1
|
Jiang T, Xu X. Protective effect of Timosaponin AIII on Escherichia coli-induced endometritis in mice through inhibiting inflammatory response and regulating uterine microbiota structure. Int Immunopharmacol 2024; 130:111649. [PMID: 38367462 DOI: 10.1016/j.intimp.2024.111649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/10/2024] [Accepted: 01/31/2024] [Indexed: 02/19/2024]
Abstract
Endometritis is a sort of general reproductive disease, which can lead to infertility in both humans and animals. Escherichia coli (E. coli) is recognised as the main bacterial etiology of endometritis among livestock and causes huge economic losses to dairy farming industry. Antibiotics are frequently used in the clinical treatment of endometritis; nevertheless, long-term use may result in adverse effects, including bacterial resistance and food safety concerns. TSAIII, one of the active pharmacological components of A. asphodeloides, has exhibited multiple biological activities, including anticancer, anti-angiogenesis, and anti-inflammatory properties. However, the protective effects of TSAIII in E. coli-challenged endometritis remain unclear. This study aimed to clarify the role of TSAIII in E. coli-induced endometritis in mice and elucidate its specific molecular mechanisms. In the present research, TSAIII treatment markedly alleviated the E. coli-induced uterine histopathological injury, and decreased myeloperoxidase (MPO) activity and pro-inflammatory cytokines levels in uterine tissue. Our results further demonstrated that TSAIII improved uterine epithelial barrier function by restoring the expressions of tight junction proteins. Furthermore, TSAIII administration noticeably suppressed the activation of the TLR4/NF-κB pathway and the NLRP3 inflammasome. Importantly, we found that TSAIII could regulate the uterine microbiota structure and composition in E. coli-induced mouse endometritis. In conclusion, these data demonstrate that treatment with TSAIII protects against E. coli-induced endometritis via modulating uterine microbiota composition, inhibiting TLR4/NF-κB pathway and NLRP3 inflammasome activation, in addition to improving uterine epithelial barrier function. Therefore, the results of this study provide a new therapeutic to potentially prevent endometritis.
Collapse
Affiliation(s)
- Tao Jiang
- China-Japan Union Hospital, Jilin University, Jilin, China
| | - Xuesong Xu
- China-Japan Union Hospital, Jilin University, Jilin, China.
| |
Collapse
|
2
|
Aksu B, Afonso AC, Akil I, Alpay H, Atmis B, Aydog O, Bayazıt AK, Bayram MT, Bilge I, Bulut IK, Buyukkaragoz B, Comak E, Demir BK, Dincel N, Donmez O, Durmus MA, Dursun H, Dusunsel R, Duzova A, Ertan P, Gedikbasi A, Goknar N, Guven S, Hacihamdioglu D, Jankauskiene A, Kalyoncu M, Kavukcu S, Kenan BU, Kucuk N, Kural B, Litwin M, Montini G, Morello W, Obrycki L, Omer B, Oner HA, Ozdemir EM, Ozkayin N, Paripovic D, Pehlivanoglu C, Saygili S, Schaefer F, Schaefer S, Sonmez F, Tabel Y, Tas N, Tasdemir M, Teixeira A, Tekcan D, Topaloglu R, Tulpar S, Turkkan ON, Uysal B, Uysalol M, Vitkevic R, Yavuz S, Yel S, Yildirim T, Yildirim ZY, Yildiz N, Yuksel S, Yurtseven E, Yilmaz A. Urine soluble TLR4 levels may contribute to predict urinary tract infection in children: the UTILISE Study. Pediatr Nephrol 2024; 39:483-491. [PMID: 37462743 DOI: 10.1007/s00467-023-06063-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND One of the most common bacterial infections in childhood is urinary tract infection (UTI). Toll-like receptors (TLRs) contribute to immune response against UTI recognizing specific pathogenic agents. Our aim was to determine whether soluble TLR4 (sTLR4), soluble TLR5 (sTLR5) and interleukin 8 (IL-8) can be used as biomarkers to diagnose UTI. We also aimed to reveal the relationship between urine Heat Shock Protein 70 (uHSP70) and those biomarkers investigated in this study. METHODS A total of 802 children from 37 centers participated in the study. The participants (n = 282) who did not meet the inclusion criteria were excluded from the study. The remaining 520 children, including 191 patients with UTI, 178 patients with non-UTI infections, 50 children with contaminated urine samples, 26 participants with asymptomatic bacteriuria and 75 healthy controls were included in the study. Urine and serum levels of sTLR4, sTLR5 and IL-8 were measured at presentation in all patients and after antibiotic treatment in patients with UTI. RESULTS Urine sTLR4 was higher in the UTI group than in the other groups. UTI may be predicted using 1.28 ng/mL as cut-off for urine sTLR4 with 68% sensitivity and 65% specificity (AUC = 0.682). In the UTI group, urine sTLR4 levels were significantly higher in pyelonephritis than in cystitis (p < 0.0001). Post-treatment urine sTLR4 levels in the UTI group were significantly lower than pre-treatment values (p < 0.0001). CONCLUSIONS Urine sTLR4 may be used as a useful biomarker in predicting UTI and subsequent pyelonephritis in children with UTI. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Bagdagul Aksu
- Department of Pediatrics Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey.
- Division of Pediatric Nephrology, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul, Turkey.
| | - Alberto Caldas Afonso
- Division of Pediatric Nephrology, Centro Materno Infantil do Norte, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Ipek Akil
- Division of Pediatric Nephrology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Harika Alpay
- Division of Pediatric Nephrology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Bahriye Atmis
- Pediatric Nephrology, Erzurum Training and Research Hospital, Erzurum, Turkey
- Division of Pediatric Nephrology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ozlem Aydog
- Division of Pediatric Nephrology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Aysun Karabay Bayazıt
- Division of Pediatric Nephrology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Meral Torun Bayram
- Division of Pediatric Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ilmay Bilge
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Koc University, Istanbul, Turkey
| | - Ipek Kaplan Bulut
- Division of Pediatric Nephrology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Bahar Buyukkaragoz
- Division of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Elif Comak
- Division of Pediatric Nephrology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Belde Kasap Demir
- Division of Pediatric Nephrology, Tepecik Training and Research Hospital, University of Health Sciences, Izmir, Turkey
- Division of Pediatric Nephrology, Izmir Katip Celebi University Faculty of Medicine, Izmir, Turkey
| | - Nida Dincel
- Division of Pediatric Nephrology, Dr. Behcet Uz Children Diseases Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Osman Donmez
- Division of Pediatric Nephrology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Mehmet Akif Durmus
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hasan Dursun
- Division of Pediatric Nephrology, Okmeydani Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ruhan Dusunsel
- Division of Pediatric Nephrology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Ali Duzova
- Division of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Pelin Ertan
- Division of Pediatric Nephrology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Asuman Gedikbasi
- Department of Rare Diseases, Institute of Child Health, Istanbul University, Istanbul, Turkey
| | - Nilufer Goknar
- Division of Pediatric Nephrology, Bagcilar Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Sercin Guven
- Division of Pediatric Nephrology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Duygu Hacihamdioglu
- Division of Pediatric Nephrology, Faculty of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Augustina Jankauskiene
- Clinic of Pediatrics, Institute of Clinical Medicine, Vilnius University, Vilnius, Lithuania
| | - Mukaddes Kalyoncu
- Division of Pediatric Nephrology, Faculty of Medicine, Karadeniz Technic University, Trabzon, Turkey
| | - Salih Kavukcu
- Division of Pediatric Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Bahriye Uzun Kenan
- Division of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Nuran Kucuk
- Division of Pediatric Nephrology, Kartal Training and Research Hospital, Istanbul, Turkey
| | - Bahar Kural
- Department of Pediatrics, Health Science University Bakirkoy Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Mieczysław Litwin
- Division of Nephrology, Kidney Transplantation and Hypertension, The Children's Memorial Health Institute, Warsaw, Poland
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - William Morello
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Lukasz Obrycki
- Division of Nephrology, Kidney Transplantation and Hypertension, The Children's Memorial Health Institute, Warsaw, Poland
| | - Beyhan Omer
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Huseyin Adil Oner
- Division of Pediatric Nephrology, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul, Turkey
| | - Ebru Misirli Ozdemir
- Department of Pediatrics, Okmeydani Training and Research Hospital, Istanbul, Turkey
| | - Nese Ozkayin
- Division of Pediatric Nephrology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Dusan Paripovic
- Division of Pediatric Nephrology, University Children's Hospital, Belgrade, Serbia
| | - Cemile Pehlivanoglu
- Division of Pediatric Nephrology, Umraniye Training and Research Hospital, Istanbul, Turkey
| | - Seha Saygili
- Division of Pediatric Nephrology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Susanne Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University, Heidelberg, Germany
| | - Ferah Sonmez
- Division of Pediatric Nephrology, Faculty of Medicine, Adnan Menderes University, Aydin, Turkey
| | - Yilmaz Tabel
- Division of Pediatric Nephrology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Nesrin Tas
- Division of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Mehmet Tasdemir
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Koc University, Istanbul, Turkey
- Department of Pediatric Nephrology, Istinye University, Faculty of Medicine, Liv Hospital Ulus, Istanbul, Turkey
| | - Ana Teixeira
- Division of Pediatric Nephrology, Centro Materno Infantil do Norte, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Demet Tekcan
- Division of Pediatric Nephrology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Rezan Topaloglu
- Division of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sebahat Tulpar
- Division of Pediatric Nephrology, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Ozde Nisa Turkkan
- Division of Pediatric Nephrology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Berfin Uysal
- Division of Pediatric Nephrology, Dortcelik Children's Hospital, Bursa, Turkey
- Division of Pediatric Nephrology, Bursa City Hospital, Bursa, Turkey
| | - Metin Uysalol
- Division of Pediatric Emergency, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Renata Vitkevic
- Clinic of Pediatrics, Institute of Clinical Medicine, Vilnius University, Vilnius, Lithuania
| | - Sevgi Yavuz
- Division of Pediatric Nephrology, Department of Pediatrics, Kanuni Sultan Suleyman Research and Training Hospital, University of Health Sciences, Istanbul, Turkey
| | - Sibel Yel
- Division of Pediatric Nephrology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Tarik Yildirim
- Department of Pediatrics, Kanuni Sultan Suleyman Research and Training Hospital, University of Health Sciences, Istanbul, Turkey
| | - Zeynep Yuruk Yildirim
- Department of Pediatrics Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Division of Pediatric Nephrology, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul, Turkey
| | - Nurdan Yildiz
- Division of Pediatric Nephrology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Selcuk Yuksel
- Division of Pediatric Nephrology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Eray Yurtseven
- Department of Biostatistics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Alev Yilmaz
- Department of Pediatrics Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Division of Pediatric Nephrology, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul, Turkey
| |
Collapse
|
3
|
Habeichi NJ, Amin G, Lakkis B, Kataya R, Mericskay M, Booz GW, Zouein FA. Potential Alternative Receptors for SARS-CoV-2-Induced Kidney Damage: TLR-4, KIM-1/TIM-1, and CD147. FRONT BIOSCI-LANDMRK 2024; 29:8. [PMID: 38287815 PMCID: PMC10924798 DOI: 10.31083/j.fbl2901008] [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: 10/17/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 01/31/2024]
Abstract
Kidney damage in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can occur even in patients with no underlying kidney disease. Signs of kidney problems can progress to a state that demands dialysis and hampering recovery. Although not without controversy, emerging evidence implicates direct infectivity of SARS-CoV-2 in the kidney. At the early stage of the pandemic, consideration was mainly on the well-recognized angiotensin-converting enzyme 2 (ACE2) receptor as being the site for viral interaction and subsequent cellular internalization. Despite the abundance of ACE2 receptors in the kidneys, researchers have expanded beyond ACE2 and identified novel viral entry pathways that could be advantageously explored as therapeutic targets. This review presents the potential involvement of toll-like receptor 4 (TLR-4), kidney injury molecule-1/T cell immunoglobulin mucin domain 1 (KIM-1/TIM-1), and cluster of differentiation 147 (CD147) in SARS-CoV-2-associated renal damage. In this context, we address the unresolved issues surrounding SARS-CoV-2 renal infectivity.
