1
|
Mennitti C, Calvanese M, Gentile A, Vastola A, Romano P, Ingenito L, Gentile L, Veneruso I, Scarano C, La Monica I, Di Lorenzo R, Frisso G, D’Argenio V, Lombardo B, Scudiero O, Pero R, Laneri S. Skin Microbiome Overview: How Physical Activity Influences Bacteria. Microorganisms 2025; 13:868. [PMID: 40284707 PMCID: PMC12029778 DOI: 10.3390/microorganisms13040868] [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: 03/03/2025] [Revised: 04/05/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
The skin cannot be considered as just a barrier that protects against physical, chemical, and biological damage; it is a complex and dynamic ecosystem that varies across lifespans. Interest in the relationship between physical activity and skin microbiota has grown significantly in recent years. The skin microbiota has a crucial role in skin functions and physiology, and an imbalance, known as dysbiosis, is correlated with several diseases, such as inflammatory bowel disease (IBD), infectious disease, obesity, allergic disorders, and type 1 diabetes mellitus. Among the causes of dysbiosis, the practice of physical exercise, especially in contact sports, including wrestling, artistic gymnastics, and boating, certainly represents a predisposing factor for infectious disease. This review aims to provide an overview of the skin microbiota and its regulation, focusing on interactions between physical exercise and skin microbiota, the antimicrobial peptides (AMPs) as regulators of skin microbiota, and the impact of probiotics supplementation on physical performance.
Collapse
Affiliation(s)
- Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
| | - Mariella Calvanese
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
| | - Alessandro Gentile
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
| | - Aniello Vastola
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
| | - Pietro Romano
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
| | - Laura Ingenito
- Integrated Department of Laboratory and Transfusion Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (L.G.)
| | - Luca Gentile
- Integrated Department of Laboratory and Transfusion Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (L.G.)
| | - Iolanda Veneruso
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
| | - Carmela Scarano
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
| | - Ilaria La Monica
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
| | - Ritamaria Di Lorenzo
- Department of Pharmacy, University of Naples Federico II, Via Montesano, 80138 Naples, Italy; (R.D.L.); (S.L.)
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
| | - Valeria D’Argenio
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Via di Val Cannuta 247, 00166 Roma, Italy
| | - Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Via G. Salvatore 486, 80145 Naples, Italy; (I.L.M.); (V.D.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Raffaela Pero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, Via Sergio Pansini 5, 80131 Napoli, Italy; (C.M.); (M.C.); (A.G.); (A.V.); (P.R.); (I.V.); (C.S.); (G.F.); (B.L.); (O.S.)
- Task Force on Microbiome Studies, University of Naples Federico II, 80100 Naples, Italy
| | - Sonia Laneri
- Department of Pharmacy, University of Naples Federico II, Via Montesano, 80138 Naples, Italy; (R.D.L.); (S.L.)
| |
Collapse
|
2
|
Salamon K, Linn-Peirano S, Simoni A, Ruiz-Rosado JDD, Becknell B, John P, Schwartz L, Spencer JD. Analysing the influence of dapagliflozin on urinary tract infection vulnerability and kidney injury in mice infected with uropathogenic Escherichia coli. Diabetes Obes Metab 2025; 27:40-53. [PMID: 39344841 PMCID: PMC11620950 DOI: 10.1111/dom.15981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
AIM Sodium-glucose co-transporter-2 (SGLT2) inhibitors have revolutionized clinical medicine, but their association with urinary tract infection (UTI) risk remains debated. This study investigates the influence of dapagliflozin on UTI outcomes, focusing on kidney injury. MATERIALS AND METHODS Female non-diabetic C57BL/6J and C3H/HeOuJ mice, along with diabetic db/db mice, were orally administered dapagliflozin (1 mg/kg or 10 mg/kg) for 7 days before transurethral uropathogenic Escherichia coli (UPEC) infection. Mice were killed either 24 h after UTI or after six additional days of dapagliflozin treatment. UPEC titers were enumerated, and kidney histopathology, injury, fibrosis and function were assessed. RESULTS Vehicle- and dapagliflozin-treated C57BL/6J mice exhibited similar urine and bladder UPEC titers, with minimal kidney burden 24 h after UTI. In C3H/HeOuJ mice, UPEC burden was comparable in vehicle- and 1 mg/kg dapagliflozin-treated groups both 24 h and 7 days after UTI. However, C3H/HeOuJ mice receiving 10 mg/kg dapagliflozin had increased UPEC titers in the urine, bladder and kidneys at both endpoints. Kidney injury and fibrosis markers, as well as kidney function, were similar in vehicle and dapagliflozin groups. In diabetic db/db mice receiving dapagliflozin, UPEC strain UTI89 titers were reduced 7 days after UTI compared to vehicle-treated mice, but no difference in UPEC titers was observed when mice were infected with UPEC strain CFT073. Kidney injury and fibrosis markers and kidney function remained similar across treatment groups. CONCLUSIONS Dapagliflozin does not consistently influence UTI susceptibility and shows limited impact on kidney injury or fibrosis, suggesting SGLT2 inhibitors have minimal effects on UTI-related kidney complications.
