuria (ABU) strain E. coli 83972, and inhibition was verified by infection of human cells. Specific repressors and activators of Pol II–dependent transcription were modified, Pol II phosphorylation was inhibited, and pathogen-specific signaling was suppressed in cell lines and inoculated patients. An increased frequency of strains inhibiting Pol II was epidemiologically verified in ABU and fecal strains compared with acute pyelonephritis, and a Pol II antagonist suppressed the disease-associated host response. These results suggest that by manipulating host gene expression, ABU strains promote tissue integrity while inhibiting pathology. Such bacterial modulation of host gene expression may be essential to sustain asymptomatic bacterial carriage by ensuring that potentially destructive immune activation will not occur.
Collapse
Affiliation(s)
- Nataliya Lutay
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lebeaux D, Chauhan A, Rendueles O, Beloin C. From in vitro to in vivo Models of Bacterial Biofilm-Related Infections. Pathogens 2013; 2:288-356. [PMID: 25437038 PMCID: PMC4235718 DOI: 10.3390/pathogens2020288] [Citation(s) in RCA: 326] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/01/2013] [Accepted: 05/08/2013] [Indexed: 12/13/2022] Open
Abstract
The influence of microorganisms growing as sessile communities in a large number of human infections has been extensively studied and recognized for 30–40 years, therefore warranting intense scientific and medical research. Nonetheless, mimicking the biofilm-life style of bacteria and biofilm-related infections has been an arduous task. Models used to study biofilms range from simple in vitro to complex in vivo models of tissues or device-related infections. These different models have progressively contributed to the current knowledge of biofilm physiology within the host context. While far from a complete understanding of the multiple elements controlling the dynamic interactions between the host and biofilms, we are nowadays witnessing the emergence of promising preventive or curative strategies to fight biofilm-related infections. This review undertakes a comprehensive analysis of the literature from a historic perspective commenting on the contribution of the different models and discussing future venues and new approaches that can be merged with more traditional techniques in order to model biofilm-infections and efficiently fight them.
Collapse
Affiliation(s)
- David Lebeaux
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Ashwini Chauhan
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Olaya Rendueles
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Christophe Beloin
- Institut Pasteur, Unité de Génétique des Biofilms, 25 rue du Dr. Roux, 75724 Paris cedex 15, France.
| |
Collapse
|
44
|
Ulett GC, Totsika M, Schaale K, Carey AJ, Sweet MJ, Schembri MA. Uropathogenic Escherichia coli virulence and innate immune responses during urinary tract infection. Curr Opin Microbiol 2013; 16:100-7. [DOI: 10.1016/j.mib.2013.01.005] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 12/19/2012] [Accepted: 01/08/2013] [Indexed: 12/17/2022]
|
45
|
Ragnarsdóttir B, Svanborg C. Susceptibility to acute pyelonephritis or asymptomatic bacteriuria: host-pathogen interaction in urinary tract infections. Pediatr Nephrol 2012; 27:2017-2029. [PMID: 22327887 DOI: 10.1007/s00467-011-2089-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 11/30/2011] [Accepted: 12/02/2011] [Indexed: 10/14/2022]
Abstract
Our knowledge of the molecular mechanisms of urinary tract infection (UTI) pathogenesis has advanced greatly in recent years. In this review, we provide a general background of UTI pathogenesis, followed by an update on the mechanisms of UTI susceptibility, with a particular focus on genetic variation affecting innate immunity. The innate immune response of the host is critically important in the antibacterial defence mechanisms of the urinary tract, and bacterial clearance normally proceeds without sequelae. However, slight dysfunctions in these mechanisms may result in acute disease and tissue destruction. The symptoms of acute pyelonephritis are caused by the innate immune response, and inflammation in the urinary tract decreases renal tubular function and may give rise to renal scarring, especially in paediatric patients. In contrast, in children with asymptomatic bacteriuria (ABU), bacteria persist without causing symptoms or pathology. Pathogenic agents trigger a response determined by their virulence factors, mediating adherence to the urinary tract mucosa, signalling through Toll-like receptors (TLRs) and activating the defence mechanisms. In ABU strains, such virulence factors are mostly not expressed. However, the influence of the host on UTI severity cannot be overestimated, and rapid progress is being made in clarifying host susceptibility mechanisms. For example, genetic alterations that reduce TLR4 function are associated with ABU, while polymorphisms reducing IRF3 or CXCR1 expression are associated with acute pyelonephritis and an increased risk for renal scarring. It should be plausible to "individualize" diagnosis and therapy by combining information on bacterial virulence and the host response.