Collapse
Affiliation(s)
- Nada J. Habeichi
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, 1107-2020 Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, 1107-2020 Beirut, Lebanon
- Department of Signaling and Cardiovascular Pathophysiology, University Paris Saclay, INSERM UMR_1180, 91400 Orsay, France
- MatriceLab Innove Laboratory, Immeuble Les Gemeaux, 94000 Creteil, France
| | - Ghadir Amin
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, 1107-2020 Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, 1107-2020 Beirut, Lebanon
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bachir Lakkis
- Division of Cardiology, Department of Internal Medicine, American University of Beirut Medical Center, 1107-2020 Beirut, Lebanon
| | - Rayane Kataya
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, 1107-2020 Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, 1107-2020 Beirut, Lebanon
| | - Mathias Mericskay
- Department of Signaling and Cardiovascular Pathophysiology, University Paris Saclay, INSERM UMR_1180, 91400 Orsay, France
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, 1107-2020 Beirut, Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, 1107-2020 Beirut, Lebanon
- Department of Signaling and Cardiovascular Pathophysiology, University Paris Saclay, INSERM UMR_1180, 91400 Orsay, France
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
4
|
Chen C, Li J, Wang J, Zhang M, Zhang L, Lin Z. Oxybutynin ameliorates LPS-induced inflammatory response in human bladder epithelial cells. J Biochem Mol Toxicol 2024; 38:e23584. [PMID: 38009396 DOI: 10.1002/jbt.23584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 10/10/2023] [Accepted: 11/10/2023] [Indexed: 11/28/2023]
Abstract
Urinary tract infection (UTI) mainly results from bacterial infections in the urinary tract and markedly impacts the normal lives of millions of patients worldwide. The infection and damage to urethral epithelial cells is the first and key step of UTI development and is a critical target for treating clinical UTI. Oxybutynin, an agent for treating urinary incontinence, is recently claimed with protective effects on bladder ultrastructure. Our study will assess the impact of Oxybutynin on inflammation in lipopolysaccharide (LPS)-stimulated bladder epithelial cells. Bladder epithelial T24 cells were treated with 1 μg/mL LPS with or without 10 and 20 μM Oxybutynin for 24 h. Increased levels of oxidative stress (OS) biomarkers, such as reactive oxygen species, 8-hydroxy-2'-deoxyguanosine, malondialdehyde, as well as upregulated inducible nitric oxide synthase and promoted release of nitric oxide, were observed in LPS-managed T24 cells, all of which were signally suppressed by Oxybutynin. Furthermore, severe inflammatory responses, including enhanced release of cytokines, upregulated matrix metallopeptidase-2 (MMP-2) and MMP-9, and raised monocyte chemoattractant protein-1 level, were found in LPS-challenged T24 cells, which were markedly reversed by Oxybutynin. Moreover, the activated toll-1ike receptor 4/nuclear factor-κB pathway observed in LPS-managed T24 cells was repressed by Oxybutynin. Collectively, Oxybutynin mitigated LPS-induced inflammatory response in human bladder epithelial cells.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Pharmacy, The First People's Hospital of Yibin, Yibin, China
| | - Jiangtao Li
- Department of Rheumatology and Immunology, The First People's Hospital of Yibin, Yibin, China
| | - Juan Wang
- Department of Pharmaceutical Engineering, School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Mao Zhang
- Department of Pharmacy, The First People's Hospital of Yibin, Yibin, China
| | - Lei Zhang
- Department of Pharmacy, The First People's Hospital of Yibin, Yibin, China
| | - Zhihua Lin
- Department of Pharmaceutical Engineering, School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| |
Collapse
|
5
|
Archer AC, DeBerry JJ, DeWitte C, Ness TJ. Neonatal Cystitis Makes Adult Female Rat Urinary Bladders More Sensitive to Low Concentration Microbial Antigens. Res Rep Urol 2023; 15:531-539. [PMID: 38106986 PMCID: PMC10723592 DOI: 10.2147/rru.s444167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
Purpose Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic pain disorder. Patients with IC/BPS often experience "flares" of symptom exacerbation throughout their lifetime, initiated by triggers, such as urinary tract infections. This study sought to determine whether neonatal bladder inflammation (NBI) alters the sensitivity of adult rat bladders to microbial antigens. Methods Female NBI rats received intravesical zymosan treatments on postnatal days P14-P16 while anesthetized; Neonatal Control Treatment (NCT) rats were anesthetized. In adults, bladder and spinal cord Toll-like receptor type 2 and 4 (TLR2, TLR4) contents were determined using ELISAs. Other rats were injected intravesically with lipopolysaccharide (LPS; mimics an E. coli infection; 25, 50, 100, or 200 μg/mL) or Zymosan (mimics yeast infection; 0.01, 0.1, 1, and 10 mg/mL) solutions on the following day. Visceromotor responses (VMRs; abdominal contractions) to graded urinary bladder distention (UBD, 10-60 mm Hg, 20s) were quantified as abdominal electromyograms (EMGs). Results Bladder TLR2 and TLR4 protein levels increased in NBI rats. These rats displayed statistically significant, dose-dependent, robustly augmented VMRs following all but the lowest doses of LPS and Zymosan tested, when compared with their adult treatment control groups. The NCT groups showed minimal responses to LPS in adults and minimally increased EMG measurements following the highest dose of Zymosan. Conclusion The microbial antigens LPS and Zymosan augmented nociceptive VMRs to UBD in rats that experienced NBI but had little effect on NCT rats at the doses tested. The greater content of bladder TLR2 and TLR4 proteins in the NBI group was consistent with increased responsiveness to their agonists, Zymosan and LPS, respectively. Given that patients with IC/BPS have a higher incidence of childhood urinary tract infections, this increased responsiveness to microbial antigens may explain the flares in symptoms following "subclinical" tract infections.
Collapse
Affiliation(s)
- Ashley C Archer
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Jennifer J DeBerry
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cary DeWitte
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Timothy J Ness
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
6
|
Almazmomi MA, Esmat A, Naeem A. Acute Kidney Injury: Definition, Management, and Promising Therapeutic Target. Cureus 2023; 15:e51228. [PMID: 38283512 PMCID: PMC10821757 DOI: 10.7759/cureus.51228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Acute kidney injury (AKI) is caused by a sudden loss of renal function, resulting in the build-up of waste products and a significant increase in mortality and morbidity. It is commonly diagnosed in critically ill patients, with its occurrence estimated at up to 50% in patients hospitalized in the intensive critical unit. Despite ongoing efforts, the death rate associated with AKI has remained high over the past half-century. Thus, it is critical to investigate novel therapy options for preventing the epidemic. Many studies have found that inflammation and Toll-like receptor-4 (TLR-4) activation have a significant role in the pathogenesis of AKI. Noteworthy, challenges in the search for efficient pharmacological therapy for AKI have arisen due to the multifaceted origin and complexity of the clinical history of people with the disease. This article focuses on kidney injury's epidemiology, risk factors, and pathophysiological processes. Specifically, it focuses on the role of TLRs especially type 4 in disease development.
Collapse
Affiliation(s)
- Meaad A Almazmomi
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Ahmed Esmat
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Anjum Naeem
- Pharmaceutical Care Department, Ministry of National Guard - Health Affairs, Jeddah, SAU
| |
Collapse
|
7
|
Peng Z, Zhuang J, Shen B. The role of microbiota in tumorigenesis, progression and treatment of bladder cancer. MICROBIOME RESEARCH REPORTS 2023; 3:5. [PMID: 38455086 PMCID: PMC10917617 DOI: 10.20517/mrr.2023.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/09/2023] [Accepted: 11/13/2023] [Indexed: 03/09/2024]
Abstract
For decades, the urinary system was regarded as a sterile environment due to the absence of any bacterial growth in clinical standard urine cultures from healthy individuals. However, a diverse array of microbes colonizes the urinary system in small quantities, exhibiting a variable compositional signature influenced by differences in sex, age, and pathological state. Increasing pieces of evidence suggest microbiota exists in tumor tissue and plays a crucial role in tumor microenvironment based on research in multiple cancer models. Current studies about microbiota and bladder cancer have preliminarily characterized the bladder cancer-related microbiota, but how the microbiota influences the biological behavior of bladder cancer remains unclarified. This review summarizes the characteristics of microbiota in bladder cancer, aims to propose possible mechanisms that microbiota acts in tumorigenesis and progression of bladder cancer based on advances in gut microbiota, and discusses the potential clinical application of microbiota in bladder cancer.
Collapse
Affiliation(s)
| | | | - Bing Shen
- Correspondence to: Prof. Bing Shen, Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, NO. 85 Wu Jin Road, Hongkou District, Shanghai 200080, China. E-mail:
| |
Collapse
|
8
|
Whelan S, Lucey B, Finn K. Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment. Microorganisms 2023; 11:2169. [PMID: 37764013 PMCID: PMC10537683 DOI: 10.3390/microorganisms11092169] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections, especially among women and older adults, leading to a significant global healthcare cost burden. Uropathogenic Escherichia coli (UPEC) are the most common cause and accounts for the majority of community-acquired UTIs. Infection by UPEC can cause discomfort, polyuria, and fever. More serious clinical consequences can result in urosepsis, kidney damage, and death. UPEC is a highly adaptive pathogen which presents significant treatment challenges rooted in a complex interplay of molecular factors that allow UPEC to evade host defences, persist within the urinary tract, and resist antibiotic therapy. This review discusses these factors, which include the key genes responsible for adhesion, toxin production, and iron acquisition. Additionally, it addresses antibiotic resistance mechanisms, including chromosomal gene mutations, antibiotic deactivating enzymes, drug efflux, and the role of mobile genetic elements in their dissemination. Furthermore, we provide a forward-looking analysis of emerging alternative therapies, such as phage therapy, nano-formulations, and interventions based on nanomaterials, as well as vaccines and strategies for immunomodulation. This review underscores the continued need for research into the molecular basis of pathogenesis and antimicrobial resistance in the treatment of UPEC, as well as the need for clinically guided treatment of UTIs, particularly in light of the rapid spread of multidrug resistance.
