1
|
Chulkina M, Tran H, Uribe G, McAninch SB, McAninch C, Seideneck A, He B, Lanza M, Khanipov K, Golovko G, Powell DW, Davenport ER, Pinchuk IV. MyD88-mediated signaling in intestinal fibroblasts regulates macrophage antimicrobial defense and prevents dysbiosis in the gut. Cell Rep 2025; 44:115553. [PMID: 40257864 DOI: 10.1016/j.celrep.2025.115553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 02/03/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
Fibroblasts that reside in the gut mucosa are among the key regulators of innate immune cells, but their role in the regulation of the defense functions of macrophages remains unknown. MyD88 is suggested to shape fibroblast responses in the intestinal microenvironment. We found that mice lacking MyD88 in fibroblasts showed a decrease in the colonic antimicrobial defense, developing dysbiosis and aggravated dextran sulfate sodium (DSS)-induced colitis. These pathological changes were associated with the accumulation of Arginase 1+ macrophages with low antimicrobial defense capability. Mechanistically, the production of interleukin (IL)-6 and CCL2 downstream of MyD88 was critically involved in fibroblast-mediated support of macrophage antimicrobial function, and IL-6/CCL2 neutralization resulted in the generation of macrophages with decreased production of the antimicrobial peptide cathelicidin and impaired bacterial clearance. Collectively, these findings revealed a critical role of fibroblast-intrinsic MyD88 signaling in regulating macrophage antimicrobial defense under colonic homeostasis, and its disruption results in dysbiosis, predisposing the host to the development of intestinal inflammation.
Collapse
Affiliation(s)
- Marina Chulkina
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA
| | - Hanh Tran
- The Pennsylvania State University, Department of Biology, Huck Institute of the Life Sciences, University Park, PA, USA
| | - Gabriela Uribe
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA
| | - Steven Bruce McAninch
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA
| | - Christina McAninch
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA
| | - Ashley Seideneck
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA
| | - Bing He
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA
| | - Matthew Lanza
- The Pennsylvania State University, College of Medicine, Department of Comparative Medicine, Hershey, PA, USA
| | - Kamil Khanipov
- The University of Texas Medical Branch, Department of Pharmacology, Galveston, TX, USA
| | - Georgiy Golovko
- The University of Texas Medical Branch, Department of Pharmacology, Galveston, TX, USA
| | - Don W Powell
- The University of Texas Medical Branch, Department of Internal Medicine, Galveston, TX, USA
| | - Emily R Davenport
- The Pennsylvania State University, Department of Biology, Huck Institute of the Life Sciences, University Park, PA, USA
| | - Irina V Pinchuk
- The Pennsylvania State University, College of Medicine, Department of Medicine, Hershey, PA, USA.
| |
Collapse
|
2
|
Ramaiah KB, Suresh I, Nesakumar N, Sai Subramanian N, Rayappan JBB. "Urinary tract infection: Conventional testing to developing Technologies". Clin Chim Acta 2025; 565:119979. [PMID: 39341530 DOI: 10.1016/j.cca.2024.119979] [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: 08/26/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Urinary tract infections (UTIs) present an escalating global health concern, precipitating increased hospitalizations and antibiotic utilization, thereby fostering the emergence of antimicrobial resistance. Current diagnostic modalities exhibit protracted timelines and substantial financial burdens, necessitating specialized infrastructures. Addressing these impediments mandates the development of a precise diagnostic paradigm to expedite identification and augment antibiotic stewardship. The application of biosensors, recognized for their transformative efficacy, emerges as a promising resolution. Recent strides in biosensor technologies have introduced pioneering methodologies, yielding pertinent biosensors and integrated systems with significant implications for point-of-care applications. This review delves into historical perspectives, furnishing a comprehensive delineation of advancements in UTI diagnostics, disease etiology, and biomarkers, underscoring the potential merits of these innovations for optimizing patient care.
Collapse
Affiliation(s)
- Kavi Bharathi Ramaiah
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India; Biofilm Biology Lab & Antimicrobial Resistance Lab, Centre for Research in Infectious Diseases, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Indhu Suresh
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India; School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India
| | - Noel Nesakumar
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India; School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - N Sai Subramanian
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India; Biofilm Biology Lab & Antimicrobial Resistance Lab, Centre for Research in Infectious Diseases, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India.
| | - John Bosco Balaguru Rayappan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India; School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613401, Tamil Nadu, India.
| |
Collapse
|
3
|
Sun J, Cheng K, Xie Y. Urinary Tract Infections Detection with Molecular Biomarkers. Biomolecules 2024; 14:1540. [PMID: 39766247 PMCID: PMC11673847 DOI: 10.3390/biom14121540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Urinary tract infection (UTI) is the most prevalent kind of pathogenic bacteria infection, and the midstream urine culture is regarded as the gold standard in UTI diagnosis. Recently, even with modern media and techniques such as polymerase chain reaction (PCR), urinary cultures still create a considerable workload for hospital laboratories. Other UTI-detecting methods, such as flow cytometry and lateral flow immunoassay, suffer from various drawbacks like long time consumption and low sensitivity. Therefore, looking for reliable biomarkers in UTI is urgently needed. In this review, the current definitions of UTI can be basically divided into two main categories: uncomplicated UTI and complicated UTI. In light of anatomical sites, it can be classified as either lower UTI or upper UTI. We take the classification of UTI as a clue and review the reported extensive literature to classify the existing studied markers into the following three categories: Biomarkers used clinically; Promising biomarkers; and Controversial biomarkers. Particularly, the nucleic acid-associated, metabolomic, and lipidomic biomarkers are highlighted. At the end, we discuss the challenges and prospects of biomarkers in UTI, hoping to further inspire the diagnosis of UTI.
Collapse
Affiliation(s)
- Jiayi Sun
- Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Kai Cheng
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
4
|
Sapkota A, Park EJ, Kim YJ, Heo JB, Nguyen TQ, Heo BE, Kim JK, Lee SH, Kim SI, Choi YJ, Roh T, Jeon SM, Jang M, Heo HJ, Whang J, Paik S, Yuk JM, Kim JM, Song GY, Jang J, Jo EK. The autophagy-targeting compound V46 enhances antimicrobial responses to Mycobacteroides abscessus by activating transcription factor EB. Biomed Pharmacother 2024; 179:117313. [PMID: 39167844 DOI: 10.1016/j.biopha.2024.117313] [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: 02/21/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Mycobacteroides abscessus (Mabc) is a rapidly growing nontuberculous mycobacterium that poses a considerable challenge as a multidrug-resistant pathogen causing chronic human infection. Effective therapeutics that enhance protective immune responses to Mabc are urgently needed. This study introduces trans-3,5,4'-trimethoxystilbene (V46), a novel resveratrol analogue with autophagy-activating properties and antimicrobial activity against Mabc infection, including multidrug-resistant strains. Among the resveratrol analogues tested, V46 significantly inhibited the growth of both rough and smooth Mabc strains, including multidrug-resistant strains, in macrophages and in the lungs of mice infected with Mabc. Additionally, V46 substantially reduced Mabc-induced levels of pro-inflammatory cytokines and chemokines in both macrophages and during in vivo infection. Mechanistic analysis showed that V46 suppressed the activation of the protein kinase B/Akt-mammalian target of rapamycin signaling pathway and enhanced adenosine monophosphate-activated protein kinase signaling in Mabc-infected cells. Notably, V46 activated autophagy and the nuclear translocation of transcription factor EB, which is crucial for antimicrobial host defenses against Mabc. Furthermore, V46 upregulated genes associated with autophagy and lysosomal biogenesis in Mabc-infected bone marrow-derived macrophages. The combination of V46 and rifabutin exerted a synergistic antimicrobial effect. These findings identify V46 as a candidate host-directed therapeutic for Mabc infection that activates autophagy and lysosomal function via transcription factor EB.
Collapse
Affiliation(s)
- Asmita Sapkota
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Eun-Jin Park
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Young Jae Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jong Beom Heo
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Thanh Quang Nguyen
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Bo Eun Heo
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University, School of Medicine, Daegu, South Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Chungbuk, South Korea
| | - Soo In Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yoon-Jung Choi
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Taylor Roh
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Sang Min Jeon
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Marnpyung Jang
- College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Hae Joon Heo
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | - Jake Whang
- Korea Mycobacterium Resource Center & Basic Research Section, The Korean Institute of Tuberculosis, Cheongju, South Korea
| | - Seungwha Paik
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jae-Min Yuk
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Infection Biology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jin-Man Kim
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Pathology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Gyu Yong Song
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; College of Pharmacy, Chungnam National University, Daejeon, South Korea.
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea.
| | - Eun-Kyeong Jo
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea; Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, South Korea; Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea; Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
5
|
Naskar M, Choi HW. A Dynamic Interplay of Innate Immune Responses During Urinary Tract Infection. Immune Netw 2024; 24:e31. [PMID: 39246616 PMCID: PMC11377947 DOI: 10.4110/in.2024.24.e31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024] Open
Abstract
Urinary tract infections (UTIs) represent one of the most prevalent bacterial infections globally, manifesting in diverse clinical phenotypes with varying degrees of severity and complications. The mechanisms underlying UTIs are gradually being elucidated, leading to an enhanced understanding of the immune responses involved. Innate immune cells play a crucial defensive role against uropathogenic bacteria through various mechanisms. Despite their significant contributions to host defense, these cells often fail to achieve complete clearance of uropathogens, necessitating the frequent prescription of antibiotics for UTI patients. However, the persistence of infections and related pathological symptoms in the absence of innate immune cells in animal models underscore the importance of innate immunity in UTIs. Therefore, the host protective functions of innate immune cells, including neutrophils, macrophages, mast cells, NK cells, innate lymphoid cells, and γδ T cells, are delicately coordinated and timely regulated by a variety of cytokines to ensure successful pathogen clearance.
Collapse
Affiliation(s)
- Manisha Naskar
- Division of Life Sciences, Korea University, Seoul 02841, Korea
| | - Hae Woong Choi
- Division of Life Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
6
|
Li T, Zhang Y, Dong R, Bi W, Wang S, Zeng K, Han L. Identification and mechanistic exploration of key anti-inflammatory molecules in American ginseng: Impacts on signal transducer and activator of transcription 3 STAT3 phosphorylation and macrophage polarization. Phytother Res 2024; 38:4307-4320. [PMID: 38973353 DOI: 10.1002/ptr.8277] [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: 03/28/2024] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024]
Abstract
American ginseng (AG) has been reported to have anti-inflammatory effects in many diseases, but the key molecules and mechanisms are unclear. This study aims to evaluate the anti-inflammatory mechanism of AG and identify the key molecules by in vivo and in vitro models. Zebrafish was employed to assess the anti-inflammatory properties of AG and the compounds. Metabolomics was utilized to identify potential anti-inflammatory molecules in AG, while molecular dynamics simulations were conducted to forecast the interaction capabilities of these compounds with inflammatory targets. Additionally, macrophage cell was employed to investigate the anti-inflammatory mechanisms of the key molecules in AG by enzyme-linked immunosorbent assay and western blotting. Seven potential anti-inflammatory molecules were discovered in AG, with ginsenoside Rg1, ginsenoside Rs3 (G-Rs3), and oleanolic acid exhibiting the strongest affinity for signal transducer and activator of transcription 3. These compounds demonstrated inhibitory effects on macrophage migration in zebrafish models and the ability to regulate ROS levels in both zebrafish and macrophages. The cell experiments found that ginsenoside Rg1, ginsenoside Rs3, and oleanolic acid could promote macrophage M2/M1 polarization ratio and inhibit phosphorylation overexpression of signal transducer and activator of transcription 3. This study revealed the key anti-inflammatory molecules and mechanisms of AG, and provided new evidence of anti-inflammatory for the scientific use of AG.