Collapse
Affiliation(s)
- Kristin Salamon
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
| | - Sarah Linn-Peirano
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
- The Ohio State University College of Veterinary Medicine, Columbus, OH USA
- Department of Biomedical and Diagnostic Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN USA
| | - Aaron Simoni
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
| | - Juan de Dios Ruiz-Rosado
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
- The Ohio State University College of Medicine, Columbus, OH USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
- The Ohio State University College of Medicine, Columbus, OH USA
| | - Preeti John
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
| | - Laura Schwartz
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
- The Ohio State University College of Medicine, Columbus, OH USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children’s, Columbus, OH USA
- The Ohio State University College of Medicine, Columbus, OH USA
| |
Collapse
|
3
|
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
|
4
|
Imamura T, Komatsu S, Nishibeppu K, Kiuchi J, Ohashi T, Konishi H, Shiozaki A, Yamamoto Y, Moriumura R, Ikoma H, Ochiai T, Otsuji E. Urinary microRNA-210-3p as a novel and non-invasive biomarker for the detection of pancreatic cancer, including intraductal papillary mucinous carcinoma. BMC Cancer 2024; 24:907. [PMID: 39069624 DOI: 10.1186/s12885-024-12676-x] [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: 08/18/2023] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND This study aims to explore novel microRNAs in urine for screening and predicting clinical characteristics in pancreatic cancer (PC) patients using a microRNA array-based approach. METHODS We used the Toray® 3D-Gene microRNA array-based approach to compare urinary levels between PC patients and healthy volunteers. RESULTS (1) Four oncogenic microRNAs (miR-744-5p, miR-572, miR-210-3p, and miR-575) that were highly upregulated in the urine of PC patients compared to healthy individuals were identified by comprehensive microRNA array analysis. (2) Test-scale analysis by quantitative RT-PCR for each group of 20 cases showed that miR-210-3p was significantly upregulated in the urine of PC patients compared to healthy individuals (P = 0.009). (3) Validation analysis (58 PC patients and 35 healthy individuals) confirmed that miR-210-3p was significantly upregulated in the urine of PC patients compared to healthy individuals (P < 0.001, area under the receiver operating characteristic curve = 0.79, sensitivity: 0.828, specificity: 0.743). We differentiated PC patients into invasive ductal carcinoma (IDCa) and intraductal papillary mucinous carcinoma (IPMC) groups. In addition to urinary miR-210-3p levels being upregulated in IDCa over healthy individuals (P = 0.009), urinary miR-210-3p levels were also elevated in IPMC over healthy individuals (P = 0.0018). Urinary miR-210-3p can differentiate IPMC from healthy individuals by a cutoff of 8.02 with an AUC value of 0.762, sensitivity of 94%, and specificity of 63%. (4) To test whether urinary miR210-3p levels reflected plasma miR-210-3p levels, we examined the correlation between urinary and plasma levels. Spearman's correlation analysis showed a moderate positive correlation (ρ = 0.64, P = 0.005) between miR-210-3p expression in plasma and urine. CONCLUSIONS Urinary miR-210-3p is a promising, non-invasive diagnostic biomarker of PC, including IPMC. TRIAL REGISTRATION Not applicable.
Collapse
MESH Headings
- Humans
- MicroRNAs/urine
- MicroRNAs/blood
- MicroRNAs/genetics
- Female
- Male
- Biomarkers, Tumor/urine
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/blood
- Pancreatic Neoplasms/urine
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/blood
- Middle Aged
- Aged
- Adenocarcinoma, Mucinous/urine
- Adenocarcinoma, Mucinous/genetics
- Adenocarcinoma, Mucinous/diagnosis
- ROC Curve
- Case-Control Studies
- Gene Expression Regulation, Neoplastic
- Adult
- Carcinoma, Pancreatic Ductal/urine
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/blood
Collapse
Affiliation(s)
- Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yusuke Yamamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Ryo Moriumura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshiya Ochiai
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii- cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
5
|
Schwartz L, Salamon K, Simoni A, Eichler T, Jackson AR, Murtha M, Becknell B, Kauffman A, Linn-Peirano S, Holdsworth N, Tyagi V, Tang H, Rust S, Cortado H, Zabbarova I, Kanai A, Spencer JD. Insulin receptor signaling engages bladder urothelial defenses that limit urinary tract infection. Cell Rep 2024; 43:114007. [PMID: 38517889 PMCID: PMC11094371 DOI: 10.1016/j.celrep.2024.114007] [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: 09/07/2023] [Revised: 02/10/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024] Open
Abstract
Urinary tract infections (UTIs) commonly afflict people with diabetes. To better understand the mechanisms that predispose diabetics to UTIs, we employ diabetic mouse models and altered insulin signaling to show that insulin receptor (IR) shapes UTI defenses. Our findings are validated in human biosamples. We report that diabetic mice have suppressed IR expression and are more susceptible to UTIs caused by uropathogenic Escherichia coli (UPEC). Systemic IR inhibition increases UPEC susceptibility, while IR activation reduces UTIs. Localized IR deletion in bladder urothelium promotes UTI by increasing barrier permeability and suppressing antimicrobial peptides. Mechanistically, IR deletion reduces nuclear factor κB (NF-κB)-dependent programming that co-regulates urothelial tight junction integrity and antimicrobial peptides. Exfoliated urothelial cells or urine samples from diabetic youths show suppressed expression of IR, barrier genes, and antimicrobial peptides. These observations demonstrate that urothelial insulin signaling has a role in UTI prevention and link IR to urothelial barrier maintenance and antimicrobial peptide expression.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA
| | - Kristin Salamon
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Aaron Simoni
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Tad Eichler
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Ashley R Jackson
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA
| | - Matthew Murtha
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA
| | - Andrew Kauffman
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Tulane University, New Orleans, LA 70118, USA
| | - Sarah Linn-Peirano
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, OH 43210, USA
| | - Natalie Holdsworth
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Vidhi Tyagi
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Hancong Tang
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Steve Rust
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Hanna Cortado
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA
| | - Irina Zabbarova
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Anthony Kanai
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH 43205, USA; Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH 43205, USA.
| |
Collapse
|
6
|
Schwaderer AL, Rajadhyaksha E, Canas J, Saxena V, Hains DS. Intercalated cell function, kidney innate immunity, and urinary tract infections. Pflugers Arch 2024; 476:565-578. [PMID: 38227050 DOI: 10.1007/s00424-024-02905-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/17/2024]
Abstract
Intercalated cells (ICs) in the kidney collecting duct have a versatile role in acid-base and electrolyte regulation along with the host immune defense. Located in the terminal kidney tubule segment, ICs are among the first kidney cells to encounter bacteria when bacteria ascend from the bladder into the kidney. ICs have developed several mechanisms to combat bacterial infections of the kidneys. For example, ICs produce antimicrobial peptides (AMPs), which have direct bactericidal activity, and in many cases are upregulated in response to infections. Some AMP genes with IC-specific kidney expression are multiallelic, and having more copies of the gene confers increased resistance to bacterial infections of the kidney and urinary tract. Similarly, studies in human children demonstrate that those with history of UTIs are more likely to have single-nucleotide polymorphisms in IC-expressed AMP genes that impair the AMP's bactericidal activity. In murine models, depleted or impaired ICs result in decreased clearance of bacterial load following transurethral challenge with uropathogenic E. coli. A 2021 study demonstrated that ICs even act as phagocytes and acidify bacteria within phagolysosomes. Several immune signaling pathways have been identified in ICs which may represent future therapeutic targets in managing kidney infections or inflammation. This review's objective is to highlight IC structure and function with an emphasis on current knowledge of IC's diverse innate immune capabilities.