Collapse
Affiliation(s)
- Shane Whelan
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Brigid Lucey
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Karen Finn
- Department of Analytical, Biopharmaceutical and Medical Sciences, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland
| |
Collapse
|
9
|
Edwards K, Lydyard PM, Kulikova N, Tsertsvadze T, Volpi EV, Chiorazzi N, Porakishvili N. The role of CD180 in hematological malignancies and inflammatory disorders. Mol Med 2023; 29:97. [PMID: 37460961 PMCID: PMC10353253 DOI: 10.1186/s10020-023-00682-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Toll-like receptors play a significant role in the innate immune system and are also involved in the pathophysiology of many different diseases. Over the past 35 years, there have been a growing number of publications exploring the role of the orphan toll-like receptor, CD180. We therefore set out to provide a narrative review of the current evidence surrounding CD180 in both health and disease. We first explore the evidence surrounding the role of CD180 in physiology including its expression, function and signaling in antigen presenting cells (APCs) (dendritic cells, monocytes, and B cells). We particularly focus on the role of CD180 as a modulator of other TLRs including TLR2, TLR4, and TLR9. We then discuss the role of CD180 in inflammatory and autoimmune diseases, as well as in hematological malignancies of B cell origin, including chronic lymphocytic leukemia (CLL). Based on this evidence we produce a current model for CD180 in disease and explore the potential role for CD180 as both a prognostic biomarker and therapeutic target. Throughout, we highlight specific areas of research which should be addressed to further the understanding of CD180 biology and the translational potential of research into CD180 in various diseases.
Collapse
Affiliation(s)
- Kurtis Edwards
- School of Life Sciences, University of Westminster, London, UK
| | - Peter M Lydyard
- School of Life Sciences, University of Westminster, London, UK.
- The University of Georgia, Tbilisi, Georgia.
- Division of Infection of Immunity, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Nino Kulikova
- Agricultural University of Georgia, Tbilisi, Georgia
| | | | | | | | | |
Collapse
|
10
|
Faustino M, Silva S, Costa EM, Pereira AM, Pereira JO, Oliveira AS, Ferreira CMH, Pereira CF, Durão J, Pintado ME, Carvalho AP. Effect of Mannan Oligosaccharides Extracts in Uropathogenic Escherichia coli Adhesion in Human Bladder Cells. Pathogens 2023; 12:885. [PMID: 37513732 PMCID: PMC10384913 DOI: 10.3390/pathogens12070885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/16/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Urinary tract infections (UTIs) are a common public health problem, mainly caused by uropathogenic Escherichia coli (UPEC). Patients with chronic UTIs are usually treated with long-acting prophylactic antibiotics, which promotes the development of antibiotic-resistant UPEC strains and may complicate their long-term management. D-mannose and extracts rich in D-mannose such as mannan oligosaccharides (MOS; D-mannose oligomers) are promising alternatives to antibiotic prophylaxis due to their ability to inhibit bacterial adhesion to urothelial cells and, therefore, infection. This highlights the therapeutic potential and commercial value of using them as health supplements. Studies on the effect of MOS in UTIs are, however, scarce. Aiming to evaluate the potential benefits of using MOS extracts in UTIs prophylaxis, their ability to inhibit the adhesion of UPEC to urothelial cells and its mechanism of action were assessed. Additionally, the expression levels of the pro-inflammatory marker interleukin 6 (IL-6) were also evaluated. After characterizing their cytotoxic profiles, the preliminary results indicated that MOS extracts have potential to be used for the handling of UTIs and demonstrated that the mechanism through which they inhibit bacterial adhesion is through the competitive inhibition of FimH adhesins through the action of mannose, validated by a bacterial growth impact assessment.
Collapse
Affiliation(s)
- Margarida Faustino
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Sara Silva
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Eduardo M Costa
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Ana Margarida Pereira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
- Amyris Bio Products Portugal, Unipessoal Lda, Rua Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - Joana Odila Pereira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
- Amyris Bio Products Portugal, Unipessoal Lda, Rua Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - Ana Sofia Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Carlos M H Ferreira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
- Amyris Bio Products Portugal, Unipessoal Lda, Rua Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - Carla F Pereira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Joana Durão
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
- Amyris Bio Products Portugal, Unipessoal Lda, Rua Diogo Botelho, 1327, 4169-005 Porto, Portugal
| | - Manuela E Pintado
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| | - Ana P Carvalho
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal
| |
Collapse
|
11
|
Colceriu MC, Aldea PL, Răchișan AL, Clichici S, Sevastre-Berghian A, Mocan T. Vesicoureteral Reflux and Innate Immune System: Physiology, Physiopathology, and Clinical Aspects. J Clin Med 2023; 12:jcm12062380. [PMID: 36983379 PMCID: PMC10058356 DOI: 10.3390/jcm12062380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Vesicoureteral reflux represents one of the most concerning topics in pediatric nephrology due to its frequency, clinical expression with the potential to evolve into chronic kidney disease, and last but not least, its socio-economic implications. The presence of vesicoureteral reflux, the occurrence of urinary tract infections, and the development of reflux nephropathy, hypertension, chronic kidney disease, and finally, end-stage renal disease represent a progressive spectrum of a single physiopathological condition. For the proper management of these patients with the best clinical outcomes, and in an attempt to prevent the spread of uropathogens' resistance to antibacterial therapy, we must better understand the physiopathology of urinary tract infections in patients with vesicoureteral reflux, and at the same time, we should acknowledge the implication and response of the innate immune system in this progressive pathological condition. The present paper focuses on theoretical aspects regarding the physiopathology of vesicoureteral reflux and the interconditionality between urinary tract infections and the innate immune system. In addition, we detailed aspects regarding cytokines, interleukins, antimicrobial peptides, and proteins involved in the innate immune response as well as their implications in the physiopathology of reflux nephropathy. New directions of study should focus on using these innate immune system effectors as diagnostic and therapeutic tools in renal pathology.
Collapse
Affiliation(s)
- Marius-Cosmin Colceriu
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Paul Luchian Aldea
- Department of Community Medicine, Discipline of Public Health and Management, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Andreea-Liana Răchișan
- Department of Mother and Child, Discipline of Pediatrics II, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Simona Clichici
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Alexandra Sevastre-Berghian
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Teodora Mocan
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, 400158 Cluj-Napoca, Romania
| |
Collapse
|
12
|
Tan A, Alsenani Q, Lanz M, Birchall C, Drage LKL, Picton D, Mowbray C, Ali A, Harding C, Pickard RS, Hall J, Aldridge PD. Evasion of toll-like receptor recognition by Escherichia coli is mediated via population level regulation of flagellin production. Front Microbiol 2023; 14:1093922. [PMID: 37032848 PMCID: PMC10078357 DOI: 10.3389/fmicb.2023.1093922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Uropathogenic Escherichia coli is a major cause of urinary tract infections. Analysis of the innate immune response in immortalized urothelial cells suggests that the bacterial flagellar subunit, flagellin, is key in inducing host defenses. A panel of 48 clinical uro-associated E. coli isolates recovered from either cystitis, pyelonephritis asymptomatic bacteriuria (ABU) or UTI-associated bacteraemia infections were characterized for motility and their ability to induce an innate response in urothelial cells stably transfected with a NF-κB luciferase reporter. Thirty-two isolates (67%) were identified as motile with strains recovered from cystitis patients exhibiting an uneven motility distribution pattern; seven of the cystitis isolates were associated with a > 5-fold increase in NF-κB signaling. To explore whether the NF-κB signaling response reflected antigenic variation, flagellin was purified from 14 different isolates. Purified flagellin filaments generated comparable NF-κB signaling responses, irrespective of either the source of the isolate or H-serotype. These data argued against any variability between isolates being related to flagellin itself. Investigations also argued that neither TLR4 dependent recognition of bacterial lipopolysaccharide nor growth fitness of the isolates played key roles in leading to the variable host response. To determine the roles, if any, of flagellar abundance in inducing these variable responses, flagellar hook numbers of a range of cystitis and ABU isolates were quantified. Images suggested that up to 60% of the isolate population exhibited flagella with the numbers averaging between 1 and 2 flagella per bacterial cell. These data suggest that selective pressures exist in the urinary tract that allow uro-associated E. coli strains to maintain motility, but exploit population heterogeneity, which together function to prevent host TLR5 recognition and bacterial killing.
Collapse
Affiliation(s)
- Aaron Tan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Qusai Alsenani
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Marcello Lanz
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher Birchall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lauren K. L. Drage
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David Picton
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Catherine Mowbray
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ased Ali
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher Harding
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Urology Department, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Robert S. Pickard
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Urology Department, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Judith Hall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Judith Hall,
| | - Phillip D. Aldridge
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Phillip D. Aldridge,
| |
Collapse
|
13
|
Martin A, Woolbright BL, Umar S, Ingersoll MA, Taylor JA. Bladder cancer, inflammageing and microbiomes. Nat Rev Urol 2022; 19:495-509. [PMID: 35798831 DOI: 10.1038/s41585-022-00611-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
Ageing is correlated with elevated bladder cancer incidence, morbidity and mortality. Advanced age is also associated with elevated markers of chronic inflammation and perturbations in gut and urinary tract microbiota. One reason for the increased incidence and mortality of bladder cancer in the elderly might be that age-associated changes in multiple microbiomes induce systemic metabolic changes that contribute to immune dysregulation with potentially tumorigenic effects. The gut and urinary microbiomes could be dysregulated in bladder cancer, although the effect of these changes is poorly understood. Each of these domains - the immune system, gut microbiome and urinary microbiome - might also influence the response of patients with bladder cancer to treatment. Improved understanding of age-related alterations to the immune system and gut and urinary microbiomes could provide possible insight into the risk of bladder cancer development and progression in the elderly. In patients with bladder cancer, improved understanding of microbiota might also provide potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Austin Martin
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Molly A Ingersoll
- Université Paris Cité, Institut Cochin, INSERM U1016, Paris, France.,Mucosal Inflammation and Immunity group, Department of Immunology, Institut Pasteur, Paris, France
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
14
|
Jafari NV, Rohn JL. The urothelium: a multi-faceted barrier against a harsh environment. Mucosal Immunol 2022; 15:1127-1142. [PMID: 36180582 PMCID: PMC9705259 DOI: 10.1038/s41385-022-00565-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 02/04/2023]
Abstract
All mucosal surfaces must deal with the challenge of exposure to the outside world. The urothelium is a highly specialized layer of stratified epithelial cells lining the inner surface of the urinary bladder, a gruelling environment involving significant stretch forces, osmotic and hydrostatic pressures, toxic substances, and microbial invasion. The urinary bladder plays an important barrier role and allows the accommodation and expulsion of large volumes of urine without permitting urine components to diffuse across. The urothelium is made up of three cell types, basal, intermediate, and umbrella cells, whose specialized functions aid in the bladder's mission. In this review, we summarize the recent insights into urothelial structure, function, development, regeneration, and in particular the role of umbrella cells in barrier formation and maintenance. We briefly review diseases which involve the bladder and discuss current human urothelial in vitro models as a complement to traditional animal studies.