Collapse
Affiliation(s)
- Taiping Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yougang Zhang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Rong Dong
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wenjie Bi
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Songsong Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Liwen Han
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Maddah R, Ghanbari F, Veisi M, Koosehlar E, Shadpirouz M, Basharat Z, Hejrati A, Amiri BS, Hejrati L. Unveiling Potential Biomarkers for Urinary Tract Infection: An Integrated Bioinformatics Approach. Adv Biomed Res 2024; 13:44. [PMID: 39224398 PMCID: PMC11368229 DOI: 10.4103/abr.abr_355_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 09/04/2024] Open
Abstract
Background Urinary tract infections (UTIs) are a widespread health concern with high recurrence rates and substantial economic impact, and they can increase the prevalence of antibiotic resistance. This study employed an integrated bioinformatics approach to identify key genes associated with UTI development, offering potential targets for interventions. Materials and Methods For this study, the microarray dataset GSE124917 from the Gene Expression Omnibus (GEO) database was selected and reanalyzed. The differentially expressed genes (DEGs) between UTIs and healthy samples were identified using the LIMMA package in R software. In this section, Enrichr database was utilized to perform functional enrichment analysis of DEGs. Subsequently, the protein-protein interaction (PPI) network of the DEGs was constructed and visualized through Cytoscape, utilizing the STRING online database. The identification of hub genes was performed using Cytoscape's cytoHubba plug-in employing various methods. Receiver operating characteristic (ROC) analysis was performed to assess the diagnostic accuracy of hub genes. Results Among the outcomes of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, the tumor necrosis factor (TNF) signaling pathway was identified as one of the notable pathways. The PPI network of the DEGs was successfully established and visualized in Cytoscape with the aid of the STRING online database. Using cytoHubba with different methods, we identified seven hub genes (STAT1, IL6, IFIT1, IFIT3, IFIH1, MX1, and IRF7). Based on the ROC analysis, all hub genes showed high diagnostic value. Conclusion These findings provide a valuable baseline for future research aimed at unraveling the intricate molecular mechanisms behind UTI.
Collapse
Affiliation(s)
- Reza Maddah
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fahimeh Ghanbari
- Applied Physiology Research Center, Isfahan University of Medical, Isfahan, Iran
| | - Maziyar Veisi
- Department of Veterinary Medicine Shahrekord, Shahrekord University, Chaharmahal and Bakhtiari Province, Shahrekord, Iran
| | - Eman Koosehlar
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Marzieh Shadpirouz
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Shahrood University of Technology, Semnan, Iran
| | | | - Alireza Hejrati
- Internal Medicine Department, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Bahareh Shateri Amiri
- Internal Medicine Department, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Lina Hejrati
- Internal Medicine Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Cao S, Gao S, Ni C, Xu Y, Pang B, Zhang J, Zhang Y, Wang Y, Geng Z, Li S, Zhao R, Han B, Cui X, Bao Y. Study on the therapeutic mechanism of HJ granules in a rat model of urinary tract infection caused by Escherichia coli. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118056. [PMID: 38490287 DOI: 10.1016/j.jep.2024.118056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Urinary tract infections (UTIs) are globally prevalent infectious diseases, predominantly caused by uropathogenic Escherichia coli (UPEC). The misuse of antibiotics has led to the emergence of several drug-resistant strains. Traditional Chinese Medicine (TCM) has its own advantages in the treatment of UTIs. HJ granules is a herbal formula used for the treatment of UTIs. However, its mechanism of action is not clear. AIM OF THE STUDY The aim of this study was to investigate the therapeutic efficacy and mechanism of action of HJ granules in a rat model of UTI caused by Escherichia coli (E coli) CFT073. MATERIALS AND METHODS SD rats were selected to establish a rat UTI model by injecting UPEC strain CFT073 into the bladder using the transurethral placement method. HJ granules were administered to rats after modelling and the efficacy of HJ granule was investigated by measuring urinary decanalogue, inflammatory factors in bladder tissue and pathological changes in the bladder after 3d of administration. Expression of sonic hedgehog (SHH), NOD-like receptor thermoprotein domain 3 (NLRP3), apoptosis-associated speck-like protein (ASC) and activation of cysteinyl aspartate specific proteinase-1 (caspase-1) were detected by western blotting and immunofluorescence staining in rat bladder tissue. NLRP3, ASC and caspase-1, a cysteine-containing aspartic protein, were expressed and activated. RESULTS The results showed that infection of rats with UPEC resulted in increased pH and erythrocytes in bladder irrigation fluid; increased expression of IL-1β, IL-6 and SHH and decreased expression of IL-10 in bladder tissue; and significant upregulation of the expression of both SHH and NLRP3 inflammasom and significant activation of NLRP3 inflammasom. HJ granules significantly increased the concentration of IL-10 in the bladder, inhibited the expression of SHH and NLRP3 inflammasom in bladder tissue, and suppressed the activation of NLRP3 inflammasom, thereby reducing inflammatory lesions in bladder tissue. CONCLUSION HJ granules may improve bladder injury and treat UTIs by inhibiting the expression and activation of NLRP3 inflammasom.
Collapse
Affiliation(s)
- Shan Cao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shuangrong Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chen Ni
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yingli Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Bo Pang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jingsheng Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yu Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yaxin Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zihan Geng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shurang Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ronghua Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Bing Han
- Heilongjiang Jiren Pharmaceutical Co., Ltd., Harbin, 150000, China.
| | - Xiaolan Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Yanyan Bao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
9
|
Cortado H, Kercsmar M, Li B, Vasquez-Martinez G, Gupta S, Ching C, Ballash G, Cotzomi-Ortega I, Sanchez-Zamora YI, Boix E, Zepeda-Orozco D, Jackson AR, Spencer JD, Ruiz-Rosado JDD, Becknell B. Murine Ribonuclease 6 Limits Bacterial Dissemination during Experimental Urinary Tract Infection. J Innate Immun 2024; 16:283-294. [PMID: 38744252 PMCID: PMC11250601 DOI: 10.1159/000539177] [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: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION The ribonuclease (RNase) A superfamily encodes cationic antimicrobial proteins with potent microbicidal activity toward uropathogenic bacteria. Ribonuclease 6 (RNase6) is an evolutionarily conserved, leukocyte-derived antimicrobial peptide with potent microbicidal activity toward uropathogenic Escherichia coli (UPEC), the most common cause of bacterial urinary tract infections (UTIs). In this study, we generated Rnase6-deficient mice to investigate the hypothesis that endogenous RNase 6 limits host susceptibility to UTI. METHODS We generated a Rnase6EGFP knock-in allele to identify cellular sources of Rnase6 and determine the consequences of homozygous Rnase6 deletion on antimicrobial activity and UTI susceptibility. RESULTS We identified monocytes and macrophages as the primary cellular sources of Rnase6 in bladders and kidneys of Rnase6EGFP/+ mice. Rnase6 deficiency (i.e., Rnase6EGFP/EGFP) resulted in increased upper urinary tract UPEC burden during experimental UTI, compared to Rnase6+/+ controls. UPEC displayed increased intracellular survival in Rnase6-deficient macrophages. CONCLUSION Our findings establish that RNase6 prevents pyelonephritis by promoting intracellular UPEC killing in monocytes and macrophages and reinforce the overarching contributions of endogenous antimicrobial RNase A proteins to host UTI defense.
Collapse
Affiliation(s)
- Hanna Cortado
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Macie Kercsmar
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Birong Li
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Gabriela Vasquez-Martinez
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Sudipti Gupta
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Christina Ching
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Department of Urology, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Gregory Ballash
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, USA
| | - Israel Cotzomi-Ortega
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Yuriko I. Sanchez-Zamora
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Ashley R. Jackson
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - John David Spencer
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Juan de Dios Ruiz-Rosado
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Brian Becknell
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
10
|
Li ZP, Li J, Li TL, Song ZY, Gong XZ. Uropathogenic Escherichia coli infection: innate immune disorder, bladder damage, and Tailin Fang II. Front Cell Infect Microbiol 2024; 14:1322119. [PMID: 38638825 PMCID: PMC11024302 DOI: 10.3389/fcimb.2024.1322119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
Background Uropathogenic Escherichia coli (UPEC) activates innate immune response upon invading the urinary tract, whereas UPEC can also enter bladder epithelial cells (BECs) through interactions with fusiform vesicles on cell surfaces and subsequently escape from the vesicles into the cytoplasm to establish intracellular bacterial communities, finally evading the host immune system and leading to recurrent urinary tract infection (RUTI). Tailin Fang II (TLF-II) is a Chinese herbal formulation composed of botanicals that has been clinically proven to be effective in treating urinary tract infection (UTI). However, the underlying therapeutic mechanisms remain poorly understood. Methods Network pharmacology analysis of TLF-II was conducted. Female Balb/C mice were transurethrally inoculated with UPEC CFT073 strain to establish the UTI mouse model. Levofloxacin was used as a positive control. Mice were randomly divided into four groups: negative control, UTI, TLF-II, and levofloxacin. Histopathological changes in bladder tissues were assessed by evaluating the bladder organ index and performing hematoxylin-eosin staining. The bacterial load in the bladder tissue and urine sample of mice was quantified. Activation of the TLR4-NF-κB pathway was investigated through immunohistochemistry and western blotting. The urinary levels of interleukin (IL)-1β and IL-6 and urine leukocyte counts were monitored. We also determined the protein expressions of markers associated with fusiform vesicles, Rab27b and Galectin-3, and levels of the phosphate transporter protein SLC20A1. Subsequently, the co-localization of Rab27b and SLC20A1 with CFT073 was examined using confocal fluorescence microscopy. Results Data of network pharmacology analysis suggested that TLF-II could against UTI through multiple targets and pathways associated with innate immunity and inflammation. Additionally, TLF-II significantly attenuated UPEC-induced bladder injury and reduced the bladder bacterial load. Meanwhile, TLF-II inhibited the expression of TLR4 and NF-κB on BECs and decreased the urine levels of IL-1β and IL-6 and urine leukocyte counts. TLF-II reduced SLC20A1 and Galectin-3 expressions and increased Rab27b expression. The co-localization of SLC20A1 and Rab27b with CFT073 was significantly reduced in the TLF-II group. Conclusion Collectively, innate immunity and bacterial escape from fusiform vesicles play important roles in UPEC-induced bladder infections. Our findings suggest that TLF-II combats UPEC-induced bladder infections by effectively mitigating bladder inflammation and preventing bacterial escape from fusiform vesicles into the cytoplasm. The findings suggest that TLF-II is a promising option for treating UTI and reducing its recurrence.
Collapse
Affiliation(s)
| | | | | | | | - Xue-zhong Gong
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
11
|
Bilsen MP, Treep MM, Aantjes MJ, van Andel E, Stalenhoef JE, van Nieuwkoop C, Leyten EMS, Delfos NM, van Uhm JIM, Sijbom M, Akintola AA, Numans ME, Achterberg WP, Mooijaart SP, van der Beek MT, Cobbaert CM, Conroy SP, Visser LG, Lambregts MMC. Diagnostic accuracy of urine biomarkers for urinary tract infection in older women: a case-control study. Clin Microbiol Infect 2024; 30:216-222. [PMID: 37805035 DOI: 10.1016/j.cmi.2023.09.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVES Urinary tract infection (UTI) is common among older women. However, diagnosis is challenging because of frequent chronic lower urinary tract symptoms, cognitive impairment, and a high prevalence of asymptomatic bacteriuria (ASB). Current urine diagnostics lack specificity, leading to unnecessary treatment and antimicrobial resistance. This study aimed to evaluate the diagnostic accuracy of 12 urine biomarkers for diagnosing UTI in older women. METHODS In this case-control study, cases were women ≥65 years with ≥2 new-onset lower urinary tract symptoms, pyuria, and one uropathogen ≥104 CFU/mL. Controls were asymptomatic and classified as ASB (one uropathogen ≥105 CFU/mL), negative culture, or mixed flora. Urine biomarker concentrations were measured through liquid chromatography-mass spectrometry and ELISA. Diagnostic accuracy parameters of individual biomarkers and a biomarker model were derived from receiver operating characteristic curves. RESULTS We included 162 community-dwelling and institutionalized older women. Five urine inflammatory biomarkers demonstrated high discriminative ability (area under the curve ≥0.80): interleukin 6, azurocidin, neutrophil gelatinase-associated lipocalin, tissue inhibitor of metalloproteinases 2, and C-X-C motif chemokine 9. Azurocidin exhibited the highest diagnostic accuracy (sensitivity 86% [95% CI 75%-93%] and specificity 89% [95% CI 82%-94%] at 16.7 ng/mmol creatinine). A combined biomarker and pyuria model showed improved diagnostic accuracy in patients with UTI and ASB, compared with pyuria alone. DISCUSSION We identified several urine biomarkers that accurately differentiated older women with UTI from asymptomatic women, including ASB. These findings represent a potential advancement towards improved diagnostics for UTI in older women and warrant validation in a diverse population.