Collapse
Affiliation(s)
- Bryndís Ragnarsdóttir
- Institute of Laboratory Medicine, Section of Microbiology, Immunology and Glycobiology (MIG), Lund University, Sölvegatan 23, 22362, Lund, Sweden
| | - Catharina Svanborg
- Institute of Laboratory Medicine, Section of Microbiology, Immunology and Glycobiology (MIG), Lund University, Sölvegatan 23, 22362, Lund, Sweden.
| |
Collapse
|
46
|
Hannan TJ, Totsika M, Mansfield KJ, Moore KH, Schembri MA, Hultgren SJ. Host-pathogen checkpoints and population bottlenecks in persistent and intracellular uropathogenic Escherichia coli bladder infection. FEMS Microbiol Rev 2012; 36:616-48. [PMID: 22404313 DOI: 10.1111/j.1574-6976.2012.00339.x] [Citation(s) in RCA: 232] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bladder infections affect millions of people yearly, and recurrent symptomatic infections (cystitis) are very common. The rapid increase in infections caused by multidrug-resistant uropathogens threatens to make recurrent cystitis an increasingly troubling public health concern. Uropathogenic Escherichia coli (UPEC) cause the vast majority of bladder infections. Upon entry into the lower urinary tract, UPEC face obstacles to colonization that constitute population bottlenecks, reducing diversity, and selecting for fit clones. A critical mucosal barrier to bladder infection is the epithelium (urothelium). UPEC bypass this barrier when they invade urothelial cells and form intracellular bacterial communities (IBCs), a process which requires type 1 pili. IBCs are transient in nature, occurring primarily during acute infection. Chronic bladder infection is common and can be either latent, in the form of the quiescent intracellular reservoir (QIR), or active, in the form of asymptomatic bacteriuria (ASB/ABU) or chronic cystitis. In mice, the fate of bladder infection, QIR, ASB, or chronic cystitis, is determined within the first 24 h of infection and constitutes a putative host-pathogen mucosal checkpoint that contributes to susceptibility to recurrent cystitis. Knowledge of these checkpoints and bottlenecks is critical for our understanding of bladder infection and efforts to devise novel therapeutic strategies.
Collapse
Affiliation(s)
- Thomas J Hannan
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
47
|
Genome-wide mapping of cystitis due to Streptococcus agalactiae and Escherichia coli in mice identifies a unique bladder transcriptome that signifies pathogen-specific antimicrobial defense against urinary tract infection. Infect Immun 2012; 80:3145-60. [PMID: 22733575 DOI: 10.1128/iai.00023-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The most common causes of urinary tract infections (UTIs) are Gram-negative pathogens such as Escherichia coli; however, Gram-positive organisms, including Streptococcus agalactiae, or group B streptococcus (GBS), also cause UTI. In GBS infection, UTI progresses to cystitis once the bacteria colonize the bladder, but the host responses triggered in the bladder immediately following infection are largely unknown. Here, we used genome-wide expression profiling to map the bladder transcriptome of GBS UTI in mice infected transurethrally with uropathogenic GBS that was cultured from a 35-year-old women with cystitis. RNA from bladders was applied to Affymetrix Gene-1.0ST microarrays; quantitative reverse transcriptase PCR (qRT-PCR) was used to analyze selected gene responses identified in array data sets. A surprisingly small significant-gene list of 172 genes was identified at 24 h; this compared to 2,507 genes identified in a side-by-side comparison with uropathogenic E. coli (UPEC). No genes exhibited significantly altered expression at 2 h in GBS-infected mice according to arrays despite high bladder bacterial loads at this early time point. The absence of a marked early host response to GBS juxtaposed with broad-based bladder responses activated by UPEC at 2 h. Bioinformatics analyses, including integrative system-level network mapping, revealed multiple activated biological pathways in the GBS bladder transcriptome that regulate leukocyte activation, inflammation, apoptosis, and cytokine-chemokine biosynthesis. These findings define a novel, minimalistic type of bladder host response triggered by GBS UTI, which comprises collective antimicrobial pathways that differ dramatically from those activated by UPEC. Overall, this study emphasizes the unique nature of bladder immune activation mechanisms triggered by distinct uropathogens.