Collapse
Affiliation(s)
- Nazila V Jafari
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK
| | - Jennifer L Rohn
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK.
| |
Collapse
|
15
|
Liu J, Feng X, Li B, Sun Y, Jin T, Feng M, Ni Y, Liu M. Lactobacillus rhamnosus GR-1 Alleviates Escherichia coli-Induced Inflammation via NF-κB and MAPKs Signaling in Bovine Endometrial Epithelial Cells. Front Cell Infect Microbiol 2022; 12:809674. [PMID: 35310848 PMCID: PMC8924357 DOI: 10.3389/fcimb.2022.809674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Escherichia coli counts as a major endometritis-causing pathogen among dairy cows, which lowered the economic benefits of dairy farming seriously. Probiotic consumption has been reported to impart beneficial effects on immunomodulation. However, the inflammatory regulation mechanism of probiotics on endometritis in dairy cows remains unexplored. The current work aimed to clarify the mechanism whereby Lactobacillus rhamnosus GR-1 (L. rhamnosus GR-1) resists bovine endometrial epithelial cells (BEECs) inflammatory injury induced by E. coli. The model of cellular inflammatory injury was established in the BEECs, which comes from the uterus of healthy dairy cows using E. coli. The outcome of L. rhamnosus GR-1 addition on inflammation was evaluated in BEECs with E. coli-induced endometritis. The underlying mechanisms of anti-inflammation by L. rhamnosus GR-1 were further explored in E. coli-stimulated BEECs. In accordance with the obtained results, the use L. rhamnosus GR-1 alone could not cause the change of inflammatory factors, while L. rhamnosus GR-1 could significantly alleviate the expression of E. coli-induced inflammatory factors. Based on further study, L. rhamnosus GR-1 significantly hindered the TLR4 and MyD88 expression stimulated by E. coli. Moreover, we observed that in BEECs, L. rhamnosus GR-1 could inhibit the E. coli-elicited expressions of pathway proteins that are associated with NF-κB and MAPKs. Briefly, L. rhamnosus GR-1 can effectively protect against E. coli-induced inflammatory response that may be closely related to the inhibition of TLR4 and MyD88 stimulating NF-κB and MAPKs.
Collapse
|
16
|
McSweeney KR, Gadanec LK, Qaradakhi T, Ali BA, Zulli A, Apostolopoulos V. Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers (Basel) 2021; 13:1572. [PMID: 33805488 PMCID: PMC8036620 DOI: 10.3390/cancers13071572] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Administration of the chemotherapeutic agent cisplatin leads to acute kidney injury (AKI). Cisplatin-induced AKI (CIAKI) has a complex pathophysiological map, which has been linked to cellular uptake and efflux, apoptosis, vascular injury, oxidative and endoplasmic reticulum stress, and inflammation. Despite research efforts, pharmaceutical interventions, and clinical trials spanning over several decades, a consistent and stable pharmacological treatment option to reduce AKI in patients receiving cisplatin remains unavailable. This has been predominately linked to the incomplete understanding of CIAKI pathophysiology and molecular mechanisms involved. Herein, we detail the extensively known pathophysiology of cisplatin-induced nephrotoxicity that manifests and the variety of pharmacological and genetic alteration studies that target them.
Collapse
|
17
|
Gill K, Horsley H, Swamy S, Khasriya R, Malone-Lee J. A prospective observational study of urinary cytokines and inflammatory response in patients with Overactive Bladder Syndrome. BMC Urol 2021; 21:39. [PMID: 33740940 PMCID: PMC7980577 DOI: 10.1186/s12894-021-00809-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/03/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Contemporary studies have discredited the methods used to exclude urinary tract infection (UTI) when treating overactive bladder (OAB). Thus we must revisit the OAB phenotype to check that UTI has not been overlooked. AIMS To examine the differences in urinary cytokines IL6 and lactoferrin in OAB patients compared to controls, with references to microscopy of urine and enhanced quantitative urine culture. METHODS A blinded, prospective cohort study with normal controls using six repeated measures, achieved two-monthly, over 12 months. RESULTS The differences between patients and controls in urine IL6 (F = 49.0, p < .001) and lactoferrin (F = 228.5, p < .001) were significant and of a magnitude to have clinical implications. These differences were for lactoferrin correlated to symptoms (9.3, p = .003); for both to pyuria (IL6 F = 66.2, p < .001, Lactoferrin F = 73.9, p < .001); and for IL6 microbial abundance (F = 5.1, p = .024). The pathological markers had been missed by urinary dipsticks and routine MSU culture. CONCLUSION The OAB phenotype may encompass patients with UTI that is being overlooked because of the failure of standard screening methods.
Collapse
Affiliation(s)
- Kiren Gill
- Women’s Health, Whittington Health NHS Trust, Magdala Avenue, London, N19 5FN UK
| | - Harry Horsley
- Bladder Infection and Immunity Group (BIIG), Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF UK
| | - Sheela Swamy
- Women’s Health, St Thomas’ Hospital, Westminster Bridge Road, London, SE1 7EH UK
| | - Rajvinder Khasriya
- Women’s Health, Whittington Health NHS Trust, Magdala Avenue, London, N19 5FN UK
| | - James Malone-Lee
- Bladder Infection and Immunity Group (BIIG), Department of Renal Medicine, Division of Medicine, University College London, 10 Harley Street, London, W1G 9PF UK
| |
Collapse
|
18
|
Jha AK, Gairola S, Kundu S, Doye P, Syed AM, Ram C, Murty US, Naidu VGM, Sahu BD. Toll-like receptor 4: An attractive therapeutic target for acute kidney injury. Life Sci 2021; 271:119155. [PMID: 33548286 DOI: 10.1016/j.lfs.2021.119155] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/14/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
Acute kidney injury (AKI) is a progressive renal complication which significantly affects the patient's life with huge economic burden. Untreated acute kidney injury eventually progresses to a chronic form and end-stage renal disease. Although significant breakthroughs have been made in recent years, there are still no effective pharmacological therapies for the treatment of acute kidney injury. Toll-like receptor 4 (TLR4) is a well-characterized pattern recognition receptor, and increasing evidence has shown that TLR4 mediated inflammatory response plays a pivotal role in the pathogenesis of acute kidney injury. The expression of TLR4 has been seen in resident renal cells, including podocytes, mesangial cells, tubular epithelial cells and endothelial cells. Activation of TLR4 signaling regulates the transcription of numerous pro-inflammatory cytokines and chemokines, resulting in renal inflammation. Therefore, targeting TLR4 and its downstream effectors could serve as an effective therapeutic intervention to prevent renal inflammation and subsequent kidney damage. For the first time, this review summarizes the literature on acute kidney injury from the perspective of TLR4 from year 2010 to 2020. In the current review, the role of TLR4 signaling pathway in AKI with preclinical evidence is discussed. Furthermore, we have highlighted several compounds of natural and synthetic origin, which have the potential to avert the renal TLR4 signaling in preclinical AKI models and have shown protection against AKI. This scientific review provides new ideas for targeting TLR4 in the treatment of AKI and provides strategies for the drug development against AKI.
Collapse
Affiliation(s)
- Ankush Kumar Jha
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Shobhit Gairola
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Sourav Kundu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Pakpi Doye
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Abu Mohammad Syed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Chetan Ram
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Upadhyayula Suryanarayana Murty
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Changsari PIN-781101, Assam, India.
| |
Collapse
|
19
|
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
|
20
|
Ren Q, Cheng L, Yi J, Ma L, Pan J, Gou SJ, Fu P. Toll-like Receptors as Potential Therapeutic Targets in Kidney Diseases. Curr Med Chem 2020; 27:5829-5854. [PMID: 31161985 DOI: 10.2174/0929867325666190603110907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/15/2019] [Accepted: 05/13/2019] [Indexed: 02/08/2023]
Abstract
Toll-like Receptors (TLRs) are members of pattern recognition receptors and serve a pivotal role in host immunity. TLRs response to pathogen-associated molecular patterns encoded by pathogens or damage-associated molecular patterns released by dying cells, initiating an inflammatory cascade, where both beneficial and detrimental effects can be exerted. Accumulated evidence has revealed that TLRs are closely associated with various kidney diseases but their roles are still not well understood. This review updated evidence on the roles of TLRs in the pathogenesis of kidney diseases including urinary tract infection, glomerulonephritis, acute kidney injury, transplant allograft dysfunction and chronic kidney diseases.
Collapse
Affiliation(s)
- Qian Ren
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lu Cheng
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jing Yi
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Liang Ma
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jing Pan
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Shen-Ju Gou
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ping Fu
- Kidney Research Laboratory, Division of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Bladder urothelium converts bacterial lipopolysaccharide information into neural signaling via an ATP-mediated pathway to enhance the micturition reflex for rapid defense. Sci Rep 2020; 10:21167. [PMID: 33273625 PMCID: PMC7713076 DOI: 10.1038/s41598-020-78398-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
When bacteria enter the bladder lumen, a first-stage active defensive mechanism flushes them out. Although urinary frequency induced by bacterial cystitis is a well-known defensive response against bacteria, the underlying mechanism remains unclear. In this study, using a mouse model of acute bacterial cystitis, we demonstrate that the bladder urothelium senses luminal extracellular bacterial lipopolysaccharide (LPS) through Toll-like receptor 4 and releases the transmitter ATP. Moreover, analysis of purinergic P2X2 and P2X3 receptor-deficient mice indicated that ATP signaling plays a pivotal role in the LPS-induced activation of L6–S1 spinal neurons through the bladder afferent pathway, resulting in rapid onset of the enhanced micturition reflex. Thus, we revealed a novel defensive mechanism against bacterial infection via an epithelial-neural interaction that induces urinary frequency prior to bacterial clearance by neutrophils of the innate immune system. Our results indicate an important defense role for the bladder urothelium as a chemical-neural transducer, converting bacterial LPS information into neural signaling via an ATP-mediated pathway, with bladder urothelial cells acting as sensory receptor cells.