Collapse
Affiliation(s)
- Manu P Bilsen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | - Maxim M Treep
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Margaretha J Aantjes
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther van Andel
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Cees van Nieuwkoop
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, The Netherlands; Department of Public Health and Primary Care, Leiden University Medical Center, Health Campus The Hague, The Netherlands
| | - Eliane M S Leyten
- Department of Internal Medicine, Haaglanden Medisch Centrum, The Hague, The Netherlands
| | - Nathalie M Delfos
- Department of Internal Medicine, Alrijne Hospital, Leiderdorp, The Netherlands
| | - Janneke I M van Uhm
- Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn Sijbom
- Department of Public Health and Primary Care, Leiden University Medical Center, Health Campus The Hague, The Netherlands
| | - Abimbola A Akintola
- Department of Public Health and Primary Care, Leiden University Medical Center, Health Campus The Hague, The Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Center, Health Campus The Hague, The Netherlands
| | - Wilco P Achterberg
- Department of Public Health and Primary Care, Leiden University Medical Center, Health Campus The Hague, The Netherlands; LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Mooijaart
- LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, The Netherlands; Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Martha T van der Beek
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Conroy
- Medical Research Council Unit for Lifelong Health and Ageing at University College London, University College London, London, United Kingdom
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Merel M C Lambregts
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
12
|
Ramal M, Corral S, Kalisz M, Lapi E, Real FX. The urothelial gene regulatory network: understanding biology to improve bladder cancer management. Oncogene 2024; 43:1-21. [PMID: 37996699 DOI: 10.1038/s41388-023-02876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
The urothelium is a stratified epithelium composed of basal cells, one or more layers of intermediate cells, and an upper layer of differentiated umbrella cells. Most bladder cancers (BLCA) are urothelial carcinomas. Loss of urothelial lineage fidelity results in altered differentiation, highlighted by the taxonomic classification into basal and luminal tumors. There is a need to better understand the urothelial transcriptional networks. To systematically identify transcription factors (TFs) relevant for urothelial identity, we defined highly expressed TFs in normal human bladder using RNA-Seq data and inferred their genomic binding using ATAC-Seq data. To focus on epithelial TFs, we analyzed RNA-Seq data from patient-derived organoids recapitulating features of basal/luminal tumors. We classified TFs as "luminal-enriched", "basal-enriched" or "common" according to expression in organoids. We validated our classification by differential gene expression analysis in Luminal Papillary vs. Basal/Squamous tumors. Genomic analyses revealed well-known TFs associated with luminal (e.g., PPARG, GATA3, FOXA1) and basal (e.g., TP63, TFAP2) phenotypes and novel candidates to play a role in urothelial differentiation or BLCA (e.g., MECOM, TBX3). We also identified TF families (e.g., KLFs, AP1, circadian clock, sex hormone receptors) for which there is suggestive evidence of their involvement in urothelial differentiation and/or BLCA. Genomic alterations in these TFs are associated with BLCA. We uncover a TF network involved in urothelial cell identity and BLCA. We identify novel candidate TFs involved in differentiation and cancer that provide opportunities for a better understanding of the underlying biology and therapeutic intervention.
Collapse
Affiliation(s)
- Maria Ramal
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sonia Corral
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Mark Kalisz
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Eleonora Lapi
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- CIBERONC, Madrid, Spain.
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
13
|
Ruiz-Rosado JDD, Cortado H, Kercsmar M, Li B, Ballash G, Cotzomi-Ortega I, Sanchez-Zamora YI, Gupta S, Ching C, Boix E, Jackson AR, Spencer JD, Becknell B. Human Ribonuclease 6 Has a Protective Role during Experimental Urinary Tract Infection. J Innate Immun 2023; 15:865-875. [PMID: 37980892 PMCID: PMC10699853 DOI: 10.1159/000534736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/18/2023] [Indexed: 11/21/2023] Open
Abstract
Mounting evidence suggests that antimicrobial peptides and proteins (AMPs) belonging to the RNase A superfamily have a critical role in defending the bladder and kidney from bacterial infection. RNase 6 has been identified as a potent, leukocyte-derived AMP, but its impact on urinary tract infection (UTI) in vivo has not been demonstrated. To test the functional role of human RNase 6, we generated RNASE6 transgenic mice and studied their susceptibility to experimental UTI. In addition, we generated bone marrow-derived macrophages to study the impact of RNase 6 on antimicrobial activity within a cellular context. When subjected to experimental UTI, RNASE6 transgenic mice developed reduced uropathogenic Escherichia coli (UPEC) burden, mucosal injury, and inflammation compared to non-transgenic controls. Monocytes and macrophages were the predominant cellular sources of RNase 6 during UTI, and RNASE6 transgenic macrophages were more proficient at intracellular UPEC killing than non-transgenic controls. Altogether, our findings indicate a protective role for human RNase 6 during experimental UTI.
Collapse
Affiliation(s)
- Juan de Dios Ruiz-Rosado
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Hanna Cortado
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Macie Kercsmar
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Birong Li
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Gregory Ballash
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Israel Cotzomi-Ortega
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Yuriko I. Sanchez-Zamora
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Sudipti Gupta
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
| | - Christina Ching
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Department of Urology, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma De Barcelona, Barcelona, Spain
| | - Ashley R. Jackson
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - John David Spencer
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Brian Becknell
- Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
14
|
Durrani B, Mohammad A, Ljubetic BM, Dobberfuhl AD. The Potential Role of Persister Cells in Urinary Tract Infections. Curr Urol Rep 2023; 24:541-551. [PMID: 37907771 DOI: 10.1007/s11934-023-01182-5] [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] [Accepted: 08/22/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE OF REVIEW This review explores the role of persister cells in urinary tract infections (UTIs). UTIs are one of the most common bacterial infections, affecting millions of people worldwide. Persister cells are a subpopulation of bacteria with dormant metabolic activity which allows survival in the presence of antibiotics. RECENT FINDINGS This review summarizes recent research on the pathogenesis of persister cell formation in UTIs, the impact of persister cells on the effectiveness of antibiotics, the challenges they pose for treatment, and the need for new strategies to target these cells. Furthermore, this review examines the current state of research on the identification and characterization of persister cells in UTIs, as well as the future directions for investigations in this field. This review highlights the importance of understanding the role of persister cells in UTIs and the potential impact of targeting these cells in the development of new treatments.
Collapse
Affiliation(s)
- Butool Durrani
- Department of Internal Medicine, Aga Khan University Hospital, National Stadium Rd, Karachi, Karachi City, Pakistan
| | - Ashu Mohammad
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA
| | - Bernardita M Ljubetic
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA
| | - Amy D Dobberfuhl
- Department of Urology, Center for Academic Medicine, Stanford University School of Medicine, 453 Quarry Road, Urology-5656, Palo Alto, CA, 94304, USA.
| |
Collapse
|
15
|
Manoharan A, Farrell J, Aldilla VR, Whiteley G, Kriel E, Glasbey T, Kumar N, Moore KH, Manos J, Das T. N-acetylcysteine prevents catheter occlusion and inflammation in catheter associated-urinary tract infections by suppressing urease activity. Front Cell Infect Microbiol 2023; 13:1216798. [PMID: 37965267 PMCID: PMC10641931 DOI: 10.3389/fcimb.2023.1216798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/19/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction Proteus mirabilis is a key pathobiont in catheter-associated urinary tract infections (CA-UTIs), which is well known to form crystalline biofilms that occlude catheters. Urease activity alkylates urine through the release of ammonia, consequentially resulting in higher levels of Mg2+ and Ca2+ and formation of crystals. In this study, we showed that N-acetyl cysteine (NAC), a thiol antioxidant, is a potent urease inhibitor that prevents crystalline biofilm formation. Methods To quantify urease activity, Berthelot's method was done on bacterial extracts treated with NAC. We also used an in vitro catheterised glass bladder model to study the effect of NAC treatment on catheter occlusion and biofilm encrustation in P. mirabilis infections. Inductively-coupled plasma mass spectrometry (ICP-MS) was performed on catheter samples to decipher elemental profiles. Results NAC inhibits urease activity of clinical P. mirabilis isolates at concentrations as low as 1 mM, independent of bacterial killing. The study also showed that NAC is bacteriostatic on P. mirabilis, and inhibited biofilm formation and catheter occlusion in an in vitro. A significant 4-8log10 reduction in viable bacteria was observed in catheters infected in this model. Additionally, biofilms in NAC treated catheters displayed a depletion of calcium, magnesium, or phosphates (>10 fold reduction), thus confirming the absence of any urease activity in the presence of NAC. Interestingly, we also showed that not only is NAC anti-inflammatory in bladder epithelial cells (BECs), but that it mutes its inflammatory response to urease and P. mirabilis infection by reducing the production of IL-6, IL-8 and IL-1b. Discussion Using biochemical, microbiological and immunological techniques, this study displays the functionality of NAC in preventing catheter occlusion by inhibiting urease activity. The study also highlights NAC as a strong anti-inflammatory antibiofilm agent that can target both bacterial and host factors in the treatment of CA-UTIs.
Collapse
Affiliation(s)
- Arthika Manoharan
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute of Infectious Disease, The University of Sydney, Sydney, NSW, Australia
| | - Jessica Farrell
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Whiteley Corporation, Tomago, NSW, Australia
| | - Vina R. Aldilla
- School of Chemistry, The University of New South Wales, Sydney, NSW, Australia
| | - Greg Whiteley
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Whiteley Corporation, Tomago, NSW, Australia
- School of Medicine, Western Sydney University, NSW, Australia
| | - Erik Kriel
- Whiteley Corporation, Tomago, NSW, Australia
| | | | - Naresh Kumar
- School of Chemistry, The University of New South Wales, Sydney, NSW, Australia
| | - Kate H. Moore
- Department of Urogynaecology, St George Hospital, University of New South Wales, Sydney, NSW, Australia
| | - Jim Manos
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute of Infectious Disease, The University of Sydney, Sydney, NSW, Australia
| | - Theerthankar Das
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute of Infectious Disease, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
16
|
Rao SS, Nelson PA, Lunde HS, Haugland GT. Evolutionary, comparative, and functional analyses of STATs and regulation of the JAK-STAT pathway in lumpfish upon bacterial and poly(I:C) exposure. Front Cell Infect Microbiol 2023; 13:1252744. [PMID: 37808912 PMCID: PMC10556531 DOI: 10.3389/fcimb.2023.1252744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Background The Janus kinase/signal transducers and activators of transcription (JAK-STAT) system regulates several biological processes by affecting transcription of genes as a response to cytokines and growth factors. In the present study, we have characterized the STAT genes in lumpfish (Cyclopterus lumpus L.), belonging to the order Perciformes, and investigated regulation of the JAK-STAT signaling pathway upon exposure to bacteria (Vibrio anguillarum) and poly(I:C), the latter mimicking antiviral responses. Methods Characterization and evolutionary analyses of the STATs were performed by phylogeny, protein domain, homology similarity and synteny analyses. Antibacterial and antiviral responses were investigated by performing KEGG pathway analysis. Results We observed that lumpfish have stat1a, 2, 3, 4, 5a, 5b, and 6. Transcriptome-wide analyses showed that most components of the JAK-STAT pathway were present in lumpfish. il-6, il-10, il-21, iκBα and stat3 were upregulated 6 hours post exposure (hpe) against bacteria while type I interferons (IFNs), irf1, irf3, irf10, stat1 and 2 were upregulated 24 hpe against poly(I:C). Conclusions Our findings shed light on the diversity and evolution of the STATs and the data show that the STAT genes are highly conserved among fish, including lumpfish. The transcriptome-wide analyses lay the groundwork for future research into the functional significance of these genes in regulating critical biological processes and make an important basis for development of prophylactic measure such as vaccination, which is highly needed for lumpfish since it is vulnerable for both bacterial and viral diseases.
Collapse
Affiliation(s)
- Shreesha S Rao
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| | - Patrick A Nelson
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| | - Harald S Lunde
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| | - Gyri T Haugland
- Department of Biological Sciences, Bergen High-Technology Centre, University of Bergen, Bergen, Norway
| |
Collapse
|
17
|
Kuhn HW, Hreha TN, Hunstad DA. Immune defenses in the urinary tract. Trends Immunol 2023; 44:701-711. [PMID: 37591712 PMCID: PMC10528756 DOI: 10.1016/j.it.2023.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 08/19/2023]
Abstract
Recent advances in preclinical modeling of urinary tract infections (UTIs) have enabled the identification of key facets of the host response that influence pathogen clearance and tissue damage. Here, we review new insights into the functions of neutrophils, macrophages, and antimicrobial peptides in innate control of uropathogens and in mammalian infection-related tissue injury and repair. We also discuss novel functions for renal epithelial cells in innate antimicrobial defense. In addition, epigenetic modifications during bacterial cystitis have been implicated in bladder remodeling, conveying susceptibility to recurrent UTI. In total, contemporary work in this arena has better defined host processes that shape UTI susceptibility and severity and might inform the development of novel preventive and therapeutic approaches for acute and recurrent UTI.