Collapse
|
48
|
Role of Uropathogenic Escherichia coli Virulence Factors in Development of Urinary Tract Infection and Kidney Damage. Int J Nephrol 2012; 2012:681473. [PMID: 22506110 PMCID: PMC3312279 DOI: 10.1155/2012/681473] [Citation(s) in RCA: 216] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 11/02/2011] [Accepted: 12/01/2011] [Indexed: 01/17/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is a causative agent in the vast majority of urinary tract infections (UTIs), including cystitis and pyelonephritis, and infectious complications, which may result in acute renal failure in healthy individuals as well as in renal transplant patients. UPEC expresses a multitude of virulence factors to break the inertia of the mucosal barrier. In response to the breach by UPEC into the normally sterile urinary tract, host inflammatory responses are triggered leading to cytokine production, neutrophil influx, and the exfoliation of infected bladder epithelial cells. Several signaling pathways activated during UPEC infection, including the pathways known to activate the innate immune response, interact with calcium-dependent signaling pathways. Some UPEC isolates, however, might possess strategies to delay or suppress the activation of components of the innate host response in the urinary tract. Studies published in the recent past provide new information regarding how virulence factors of uropathogenic E. coli are involved in activation of the innate host response. Despite numerous host defense mechanisms, UPEC can persist within the urinary tract and may serve as a reservoir for recurrent infections and serious complications. Presentation of the molecular details of these events is essential for development of successful strategies for prevention of human UTIs and urological complications associated with UTIs.
Collapse
|
49
|
Akbarshahi H, Axelsson JBF, Said K, Malmström A, Fischer H, Andersson R. TLR4 dependent heparan sulphate-induced pancreatic inflammatory response is IRF3-mediated. J Transl Med 2011; 9:219. [PMID: 22188870 PMCID: PMC3286488 DOI: 10.1186/1479-5876-9-219] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 12/21/2011] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Degraded extracellular matrix can stimulate the innate immune system via the Toll-Like Receptor-4 (TLR4). In the pancreas, syndecan-anchored heparan sulphate (HS) on the ductal epithelium can be cleaved off its protein cores by the proteases (trypsin and elastase) and potentially activate TLR4 signalling. METHODS To investigate this signalling event, a low sulphated HS (500 μg/ml) was infused into the biliary-pancreatic duct of C57BL/6J wild-type mice. Phosphate buffered saline (PBS) and lipopolysaccharide (LPS) were used as negative and positive controls, respectively. Mice were sacrificed after 1, 3, 6, 9, and 48 hours and tissues were analysed for neutrophil and cytokine contents. In order to study the TLR4 signalling pathway of HS in the pancreas, genetically engineered mice lacking TLR4, Myeloid Differentiation primary response gene (88) (MyD88) or Interferon Regulatory Factor 3 (IRF3) were subjected to pancreatic infusion of HS. RESULTS Neutrophil sequestration and corresponding myeloperoxidase (MPO) activity in the pancreas were increased 9 hours following HS challenge. In wild-type mice, the monocyte chemoattractant protein-1(MCP-1) increased at 3 hours after infusion, while RANTES increased after 9 hours.TLR4, MyD88, and IRF3 knockout mice showed an abrogated neutrophil recruitment and myeloperoxidase activity in the HS group, while the LPS response was only abolished in TLR4 and MyD88 knockouts. CONCLUSIONS The results of this study show that HS is capable of initiating a TLR4-dependent innate immune response in the pancreas which is distinctly different from that induced by LPS. This inflammatory response was mediated predominantly through IRF3- dependent pathway. Release of HS into the pancreatic duct may be one important mediator in the pancreatic ductal defence.