Collapse
|
22
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
23
|
Wang P, Wang J, Xie Z, Zhou J, Lu Q, Zhao Y, Dong C, Zou L. Depletion of multidrug-resistant uropathogenic Escherichia coli BC1 by ebselen and silver ion. J Cell Mol Med 2020; 24:13139-13150. [PMID: 32975381 PMCID: PMC7701569 DOI: 10.1111/jcmm.15920] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/19/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Ebselen, an organo‐selenium compound with well‐characterized toxicology and pharmacology, recently exhibited potent antibacterial activity against glutathione (GSH)‐negative bacteria by disrupting redox homeostasis. In this paper, we show that ebselen and silver ion in combination exert strong bactericidal activity against multidrug‐resistant (MDR) uropathogenic Escherichia coli (UPEC) BC1, a model MDR GSH‐positive bacterium. The mechanisms were found to involve consumption of total intracellular GSH and inhibition of thioredoxin reductase activity, which was highly related to reactive oxygen species up‐regulation. Furthermore, the therapeutic efficacy of ebselen and silver ion against UPEC‐induced cystitis was assessed in a mouse model. Treatment with ebselen and silver ion significantly reduced bacterial loads, down‐regulated the expression levels of tumour necrosis factor‐α (TNF‐α) and interferon‐γ (IFN‐γ) on‐site and decreased white/red blood cell counts in mild cystitis model mice, which demonstrated the anti‐inflammatory property of these agents. In addition, ebselen and silver ion also exhibited significantly high protective ability (100%) against acute cystitis infections. These results together may lay the foundation for further analysis and development of ebselen and silver ion as antibacterial agents for treatment of MDR UPEC infections.
Collapse
Affiliation(s)
- Peng Wang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China.,The Institute of Infection and Inflammation, Medical College, China Three Gorges University, Yichang, Hubei, China.,Central Laboratory, The People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Jun Wang
- The Institute of Infection and Inflammation, Medical College, China Three Gorges University, Yichang, Hubei, China.,Central Laboratory, The People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Zonglan Xie
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Jingxuan Zhou
- The Institute of Infection and Inflammation, Medical College, China Three Gorges University, Yichang, Hubei, China.,Central Laboratory, The People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Qianqian Lu
- The Institute of Infection and Inflammation, Medical College, China Three Gorges University, Yichang, Hubei, China.,Central Laboratory, The People's Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Ying Zhao
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Chuanjiang Dong
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China
| | - Lili Zou
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei, China.,The Institute of Infection and Inflammation, Medical College, China Three Gorges University, Yichang, Hubei, China.,Central Laboratory, The People's Hospital of China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
24
|
Desvaux M, Dalmasso G, Beyrouthy R, Barnich N, Delmas J, Bonnet R. Pathogenicity Factors of Genomic Islands in Intestinal and Extraintestinal Escherichia coli. Front Microbiol 2020; 11:2065. [PMID: 33101219 PMCID: PMC7545054 DOI: 10.3389/fmicb.2020.02065] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022] Open
Abstract
Escherichia coli is a versatile bacterial species that includes both harmless commensal strains and pathogenic strains found in the gastrointestinal tract in humans and warm-blooded animals. The growing amount of DNA sequence information generated in the era of "genomics" has helped to increase our understanding of the factors and mechanisms involved in the diversification of this bacterial species. The pathogenic side of E. coli that is afforded through horizontal transfers of genes encoding virulence factors enables this bacterium to become a highly diverse and adapted pathogen that is responsible for intestinal or extraintestinal diseases in humans and animals. Many of the accessory genes acquired by horizontal transfers form syntenic blocks and are recognized as genomic islands (GIs). These genomic regions contribute to the rapid evolution, diversification and adaptation of E. coli variants because they are frequently subject to rearrangements, excision and transfer, as well as to further acquisition of additional DNA. Here, we review a subgroup of GIs from E. coli termed pathogenicity islands (PAIs), a concept defined in the late 1980s by Jörg Hacker and colleagues in Werner Goebel's group at the University of Würzburg, Würzburg, Germany. As with other GIs, the PAIs comprise large genomic regions that differ from the rest of the genome by their G + C content, by their typical insertion within transfer RNA genes, and by their harboring of direct repeats (at their ends), integrase determinants, or other mobility loci. The hallmark of PAIs is their contribution to the emergence of virulent bacteria and to the development of intestinal and extraintestinal diseases. This review summarizes the current knowledge on the structure and functional features of PAIs, on PAI-encoded E. coli pathogenicity factors and on the role of PAIs in host-pathogen interactions.
Collapse
Affiliation(s)
- Mickaël Desvaux
- Université Clermont Auvergne, INRAE, MEDiS, Clermont-Ferrand, France
| | - Guillaume Dalmasso
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Racha Beyrouthy
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Nicolas Barnich
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Julien Delmas
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Richard Bonnet
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
25
|
|
26
|
The pathogenesis and management of renal scarring in children with vesicoureteric reflux and pyelonephritis. Pediatr Nephrol 2020; 35:349-357. [PMID: 30847554 DOI: 10.1007/s00467-018-4187-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 10/27/2022]
Abstract
Bacterial urinary tract infections (UTIs) are one of the most common reasons for children to be admitted to hospital. Bacteria infect and invade the bladder (the lower urinary tract) and if the infection disseminates to the upper urinary tract, significant inflammation in the kidneys may arise. Inflammation is a double-edged sword: it is needed to clear bacteria, but if excessive, kidney tissue is injured. During injury, nephrons are destroyed and replaced with deposition of extracellular matrix and a renal scar. In this review, we explore the pathogenesis of UTIs and discuss the risk factors that result in dissemination of bladder infection to the kidneys. Three major risk factors predispose to kidney infections: the presence of vesicoureteric reflux, the presence of bladder and bowel dysfunction, and defects in the ability of the host immune response to clear bacteria. In this review, we will discuss these factors, their relationship to renal scarring, and potential treatments that might be beneficial to prevent renal scar formation in children.
Collapse
|
27
|
Simões E Silva AC, Oliveira EA, Mak RH. Urinary tract infection in pediatrics: an overview. J Pediatr (Rio J) 2020; 96 Suppl 1:65-79. [PMID: 31783012 PMCID: PMC9432043 DOI: 10.1016/j.jped.2019.10.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/12/2019] [Accepted: 10/16/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE This review aimed to provide a critical overview on the pathogenesis, clinical findings, diagnosis, imaging investigation, treatment, chemoprophylaxis, and complications of urinary tract infection in pediatric patients. SOURCE OF DATA Data were obtained independently by two authors, who carried out a comprehensive and non-systematic search in public databases. SUMMARY OF FINDINGS Urinary tract infection is the most common bacterial infection in children. Urinary tract infection in pediatric patients can be the early clinical manifestation of congenital anomalies of the kidney and urinary tract (CAKUT) or be related to bladder dysfunctions. E. coli is responsible for 80-90% of community-acquired acute pyelonephritis episodes, especially in children. Bacterial virulence factors and the innate host immune systems may contribute to the occurrence and severity of urinary tract infection. The clinical presentation of urinary tract infections in children is highly heterogeneous, with symptoms that can be quite obscure. Urine culture is still the gold standard for diagnosing urinary tract infection and methods of urine collection in individual centers should be determined based on the accuracy of voided specimens. The debate on the ideal imaging protocol is still ongoing and there is tendency of less use of prophylaxis. Alternative measures and management of risk factors for recurrent urinary tract infection should be emphasized. However, in selected patients, prophylaxis can protect from recurrent urinary tract infection and long-term consequences. According to population-based studies, hypertension and chronic kidney disease are rarely associated with urinary tract infection. CONCLUSION Many aspects regarding urinary tract infection in children are still matters of debate, especially imaging investigation and indication of antibiotic prophylaxis. Further longitudinal studies are needed to establish tailored approach of urinary tract infection in childhood.
Collapse
Affiliation(s)
- Ana Cristina Simões E Silva
- Universidade Federal de Minas Gerais (UFMG), Faculdade de Medicina, Laboratório Interdisciplinar de Investigação Médica, Departamento de Pediatria, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG, Brazil.
| | - Eduardo A Oliveira
- Universidade Federal de Minas Gerais (UFMG), Faculdade de Medicina, Laboratório Interdisciplinar de Investigação Médica, Departamento de Pediatria, Unidade de Nefrologia Pediátrica, Belo Horizonte, MG, Brazil
| | - Robert H Mak
- University of California, Rady Children's Hospital San Diego, Division of Pediatric Nephrology, San Diego, United States
| |
Collapse
|
28
|
Luna-Pineda VM, Moreno-Fierros L, Cázares-Domínguez V, Ilhuicatzi-Alvarado D, Ochoa SA, Cruz-Córdova A, Valencia-Mayoral P, Rodríguez-Leviz A, Xicohtencatl-Cortes J. Curli of Uropathogenic Escherichia coli Enhance Urinary Tract Colonization as a Fitness Factor. Front Microbiol 2019; 10:2063. [PMID: 31551993 PMCID: PMC6733918 DOI: 10.3389/fmicb.2019.02063] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/21/2019] [Indexed: 01/24/2023] Open
Abstract
Curli, a type of fimbriae widely distributed in uropathogenic Escherichia coli (UPEC), are involved in adhesion to human bladder cell surfaces and biofilm development. The role of UPEC curli was evaluated in a murine model of urinary tract infection. The aim of this study was to establish the role of curli in C57BL/6 mice transurethrally infected with curli-producing and non-curli-producing UPEC strains. We confirmed that curli enhanced UPEC colonization in the urinary tract, resulting in damage to both the bladder and kidney. Intranasal immunization with recombinant CsgA protein protected against colonization by curli-producing UPEC in the urinary tract. Quantification of cytokines from urinary tract organs showed increases in interleukin-6 and tumor necrosis factor (TNF) release in the kidneys 48 h postinfection with curli-producing UPEC. By contrast, mice infected with non-curli-producing UPEC showed the highest release of interleukin-6, -10, and -17A and TNF. Curli may obscure other fimbriae and LPS, preventing interactions with Toll-like receptors. When intranasal immunization with recombinant FimH and PapG proteins and subsequent infection with this strain were performed, cytokine quantification showed a decrease in the stimulation and release by the uroepithelium. Thus, curli are amyloid-like fimbriae that enhances colonization in the urinary tract and a possible fitness factor.
Collapse
Affiliation(s)
- Víctor M Luna-Pineda
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | - Leticia Moreno-Fierros
- Laboratorio de Inmunidad en Mucosas, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Vicenta Cázares-Domínguez
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | - Damaris Ilhuicatzi-Alvarado
- Laboratorio de Inmunidad en Mucosas, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Sara A Ochoa
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | - Ariadnna Cruz-Córdova
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | - Pedro Valencia-Mayoral
- Departamento de Patología, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| | | | - Juan Xicohtencatl-Cortes
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de México "Federico Gómez", Mexico City, Mexico
| |
Collapse
|
29
|
Engelsöy U, Rangel I, Demirel I. Impact of Proinflammatory Cytokines on the Virulence of Uropathogenic Escherichia coli. Front Microbiol 2019; 10:1051. [PMID: 31143172 PMCID: PMC6520761 DOI: 10.3389/fmicb.2019.01051] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/26/2019] [Indexed: 12/17/2022] Open
Abstract
The effect of a urinary tract infection on the host is a well-studied research field. However, how the host immune response affects uropathogenic Escherichia coli (CFT073) virulence is less studied. The aim of the present study was to investigate the impact of proinflammatory cytokine exposure on the virulence of uropathogenic Escherichia coli. We found that all tested proinflammatory cytokines (TNF-α, IL-1β, IL-6, IL-8 and IFN-γ) induced an increased CFT073 growth. We also found that biofilm formation and hemolytic activity was reduced in the presence of all proinflammatory cytokines. However, a reduction in siderophore release was only observed in the presence of IL-1β, IL-6 and IL-8. Real time-qPCR showed that all proinflammatory cytokines except TNF-α significantly increased genes associated with the iron acquisition system in CFT073. We also found that the proinflammatory cytokines induced significant changes in type-1 fimbriae, P-fimbriae and gluconeogenetic genes. Furthermore, we also showed, using a Caenorhabditis elegans (C. elegans) killing assay that all cytokines decreased the survival of C. elegans worms significantly. Taken together, our findings show that proinflammatory cytokines have the ability to alter the virulence traits of UPEC.