Collapse
Affiliation(s)
- Hunter W Kuhn
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Teri N Hreha
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Hunstad
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
18
|
Gao M, Zhao T, Zhang C, Li P, Wang J, Han J, Zhang N, Pang B, Liu S. Ferritinophagy-mediated iron competition in RUTIs: Tug-of-war between UPEC and host. Biomed Pharmacother 2023; 163:114859. [PMID: 37167722 DOI: 10.1016/j.biopha.2023.114859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the main pathogen of recurrent urinary tract infections (RUTIs). Urinary tract infection is a complicated interaction between UPEC and the host. During infection, UPEC can evade the host's immune response and retain in bladder epithelial cells, which requires adequate nutritional support. Iron is the first necessary trace element in life and a key nutritional factor, making it an important part of the competition between UPEC and the host. On the one hand, UPEC grabs iron to satisfy its reproduction, on the other hand, the host relies on iron to build nutritional immunity defenses against UPEC. Ferritinophagy is a selective autophagy of ferritin mediated by nuclear receptor coactivator 4, which is not only a way for the host to regulate iron metabolism to maintain iron homeostasis, but also a key point of competition between the host and UPEC. Although recent studies have confirmed the role of ferritinophagy in the progression of many diseases, the mechanism of potential interactions between ferritinophagy in UPEC and the host is poorly understood. In this paper, we reviewed the potential mechanisms of ferritinophagy-mediated iron competition in the UPEC-host interactions. This competitive relationship, like a tug-of-war, is a confrontation between the capability of UPEC to capture iron and the host's nutritional immunity defense, which could be the trigger for RUTIs. Therefore, understanding ferritinophagy-mediated iron competition may provide new strategies for exploring effective antibiotic alternative therapies to prevent and treat RUTIs.
Collapse
Affiliation(s)
- Mengqi Gao
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Tingting Zhao
- Department of Nephrology, Beijing Key Laboratory for Immune-Mediated Inflammatory 9 Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Chuanlong Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ping Li
- Department of Nephrology, Beijing Key Laboratory for Immune-Mediated Inflammatory 9 Diseases, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jiazhe Wang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiatong Han
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ning Zhang
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Bo Pang
- International Medical Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Shiwei Liu
- Department of Nephrology and Endocrinology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China.
| |
Collapse
|
19
|
Egozi A, Olaloye O, Werner L, Silva T, McCourt B, Pierce RW, An X, Wang F, Chen K, Pober JS, Shouval D, Itzkovitz S, Konnikova L. Single-cell atlas of the human neonatal small intestine affected by necrotizing enterocolitis. PLoS Biol 2023; 21:e3002124. [PMID: 37205711 PMCID: PMC10234541 DOI: 10.1371/journal.pbio.3002124] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/01/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a gastrointestinal complication of premature infants with high rates of morbidity and mortality. A comprehensive view of the cellular changes and aberrant interactions that underlie NEC is lacking. This study aimed at filling in this gap. We combine single-cell RNA sequencing (scRNAseq), T-cell receptor beta (TCRβ) analysis, bulk transcriptomics, and imaging to characterize cell identities, interactions, and zonal changes in NEC. We find an abundance of proinflammatory macrophages, fibroblasts, endothelial cells as well as T cells that exhibit increased TCRβ clonal expansion. Villus tip epithelial cells are reduced in NEC and the remaining epithelial cells up-regulate proinflammatory genes. We establish a detailed map of aberrant epithelial-mesenchymal-immune interactions that are associated with inflammation in NEC mucosa. Our analyses highlight the cellular dysregulations of NEC-associated intestinal tissue and identify potential targets for biomarker discovery and therapeutics.
Collapse
Affiliation(s)
- Adi Egozi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Lael Werner
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel, affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tatiana Silva
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Blake McCourt
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Richard W. Pierce
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Xiaojing An
- Department of Medicine, University of Pittsburgh Medical Center Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Fujing Wang
- Department of Medicine, University of Pittsburgh Medical Center Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Kong Chen
- Department of Medicine, University of Pittsburgh Medical Center Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Jordan S. Pober
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Dror Shouval
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children’s Medical Center of Israel, Petah Tikva, Israel, affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut, United States of America
- Program in Human and Translational Immunology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
20
|
Fashemi BE, Wang C, Chappidi RR, Morsy H, Mysorekar IU. Supraphysiologic Vaginal Estrogen Therapy in Aged Mice Mitigates Age-Associated Bladder Inflammatory Response to Urinary Tract Infections. UROGYNECOLOGY (PHILADELPHIA, PA.) 2023; 29:430-442. [PMID: 36384972 PMCID: PMC10117622 DOI: 10.1097/spv.0000000000001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
IMPORTANCE Bladder diseases characterized by chronic inflammation are highly prevalent in older women, as are recurrent urinary tract infections (rUTIs). Recurrent urinary tract infections lead to chronic inflammation of the bladder mucosa and cause lower urinary tract symptoms that persist even after the infection is cleared. Vaginal estrogen therapy (VET) has long been used for the treatment of rUTIs; however, its mechanism of action remains unclear. OBJECTIVES The objective of this study was to examine the mechanism(s) by which VET affects bladder inflammation and response to rUTIs. STUDY DESIGN Here, we induced surgical menopause in aged (18 months old) mice followed by VET. Mice were then infected with uropathogenic Escherichia coli , and course of infection was investigated. Inflammatory cytokine response was assessed before and during infection using enzyme-linked immunosorbent assay. RNA sequencing analysis was used to compare the inflammatory status of the young versus aged bladder and principal changes confirmed via quantitative reverse transcriptase-polymerase chain reaction to determine the effects of VET on bladder inflammation. Impact on age-associated bladder tertiary lymphoid tissue formation was evaluated histologically. RESULTS In the ovariectomized aged model, VET not only mitigated uterine atrophy but was also associated with reduced rUTIs, number of bacterial reservoirs, dampened immune response, and promotion of terminal differentiation of urothelial cells. Bladder tertiary lymphoid tissue lesions were also reduced with VET, with an associated decrease in signals important for bladder tertiary lymphoid tissue formation. Finally, we determined that VET reverses age-associated upregulation of inflammatory genes and pathways. CONCLUSIONS Our data suggest that VET is effective by reducing age-associated hyperinflammatory conditions in bladder mucosa and in enhancing the host response to infection.
Collapse
Affiliation(s)
- Bisiayo E Fashemi
- From the Center for Reproductive Health Sciences, Division of Basic Research
| | - Caihong Wang
- From the Center for Reproductive Health Sciences, Division of Basic Research
| | - Rayvanth R Chappidi
- From the Center for Reproductive Health Sciences, Division of Basic Research
| | - Haidy Morsy
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics & Gynecology, Washington University School of Medicine
| | | |
Collapse
|
21
|
Wu R, Kumawat AK, Demirel I. Trimethylamine N-Oxide (TMAO) Mediates Increased Inflammation and Colonization of Bladder Epithelial Cells during a Uropathogenic E. coli Infection In Vitro. Pathogens 2023; 12:pathogens12040523. [PMID: 37111409 PMCID: PMC10141729 DOI: 10.3390/pathogens12040523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common infections in humans and are often caused by uropathogenic E. coli (UPEC). Trimethylamine N-oxide (TMAO) is a proinflammatory metabolite that has been linked to vascular inflammation, atherosclerosis, and chronic kidney disease. As of today, no studies have investigated the effects of TMAO on infectious diseases like UTIs. The aim of this study was to investigate whether TMAO can aggravate bacterial colonization and the release of inflammatory mediators from bladder epithelial cells during a UPEC infection. We found that TMAO aggravated the release of several key cytokines (IL-1β and IL-6) and chemokines (IL-8, CXCL1 and CXCL6) from bladder epithelial cells during a CFT073 infection. We also found that CFT073 and TMAO mediate increased release of IL-8 from bladder epithelial cells via ERK 1/2 signaling and not bacterial growth. Furthermore, we showed that TMAO enhances UPEC colonization of bladder epithelial cells. The data suggest that TMAO may also play a role in infectious diseases. Our results can be the basis of further research to investigate the link between diet, gut microbiota, and urinary tract infection.
Collapse
|
22
|
Peskar D, Kuret T, Lakota K, Erman A. Molecular Profiling of Inflammatory Processes in a Mouse Model of IC/BPS: From the Complete Transcriptome to Major Sex-Related Histological Features of the Urinary Bladder. Int J Mol Sci 2023; 24:ijms24065758. [PMID: 36982831 PMCID: PMC10058956 DOI: 10.3390/ijms24065758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Animal models are invaluable in the research of the pathophysiology of interstitial cystitis/bladder pain syndrome (IC/BPS), a chronic aseptic urinary bladder disease of unknown etiology that primarily affects women. Here, a mouse model of IC/BPS was induced with multiple low-dose cyclophosphamide (CYP) applications and thoroughly characterized by RNA sequencing, qPCR, Western blot, and immunolabeling to elucidate key inflammatory processes and sex-dependent differences in the bladder inflammatory response. CYP treatment resulted in the upregulation of inflammatory transcripts such as Ccl8, Eda2r, and Vegfd, which are predominantly involved in innate immunity pathways, recapitulating the crucial findings in the bladder transcriptome of IC/BPS patients. The JAK/STAT signaling pathway was analyzed in detail, and the JAK3/STAT3 interaction was found to be most activated in cells of the bladder urothelium and lamina propria. Sex-based data analysis revealed that cell proliferation was more pronounced in male bladders, while innate immunity and tissue remodeling processes were the most distinctive responses of female bladders to CYP treatment. These processes were also reflected in prominent histological changes in the bladder. The study provides an invaluable reference dataset for preclinical research on IC/BPS and an insight into the sex-specific mechanisms involved in the development of IC/BPS pathology, which may explain the more frequent occurrence of this disease in women.
Collapse
Affiliation(s)
- Dominika Peskar
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadeja Kuret
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Katja Lakota
- Department of Rheumatology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
| | - Andreja Erman
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
23
|
Tan Z, Liu Q, Chen H, Zhang Z, Wang Q, Mu Y, Li Y, Hu T, Yang Y, Yan X. Pectolinarigenin alleviated septic acute kidney injury via inhibiting Jak2/Stat3 signaling and mitochondria dysfunction. Biomed Pharmacother 2023; 159:114286. [PMID: 36706631 DOI: 10.1016/j.biopha.2023.114286] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Sepsis is a systemic inflammatory response to infection, where sepsis-associated acute kidney injury (AKI) is a common morbid disease with a high morbidity and mortality, and however at present no effective therapy exists. Increasing evidence have shown that mitochondrial damage and inflammatory response are important initiating factors in pathogenesis of septic AKI. Natural flavonoid pectolinarigenin exerted anti-inflammatory properties in previous studies, while its role in septic AKI remains unknown. In the study, pectolinarigenin administration significantly ameliorated the dramatic rise of serum creatinine and blood urea nitrogen in lipopolysaccharide (LPS)- and cecal ligation/puncture (CLP)-induced septic mice, respectively. Consistently, LPS/CLP-induced renal damage as implied by histopathological score and the increased injury markers NGAL and KIM-1, which was attenuated by pectolinarigenin. Meanwhile, LPS/CLP triggered proinflammatory cytokine production and inflammation related proteins in the kidneys. However, pectolinarigenin inhibited renal expression of IL-6, IL-1β, TNF-α, and MCP-1 to improve inflammatory response. Furthermore, pectolinarigenin upregulated Bcl-2 protein expression and suppressed apoptotic protein of BAX and cleaved caspase-3 in the kidneys of CLP-induced septic AKI. Mechanistically, LPS could induce the high expression of IL-6 and trigger the phosphorylation of Jak2 and Stat3, while pectolinarigenin remarkably reduced their corresponding levels. Notably, CLP-induced kidney injury of mice significantly reduced the expression of PGC-1α, OPA1 and increased the expression of Drp1, Cyt-C, where pectolinarigenin pretreatment significantly restored their corresponding expression in mice. In summary, pectolinarigenin improved septic AKI via inhibiting JAK2/STAT3 signaling and mitochondria dysfunction.