Collapse
Affiliation(s)
- Hamid Akbarshahi
- Department of Clinical Sciences Lund, Lund University, BMC, D12, SE-221 84 Lund, Sweden
| | - Jakob BF Axelsson
- Department of Clinical Sciences Lund, Lund University, BMC, D12, SE-221 84 Lund, Sweden
| | - Katarzyna Said
- Department of Clinical Sciences Lund, Lund University, BMC, D12, SE-221 84 Lund, Sweden
| | - Anders Malmström
- Department of Experimental Medical Sciences, Lund University, BMC, D12, SE-221 84 Lund, Sweden
| | - Hans Fischer
- Department of Experimental Medical Sciences, Lund University, BMC, D12, SE-221 84 Lund, Sweden
| | - Roland Andersson
- Department of Clinical Sciences Lund, Lund University, BMC, D12, SE-221 84 Lund, Sweden
| |
Collapse
|
50
|
Duell BL, Carey AJ, Tan CK, Cui X, Webb RI, Totsika M, Schembri MA, Derrington P, Irving-Rodgers H, Brooks AJ, Cripps AW, Crowley M, Ulett GC. Innate transcriptional networks activated in bladder in response to uropathogenic Escherichia coli drive diverse biological pathways and rapid synthesis of IL-10 for defense against bacterial urinary tract infection. THE JOURNAL OF IMMUNOLOGY 2011; 188:781-92. [PMID: 22184725 DOI: 10.4049/jimmunol.1101231] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Early transcriptional activation events that occur in bladder immediately following bacterial urinary tract infection (UTI) are not well defined. In this study, we describe the whole bladder transcriptome of uropathogenic Escherichia coli (UPEC) cystitis in mice using genome-wide expression profiling to define the transcriptome of innate immune activation stemming from UPEC colonization of the bladder. Bladder RNA from female C57BL/6 mice, analyzed using 1.0 ST-Affymetrix microarrays, revealed extensive activation of diverse sets of innate immune response genes, including those that encode multiple IL-family members, receptors, metabolic regulators, MAPK activators, and lymphocyte signaling molecules. These were among 1564 genes differentially regulated at 2 h postinfection, highlighting a rapid and broad innate immune response to bladder colonization. Integrative systems-level analyses using InnateDB (http://www.innatedb.com) bioinformatics and ingenuity pathway analysis identified multiple distinct biological pathways in the bladder transcriptome with extensive involvement of lymphocyte signaling, cell cycle alterations, cytoskeletal, and metabolic changes. A key regulator of IL activity identified in the transcriptome was IL-10, which was analyzed functionally to reveal marked exacerbation of cystitis in IL-10-deficient mice. Studies of clinical UTI revealed significantly elevated urinary IL-10 in patients with UPEC cystitis, indicating a role for IL-10 in the innate response to human UTI. The whole bladder transcriptome presented in this work provides new insight into the diversity of innate factors that determine UTI on a genome-wide scale and will be valuable for further data mining. Identification of protective roles for other elements in the transcriptome will provide critical new insight into the complex cascade of events that underpin UTI.
Collapse
Affiliation(s)
- Benjamin L Duell
- School of Medical Sciences, Centre for Medicine and Oral Health, Griffith University Gold Coast Campus, Queensland 4222, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|