Collapse
Affiliation(s)
- Andrew L Schwaderer
- Division of Nephrology, Department of Pediatrics, Indiana University, 699 Riley Hospital Drive, STE 230, Indianapolis, IN, 46202, USA.
| | - Evan Rajadhyaksha
- Division of Nephrology, Department of Pediatrics, Indiana University, 699 Riley Hospital Drive, STE 230, Indianapolis, IN, 46202, USA
| | - Jorge Canas
- Division of Nephrology, Department of Pediatrics, Indiana University, 699 Riley Hospital Drive, STE 230, Indianapolis, IN, 46202, USA
| | - Vijay Saxena
- Division of Nephrology, Department of Pediatrics, Indiana University, 699 Riley Hospital Drive, STE 230, Indianapolis, IN, 46202, USA
| | - David S Hains
- Division of Nephrology, Department of Pediatrics, Indiana University, 699 Riley Hospital Drive, STE 230, Indianapolis, IN, 46202, USA
| |
Collapse
|
7
|
Kumar R, Tyagi N, Nagpal A, Kaushik JK, Mohanty AK, Kumar S. Peptidome Profiling of Bubalus bubalis Urine and Assessment of Its Antimicrobial Activity against Mastitis-Causing Pathogens. Antibiotics (Basel) 2024; 13:299. [PMID: 38666975 PMCID: PMC11047597 DOI: 10.3390/antibiotics13040299] [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: 12/29/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 04/29/2024] Open
Abstract
Urinary proteins have been studied quite exhaustively in the past, however, the small sized peptides have remained neglected for a long time in dairy cattle. These peptides are the products of systemic protein turnover, which are excreted out of the body and hence can serve as an important biomarker for various pathophysiologies. These peptides in other species of bovine have been reported to possess several bioactive properties. To investigate the urinary peptides in buffalo and simultaneously their bioactivities, we generated a peptidome profile from the urine of Murrah Buffaloes (n = 10). Urine samples were processed using <10 kDa MWCO filter and filtrate obtained was used for peptide extraction using Solid Phase Extraction (SPE). The nLC-MS/MS of the aqueous phase from ten animals resulted in the identification of 8165 peptides originating from 6041 parent proteins. We further analyzed these peptide sequences to identify bioactive peptides and classify them into anti-cancerous, anti-hypertensive, anti-microbial, and anti-inflammatory groups with a special emphasis on antimicrobial properties. With this in mind, we simultaneously conducted experiments to evaluate the antimicrobial properties of urinary aqueous extract on three pathogenic bacterial strains: S. aureus, E. coli, and S. agalactiae. The urinary peptides observed in the study are the result of the activity of possibly 76 proteases. The GO of these proteases showed the significant enrichment of the antibacterial peptide production. The total urinary peptide showed antimicrobial activity against the aforementioned pathogenic bacterial strains with no significant inhibitory effects against a buffalo mammary epithelial cell line. Just like our previous study in cows, the present study suggests the prime role of the antimicrobial peptides in the maintenance of the sterility of the urinary tract in buffalo by virtue of their amino acid composition.
Collapse
Affiliation(s)
- Rohit Kumar
- Cell Biology and Proteomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Nikunj Tyagi
- Cell Biology and Proteomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Anju Nagpal
- Cell Biology and Proteomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Jai Kumar Kaushik
- Cell Biology and Proteomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Ashok Kumar Mohanty
- ICAR-Indian Veterinary Research Institute, Mukteshwar 263138, Uttarakhand, India
| | - Sudarshan Kumar
- Cell Biology and Proteomics Lab., Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal 132001, Haryana, India
| |
Collapse
|
8
|
Jebbia M, Gupta S, Klamer BG, Pavlek L, Ching CB, Mohamed TH, Becknell B. Concentration of novel urinary tract infection biomarkers in neonates. Sci Rep 2024; 14:2996. [PMID: 38316971 PMCID: PMC10844638 DOI: 10.1038/s41598-024-53486-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/01/2024] [Indexed: 02/07/2024] Open
Abstract
Urinary tract infections (UTIs) are a common comorbidity in hospitalized neonates. The current UTI diagnostics have several limitations including invasive collection of urinary samples to ensure sterility, risk of contamination and lack of consensus definitions of UTI based on urine culture. Antimicrobial peptides (AMPs) have been recently utilized as novel biomarkers that can efficiently and accurately diagnose pediatric UTI. However, the concentration of AMPs in neonatal urine is not well-defined. Urine from neonates admitted to a single level IV neonatal intensive care unit was obtained to determine baseline concentration of two AMPs, Ribonuclease 7 (RNase 7) and Beta Defensin-1 (BD-1) and to define the relationship between AMP concentration and gestational age (GA). AMP levels were normalized to urine creatinine. RNase 7 and BD-1 were expressed in neonatal urine (n = 66) regardless of GA and as early as 22 weeks gestation. Urinary concentrations of both AMPs decreased as GA and birthweight increased. The overall median urinary RNase 7/UCr and BD-1/UCr values were 271 ng/mg, and 116 ng/mg, respectively. Median urinary concentrations of RNase 7/UCr for infants born at < 27, 27-32, 33-35 and ≥ 36 weeks were 569, 308, 254, and 124 ng/mg respectively. Similarly, the concentrations of BD-1/UCr at these GA were 166, 115, 108, and 14 ng/mg, respectively. Baseline neonatal urinary concentration of two AMPs (RNase 7 and BD-1) and the variation by GA were identified. This is an essential first step toward the potential utilization of AMPs in improving neonatal UTI diagnostics.