Collapse
Affiliation(s)
- Zhouke Tan
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China; Organ Transplant Center, Affiliated Hospital of ZunYi Medical University, ZunYi 563000, China.
| | - Qianqian Liu
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - Hongjun Chen
- Department of Critical Care Medicine, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - Ziyang Zhang
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - Qin Wang
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - Yingsong Mu
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - Yiman Li
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - TingTing Hu
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China
| | - Yibin Yang
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China.
| | - Xiaoyong Yan
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi 563003, China.
| |
Collapse
|
24
|
Huang Y, Chen J, Jiang Q, Huang N, Ding X, Peng L, Deng X. The molybdate-binding protein ModA is required for Proteus mirabilis-induced UTI. Front Microbiol 2023; 14:1156273. [PMID: 37180242 PMCID: PMC10174112 DOI: 10.3389/fmicb.2023.1156273] [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: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 05/16/2023] Open
Abstract
Background Proteus mirabilis is one of the pathogens commonly causing urinary tract infections (UTIs). The molybdate-binding protein ModA encoded by modA binds molybdate with high affinity and transports it. Increasing evidence shows that ModA promotes the survival of bacteria in anaerobic environments and participates in bacterial virulence by obtaining molybdenum. However, the role of ModA in the pathogenesis of P. mirabilis remains unknown. Results In this study, a series of phenotypic assays and transcriptomic analyses were used to study the role of ModA in the UTIs induced by P. mirabilis. Our data showed that ModA absorbed molybdate with high affinity and incorporated it into molybdopterin, thus affecting the anaerobic growth of P. mirabilis. Loss of ModA enhanced bacterial swarming and swimming and up-regulated the expression of multiple genes in flagellar assembly pathway. The loss of ModA also resulted in decreased biofilm formation under anaerobic growth conditions. The modA mutant significantly inhibited bacterial adhesion and invasion to urinary tract epithelial cells and down-regulated the expression of multiple genes associated with pilus assembly. Those alterations were not due to anaerobic growth defects. In addition, the decreased bacteria in the bladder tissue, the weakened inflammatory damage, the low level of IL-6, and minor weight change was observed in the UTI mouse model infected with modA mutant. Conclusion Here, we reported that in P. mirabilis, ModA mediated the transport of molybdate, thereby affecting the activity of nitrate reductase and thus affecting the growth of bacteria under anaerobic conditions. Overall, this study clarified the indirect role of ModA in the anaerobic growth, motility, biofilm formation, and pathogenicity of P. mirabilis and its possible pathway, and emphasized the importance of the molybdate-binding protein ModA to P. mirabilis in mediating molybdate uptake, allowing the bacterium to adapt to complex environmental conditions and cause UTIs. Our results provided valuable information on the pathogenesis of ModA-induced P. mirabilis UTIs and may facilitate the development of new treatment strategies.
Collapse
Affiliation(s)
- Yi Huang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinbin Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiao Jiang
- Guangdong 999 Brain Hospital, Guangzhou, Guangdong, China
| | - Nan Huang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Ding
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liang Peng
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- *Correspondence: Xiaoyan Deng, ; Liang Peng,
| | - Xiaoyan Deng
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
- *Correspondence: Xiaoyan Deng, ; Liang Peng,
| |
Collapse
|
25
|
DIA mass spectrometry characterizes urinary proteomics in neonatal and adult donkeys. Sci Rep 2022; 12:22590. [PMID: 36585464 PMCID: PMC9803668 DOI: 10.1038/s41598-022-27245-0] [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: 01/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Health monitoring is critical for newborn animals due to their vulnerability to diseases. Urine can be not only a useful and non-invasive tool (free-catch samples) to reflect the physiological status of animals but also to help monitor the progression of diseases. Proteomics involves the study of the whole complement of proteins and peptides, including structure, quantities, functions, variations and interactions. In this study, urinary proteomics of neonatal donkeys were characterized and compared to the profiles of adult donkeys to provide a reference database for healthy neonatal donkeys. The urine samples were collected from male neonatal donkeys on their sixth to tenth days of life (group N) and male adult donkeys aging 4-6 years old (group A). Library-free data-independent acquisition (direct DIA) mass spectrometry-based proteomics were applied to analyze the urinary protein profiles. Total 2179 urinary proteins were identified, and 411 proteins were differentially expressed (P < 0.05) between the two groups. 104 proteins were exclusively expressed in group N including alpha fetoprotein (AFP), peptidase-mitochondrial processing data unit (PMPCB), and upper zone of growth plate and cartilage matrix associated (UCMA), which might be used to monitor the health status of neonatal donkeys. In functional analysis, some differentially expressed proteins were identified related to immune system pathways, which might provide more insight in the immature immunity of neonatal donkeys. To the best of our knowledge, this is the first time to report donkey urinary proteome and our results might provide reference for urinary biomarker discovery used to monitor and evaluate health status of neonatal donkeys.
Collapse
|
26
|
Hemmati S, Rasekhi Kazerooni H. Polypharmacological Cell-Penetrating Peptides from Venomous Marine Animals Based on Immunomodulating, Antimicrobial, and Anticancer Properties. Mar Drugs 2022; 20:md20120763. [PMID: 36547910 PMCID: PMC9787916 DOI: 10.3390/md20120763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/09/2022] Open
Abstract
Complex pathological diseases, such as cancer, infection, and Alzheimer's, need to be targeted by multipronged curative. Various omics technologies, with a high rate of data generation, demand artificial intelligence to translate these data into druggable targets. In this study, 82 marine venomous animal species were retrieved, and 3505 cryptic cell-penetrating peptides (CPPs) were identified in their toxins. A total of 279 safe peptides were further analyzed for antimicrobial, anticancer, and immunomodulatory characteristics. Protease-resistant CPPs with endosomal-escape ability in Hydrophis hardwickii, nuclear-localizing peptides in Scorpaena plumieri, and mitochondrial-targeting peptides from Synanceia horrida were suitable for compartmental drug delivery. A broad-spectrum S. horrida-derived antimicrobial peptide with a high binding-affinity to bacterial membranes was an antigen-presenting cell (APC) stimulator that primes cytokine release and naïve T-cell maturation simultaneously. While antibiofilm and wound-healing peptides were detected in Synanceia verrucosa, APC epitopes as universal adjuvants for antiviral vaccination were in Pterois volitans and Conus monile. Conus pennaceus-derived anticancer peptides showed antiangiogenic and IL-2-inducing properties with moderate BBB-permeation and were defined to be a tumor-homing peptide (THP) with the ability to inhibit programmed death ligand-1 (PDL-1). Isoforms of RGD-containing peptides with innate antiangiogenic characteristics were in Conus tessulatus for tumor targeting. Inhibitors of neuropilin-1 in C. pennaceus are proposed for imaging probes or therapeutic delivery. A Conus betulinus cryptic peptide, with BBB-permeation, mitochondrial-targeting, and antioxidant capacity, was a stimulator of anti-inflammatory cytokines and non-inducer of proinflammation proposed for Alzheimer's. Conclusively, we have considered the dynamic interaction of cells, their microenvironment, and proportional-orchestrating-host- immune pathways by multi-target-directed CPPs resembling single-molecule polypharmacology. This strategy might fill the therapeutic gap in complex resistant disorders and increase the candidates' clinical-translation chance.
Collapse
Affiliation(s)
- Shiva Hemmati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz 71345-1583, Iran
- Correspondence: ; Tel.: +98-7132-424-128
| | | |
Collapse
|
27
|
Human neutrophil peptides 1-3 protect the murine urinary tract from uropathogenic Escherichia coli challenge. Proc Natl Acad Sci U S A 2022; 119:e2206515119. [PMID: 36161923 PMCID: PMC9546544 DOI: 10.1073/pnas.2206515119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antimicrobial peptides (AMPs) are critical to the protection of the urinary tract of humans and other animals from pathogenic microbial invasion. AMPs rapidly destroy pathogens by disrupting microbial membranes and/or augmenting or inhibiting the host immune system through a variety of signaling pathways. We have previously demonstrated that alpha-defensins 1-3 (DEFA1A3) are AMPs expressed in the epithelial cells of the human kidney collecting duct in response to uropathogens. We also demonstrated that DNA copy number variations in the DEFA1A3 locus are associated with UTI and pyelonephritis risk. Because DEFA1A3 is not expressed in mice, we utilized human DEFA1A3 gene transgenic mice (DEFA4/4) to further elucidate the biological relevance of this locus in the murine urinary tract. We demonstrate that the kidney transcriptional and translational expression pattern is similar in humans and the human gene transgenic mouse upon uropathogenic Escherichia coli (UPEC) stimulus in vitro and in vivo. We also demonstrate transgenic human DEFA4/4 gene mice are protected from UTI and pyelonephritis under various UPEC challenges. This study serves as the foundation to start the exploration of manipulating the DEFA1A3 locus and alpha-defensins 1-3 expression as a potential therapeutic target for UTIs and other infectious diseases.
Collapse
|
28
|
Li L, Li Y, Yang J, Xie X, Chen H. The immune responses to different Uropathogens call individual interventions for bladder infection. Front Immunol 2022; 13:953354. [PMID: 36081496 PMCID: PMC9445553 DOI: 10.3389/fimmu.2022.953354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Urinary tract infection (UTI) caused by uropathogens is the most common infectious disease and significantly affects all aspects of the quality of life of the patients. However, uropathogens are increasingly becoming antibiotic-resistant, which threatens the only effective treatment option available-antibiotic, resulting in higher medical costs, prolonged hospital stays, and increased mortality. Currently, people are turning their attention to the immune responses, hoping to find effective immunotherapeutic interventions which can be alternatives to the overuse of antibiotic drugs. Bladder infections are caused by the main nine uropathogens and the bladder executes different immune responses depending on the type of uropathogens. It is essential to understand the immune responses to diverse uropathogens in bladder infection for guiding the design and development of immunotherapeutic interventions. This review firstly sorts out and comparatively analyzes the immune responses to the main nine uropathogens in bladder infection, and summarizes their similarities and differences. Based on these immune responses, we innovatively propose that different microbial bladder infections should adopt corresponding immunomodulatory interventions, and the same immunomodulatory intervention can also be applied to diverse microbial infections if they share the same effective therapeutic targets.
Collapse
Affiliation(s)
- Linlong Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Yangyang Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jiali Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- *Correspondence: Xiang Xie, ; Huan Chen,
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- *Correspondence: Xiang Xie, ; Huan Chen,
| |
Collapse
|
29
|
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: 39] [Impact Index Per Article: 13.0] [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
|
30
|
Butler D, Ambite I, Wan MLY, Tran TH, Wullt B, Svanborg C. Immunomodulation therapy offers new molecular strategies to treat UTI. Nat Rev Urol 2022; 19:419-437. [PMID: 35732832 PMCID: PMC9214477 DOI: 10.1038/s41585-022-00602-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
Abstract
Innovative solutions are needed for the treatment of bacterial infections, and a range of antibacterial molecules have been explored as alternatives to antibiotics. A different approach is to investigate the immune system of the host for new ways of making the antibacterial defence more efficient. However, the immune system has a dual role as protector and cause of disease: in addition to being protective, increasing evidence shows that innate immune responses can become excessive and cause acute symptoms and tissue pathology during infection. This role of innate immunity in disease suggests that the immune system should be targeted therapeutically, to inhibit over-reactivity. The ultimate goal is to develop therapies that selectively attenuate destructive immune response cascades, while augmenting the protective antimicrobial defence but such treatment options have remained underexplored, owing to the molecular proximity of the protective and destructive effects of the immune response. The concept of innate immunomodulation therapy has been developed successfully in urinary tract infections, based on detailed studies of innate immune activation and disease pathogenesis. Effective, disease-specific, immunomodulatory strategies have been developed by targeting specific immune response regulators including key transcription factors. In acute pyelonephritis, targeting interferon regulatory factor 7 using small interfering RNA or treatment with antimicrobial peptide cathelicidin was protective and, in acute cystitis, targeting overactive effector molecules such as IL-1β, MMP7, COX2, cAMP and the pain-sensing receptor NK1R has been successful in vivo. Furthermore, other UTI treatment strategies, such as inhibiting bacterial adhesion and vaccination, have also shown promise. Hyperactivation of innate immunity is a disease determinant in urinary tract infections (UTIs). Modulation of innate immunity has promise as a therapy for UTIs. In this Review, the authors discuss potential mechanisms and immunomodulatory therapeutic strategies in UTIs. Excessive innate immune responses to infection cause symptoms and pathology in acute pyelonephritis and acute cystitis. Innate immunomodulation therapy is, therefore, a realistic option for treating these conditions. Targeting excessive innate immune responses at the level of transcription has been successful in animal models. Innate immunomodulation therapy reduces excessive inflammation and tissue pathology and accelerates bacterial clearance from infected kidneys and bladders in mice. Innate immunomodulation therapy also accelerates the clearance of antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
- Daniel Butler
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ines Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Björn Wullt
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
31
|
Finjan NM, Mahmood AS. Gene characterization of extended-spectrum-β-lactamase producing Klebsiella pneumoniae isolates and analysis of interleukin-11 in patients with urinary tract infection. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Wang AS, Steers NJ, Parab AR, Gachon F, Sweet MJ, Mysorekar IU. Timing is everything: impact of development, ageing and circadian rhythm on macrophage functions in urinary tract infections. Mucosal Immunol 2022; 15:1114-1126. [PMID: 36038769 DOI: 10.1038/s41385-022-00558-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 02/04/2023]
Abstract
The bladder supports a diversity of macrophage populations with functional roles related to homeostasis and host defense, including clearance of cell debris from tissue, immune surveillance, and inflammatory responses. This review examines these roles with particular attention given to macrophage origins, differentiation, recruitment, and engagement in host defense against urinary tract infections (UTIs), where these cells recognize uropathogens through a combination of receptor-mediated responses. Time is an important variable that is often overlooked in many clinical and biological studies, including in relation to macrophages and UTIs. Given that ageing is a significant factor in urinary tract infection pathogenesis and macrophages have been shown to harbor their own circadian system, this review also explores the influence of age on macrophage functions and the role of diurnal variations in macrophage functions in host defense and inflammation during UTIs. We provide a conceptual framework for future studies that address these key knowledge gaps.