Collapse
Affiliation(s)
- Ulrik Engelsöy
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Ignacio Rangel
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Nutrition-Gut-Brain Interactions Research Centre, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Örebro University, Örebro, Sweden.,iRiSC - Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
30
|
Ben-David H, Livneh A, Lidar M, Feld O, Haj Yahia S, Grossman C, Ben-Zvi I. Toll-like receptor 2 is overexpressed in Familial Mediterranean fever patients and is inhibited by colchicine treatment. Best Pract Res Clin Rheumatol 2019; 32:651-661. [PMID: 31203923 DOI: 10.1016/j.berh.2019.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIM To study the role of Toll-like receptor (TLR) 2 in Familial Mediterranean fever (FMF) inflammatory process. METHODS TLR2 expression on monocytes of FMF attack-free patients (n = 20) and the effect of sera of FMF patients with an acute attack (n = 9) on TLR2 expression on monocytes of healthy donors were studied by flow cytometry (FACS). TLR2 expression was also studied in THP-1 cells, and TLR2 downstream signaling was studied by ELISA for the secretion of IL-1β and pro-inflammatory cytokines or by western blotting to measure nuclear factor (NF)-κB. RESULTS FMF attack-free patients had increased CD14 + TLR2+ cell count as compared to healthy donors. High-dose colchicine treatment (≥2 mg/d) inhibited this increased expression in FMF patients. Colchicine in vitro also inhibited TLR2 expression on THP-1 cells. Sera from FMF patients with an acute attack induced TLR2 expression by both monocytes of healthy donors and THP-1 cells as well as pro-inflammatory cytokine secretion by healthy monocytes, while colchicine inhibited this induction. Pam2CSK4 increased interleukin-1β (IL-1β) secretion by peripheral blood mononuclear cells (PBMCs) of healthy donors, and this activation was inhibited by colchicine. THP-1 cells presented elevated NF-κB expression when cultured with Pam2CSK4, whereas colchicine inhibited this elevation. CONCLUSIONS TLR2 activation was upregulated in monocytes of FMF patients, and colchicine inhibited this upregulation both in -vitro and in -vivo. This indicates that elevated expression of TLR2 promotes the production of pro-inflammatory cytokines, which may contribute to uncontrolled inflammation in FMF.
Collapse
Affiliation(s)
- Hava Ben-David
- Heller Institute of Medical Research, Tel-Aviv University, Tel-Aviv, Israel; Department of Internal Medicine F, The Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Avi Livneh
- Heller Institute of Medical Research, Tel-Aviv University, Tel-Aviv, Israel; Rheumatology Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; Department of Internal Medicine F, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Merav Lidar
- Rheumatology Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Olga Feld
- Department of Internal Medicine F, The Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Souad Haj Yahia
- Department of Internal Medicine F, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Chagai Grossman
- Rheumatology Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; Department of Internal Medicine F, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ilan Ben-Zvi
- Heller Institute of Medical Research, Tel-Aviv University, Tel-Aviv, Israel; Rheumatology Unit, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; Department of Internal Medicine F, The Chaim Sheba Medical Center, Tel-Hashomer, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; The Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel.
| |
Collapse
|
31
|
Dahiya A, Goldman RD. [Not Available]. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2018; 64:e483-e485. [PMID: 30429192 PMCID: PMC6234933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Question Je connais la fréquence des infections des voies urinaires (IVU) chez les enfants, de même que leurs séquelles potentielles si elles ne sont pas traitées. Par conséquent, dans notre clinique, nous traitons par antibiothérapie tous les enfants souffrant d’IVU symptomatiques. Par ailleurs, devrions-nous en faire autant chez les enfants qui ont une bactériurie asymptomatique? Réponse La bactériurie asymptomatique (BUA) était habituellement traitée avec des antibiotiques dans toutes les populations, y compris les enfants. Par ailleurs, selon les données probantes plus récentes, l’antibiothérapie ne s’est pas révélée bénéfique et entraîne même souvent des préjudices dans le traitement de la BUA chez l’enfant. Certaines études font valoir qu’en raison de la microbiologie différente en cause dans la BUA, celle-ci ne devrait pas être considérée comme appartenant au spectre des IVU. Ces enfants ne devraient pas recevoir d’antibiothérapie à moins d’avoir subi une greffe de rein ou des interventions urologiques invasives.
Collapse
|
32
|
Dahiya A, Goldman RD. Management of asymptomatic bacteriuria in children. CANADIAN FAMILY PHYSICIAN MEDECIN DE FAMILLE CANADIEN 2018. [PMID: 30429177 DOI: 10.19538/j.ek2018110602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Question I am aware of how common pediatric urinary tract infection (UTI) is, and of the potential long-term sequelae if left untreated. Therefore, in our practice we treat every child who presents with symptomatic UTI with antibiotics. However, should the same practice be applied to children with bacteriuria that is asymptomatic?Answer Historically, asymptomatic bacteriuria (ABU) was treated with antibiotics in all populations, including in children. However, more recent evidence has shown no benefit and often harm associated with the use of antibiotics to treat pediatric ABU. Some studies suggest that owing to the different microbiology associated with ABU it should not be considered in the spectrum of UTI. These children should not be treated with antibiotics unless they have received a renal transplant or have undergone invasive urologic procedures.
Collapse
|
33
|
Andersen-Civil AIS, Ahmed S, Guerra PR, Andersen TE, Hounmanou YMG, Olsen JE, Herrero-Fresno A. The impact of inactivation of the purine biosynthesis genes, purN and purT, on growth and virulence in uropathogenic E. coli. Mol Biol Rep 2018; 45:2707-2716. [PMID: 30377949 DOI: 10.1007/s11033-018-4441-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022]
Abstract
De novo synthesis of purines has been suggested to be an important factor for the pathogenesis of uropathogenic E. coli (UPEC). We analyzed the role of the redundant purine biosynthesis genes purN and purT, responsible for the third step in the purine biosynthesis, during UPEC infection. Growth experiments in M9 (minimal media), MOPS (rich media), filtered urine, and human serum with E. coli UTI89 and ΔpurN, ΔpurT, and ΔpurN/T mutants revealed that UPEC relies on de novo purine synthesis for growth in minimal medium. Mutants in individual genes as well as the double mutant grew equally well as the wild type in urine, rich media, and serum. However, during competition for growth in urine, the wild type UTI89 strain significantly outcompeted the purine auxotrophic ΔpurN/T mutant from late exponential growth phase. Inactivation of purN and/or purT significantly affected UPEC invasion of human bladder cells, but not the intracellular survival. Cytotoxicity levels to bladder cells were also diminished when both purN and purT were deleted, while single gene mutants did not differ from the wild type. When infecting human macrophages, no differences were observed between UTI89 and mutants in uptake, survival or cytotoxicity. Finally, the lack of the pur-gene(s), whether analysed as single or double gene knock-out, did not affect recovery rates after in vivo infection in a mouse model of UTI. These findings suggest that de novo synthesis of purines might be required only when UPEC is fully deprived of nucleotides and when grown in competition with other microorganisms in urine.
Collapse
Affiliation(s)
- Audrey Inge Schytz Andersen-Civil
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Shahana Ahmed
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Priscila Regina Guerra
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Thomas Emil Andersen
- Research Unit of Clinical Microbiology, Odense University Hospital, Odense, Denmark
| | - Yaovi Mahuton Gildas Hounmanou
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - John Elmerdahl Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ana Herrero-Fresno
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
34
|
Lee JH, Jang SJ, Rhie S. Antinatriuretic phenomena seen in children with acute pyelonephritis may be related to the activation of intrarenal RAAS. Medicine (Baltimore) 2018; 97:e12152. [PMID: 30200111 PMCID: PMC6133464 DOI: 10.1097/md.0000000000012152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We investigated whether antinatriuretic phenomena [decreases in urinary sodium (uNa) and fractional excretion of sodium (FENa)] seen in children with acute pyelonephritis (APN) are associated with the renin-angiotensin-aldosterone system (RAAS).We examined 114 children experiencing their first episode of febrile urinary tract infection (fUTI) consecutively admitted to our hospital from July 2012 to June 2014. Blood tests [C-reactive protein, white blood cell count, erythrocyte sedimentation rate, and aldosterone (Aldo)] and urine tests [uNa, urine potassium (uK) and FENa] were performed upon admission. All enrolled children underwent a 99m-dimercaptosuccinic acid renal scanning (DMSA) at admission. Areas with cortical defects (AreaCD) and uptake counts (UptakeCD) on their DMSA scans were calculated. Data were compared between children with positive DMSA results (APN), lower urinary tract infection (L-UTI), and controls; and between children with high and low Aldo levels.uNa, uNa/K, and FENa negatively correlated with AreaCD%, UptakeCD, and Aldo; were significantly lower in APN patients than in LUTIs and controls regardless of Aldo level; were lower in the high Aldo group than in the low Aldo group. However, there is no difference in AreaCD% and UptakeCD between APN children with the high and low Aldo level.Decreases in uNa, uNa/K, and FENa in children with APN may result from an antinatriuretic effect of RAAS and be related to the activation of the intrarenal RAAS.
Collapse
Affiliation(s)
| | - Su Jin Jang
- Department of Nuclear Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | | |
Collapse
|
35
|
Abstract
Urinary tract infection (UTI) is one of the most common bacterial infections in humans, and the majority are caused by uropathogenic Escherichia coli (UPEC). The rising antibiotic resistance among UPEC and the frequent failure of antibiotics to effectively treat recurrent UTI and catheter-associated UTI motivate research on alternative ways of managing UTI. Abundant evidence indicates that the toxic radical nitric oxide (NO), formed by activation of the inducible nitric oxide synthase, plays an important role in host defence to bacterial infections, including UTI. The major source of NO production during UTI is from inflammatory cells, especially neutrophils, and from the uroepithelial cells that are known to orchestrate the innate immune response during UTI. NO and reactive nitrogen species have a wide range of antibacterial targets, including DNA, heme proteins, iron-sulfur clusters, and protein thiol groups. However, UPEC have acquired a variety of defence mechanisms for protection against NO, such as the NO-detoxifying enzyme flavohemoglobin and the NO-tolerant cytochrome bd-I respiratory oxidase. The cytotoxicity of NO-derived intermediates is nonspecific and may be detrimental to host cells, and a balanced NO production is crucial to maintain the tissue integrity of the urinary tract. In this review, we will give an overview of how NO production from host cells in the urinary tract is activated and regulated, the effect of NO on UPEC growth and colonization, and the ability of UPEC to protect themselves against NO. We also discuss the attempts that have been made to develop NO-based therapeutics for UTI treatment.