Collapse
Affiliation(s)
- Maria Jebbia
- The Kidney and Urinary Tract Center at Nationwide Children's Hospital, 700 Children's Dr, Columbus, OH, USA
- Division of Perinatal Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Sudipti Gupta
- The Kidney and Urinary Tract Center at Nationwide Children's Hospital, 700 Children's Dr, Columbus, OH, USA
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Brett G Klamer
- Biostatistics Resource at Nationwide Children's Hospital, Columbus, OH, USA
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Leeann Pavlek
- Division of Perinatal Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christina B Ching
- The Kidney and Urinary Tract Center at Nationwide Children's Hospital, 700 Children's Dr, Columbus, OH, USA
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Pediatric Urology, Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Tahagod H Mohamed
- The Kidney and Urinary Tract Center at Nationwide Children's Hospital, 700 Children's Dr, Columbus, OH, USA.
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA.
| | - Brian Becknell
- The Kidney and Urinary Tract Center at Nationwide Children's Hospital, 700 Children's Dr, Columbus, OH, USA
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
9
|
Dickson K, Zhou J, Lehmann C. Lower Urinary Tract Inflammation and Infection: Key Microbiological and Immunological Aspects. J Clin Med 2024; 13:315. [PMID: 38256450 PMCID: PMC10816374 DOI: 10.3390/jcm13020315] [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: 11/27/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
The urinary system, primarily responsible for the filtration of blood and waste, is affected by several infectious and inflammatory conditions. Focusing on the lower tract, this review outlines the physiological and immune landscape of the urethra and bladder, addressing key immunological and microbiological aspects of important infectious/inflammatory conditions. The conditions addressed include urethritis, interstitial cystitis/bladder pain syndrome, urinary tract infections, and urosepsis. Key aspects of each condition are addressed, including epidemiology, pathophysiology, and clinical considerations. Finally, therapeutic options are outlined, highlighting gaps in the knowledge and novel therapeutic approaches.
Collapse
Affiliation(s)
- Kayle Dickson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Juan Zhou
- Department of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Christian Lehmann
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Anesthesiology, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
10
|
Saxena V, Arregui S, Zhang S, Canas J, Qin X, Hains DS, Schwaderer AL. Generation of Atp6v1g3-Cre mice for investigation of intercalated cells and the collecting duct. Am J Physiol Renal Physiol 2023; 325:F770-F778. [PMID: 37823193 PMCID: PMC10881235 DOI: 10.1152/ajprenal.00137.2023] [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: 05/22/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
Kidney intercalated cells (ICs) maintain acid-base homeostasis and recent studies have demonstrated that they function in the kidney's innate defense. To study kidney innate immune function, ICs have been enriched using vacuolar ATPase (V-ATPase) B1 subunit (Atp6v1b1)-Cre (B1-Cre) mice. Although Atp6v1b1 is considered kidney specific, it is expressed in multiple organ systems, both in mice and humans, raising the possibility of off-target effects when using the Cre-lox system. We have recently shown using single-cell RNA sequencing that the gene that codes for the V-ATPase G3 subunit (mouse gene: Atp6v1g3; human gene: ATP6V1G3; protein abbreviation: G3) mRNA is selectively enriched in human kidney ICs. In this study, we generated Atp6v1g3-Cre (G3-Cre) reporter mice using CRISPR/CAS technology and crossed them with Tdtomatoflox/flox mice. The resultant G3-Cre+Tdt+ progeny was evaluated for kidney specificity in multiple tissues and found to be highly specific to kidney cells with minimal or no expression in other organs evaluated compared with B1-Cre mice. Tdt+ cells were flow sorted and were enriched for IC marker genes on RT-PCR analysis. Next, we crossed these mice to ihCD59 mice to generate an IC depletion mouse model (G3-Cre+ihCD59+/+). ICs were depleted in these mice using intermedilysin, which resulted in lower blood pH, suggestive of a distal renal tubular acidosis phenotype. The G3-Cre mice were healthy, bred normally, and produce regular-sized litter. Thus, this new "IC reporter" mice can be a useful tool to study ICs.NEW & NOTEWORTHY This study details the development, validation, and experimental use of a new mouse model to study the collecting duct and intercalated cells. Kidney intercalated cells are a cell type increasingly recognized to be important in several human diseases including kidney infections, acid-base disorders, and acute kidney injury.
Collapse
Affiliation(s)
- Vijay Saxena
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Samuel Arregui
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Shaobo Zhang
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Jorge Canas
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States
| | - David S Hains
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Andrew L Schwaderer
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
11
|
Schwartz L, de Dios Ruiz-Rosado J, Stonebrook E, Becknell B, Spencer JD. Uropathogen and host responses in pyelonephritis. Nat Rev Nephrol 2023; 19:658-671. [PMID: 37479904 PMCID: PMC10913074 DOI: 10.1038/s41581-023-00737-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections seen in clinical practice. The ascent of UTI-causing pathogens to the kidneys results in pyelonephritis, which can trigger kidney injury, scarring and ultimately impair kidney function. Despite sizable efforts to understand how infections develop or are cleared in the bladder, our appreciation of the mechanisms by which infections develop, progress or are eradicated in the kidney is limited. The identification of virulence factors that are produced by uropathogenic Escherichia coli to promote pyelonephritis have begun to fill this knowledge gap, as have insights into the mechanisms by which kidney tubular epithelial cells oppose uropathogenic E. coli infection to prevent or eradicate UTIs. Emerging data also illustrate how specific cellular immune responses eradicate infection whereas other immune cell populations promote kidney injury. Insights into the mechanisms by which uropathogenic E. coli circumvent host immune defences or antibiotic therapy to cause pyelonephritis is paramount to the development of new prevention and treatment strategies to mitigate pyelonephritis and its associated complications.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Juan de Dios Ruiz-Rosado
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Stonebrook
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
12
|
Chelangarimiyandoab F, Mungara P, Batta M, Cordat E. Urinary Tract Infections: Renal Intercalated Cells Protect against Pathogens. J Am Soc Nephrol 2023; 34:1605-1614. [PMID: 37401780 PMCID: PMC10561816 DOI: 10.1681/asn.0000000000000187] [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: 01/19/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023] Open
Abstract
Urinary tract infections affect more than 1 in 2 women during their lifetime. Among these, more than 10% of patients carry antibiotic-resistant bacterial strains, highlighting the urgent need to identify alternative treatments. While innate defense mechanisms are well-characterized in the lower urinary tract, it is becoming evident that the collecting duct (CD), the first renal segment encountered by invading uropathogenic bacteria, also contributes to bacterial clearance. However, the role of this segment is beginning to be understood. This review summarizes the current knowledge on CD intercalated cells in urinary tract bacterial clearance. Understanding the innate protective role of the uroepithelium and of the CD offers new opportunities for alternative therapeutic strategies.