Collapse
Affiliation(s)
- Alison S Wang
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Nicholas J Steers
- Division of Nephrology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Adwaita R Parab
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Frédéric Gachon
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD, Australia. .,Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia.
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA. .,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
33
|
Anti-inflammatory effects of Abelmoschus manihot (L.) Medik. on LPS-induced cystitis in mice: potential candidate for cystitis treatment based on classic use. Chin J Nat Med 2022; 20:321-331. [DOI: 10.1016/s1875-5364(22)60140-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Indexed: 11/19/2022]
|
34
|
Cetin N, Kiraz ZK, Gencler A. Diagnostic Value of Urine Ribonuclease 7 (RNase 7) to Creatinine Ratio for Detecting Urinary Tract Infection in Children with Pyuria. J PEDIAT INF DIS-GER 2022. [DOI: 10.1055/s-0042-1748922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Abstract
Objective Ribonuclease 7 (RNase 7) is one of the members of the antimicrobial peptides playing a role in maintaining urinary tract sterility. We aimed to investigate the predictive value of the urine RNase 7 levels in children with pyuria and associations between RNase 7 and vesicoureteral reflux (VUR) and renal scarring.
Methods This study included 109 children with pyuria (46 febrile urinary tract infections [UTIs], 38 nonfebrile UTIs, and 25 sterile pyuria) whose RNase 7 levels were measured by enzyme-linked immunoassay. The results for urine RNase 7 concentrations were expressed as micrograms per milligrams creatinine.
Results RNase 7/Cr levels were higher in patients with both febrile and nonfebrile UTIs than the patients with sterile pyuria (p = 0.001). RNase 7/Cr had predictive values of diagnosis of febrile and nonfebrile UTIs (cut-off value: 2.92 µg/mg, p = 0.003; cut-off value: 3.67 µg/mg, p < 0.001, respectively). RNase 7/Cr had higher levels in the patients with VUR than without VUR (cut-off value: 4.28 µg/mg, p = 0.037). The patients with renal scarring had higher urine RNase 7/Cr than those without scarring (cut-off value: 4.54 µg/mg, p = 0.041).
Conclusion The evaluation of RNase 7/Cr may help prevent unnecessary and/or inappropriate antibiotic use in children with pyuria. The higher RNase 7 levels in patients with VUR and renal scarring may reflect long-term inflammation or greater inflammatory response during acute infection.
Collapse
Affiliation(s)
- Nuran Cetin
- Department of Pediatric Nephrology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Zeynep Kusku Kiraz
- Department of Biochemistry, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Aylin Gencler
- Department of Pediatric Nephrology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
35
|
Lindblad A, Johansson C, Persson K, Demirel I. The role of caspase-1, caspase-4 and NLRP3 in regulating the host cell response evoked by uropathogenic Escherichia coli. Sci Rep 2022; 12:2005. [PMID: 35132157 PMCID: PMC8821701 DOI: 10.1038/s41598-022-06052-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The inflammasome-associated proteins caspase-1, caspase-4 and NLRP3 have been emphasised to be essential in the host cell response during urinary tract infection (UTI) by regulating IL-1β release. Our aim was to investigate how the inflammasome-associated proteins regulate the cell response of bladder epithelial cells during infection with uropathogenic Escherichia coli (UPEC). Human bladder epithelial cells (5637) and CRISPR/Cas9 generated caspase-1, caspase-4 and NLRP3 knockdown cells were stimulated with the UPEC strain CFT073. Using Olink proteomics and real time RT-PCR, we showed that caspase-1, caspase-4 and NLRP3 are vital for the expression of many inflammatory genes and proteins from bladder epithelial cells. When investigating the effect of inflammasome-associated proteins on neutrophils, we found that conditioned medium from UPEC-infected caspase-4 knockdown cells significantly increased phagocytosis of CFT073 and significantly decreased ROS production from neutrophils. In contrast, conditioned medium from UPEC-infected NLRP3 knockdown cells significantly decreased the phagocytosis of CFT073 and significantly increased the ROS production from neutrophils. In conclusion, we showed that the inflammasome-associated proteins contribute to the host cell response during UPEC infection.
Collapse
Affiliation(s)
- Anna Lindblad
- School of Medical Sciences, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden
| | - Charlotte Johansson
- School of Medical Sciences, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden
| | - Katarina Persson
- School of Medical Sciences, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, 701 82, Örebro, Sweden.
| |
Collapse
|
36
|
Campbell C, Andersson MI, Ansari MA, Moswela O, Misbah SA, Klenerman P, Matthews PC. Risk of Reactivation of Hepatitis B Virus (HBV) and Tuberculosis (TB) and Complications of Hepatitis C Virus (HCV) Following Tocilizumab Therapy: A Systematic Review to Inform Risk Assessment in the COVID-19 Era. Front Med (Lausanne) 2021; 8:706482. [PMID: 34490299 PMCID: PMC8417527 DOI: 10.3389/fmed.2021.706482] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Objectives: Tocilizumab (TCZ), an IL-6 receptor antagonist, is used in the treatment of severe COVID-19 caused by infection with SARS-CoV-2. However, unintended consequences of TCZ therapy include reactivation of tuberculosis (TB) or hepatitis B virus (HBV), and worsening of hepatitis C virus (HCV). We set out to assimilate existing data for these complications, in order to help inform evidence-based risk assessments for the use of TCZ, and thus to reduce the risk of serious but preventable complications. Methods: We searched the global WHO database of Individual Case Safety Reports (ICSRs) and adverse drug reactions (ADRs) ("VigiBase") and undertook a systematic literature review, in accordance with PRISMA guidelines. We generated mean cumulative incidence estimates for infection complications. Results: Mean cumulative incidence of HBV and TB were 3.3 and 4.3%, respectively, in patients receiving TCZ. Insufficient data were available to generate estimates for HCV. These estimates derive from heterogeneous studies pre-dating SARS-CoV-2, with differing epidemiology and varied approaches to screening and prophylaxis, so formal meta-analysis was not possible. Conclusions: We underline the need for careful individual risk assessment prior to TCZ prescription, and present an algorithm to guide clinical stratification. There is an urgent need for ongoing collation of safety data as TCZ therapy is used in COVID.
Collapse
Affiliation(s)
- Cori Campbell
- Nuffield Department of Medicine, University of Oxford, Medawar Building for Pathogen Research, Oxford, United Kingdom
| | - Monique I. Andersson
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, Oxford, United Kingdom
| | - M. Azim Ansari
- Nuffield Department of Medicine, University of Oxford, Medawar Building for Pathogen Research, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Olivia Moswela
- Pharmacy Department, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Siraj A. Misbah
- Department of Clinical Immunology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Medawar Building for Pathogen Research, Oxford, United Kingdom
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Philippa C. Matthews
- Nuffield Department of Medicine, University of Oxford, Medawar Building for Pathogen Research, Oxford, United Kingdom
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
37
|
Kula BE, Clancy CJ, Hong Nguyen M, Schwartz IS. Invasive mould disease in fatal COVID-19: a systematic review of autopsies. LANCET MICROBE 2021; 2:e405-e414. [PMID: 34189490 PMCID: PMC8221729 DOI: 10.1016/s2666-5247(21)00091-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Invasive mould disease (IMD) might affect up to a third of critically ill patients with COVID-19. COVID-19-associated pulmonary aspergillosis (CAPA) is typically diagnosed on the basis of a combination of non-specific clinical, radiographical, and mycological findings, but whether most cases represent invasive disease is unresolved. We systematically reviewed autopsy series of three or more decedents with COVID-19 for evidence of IMD. We searched PubMed, Web of Science, OVID (Embase), and medRxiv for studies in English or French published from Jan 1, 2019, to Sept 26, 2020. We identified 1070 references, of which 50 studies met the criteria. These studies described autopsies from 677 decedents, with individual-level data for 443 decedents. The median age was 70·0 years (IQR 57·0–79·0). Of decedents with individual-level data, 133 (30%) had diabetes, 97 (22%) had pre-existing lung disease, and 27 (6%) had immunocompromising conditions. Of 548 decedents with such data, 320 (58%) received invasive mechanical ventilation; among 140 decedents for whom this was known, ventilation was for a median of 9·0 days (IQR 5·0–20·0). Treatment included immunomodulation in 60 decedents and antifungals in 50 decedents. Autopsy-proven IMD occurred in 11 (2%) of 677 decedents, including eight CAPA, two unspecified IMD, and one disseminated mucormycosis. Among 320 decedents who received mechanical ventilation, six (2%) had IMD. We conclude that IMD, including CAPA, is an uncommon autopsy finding in COVID-19.
Collapse
Affiliation(s)
- Brittany E Kula
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cornelius J Clancy
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Hong Nguyen
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ilan S Schwartz
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
38
|
Albracht CD, Hreha TN, Hunstad DA. Sex effects in pyelonephritis. Pediatr Nephrol 2021; 36:507-515. [PMID: 32040629 PMCID: PMC7415591 DOI: 10.1007/s00467-020-04492-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/30/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
Abstract
Urinary tract infections (UTIs) are generally considered a disease of women. However, UTIs affect females throughout the lifespan, and certain male populations (including infants and elderly men) are also susceptible. Epidemiologically, pyelonephritis is more common in women but carries increased morbidity when it does occur in men. Among children, high-grade vesicoureteral reflux is a primary risk factor for upper-tract UTI in both sexes. However, among young infants with UTI, girls are outnumbered by boys; risk factors include posterior urethral valves and lack of circumcision. Recent advances in mouse models of UTI reveal sex differences in innate responses to UTI, which vary somewhat depending on the system used. Moreover, male mice and androgenized female mice suffer worse outcomes in experimental pyelonephritis; evidence suggests that androgen exposure may suppress innate control of infection in the urinary tract, but additional androgen effects, as well as non-hormonal sex effects, may yet be specified. Among other intriguing directions, recent experiments raise the hypothesis that the postnatal testosterone surge that occurs in male infants may represent an additional factor driving the higher incidence of UTI in males under 6 months of age. Ongoing work in contemporary models will further illuminate sex- and sex-hormone-specific effects on UTI pathogenesis and immune responses.