Collapse
|
36
|
Effects of Periostracum Cicadae on Cytokines and Apoptosis Regulatory Proteins in an IgA Nephropathy Rat Model. Int J Mol Sci 2018; 19:ijms19061599. [PMID: 29844269 PMCID: PMC6032130 DOI: 10.3390/ijms19061599] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/04/2018] [Accepted: 05/22/2018] [Indexed: 11/20/2022] Open
Abstract
Periostracum cicadae, the cast-off shell of the cicada Cryptotympana pustulata Fabricius, is used in traditional Chinese medicine for its diaphoretic, anticonvulsive, sedative, antipyretic, and antiallergic effects. However, the exact pathogenesis of immunoglobulin A nephropathy (IgAN) remains unclear, thereby hindering investigations to identify novel therapeutic agents. A rat IgAN model was established by administration of bovine serum albumin, lipopolysaccharide, and carbon tetrachloride, which simultaneously established blood stasis and a heat syndrome model. The animals were sacrificed to detect changes in protein levels in urine and blood. Immunofluorescence was performed to assess IgA deposition in the glomeruli. Tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin 6 (IL-6) levels were measured in bronchoalveolar lavage fluid (BALF) by enzyme-linked immunosorbent assay. Hematology and eosin, periodic acid-Schiff, TUNEL (TdT-mediated dUTP Nick-End Labeling), and immunohistochemical staining were performed to evaluate histopathological changes in kidney tissues. Additionally, target-related proteins were measured by Western blotting. Periostracum cicadae resulted in a reduction in blood and urine protein levels. Serum TNF-α, IL-1β, and IL-6 levels significantly decreased in the periostracum cicadae-treated groups compared to the IgAN group. Furthermore, a reduction in MCP-1 (Monocyte Chemotactic Protein-1), TLR4 ((Toll-Like Receptor 4)), and IgA expression levels and a dose-dependent increase in caspase 3 expression were observed in response to periostracum cicadae treatment. TGF-β1(Transforming Growth Factor-β) levels decreased, whereas that of Fas increased in the kidney tissues of the periostracum cicadae-treated groups. The findings of the present study indicate that periostracum cicadae induces apoptosis and improves kidney inflammation and fibrosis in IgA nephropathy rat models.
Collapse
|
37
|
Mitochondrial DNA induces Foley catheter related bladder inflammation via Toll-like receptor 9 activation. Sci Rep 2018; 8:6377. [PMID: 29686303 PMCID: PMC5913242 DOI: 10.1038/s41598-018-24818-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
Bladder instrumentation engages the innate immune system via neutrophil activation, promoting inflammation and pain. Elevated levels of mitochondrial DNA (mtDNA) have been associated with tissue damage and organ dysfunction. We hypothesized that local bladder trauma induced by a Foley catheter (FC) will result in mtDNA release, migration of neutrophils into the bladder lumen, and activation of the Toll-like receptor 9 (TLR9) and nuclear factor kappa B (NF-κB) pathway leading to bladder tissue damage. We randomized 10 swine into two groups receiving uncoated, or chloroquine/N-Acetylcysteine (CQ/NAC)-coated FCs. Urine samples were analyzed for mtDNA activation of TLR9/NF-κB as demonstrated by indicators of neutrophil adhesion, migration, and activation. We found that uncoated FCs resulted in a unique active neutrophil phenotype that correlated with bladder epithelial injury, neutrophilia, necrosis, mtDNA release, TLR9/NF-κB activation, transcription and secretion of pro-inflammatory cytokines, and enhanced respiratory burst. In our study we observed that the high levels of mtDNA and elevated TLR9/NF-κB activity were ameliorated in the CQ/NAC-coated FC group. These findings suggest that post-migrated bladder luminal neutrophils are involved in local tissue damage and amelioration of the mtDNA/TLR9/NF-κB inflammatory axis may represent a therapeutic target to prevent inflammation, and bladder tissue injury.
Collapse
|
38
|
Ching CB, Gupta S, Li B, Cortado H, Mayne N, Jackson AR, McHugh KM, Becknell B. Interleukin-6/Stat3 signaling has an essential role in the host antimicrobial response to urinary tract infection. Kidney Int 2018; 93:1320-1329. [PMID: 29475562 DOI: 10.1016/j.kint.2017.12.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 12/09/2017] [Accepted: 12/14/2017] [Indexed: 01/03/2023]
Abstract
The signaling networks regulating antimicrobial activity during urinary tract infection (UTI) are incompletely understood. Interleukin-6 (IL-6) levels increase with UTI severity, but the specific contributions of IL-6 to host immunity against bacterial uropathogens are unknown. To clarify this we tested whether IL-6 activates the Stat3 transcription factor, to drive a program of antimicrobial peptide gene expression in infected urothelium during UTI. Transurethral inoculation of uropathogenic Escherichia coli led to IL-6 secretion, urothelial Stat3 phosphorylation, and activation of antimicrobial peptide transcription, in a Toll-like receptor 4-dependent manner in a murine model of cystitis. Recombinant IL-6 elicited Stat3 phosphorylation in primary urothelial cells in vitro, and systemic IL-6 administration promoted urothelial Stat3 phosphorylation and antimicrobial peptide expression in vivo. IL-6 deficiency led to decreased urothelial Stat3 phosphorylation and antimicrobial peptide mRNA expression following UTI, a finding mirrored by conditional Stat3 deletion. Deficiency in IL-6 or Stat3 was associated with increased formation of intracellular bacterial communities, and exogenous IL-6 reversed this phenotype in IL-6 knockout mice. Moreover, chronic IL-6 depletion led to increased renal bacterial burden and severe pyelonephritis in C3H/HeOuJ mice. Thus, IL-6/Stat3 signaling drives a transcriptional program of antimicrobial gene expression in infected urothelium, with key roles in limiting epithelial invasion and ascending infection.
Collapse
Affiliation(s)
- Christina B Ching
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA; Department of Surgery, Division of Pediatric Urology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sudipti Gupta
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA
| | - Birong Li
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA
| | - Hanna Cortado
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA
| | - Nicholas Mayne
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA
| | - Ashley R Jackson
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA
| | - Kirk M McHugh
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA; Department of Anatomy, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Brian Becknell
- Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio, USA; Nephrology Section, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA.
| |
Collapse
|
39
|
Sessile Innate Immune Cells. DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7123606 DOI: 10.1007/978-3-319-78655-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this chapter, sessile cells of the innate immune system are briefly introduced. Defined as cells equipped with diverse pattern recognition molecules capable of detecting MAMPs and DAMPs, they encompass cells such as epithelial cells, fibroblasts, vascular cells, chondrocytes, osteoblasts, and adipocytes. Located at the body surfaces, epithelial cells represent the first line of innate immune defense against invading microbial pathogens. They are significant contributors to innate mucosal immunity and generate various antimicrobial defense mechanisms. Also, epithelial cells critically contribute to tissue repair via the phenomenon of re-epithelialization. Fibroblasts operate as classical sentinel cells of the innate immune system dedicated to responding to MAMPs and DAMPs emitted upon any tissue injury. Typically, fibroblasts synthesize most of the extracellular matrix of connective tissues, thereby playing a crucial role in tissue repair processes. Vascular cells of the innate immune system represent an evolutionarily developed first-line defense against any inciting insult hitting the vessel walls from the luminal side including bacteria, viruses, microbial toxins, and chemical noxa such as nicotine. Upon such insults and following recognition of MAMPs and DAMPs, vascular cells react with an innate immune response to create an acute inflammatory milieu in the vessel wall aimed at curing the vascular injury concerned. Chondrocytes, osteoblasts, and osteoclasts represent other vital cells of the skeletal system acting as cells of the innate immune system in its wider sense. These cells mediate injury-promoted DAMP-induced inflammatory and regenerative processes specific for the skeletal systems. Finally, adipocytes are regarded as highly active cells of the innate immune system. As white, brown, and beige adipocytes, they operate as a dynamic metabolic organ that can secrete certain bioactive molecules which have endocrine, paracrine, and autocrine actions.
Collapse
|
40
|
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
|
41
|
Quintar AA, Maldonado CA. Androgen regulation of host defenses and response to inflammatory stimuli in the prostate gland. Cell Biol Int 2017; 41:1223-1233. [PMID: 28244686 DOI: 10.1002/cbin.10755] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/24/2017] [Indexed: 12/19/2022]
Abstract
The prostate gland is a strictly androgen-dependent organ which is also the main target of infectious and inflammatory diseases in the male reproductive tract. Host defenses and immunity of the gland have unique features to maintain a constant balance between response and tolerance to diverse antigens. In this context, the effects of reproductive hormones on the male tract are thus complex and have just started to be defined. From the classical description of "the prostatic antibacterial factor," many host defense proteins with potent microbicidal and anti-tumoral activities have been described in the organ. Indeed, it has been proposed a central role for resident cells, that is, epithelial and smooth muscle cells, in the prostatic response against injuries. However, these cells also represent the target of the inflammatory damage, leading to the development of a Proliferative Inflammatory Atrophy-like process in the epithelium and a myofibroblastic-like reactive stroma. Available data on androgen regulation of inflammation led to a model of the complex control, in which the final effect will depend on the tissue microenvironment, the cause of inflammation, and the levels of androgens among other factors. In this paper, we review the current scientific literature about the inflammatory process in the gland, the modulation of host defense proteins, and the influence of testosterone on the resolution of prostatitis.
Collapse
Affiliation(s)
- Amado A Quintar
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Pabellón de Biología Celular. E. Barros esq. Enfermera Gordillo 1 piso Ciudad Universitaria, 5000, Córdoba, Argentina
| | - Cristina A Maldonado
- Centro de Microscopía Electrónica, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Pabellón de Biología Celular. E. Barros esq. Enfermera Gordillo 1 piso Ciudad Universitaria, 5000, Córdoba, Argentina
| |
Collapse
|
42
|
Anderberg SB, Luther T, Frithiof R. Physiological aspects of Toll-like receptor 4 activation in sepsis-induced acute kidney injury. Acta Physiol (Oxf) 2017; 219:573-588. [PMID: 27602552 PMCID: PMC5324638 DOI: 10.1111/apha.12798] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/08/2016] [Accepted: 09/05/2016] [Indexed: 12/21/2022]
Abstract
Sepsis‐induced acute kidney injury (SI‐AKI) is common and associated with high mortality. Survivors are at increased risk of chronic kidney disease. The precise mechanism underlying SI‐AKI is unknown, and no curative treatment exists. Toll‐like receptor 4 (TLR4) activates the innate immune system in response to exogenous microbial products. The result is an inflammatory reaction aimed at clearing a potential infection. However, the consequence may also be organ dysfunction as the immune response can cause collateral damage to host tissue. The purpose of this review is to describe the basis for how ligand binding to TLR4 has the potential to cause renal dysfunction and the mechanisms by which this may take place in gram‐negative sepsis. In addition, we highlight areas for future research that can further our knowledge of the pathogenesis of SI‐AKI in relation to TLR4 activation. TLR4 is expressed in the kidney. Activation of TLR4 causes cytokine and chemokine release as well as renal leucocyte infiltration. It also results in endothelial and tubular dysfunction in addition to altered renal metabolism and circulation. From a physiological standpoint, inhibiting TLR4 in large animal experimental SI‐AKI significantly improves renal function. Thus, current evidence indicates that TLR4 has the ability to mediate SI‐AKI by a number of mechanisms. The strong experimental evidence supporting a role of TLR4 in the pathogenesis of SI‐AKI in combination with the availability of pharmacological tools to target TLR4 warrants future human studies.