Collapse
Affiliation(s)
- Forough Chelangarimiyandoab
- Department of Physiology and Membrane Protein Disease Research Group, Faculty of Medicine & Dentistry, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
13
|
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
|
14
|
Colceriu MC, Aldea PL, Răchișan AL, Clichici S, Sevastre-Berghian A, Mocan T. Vesicoureteral Reflux and Innate Immune System: Physiology, Physiopathology, and Clinical Aspects. J Clin Med 2023; 12:jcm12062380. [PMID: 36983379 PMCID: PMC10058356 DOI: 10.3390/jcm12062380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Vesicoureteral reflux represents one of the most concerning topics in pediatric nephrology due to its frequency, clinical expression with the potential to evolve into chronic kidney disease, and last but not least, its socio-economic implications. The presence of vesicoureteral reflux, the occurrence of urinary tract infections, and the development of reflux nephropathy, hypertension, chronic kidney disease, and finally, end-stage renal disease represent a progressive spectrum of a single physiopathological condition. For the proper management of these patients with the best clinical outcomes, and in an attempt to prevent the spread of uropathogens' resistance to antibacterial therapy, we must better understand the physiopathology of urinary tract infections in patients with vesicoureteral reflux, and at the same time, we should acknowledge the implication and response of the innate immune system in this progressive pathological condition. The present paper focuses on theoretical aspects regarding the physiopathology of vesicoureteral reflux and the interconditionality between urinary tract infections and the innate immune system. In addition, we detailed aspects regarding cytokines, interleukins, antimicrobial peptides, and proteins involved in the innate immune response as well as their implications in the physiopathology of reflux nephropathy. New directions of study should focus on using these innate immune system effectors as diagnostic and therapeutic tools in renal pathology.
Collapse
Affiliation(s)
- Marius-Cosmin Colceriu
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Paul Luchian Aldea
- Department of Community Medicine, Discipline of Public Health and Management, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Andreea-Liana Răchișan
- Department of Mother and Child, Discipline of Pediatrics II, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Simona Clichici
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Alexandra Sevastre-Berghian
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Teodora Mocan
- Department of Functional Biosciences, Discipline of Physiology, Iuliu Haţieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology, 400158 Cluj-Napoca, Romania
| |
Collapse
|
15
|
Saxena V, Arregui S, Kamocka MM, Hains DS, Schwaderer A. MAP3K7 is an innate immune regulatory gene with increased expression in human and murine kidney intercalated cells following uropathogenic Escherichia coli exposure. J Cell Biochem 2022; 123:1817-1826. [PMID: 35959632 PMCID: PMC9671826 DOI: 10.1002/jcb.30318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/28/2022] [Accepted: 08/01/2022] [Indexed: 01/09/2023]
Abstract
Understanding the mechanisms responsible for the kidney's defense against ascending uropathogen is critical to devise novel treatment strategies against increasingly antibiotic resistant uropathogen. Growing body of evidence indicate Intercalated cells of the kidney as the key innate immune epithelial cells against uropathogen. The aim of this study was to find orthologous and differentially expressed bacterial defense genes in human versus murine intercalated cells. We simultaneously analyzed 84 antibacterial genes in intercalated cells enriched from mouse and human kidney samples. Intercalated cell "reporter mice" were exposed to saline versus uropathogenic Escherichia coli (UPEC) transurethrally for 1 h in vivo, and intercalated cells were flow sorted. Human kidney intercalated cells were enriched from kidney biopsy using magnetic-activated cell sorting and exposed to UPEC in vitro for 1 h. RT2 antibacterial PCR array was performed. Mitogen-activated protein kinase kinase kinase 7 (MAP3K7) messenger RNA (mRNA) expression increased in intercalated cells of both humans and mice following UPEC exposure. Intercalated cell MAP3K7 protein expression was defined by immunofluorescence and confocal imaging analysis, was consistent with the increased MAP3K7 mRNA expression profiles defined by PCR. The presence of the orthologous innate immune gene MAP3K7/TAK1 suggests that it may be a key regulator of the intercalated cell antibacterial response and demands further investigation of its role in urinary tract infection pathogenesis.
Collapse
Affiliation(s)
- Vijay Saxena
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Samuel Arregui
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Malgorzata Maria Kamocka
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - David S. Hains
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA,Department of Pediatrics, Division of NephrologyRiley Hospital for ChildrenIndianapolisIndianaUSA
| | - Andrew Schwaderer
- Department of Pediatric NephrologyIndiana University School of MedicineIndianapolisIndianaUSA,Department of Pediatrics, Division of NephrologyRiley Hospital for ChildrenIndianapolisIndianaUSA
| |
Collapse
|
16
|
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
|
17
|
Iksanova AM, Arzumanian VG, Konanykhina SY, Samoylikov PV. Antimicrobial peptides and proteins in human biological fluids. MICROBIOLOGY INDEPENDENT RESEARCH JOURNAL 2022. [DOI: 10.18527/2500-2236-2022-9-1-37-55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Antimicrobial peptides and proteins (AMPs) are endogenous compounds that have a direct antimicrobial effect on bacteria (e. g., by disrupting bacterial membranes), as well as on fungi and viruses. AMPs are the main component of the innate immunity of living organisms and are produced by both epithelial cells (skin cells, cells of respiratory tract, intestine, urinary and genital tracts) and cells of the immune system and are secreted into secretory fluids. AMPs can also act as chemoattractants for immunocompetent cells (neutrophils, monocytes, T lymphocytes, dendritic cells) in the inflammation site and affect the antigen presenting cells by modulating adaptive T cell immune responses. The representatives of the main 15 AMP classes, that we describe in this review, are the most studied group of the large pool of these compounds. We discuss their localization, expression, and concentration in various biofluids of humans under normal and pathological conditions.