Collapse
Affiliation(s)
- Clayton D Albracht
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO, 63110, USA
| | - Teri N Hreha
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO, 63110, USA
| | - David A Hunstad
- Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8208, St. Louis, MO, 63110, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, 660 S. Euclid Ave., Campus Box 8230, St. Louis, MO, 63110, USA.
| |
Collapse
|
39
|
Lin L, Huang K, Guo W, Zhou C, Wang G, Zhao Q. Conditioned medium of the osteosarcoma cell line U2OS induces hBMSCs to exhibit characteristics of carcinoma-associated fibroblasts via activation of IL-6/STAT3 signalling. J Biochem 2021; 168:265-271. [PMID: 32302384 DOI: 10.1093/jb/mvaa044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/31/2020] [Indexed: 12/14/2022] Open
Abstract
As a research hotspot in recent years, bone mesenchymal stem cells (BMSCs) play an important role in the process of a variety of human diseases, including cancers. However, in osteosarcoma, the role of BMSCs and their communication with tumour cells are not clear. In this study, we validated the communication of osteosarcoma (OS) cells with BMSCs. The results showed that the conditioned medium of osteosarcoma cell line U2OS (U2OS-CM) induces the carcinoma-associated fibroblasts (CAFs)-like transformation of BMSCs and promotes the proliferation, migration and invasion of BMSCs. Mechanistically, treatment of human bone mesenchymal stem cells (hBMSCs) with U2OS-CM results in a significant increase in the IL-6 expression and phosphorylation of STAT3. Furthermore, blockade of the IL-6/STAT3 signalling in hBMSCs rescues the transformation of CAF phenotype induced by U2OS-CM. And, human IL-6 can directly increase the expression of the CAF marker genes in hMSCs. Meanwhile, IL-6/STAT3 signalling involves in promoting effects of U2OS-CM on the proliferation, migration and invasion of BMSCs. In summary, our results suggest that BMSCs communicate with OS cells through IL-6/STAT3 signalling and play an important role in the progress of osteosarcoma.
Collapse
Affiliation(s)
- Longshuai Lin
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Kai Huang
- Department of Orthopedics, Shanghai Jingan Zhaibei Hospital, Shanghai 200070, China
| | - Weihong Guo
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Chenghao Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Gangyang Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qinghua Zhao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
40
|
Yue R, Wei X, Zhao J, Zhou Z, Zhong W. Essential Role of IFN-γ in Regulating Gut Antimicrobial Peptides and Microbiota to Protect Against Alcohol-Induced Bacterial Translocation and Hepatic Inflammation in Mice. Front Physiol 2021; 11:629141. [PMID: 33536944 PMCID: PMC7848155 DOI: 10.3389/fphys.2020.629141] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
The mechanisms by which alcohol provokes bacterial translocation in the development of alcoholic liver disease (ALD) remain incompletely defined. Our previous study demonstrates that impaired gut epithelial antimicrobial defense is critically involved in the pathogenesis of ALD. The study was set to determine the mechanisms of how alcohol inhibits the antimicrobial ability of intestinal epithelial cells (IECs) and to explore possible solutions to this issue. C57BL/6J mice were fed either alcohol or isocaloric dextrin liquid diet for 8 weeks, and intestinal IFN-γ-signal transducer and activator of transcription (STAT) signaling was analyzed. We found that chronic alcohol exposure led to a significant reduction in intestinal IFN-γ levels compared to a control; the protein levels of phosphorylated STAT1 (p-STAT1) and p-STAT3 were both declined by alcohol. We then tested the effects of IFN-γ-STAT signaling on regulating antimicrobial peptides (AMPs), gut microbiota, and disease progression of ALD in a mouse model of chronic alcohol feeding, time-course acute IFN-γ treatment, and in vivo and in vitro IEC-specific STAT1 or STAT3 knockout mouse models, respectively. Administration of IFN-γ activated intestinal STAT1 and STAT3, upregulated the expression of Reg3 and α-defensins, orchestrated gut microbiota, and reversed alcohol-induced intestinal ZO-1 disruption and systemic endotoxin elevation as well as hepatic inflammation. Meanwhile, acute IFN-γ treatment time-dependently induced AMP expression and α-defensin activation. We then dissected the roles of STAT1 and STAT3 in this progress. Lack of IEC-specific STAT3 inhibited IFN-γ-induced expression of Reg3 and α-defensins and hindered activation of α-defensins via inactivating matrix metallopeptidase 7 (MMP7), whereas lack of IEC-specific STAT1 impaired IFN-γ-stimulated expression of α-defensins and the IEC marker, sodium-hydrogen exchanger 3. Lastly, we found that interleukin (IL)-18, a known IFN-γ inducer, was also reduced by alcohol in mice. IL-18 treatment to alcohol-fed mice normalized gut IFN-γ levels and ameliorated organ damages in both the intestine and liver. Taken together, the study reveals that IFN-γ is critically involved in the regulation of AMPs through regulation of STAT1 and STAT3; impaired IFN-γ-STAT signaling provides an explanation for alcohol-induced gut antimicrobial dysfunction and microbial dysbiosis. Therefore, IFN-γ remains a promising host defense-enhancing cytokine with unexplored clinical potential in ALD therapy.
Collapse
Affiliation(s)
- Ruichao Yue
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| | - Xiaoyuan Wei
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, United States
| | - Jiangchao Zhao
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, United States
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States.,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| | - Wei Zhong
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States.,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC, United States
| |
Collapse
|
41
|
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
|
42
|
Horváth J, Wullt B, Naber KG, Köves B. Biomarkers in urinary tract infections - which ones are suitable for diagnostics and follow-up? GMS INFECTIOUS DISEASES 2020; 8:Doc24. [PMID: 33299741 PMCID: PMC7705555 DOI: 10.3205/id000068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Urinary tract infections (UTIs) are one of the most common infections worldwide. Under special circumstances, clinicians must rely on laboratory findings, which might have a weak predicting value, misguiding the practitioners and leading to incorrect diagnosis and overuse of antibiotics. Therefore, there is an urgent need for reliable biomarkers in UTIs. Methods: We performed a literature search for biomarkers used in UTIs from January 1999 until May 2020. We used "urinary tract infection" and "biomarker" as the main key words in the PubMed, Medline and Cochrane databases. After peer review, we excluded the duplicates and identified the suitable articles, from which we collected the data and divided the available biomarkers into 5 groups: i) conventional markers; ii) promising, thoroughly studied biomarkers; iii) promising biomarkers that need further studies; iv) biomarkers of unknown significance; v) controversial, not useful markers. Results: We found 131 articles, mostly from the paediatric population. Neutrophil gelatinase-associated lipocalin (NGAL) and interleukins (IL) have a leading role in diagnosing and differentiating UTIs based on a lot of observational, comparative trials. Heparin Binding Protein (HBP), Lactoferrin (LF), Heat-Shock Protein-70 (HSP-70), Human Defensin-5 (HD-5), Lipopolysaccharide Binding Protein (LBP) and mass spectrometry studies are promising, but confirming data are lacking. The measurable components of the innate immune system and local host cell response could be appropriate biomarkers, but their significance is currently unknown. Conclusions: Conventional biomarkers for UTIs have low specificity. The use of urinary NGAL and interleukins could improve the sensitivity and specificity of laboratory diagnosis of UTIs.
Collapse
Affiliation(s)
- József Horváth
- BKMK SZTE ÁOK Okt. Kh. Urológiai Osztálya, Kecskemét, Hungary
| | - Björn Wullt
- Division of Microbiology, Immunology and Glycobiology, Lund University, Lund, Sweden
| | - Kurt G. Naber
- Department of Urology, Technical University of Munich, Munich, Germany
| | - Béla Köves
- Jahn Ferenc Dél-pesti Kórház és Rendelőintézet, Budapest, Hungary
| |
Collapse
|
43
|
Yu Y, Yang W, Bilotta AJ, Yu Y, Zhao X, Zhou Z, Yao S, Xu J, Zhou J, Dann SM, Li Y, Cong Y. STING controls intestinal homeostasis through promoting antimicrobial peptide expression in epithelial cells. FASEB J 2020; 34:15417-15430. [PMID: 32969062 PMCID: PMC7606834 DOI: 10.1096/fj.202001524r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022]
Abstract
Stimulator of interferon genes (STING) has been shown to play a critical role in orchestrating immune responses to various pathogens through sensing cyclic dinucleotides. However, how STING regulates intestinal homeostasis is still not completely understood. In this study, we found that STING-/- mice were more susceptible to enteric infection with Citrobacter rodentium compared to wild-type (WT) mice evidenced by more severe intestinal inflammation and impaired bacterial clearance. STING-/- mice demonstrated lower expression of REG3γ but not β-defensins and Cramp in IECs. Consistently, STING-/- IECs showed reduced capacity to inhibit bacterial growth. STING agonists, both 10-carboxymethyl-9-acridanone (CMA) and 5,6-dimethylxanthenone-4-acetic acid (DMXAA), promoted REG3γ expression IECs. Furthermore, STING agonists promoted WT but not REG3γ-deficient IEC bacterial killing. Mechanistically, STING agonists activated STAT3 and promoted glycolysis in IECs. Inhibition of STAT3 pathway and glycolysis suppressed STING-induced REG3γ production in IECs, and abrogated STING-mediated IEC killing of C. rodentium. Additionally, treatment with the STING ligand, 2,3-cGAMP, inhibited C. rodentium-induced colitis in vivo. Overall, STING promotes IEC REG3γ expression to inhibit enteric infection and intestinal inflammation, thus, maintaining the intestinal homeostasis.
Collapse
Affiliation(s)
- Yanbo Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch
| | - Anthony J Bilotta
- Department of Microbiology and Immunology, University of Texas Medical Branch
| | - Yu Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Xiaojing Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch
| | - Zheng Zhou
- Department of Microbiology and Immunology, University of Texas Medical Branch
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch
| | - Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch
| | - Sara M Dann
- Department of Internal Medicine, University of Texas Medical Branch
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, P.R. China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch
- Department of Pathology, University of Texas Medical Branch
| |
Collapse
|
44
|
Ho CH, Lu YC, Fan CK, Yu HJ, Liu HT, Wu CC, Chen KC, Liu SP, Cheng PC. Testosterone regulates the intracellular bacterial community formation of uropathogenic Escherichia coli in prostate cells via STAT3. Int J Med Microbiol 2020; 310:151450. [PMID: 33092696 DOI: 10.1016/j.ijmm.2020.151450] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 08/06/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND UPEC can internalize clonally in prostate to form biofilm-like intracellular bacterial communities (IBCs) for recurrent or chronic infection. We previously indicated that the exposure of prostate cells to testosterone could suppress UPEC invasion and their persistent survival within cells by effectively inhibiting the JAK/STAT1 signaling pathway. However, the regulatory mechanism by which testosterone affects UPEC-induced prostatitis via STAT3, another latent transcription factor signaling pathway is still unclear. The present study aimed to clarify the role of STAT3 in the process of UPEC-induced inflammation and colonization in prostate epithelial cells. METHODS The effects of testosterone-mediated inhibition were compared between the prostatitis by different UPEC strains (CFT073 and J96) through the specific GFP-UPEC-infected prostate cell model. Fluorescence microscopy was used for UPEC IBCs detection and quantifying, and Flow cytometry, RT-PCR and western blotting were used for analyzing related gene and protein expressions. Pretreatment of JAK and STAT3 inhibitors were also applied to verify the regulation of transduction pathway in testosterone-mediated anti-UPEC infection. RESULTS This study revealed that testosterone effectively suppresses UPEC infection and IBC formation in prostate cells through the JAK/STAT3 pathway. The results show that CFT073 and J96 UPEC infection rates and colony numbers were dose-dependently reduced in RWPE-1 cells pretreated with 5 and 20 μg/mL testosterone at 0 and 24 h post-infection. Further, testosterone reduced the amounts of UPEC infecting and surviving within the prostate cells, as well as suppressed the size of IBCs formed. We demonstrated that pretreating testosterone effectively inhibited UPEC infection along with dose-dependent suppression of STAT3 and the phosphorylated-STAT3 expression in prostate cells, especially in 24 h J96 UPEC infected groups. The STAT inhibitor, SOCS3 also up-regulated at the same time. In addition, we pretreated the JAK1 or STAT3 inhibitor with testosterone to block the signaling transduction before CFT073 and J96 UPEC infection, and found the significant restoring in both the sizes of IBCs and bacterial numbers in RWPE-1 cells. Therefore, our results suggest that the suppression of STAT3 by testosterone treatment attenuate UPEC growing within IBCs and interfere with their infection to prostate cells. CONCLUSIONS Overall, our study demonstrates that testosterone suppresses the initial infection of prostate epithelial cells by UPEC and reduces the survival of UPEC within IBCs after infection. These results indicate a critical role for STAT3 in facilitating UPEC infection and persistence, and its participation in driving testosterone-suppressive responses in prostate epithelial cells. In conclusion, this study suggests that testosterone may be beneficial in treating clinically recurrent UPEC infections and, thus, the persistent recurrence of prostatic inflammation.