Collapse
Affiliation(s)
- S. B. Anderberg
- Department of Surgical Sciences; Section of Anesthesia & Intensive Care; Uppsala University; Uppsala Sweden
| | - T. Luther
- Department of Surgical Sciences; Section of Anesthesia & Intensive Care; Uppsala University; Uppsala Sweden
| | - R. Frithiof
- Department of Surgical Sciences; Section of Anesthesia & Intensive Care; Uppsala University; Uppsala Sweden
| |
Collapse
|
43
|
The Adherent/Invasive Escherichia coli Strain LF82 Invades and Persists in Human Prostate Cell Line RWPE-1, Activating a Strong Inflammatory Response. Infect Immun 2016; 84:3105-3113. [PMID: 27600504 PMCID: PMC5067744 DOI: 10.1128/iai.00438-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/30/2016] [Indexed: 01/26/2023] Open
Abstract
Adherent/invasive Escherichia coli (AIEC) strains have recently been receiving increased attention because they are more prevalent and persistent in the intestine of Crohn's disease (CD) patients than in healthy subjects. Since AIEC strains show a high percentage of similarity to extraintestinal pathogenic E. coli (ExPEC), neonatal meningitis-associated E. coli (NMEC), and uropathogenic E. coli (UPEC) strains, here we compared AIEC strain LF82 with a UPEC isolate (strain EC73) to assess whether LF82 would be able to infect prostate cells as an extraintestinal target. The virulence phenotypes of both strains were determined by using the RWPE-1 prostate cell line. The results obtained indicated that LF82 and EC73 are able to adhere to, invade, and survive within prostate epithelial cells. Invasion was confirmed by immunofluorescence and electron microscopy. Moreover, cytochalasin D and colchicine strongly inhibited bacterial uptake of both strains, indicating the involvement of actin microfilaments and microtubules in host cell invasion. Moreover, both strains belong to phylogenetic group B2 and are strong biofilm producers. In silico analysis reveals that LF82 shares with UPEC strains several virulence factors: namely, type 1 pili, the group II capsule, the vacuolating autotransporter toxin, four iron uptake systems, and the pathogenic island (PAI). Furthermore, compared to EC73, LF82 induces in RWPE-1 cells a marked increase of phosphorylation of mitogen-activated protein kinases (MAPKs) and of NF-κB already by 5 min postinfection, thus inducing a strong inflammatory response. Our in vitro data support the hypothesis that AIEC strains might play a role in prostatitis, and, by exploiting host-cell signaling pathways controlling the innate immune response, likely facilitate bacterial multiplication and dissemination within the male genitourinary tract.
Collapse
|
44
|
Giglio D, Wasén C, Mölne J, Suchy D, Swanpalmer J, Jabonero Valbuena J, Tobin G, Ny L. Downregulation of toll-like receptor 4 and IL-6 following irradiation of the rat urinary bladder. Clin Exp Pharmacol Physiol 2016; 43:698-705. [DOI: 10.1111/1440-1681.12583] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/15/2016] [Accepted: 04/24/2016] [Indexed: 11/26/2022]
Affiliation(s)
- D Giglio
- Department of Pharmacology; University of Gothenburg; Gothenburg Sweden
- Department of Oncology; University of Gothenburg; Gothenburg Sweden
| | - C Wasén
- Department of Rheumatology and Inflammation Research; University of Gothenburg; Gothenburg Sweden
| | - J Mölne
- Department of Pathology; University of Gothenburg; Gothenburg Sweden
| | - D Suchy
- Department of Pharmacology; University of Gothenburg; Gothenburg Sweden
| | - J Swanpalmer
- Department of Radiation Physics; The Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
| | | | - G Tobin
- Department of Pharmacology; University of Gothenburg; Gothenburg Sweden
| | - L Ny
- Department of Oncology; University of Gothenburg; Gothenburg Sweden
| |
Collapse
|
45
|
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
|
46
|
Olson PD, Hunstad DA. Subversion of Host Innate Immunity by Uropathogenic Escherichia coli. Pathogens 2016; 5:E2. [PMID: 26742078 PMCID: PMC4810123 DOI: 10.3390/pathogens5010002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/03/2015] [Accepted: 12/29/2015] [Indexed: 01/09/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) cause the majority of community-onset urinary tract infections (UTI) and represent a major etiologic agent of healthcare-associated UTI. Introduction of UPEC into the mammalian urinary tract evokes a well-described inflammatory response, comprising pro-inflammatory cytokines and chemokines as well as cellular elements (neutrophils and macrophages). In human UTI, this inflammatory response contributes to symptomatology and provides means for diagnosis by standard clinical testing. Early in acute cystitis, as demonstrated in murine models, UPEC gains access to an intracellular niche that protects a population of replicating bacteria from arriving phagocytes. To ensure the establishment of this protected niche, UPEC employ multiple strategies to attenuate and delay the initiation of host inflammatory components, including epithelial secretion of chemoattractants. Recent work has also revealed novel mechanisms by which UPEC blunts neutrophil migration across infected uroepithelium. Taken together, these attributes distinguish UPEC from commensal and nonpathogenic E. coli strains. This review highlights the unique immune evasion and suppression strategies of this bacterial pathogen and offers directions for further study; molecular understanding of these mechanisms will inform the development of adjunctive, anti-virulence therapeutics for UTI.
Collapse
Affiliation(s)
- Patrick D Olson
- Medical Scientist Training Program, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO 63110, USA.
| | - David A Hunstad
- Departments of Pediatrics and Molecular Microbiology, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO 63110, USA.
| |
Collapse
|
47
|
Abstract
Urinary tract infections (UTI) are among the most common bacterial infections of humans. The mouse provides an excellent and tractable model system for cystitis and pyelonephritis caused by Escherichia coli and other uropathogens. Using a well-established model of experimental cystitis in which the bladders of female mice are infected via transurethral catheterization, the molecular details of the pathogenesis of bacterial cystitis have been substantially illuminated in the last decade. Uropathogenic E. coli attach to bladder epithelium (both in human and mouse) via adhesive type 1 pili, establish a replicative niche within epithelial cell cytoplasm, and form intracellular bacterial communities that are protected from antibiotic effects and immune clearance. The use of different inbred and mutant mouse strains offers the opportunity to study outcomes of infection, including resolution, formation of quiescent intracellular bacterial reservoirs, chronic bacterial cystitis, and recurrent infections. Urine, bladder, and kidney tissues can be analyzed by bacterial culture, histology, immunohistochemistry, immunofluorescent and confocal microscopy, electron microscopy, and flow cytometry, while a broad array of soluble markers (e.g., cytokines) can also be profiled in serum, urine, and tissue homogenates by ELISA, Western blotting, multiplex bead array, and other approaches. This model promises to afford continued opportunity for discovery of pathogenic mechanisms and evaluation of therapeutic and preventive strategies for acute, chronic, and recurrent UTI.
Collapse
Affiliation(s)
- Thomas J Hannan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Hunstad
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, 8208, St. Louis, MO, 63110, USA. .,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
48
|
Li D, Zhang X, Chen B. SIGIRR participates in negative regulation of LPS response and tolerance in human bladder epithelial cells. BMC Immunol 2015; 16:73. [PMID: 26634342 PMCID: PMC4669620 DOI: 10.1186/s12865-015-0137-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/27/2015] [Indexed: 12/25/2022] Open
Abstract
Background The innate immune response of urinary tract is critically important in the defense to microbial attack. Toll-like receptor 4 (TLR4) controls initial mucosal response to uropathogenic Escherichia coli (UPEC). However, excessive and dysfunctional TLR signaling may result in severe inflammation and inappropriate tissue damage. Previous studies have demonstrated that single immunoglobulin IL-1R-related receptor/Toll IL-1 receptor 8 (SIGIRR/TIR8) is a member of the toll-interleukin-1 receptor (TIR) family that can negatively modulate TLR4 mediated signaling, but its role in the innate immunity of urinary tract infection remains incompletely defined. In this study, we investigated its cellular distribution and mechanisms involved within the human bladder epithelial cells after LPS stimulation. Results Immunostaining, reverse transcription PCR and Western blot results showed that SIGIRR was constitutively expressed in the human bladder epithelial cell lines and was downregulated after LPS stimulation. To further define the role of SIGIRR, cells were transiently transfected with SIGIRR siRNA and stimulated with LPS. SIGIRR gene silencing augmented chemokine expression in response to LPS, as indicated by increased levels of IL-6 and IL-8 secretions in the supernatants compared with negative control siRNA. Furthermore, LPS tolerance, a protective mechanism against second LPS stimulation, was significantly reduced in SIGIRR siRNA transfected cells. Moreover, transient gene silencing augmented LPS-induced NF-κB and MAPK activation. Conclusions In conclusion, our results suggest that SIGIRR plays an important role in the negative regulation of LPS response and tolerance in human bladder epithelial cells, possibly through its impact on TLR-mediated signaling.
Collapse
Affiliation(s)
- Dan Li
- Department of Infectious Diseases, the First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| | - Xin Zhang
- Department of Infectious Diseases, the First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| | - Baiyi Chen
- Department of Infectious Diseases, the First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
49
|
Abstract
Pyelonephritis represents a subset of urinary tract infections that occur from bacteria ascending from the lower to the upper reaches of the genitourinary system, such as the kidney. The renal system contains a range of hydrodynamically and immunologically challenging, interconnected microenvironments where the invading pathogen may populate during the course of the infection. The situation at the infection foci changes dynamically, vacillating between bacterial colonization and clearance, to which the outcome is a summation of all host-pathogen elements in play. A selection of important determinants includes factors of microbial origin, effects of eukaryotic cell signaling, physiological facets of the infected organ, and signals from distal organs. Improved understanding of the multifactorial aspects of molecular pathogenesis of infection requires intravital, cross-disciplinary approaches with high spatio-temporal resolution. The advancement of such approaches promises to eventually provide a comprehensive understanding of the integrated pathophysiology of pyelonephritis.
Collapse
|
50
|
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
|