Collapse
|
18
|
Pierce KR, Eichler T, Mosquera Vasquez C, Schwaderer AL, Simoni A, Creacy S, Hains DS, Spencer JD. Ribonuclease 7 polymorphism rs1263872 reduces antimicrobial activity and associates with pediatric urinary tract infections. J Clin Invest 2021; 131:149807. [PMID: 34779412 DOI: 10.1172/jci149807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/28/2021] [Indexed: 01/18/2023] Open
Abstract
Ribonuclease 7 (RNase 7) is an antimicrobial peptide that prevents urinary tract infections (UTI); however, it is yet unknown how RNASE7 genetic variations affect its antimicrobial activity and its mitigation of UTI risk. This study determined whether the RNASE7 SNP rs1263872 is more prevalent in children with UTI and defined how rs1263872 affects RNase 7's antimicrobial activity against uropathogenic E. coli (UPEC). We performed genotyping for rs1263872 in 2 national UTI cohorts, including children enrolled in the Randomized Intervention for Children with Vesicoureteral Reflux trial or the Careful Urinary Tract Infection Evaluation study. Genotypes from these cohorts were compared with those of female controls with no UTI. To assess whether rs1263872 affects RNase 7's antimicrobial activity, we generated RNase 7 peptides and genetically modified urothelial cultures encoding wild-type RNase 7 and its variant. Compared with controls, girls in both UTI cohorts had an increased prevalence of the RNASE7 variant. Compared with the missense variant, wild-type RNase 7 peptide showed greater bactericidal activity against UPEC. Wild-type RNase 7 overexpression in human urothelial cultures reduced UPEC invasive infection compared with mutant overexpression. These results show that children with UTI have an increased prevalence of RNASE7 rs1263872, which may increase UTI susceptibility by suppressing RNase 7's antibacterial activity.
Collapse
Affiliation(s)
- Keith R Pierce
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tad Eichler
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Claudia Mosquera Vasquez
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | - Andrew L Schwaderer
- Division of Nephrology, Department of Pediatrics, and.,Kidney and Urology Translational Research Group, Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aaron Simoni
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA
| | | | - David S Hains
- Division of Nephrology, Department of Pediatrics, and.,Kidney and Urology Translational Research Group, Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - John D Spencer
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, Ohio, USA.,Division of Nephrology and Hypertension, Department of Pediatrics, The Ohio State University College of Medicine and Nationwide Children's, Columbus, Ohio, USA
| |
Collapse
|
19
|
Rademacher F, Gläser R, Harder J. Antimicrobial peptides and proteins: Interaction with the skin microbiota. Exp Dermatol 2021; 30:1496-1508. [PMID: 34310774 DOI: 10.1111/exd.14433] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/25/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022]
Abstract
The cutaneous microbiota comprises all living skin microorganisms. There is increasing evidence that the microbiota plays a crucial role in skin homeostasis. Accordingly, a dysbiosis of the microbiota may trigger cutaneous inflammation. The need for a balanced microbiota requires specific regulatory mechanisms that control and shape the microbiota. In this review, we highlight the present knowledge suggesting that antimicrobial peptides (AMPs) may exert a substantial influence on the microbiota by controlling their growth. This is supported by own data showing the differential influence of principal skin-derived AMPs on commensal staphylococci. Vice versa, we also illuminate how the cutaneous microbiota interacts with skin-derived AMPs by modulating AMP expression and how microbiota members protect themselves from the antimicrobial activity of AMPs. Taken together, the current picture suggests that a fine-tuned and well-balanced AMP-microbiota interplay on the skin surface may be crucial for skin health.
Collapse
Affiliation(s)
| | - Regine Gläser
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, Kiel University, Kiel, Germany
| |
Collapse
|
20
|
Pham CVA, Rademacher F, Hinrichs H, Beck-Jendroschek V, Harder M, Brasch J, Gläser R, Harder J. Expression of epidermal antimicrobial peptides is increased in tinea pedis. Mycoses 2021; 64:763-770. [PMID: 33797129 DOI: 10.1111/myc.13279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Tinea pedis is often chronic or recurrent, but not all individuals are equally susceptible to this infection. Dermatophytes are able to induce the expression of antimicrobial peptides and proteins (AMPs) in human keratinocytes and certain AMPs can inhibit the growth of dermatophytes. OBJECTIVE The focus of this study was to analyse the secretion of relevant AMPs, especially RNase 7, human beta-defensin-2 (hBD-2) and the S-100 protein psoriasin (S100A7), in patients with confirmed tinea pedis. METHODS To verify the diagnosis, skin scales were obtained from all patients (n = 13) and the dermatophytes were identified by potassium hydroxide mount, culture and molecular analysis. To determine the AMP concentrations, the lesional skin area of the foot was rinsed with a buffer that was subsequently analysed by ELISA. The corresponding area of the other unaffected foot as well as defined healthy skin areas of the forearm and forehead and samples from age and gender-matched healthy volunteers served as controls. RESULTS In tinea pedis patients the AMP concentrations were higher in lesional skin than in non-lesional skin and in healthy skin of controls. In particular, concentrations of hBD-2 and psoriasin were significantly elevated. CONCLUSIONS The induction of AMPs in tinea pedis might be triggered directly by the dermatophytes; furthermore, attendant inflammation and/or differentiation processes may play a role. Our results indicate that there is no defect in the constitutive expression and induction of the analysed AMPs by dermatophytes in the epidermis of affected patients. However, it is not known why the elevated AMP concentrations fail to efficiently combat dermatophyte growth.