Collapse
Affiliation(s)
- Chen-Hsun Ho
- Division of Urology, Department of Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Chuan Lu
- Department of Urology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chia-Kwung Fan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Center for International Tropical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Jeng Yu
- Department of Urology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hsin-Tien Liu
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Chang Wu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kuan-Chou Chen
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shih-Ping Liu
- Department of Urology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.
| | - Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Center for International Tropical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
45
|
Gupta S, Junquera GY, Nicassio L, Becknell B, Ching CB. Trans IL-6 signaling does not appear to play a role in renal scarring after urinary tract infection. J Pediatr Urol 2020; 16:586-591. [PMID: 32563691 PMCID: PMC7686088 DOI: 10.1016/j.jpurol.2020.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 04/17/2020] [Accepted: 05/09/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION While inflammation is an important innate defense mechanism against infection, it can also lead to local tissue damage. The trans signaling pathway of interleukin (IL)-6 is a known mediator of inflammation. We hypothesized that the trans IL-6 signaling pathway is associated with the development of post febrile urinary tract infection (UTI) renal scarring. OBJECTIVE To compare soluble regulators of trans IL-6 signaling between patients with a history of febrile UTI who do or do not have renal scarring. STUDY DESIGN After IRB-approval, we collected urine samples in pediatric patients with a history of febrile (≥38 °C) UTI (urine culture >50 K uropathogen) with documented presence or absence of renal scarring on imaging. Samples were collected at a time when patients were not actively infected. Enzyme-linked immunosorbent assays were performed on samples for markers of trans IL-6 signaling: IL-6, soluble (s) IL-6 receptor (R), and soluble (s)gp130, a buffer in trans IL-6 signaling. Values were normalized to urine creatinine. Results were analyzed by t-test or Mann-Whitney U. Spearman rank correlation was used. A p-value of <0.05 was considered significant. RESULTS A total of 50 urines from patients with a history of febrile UTI were collected: 23 with and 27 without scarring. There was no difference between groups regarding age or gender. There was no significant difference in urine IL-6, sIL-6R, or sgp130 between those with and without scarring (Figure). While IL-6 values significantly correlated with sIL-6R and sgp130 in those without renal scarring, IL-6 did not correlate with sgp130 in those with scarring. Ratios of IL-6 to sgp130 and sIL-6R to sgp130 were not different between groups. DISCUSSION The inflammatory response generated in response to infection is believed to be largely responsible for the development of renal scarring after UTI. IL-6 is a cytokine known to be induced during UTI with a pro-inflammatory pathway, known as trans signaling. This study investigated for differences in markers of trans IL-6 signaling between patients with a history of febrile UTI with and without renal scarring. There was no significant difference between the absolute values or ratio of these markers between groups. CONCLUSIONS Markers of trans IL-6 signaling are not different between individuals with a history of febrile UTI with and without renal scarring in the non-acute setting.
Collapse
Affiliation(s)
- Sudipti Gupta
- Department of Pediatric Urology, Nationwide Children's Hospital, Columbus OH, USA; Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA
| | | | - Lauren Nicassio
- Department of Pediatric Urology, Nationwide Children's Hospital, Columbus OH, USA
| | - Brian Becknell
- Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA; Division of Pediatric Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus OH, USA
| | - Christina B Ching
- Department of Pediatric Urology, Nationwide Children's Hospital, Columbus OH, USA; Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus OH, USA.
| |
Collapse
|
46
|
Moazzezy N, Asadi Karam MR, Rafati S, Bouzari S, Oloomi M. A Synthetic Peptide 2Abz 23S 29 Reduces Bacterial Titer and Induces Pro-Inflammatory Cytokines in a Murine Model of Urinary Tract Infection. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2797-2807. [PMID: 32764879 PMCID: PMC7381768 DOI: 10.2147/dddt.s259937] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/06/2020] [Indexed: 01/19/2023]
Abstract
Introduction A urinary tract infection (UTI), which is often caused by uropathogenic E. coli (UPEC) strains, affects many people worldwide annually. UPEC causes the production of pro-inflammatory cytokines by the bladder epithelial cells; however, it has been proven that the UPEC can inhibit the early activation of the innate immune system. Methods This study aimed to examine the antibacterial and immunomodulatory effects of different doses of truncated alpha-defensins (human neutrophil peptide (HNP)-1) analog 2Abz23S29 on the mouse UTI model. Experimentally uropathogenic E. coli CFT073-infected mice were treated with low-dose 2Abz23S29 (250µg/mL), high-dose 2Abz23S29 (750µg/mL), ciprofloxacin (cip) (800µg/mL), or high-dose 2Abz23S29plus cip once a day 24 h post-infection. The 2Abz23S29 and cip treatment were given for two consecutive days. Results The in vivo results showed that fewer UPEC were recovered from the bladders of mice treated transurethrally with 2Abz23S29. Moreover, low-dose 2Abz23S29 significantly decreased the level of the interleukin-6 (IL-6), whereas high-dose 2Abz23S29 increased pro-inflammatory cytokines including IL-6, macrophage inflammatory protein/2 (MIP/2), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) in infected bladders of mice. Besides, the levels of cytokines IL-6 and MIP/2 in infected mice treated with a combination of high-dose 2Abz23S29 and cip were significantly higher than the untreated mice. In contrast, CFT073-infected mice treated with a combination of high-dose 2Abz23S29 and cip showed no changes in cytokines TNF-α and IL-1β levels, indicating that ciprofloxacin may play an anti-inflammatory role. Conclusion Collectively, apart from the direct antibacterial role of 2Abz23S29, our data illustrated that 2Abz23S29 modulates pro-inflammatory cytokine production of bladder in a dose-dependent manner, which has implications for the development of new anti-infective agents.
Collapse
Affiliation(s)
- Neda Moazzezy
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Sima Rafati
- Immunotherapy and Leishmania Vaccine Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Saeid Bouzari
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mana Oloomi
- Molecular Biology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
47
|
Ching C, Schwartz L, Spencer JD, Becknell B. Innate immunity and urinary tract infection. Pediatr Nephrol 2020; 35:1183-1192. [PMID: 31197473 PMCID: PMC6908784 DOI: 10.1007/s00467-019-04269-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/17/2019] [Accepted: 04/25/2019] [Indexed: 01/31/2023]
Abstract
Urinary tract infections are a severe public health problem. The emergence and spread of antimicrobial resistance among uropathogens threaten to further compromise the quality of life and health of people who develop acute and recurrent upper and lower urinary tract infections. The host defense mechanisms that prevent invasive bacterial infection are not entirely delineated. However, recent evidence suggests that versatile innate immune defenses play a key role in shielding the urinary tract from invading uropathogens. Over the last decade, considerable advances have been made in defining the innate mechanisms that maintain immune homeostasis in the kidney and urinary tract. When these innate defenses are compromised or dysregulated, pathogen susceptibility increases. The objective of this review is to provide an overview of how basic science discoveries are elucidating essential innate host defenses in the kidney and urinary tract. In doing so, we highlight how these findings may ultimately translate into the clinic as new biomarkers or therapies for urinary tract infection.
Collapse
Affiliation(s)
- Christina Ching
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Urology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Laura Schwartz
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - John David Spencer
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Pediatric Nephrology, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA
| | - Brian Becknell
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, OH, USA.
- Center of Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
- Division of Pediatric Nephrology, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, 43205, USA.
| |
Collapse
|
48
|
Unique IL-13Rα2/STAT3 mediated IL-13 regulation detected in lung conventional dendritic cells, 24 h post viral vector vaccination. Sci Rep 2020; 10:1017. [PMID: 31974500 PMCID: PMC6978450 DOI: 10.1038/s41598-020-57815-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/30/2019] [Indexed: 12/19/2022] Open
Abstract
This study demonstrates that 24 h following viral vector-based vaccination IL-13Rα2 functions as a master sensor on conventional dendritic cells (cDCs), abetted by high protein stability coupled with minimal mRNA expression, to rapidly regulate DC mediated IL-13 responses at the lung mucosae, unlike IL-13Rα1. Under low IL-13, IL-13Rα2 performs as a primary signalling receptor, whilst under high IL-13, acts to sequester IL-13 to maintain homeostasis, both in a STAT3-dependent manner. Likewise, we show that viral vector-derived IL-13 levels at the vaccination site can induce differential STAT3/STAT6 paradigms in lung cDC, that can get regulated collaboratively or independently by TGF-β1 and IFN-γ. Specifically, low IL-13 responses associated with recombinant Fowlpox virus (rFPV) is regulated by early IL-13Rα2, correlated with STAT3/TGF-β1 expression. Whilst, high IL-13 responses, associated with recombinant Modified Vaccinia Ankara (rMVA) is regulated in an IL-13Rα1/STAT6 dependent manner associated with IFN-γR expression bias. Different viral vaccine vectors have previously been shown to induce unique adaptive immune outcomes. Taken together current observations suggest that IL-13Rα2-driven STAT3/STAT6 equilibrium at the cDC level may play an important role in governing the efficacy of vector-based vaccines. These new insights have high potential to be exploited to improve recombinant viral vector-based vaccine design, according to the pathogen of interest and/or therapies against IL-13 associated disease conditions.
Collapse
|
49
|
Spatial proteomics revealed a CX 3CL1-dependent crosstalk between the urothelium and relocated macrophages through IL-6 during an acute bacterial infection in the urinary bladder. Mucosal Immunol 2020; 13:702-714. [PMID: 32112048 PMCID: PMC7312419 DOI: 10.1038/s41385-020-0269-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 02/04/2023]
Abstract
The urothelium of the urinary bladder represents the first line of defense. However, uropathogenic E. coli (UPEC) damage the urothelium and cause acute bacterial infection. Here, we demonstrate the crosstalk between macrophages and the urothelium stimulating macrophage migration into the urothelium. Using spatial proteomics by MALDI-MSI and LC-MS/MS, a novel algorithm revealed the spatial activation and migration of macrophages. Analysis of the spatial proteome unravelled the coexpression of Myo9b and F4/80 in the infected urothelium, indicating that macrophages have entered the urothelium upon infection. Immunofluorescence microscopy additionally indicated that intraurothelial macrophages phagocytosed UPEC and eliminated neutrophils. Further analysis of the spatial proteome by MALDI-MSI showed strong expression of IL-6 in the urothelium and local inhibition of this molecule reduced macrophage migration into the urothelium and aggravated the infection. After IL-6 inhibition, the expression of matrix metalloproteinases and chemokines, such as CX3CL1 was reduced in the urothelium. Accordingly, macrophage migration into the urothelium was diminished in the absence of CX3CL1 signaling in Cx3cr1gfp/gfp mice. Conclusively, this study describes the crosstalk between the infected urothelium and macrophages through IL-6-induced CX3CL1 expression. Such crosstalk facilitates the relocation of macrophages into the urothelium and reduces bacterial burden in the urinary bladder.
Collapse
|
50
|
Wang C, Li Q, Lv J, Sun X, Cao Y, Yu K, Miao C, Zhang ZS, Yao Z, Wang Q. Alpha-hemolysin of uropathogenic Escherichia coli induces GM-CSF-mediated acute kidney injury. Mucosal Immunol 2020; 13:22-33. [PMID: 31719643 PMCID: PMC6914670 DOI: 10.1038/s41385-019-0225-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 02/04/2023]
Abstract
Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs), inducing acute pyelonephritis and may result in permanent renal scarring and failure. Alpha-hemolysin (HlyA), a key UPEC toxin, causes serious tissue damage; however, the mechanism through which HlyA induces kidney injury remains unclear. In the present study, granulocyte-macrophage colony-stimulating factor (GM-CSF) secreted by renal epithelial cells was upregulated by HlyA in vitro and in vivo, which induced M1 macrophage accumulation in kidney, and ADAM10 was found involved in HlyA-induced GM-CSF. Macrophage elimination or GM-CSF neutralization protected against acute kidney injury in mice, and increased GM-CSF was detected in urine of patients infected by hlyA-positive UPEC. In addition, HlyA was found to promote UPEC invasion into renal epithelial cells by interacting with Nectin-2 in vitro. However, HlyA did not affect bacterial titers during acute kidney infections, and HlyA-induced invasion did not contribute to GM-CSF upregulation in vitro, which indicate that HlyA-induced GM-CSF is independent of bacteria invasion. The role of GM-CSF in HlyA-mediated kidney injury may lead to novel strategies to treat acute pyelonephritis.
Collapse
Affiliation(s)
- Changying Wang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Qianqian Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Junqiang Lv
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Xuan Sun
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Yang Cao
- Department of Clinical Laboratory, The Second Hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Kaiyuan Yu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Chunhui Miao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Zhi-Song Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Collaborative Innovation Center for Biotherapy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, 300350, Tianjin, China
| | - Zhi Yao
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China.
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, 300070, Tianjin, China.
| | - Quan Wang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry of China, Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China.
| |
Collapse
|