Collapse
Affiliation(s)
- Christina Van Anh Pham
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Franziska Rademacher
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Heilwig Hinrichs
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Vera Beck-Jendroschek
- Department of Dermatology, Venerology and Allergology, Mycological Laboratory, University Hospitals of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Melanie Harder
- EUROIMMUN Medical Laboratory Diagnostic AG, Lübeck, Germany
| | - Jochen Brasch
- Department of Dermatology, Venerology and Allergology, Mycological Laboratory, University Hospitals of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Regine Gläser
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
21
|
Bender K, Schwartz LL, Cohen A, Vasquez CM, Murtha MJ, Eichler T, Thomas JP, Jackson A, Spencer JD. Expression and function of human ribonuclease 4 in the kidney and urinary tract. Am J Physiol Renal Physiol 2021; 320:F972-F983. [PMID: 33818125 PMCID: PMC8174806 DOI: 10.1152/ajprenal.00592.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
Antimicrobial peptides are essential host defense mechanisms that prevent urinary tract infections. Recent studies have demonstrated that peptides in the ribonuclease A superfamily have antimicrobial activity against uropathogens and protect the urinary tract from uropathogenic Escherichia coli (UPEC). Little is known about the antibacterial function or expression of ribonuclease 4 (RNase 4) in the human urinary tract. Here, we show that full-length recombinant RNase 4 peptide and synthetic amino-terminal RNase 4 peptide fragment have antibacterial activity against UPEC and multidrug-resistant (MDR)-UPEC. RNASE4 transcript expression was detected in human kidney and bladder tissue using quantitative real-time PCR. Immunostaining or in situ hybridization localized RNase 4 expression to proximal tubules, principal and intercalated cells in the kidney's collecting duct, and the bladder urothelium. Urinary RNase 4 concentrations were quantified in healthy controls and females with a history of urinary tract infection. Compared with controls, urinary RNase 4 concentrations were significantly lower in females with a history of urinary tract infection. When RNase 4 was neutralized in human urine or silenced in vitro using siRNA, urinary UPEC replication or attachment to and invasion of urothelial and kidney medullary cells increased. These data show that RNase 4 has antibacterial activity against UPEC, is expressed in the human urinary tract, and can contribute to host defense against urinary tract infections.NEW & NOTEWORTHY Ribonuclease 4 (RNase 4) is a newly identified host defense peptide in the human kidney and bladder. RNase 4 kills uropathogenic Escherichia coli (UPEC) and multidrug-resistant UPEC. RNase 4 prevents invasive UPEC infection and suppressed RNase 4 expression may be a risk factor for more severe or recurrent urinary tract infection.
Collapse
Affiliation(s)
- Kristin Bender
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Laura L Schwartz
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Ariel Cohen
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Claudia Mosquera Vasquez
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Matthew J Murtha
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Ohio State University College of Medicine, Columbus, Ohio
| | - Tad Eichler
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio
| | - Jason P Thomas
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| | - Ashley Jackson
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio
- The Ohio State University College of Medicine, Columbus, Ohio
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| | - John David Spencer
- Nephrology and Urology Research Affinity Group, Nationwide Children's Hospital, Columbus, Ohio
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Clinical and Translational Research, Columbus, Ohio
- The Ohio State University College of Medicine, Columbus, Ohio
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
22
|
Saxena V, Gao H, Arregui S, Zollman A, Kamocka MM, Xuei X, McGuire P, Hutchens M, Hato T, Hains DS, Schwaderer AL. Kidney intercalated cells are phagocytic and acidify internalized uropathogenic Escherichia coli. Nat Commun 2021; 12:2405. [PMID: 33893305 PMCID: PMC8065053 DOI: 10.1038/s41467-021-22672-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/18/2021] [Indexed: 02/02/2023] Open
Abstract
Kidney intercalated cells are involved in acid-base homeostasis via vacuolar ATPase expression. Here we report six human intercalated cell subtypes, including hybrid principal-intercalated cells identified from single cell transcriptomics. Phagosome maturation is a biological process that increases in biological pathway analysis rank following exposure to uropathogenic Escherichia coli in two of the intercalated cell subtypes. Real time confocal microscopy visualization of murine renal tubules perfused with green fluorescent protein expressing Escherichia coli or pHrodo Green E. coli BioParticles demonstrates that intercalated cells actively phagocytose bacteria then acidify phagolysosomes. Additionally, intercalated cells have increased vacuolar ATPase expression following in vivo experimental UTI. Taken together, intercalated cells exhibit a transcriptional response conducive to the kidney's defense, engulf bacteria and acidify the internalized bacteria. Intercalated cells represent an epithelial cell with characteristics of professional phagocytes like macrophages.
Collapse
Affiliation(s)
- Vijay Saxena
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA.
| | - Hongyu Gao
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Samuel Arregui
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA
| | - Amy Zollman
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis, IN, USA
| | - Malgorzata Maria Kamocka
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis, IN, USA
| | - Xiaoling Xuei
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Patrick McGuire
- Indiana University School of Medicine, Department of Medical & Molecular Genetics, Indianapolis, IN, USA
| | - Michael Hutchens
- Oregon Health and Science University, Department of Anesthesiology & Perioperative Medicine, Portland, OR, USA
| | - Takashi Hato
- Indiana University School of Medicine, Department of Medicine, Division of Nephrology, Indianapolis, IN, USA
| | - David S Hains
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA
| | - Andrew L Schwaderer
- Indiana University School of Medicine, Department of Pediatrics, Division of Nephrology, Indianapolis, IN, USA.
| |
Collapse
|
23
|
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
|
24
|
Vrancianu CO, Gheorghe I, Dobre EG, Barbu IC, Cristian RE, Popa M, Lee SH, Limban C, Vlad IM, Chifiriuc MC. Emerging Strategies to Combat β-Lactamase Producing ESKAPE Pathogens. Int J Mol Sci 2020; 21:E8527. [PMID: 33198306 PMCID: PMC7697847 DOI: 10.3390/ijms21228527] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Since the discovery of penicillin by Alexander Fleming in 1929 as a therapeutic agent against staphylococci, β-lactam antibiotics (BLAs) remained the most successful antibiotic classes against the majority of bacterial strains, reaching a percentage of 65% of all medical prescriptions. Unfortunately, the emergence and diversification of β-lactamases pose indefinite health issues, limiting the clinical effectiveness of all current BLAs. One solution is to develop β-lactamase inhibitors (BLIs) capable of restoring the activity of β-lactam drugs. In this review, we will briefly present the older and new BLAs classes, their mechanisms of action, and an update of the BLIs capable of restoring the activity of β-lactam drugs against ESKAPE (Enterococcus spp., Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.) pathogens. Subsequently, we will discuss several promising alternative approaches such as bacteriophages, antimicrobial peptides, nanoparticles, CRISPR (clustered regularly interspaced short palindromic repeats) cas technology, or vaccination developed to limit antimicrobial resistance in this endless fight against Gram-negative pathogens.
Collapse
Affiliation(s)
- Corneliu Ovidiu Vrancianu
- Microbiology Immunology Department and The Research Institute of the University of Bucharest, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania; (C.O.V.); (E.-G.D.); (I.C.B.); (M.P.); (M.C.C.)
| | - Irina Gheorghe
- Microbiology Immunology Department and The Research Institute of the University of Bucharest, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania; (C.O.V.); (E.-G.D.); (I.C.B.); (M.P.); (M.C.C.)
| | - Elena-Georgiana Dobre
- Microbiology Immunology Department and The Research Institute of the University of Bucharest, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania; (C.O.V.); (E.-G.D.); (I.C.B.); (M.P.); (M.C.C.)
| | - Ilda Czobor Barbu
- Microbiology Immunology Department and The Research Institute of the University of Bucharest, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania; (C.O.V.); (E.-G.D.); (I.C.B.); (M.P.); (M.C.C.)
| | - Roxana Elena Cristian
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania;
| | - Marcela Popa
- Microbiology Immunology Department and The Research Institute of the University of Bucharest, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania; (C.O.V.); (E.-G.D.); (I.C.B.); (M.P.); (M.C.C.)
| | - Sang Hee Lee
- Department of Biological Sciences, Myongji University, 03674 Myongjiro, Yongin 449-728, Gyeonggido, Korea;
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin 17058, Gyeonggido, Korea
| | - Carmen Limban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia no.6, 020956 Bucharest, Romania; (C.L.); (I.M.V.)
| | - Ilinca Margareta Vlad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia no.6, 020956 Bucharest, Romania; (C.L.); (I.M.V.)
| | - Mariana Carmen Chifiriuc
- Microbiology Immunology Department and The Research Institute of the University of Bucharest, Faculty of Biology, University of Bucharest, 020956 Bucharest, Romania; (C.O.V.); (E.-G.D.); (I.C.B.); (M.P.); (M.C.C.)
- Academy of Romanian Scientists, 030167 Bucharest, Romania
| |
Collapse
|
25
|
Lacerda Mariano L, Ingersoll MA. The immune response to infection in the bladder. Nat Rev Urol 2020; 17:439-458. [PMID: 32661333 DOI: 10.1038/s41585-020-0350-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2020] [Indexed: 12/22/2022]
Abstract
The bladder is continuously protected by passive defences such as a mucus layer, antimicrobial peptides and secretory immunoglobulins; however, these defences are occasionally overcome by invading bacteria that can induce a strong host inflammatory response in the bladder. The urothelium and resident immune cells produce additional defence molecules, cytokines and chemokines, which recruit inflammatory cells to the infected tissue. Resident and recruited immune cells act together to eradicate bacteria from the bladder and to develop lasting immune memory against infection. However, urinary tract infection (UTI) is commonly recurrent, suggesting that the induction of a memory response in the bladder is inadequate to prevent reinfection. Additionally, infection seems to induce long-lasting changes in the urothelium, which can render the tissue more susceptible to future infection. The innate immune response is well-studied in the field of UTI, but considerably less is known about how adaptive immunity develops and how repair mechanisms restore bladder homeostasis following infection. Furthermore, data demonstrate that sex-based differences in immunity affect resolution and infection can lead to tissue remodelling in the bladder following resolution of UTI. To combat the rise in antimicrobial resistance, innovative therapeutic approaches to bladder infection are currently in development. Improving our understanding of how the bladder responds to infection will support the development of improved treatments for UTI, particularly for those at risk of recurrent infection.
Collapse
Affiliation(s)
- Livia Lacerda Mariano
- Department of Immunology, Institut Pasteur, Paris, France.,Inserm, U1223, Paris, France
| | - Molly A Ingersoll
- Department of Immunology, Institut Pasteur, Paris, France. .,Inserm, U1223, Paris, France.
| |
Collapse
|
26
|
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
|
27
|
Savolainen K, Scaravilli M, Ilvesmäki A, Staff S, Tolonen T, Mäenpää JU, Visakorpi T, Auranen A. Expression of the miR-200 family in tumor tissue, plasma and urine of epithelial ovarian cancer patients in comparison to benign counterparts. BMC Res Notes 2020; 13:311. [PMID: 32611374 PMCID: PMC7329467 DOI: 10.1186/s13104-020-05155-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023] Open
Abstract
Objective Plasma, but also urine sample could represent a simple liquid biopsy for ovarian cancer biomarker detection. The miRNA-200 family has been shown to be dysregulated in ovarian cancer. The aim of this study was to isolate three members of miR-200 family from tumor tissue, plasma and urine of high-grade serous ovarian cancer patients in comparison with samples from patients with benign ovarian tumors. This is a methodological pilot study of a prospective ovarian cancer patient cohort investigating the potential of liquid biopsies and the role of miRNAs in ovarian cancer treatment. Results MiR-200a, miR-200b and miR-200c were isolated from samples of nine ovarian cancer patients and seven patients with benign ovarian tumor. The most significant finding is that all three miRNAs were detectable in all sample types. Tumor tissue and plasma, but not urine analysis was able to discriminate malignant and benign samples. A correlation between the miRNA-200 expression in urine and plasma was observed in malignant samples only. Plasma and urine with respect to miRNA detection show potential according to this study, but larger studies are needed to clarify the usefulness of these liquid biopsies in ovarian cancer. Trial registration: ClinicalTrials.gov NCT02758652, May 2, 2016.
Collapse
Affiliation(s)
- Kalle Savolainen
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O.box 2000, 33521, Tampere, Finland. .,Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland.
| | - Mauro Scaravilli
- Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland.,Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Ilvesmäki
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O.box 2000, 33521, Tampere, Finland
| | - Synnöve Staff
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O.box 2000, 33521, Tampere, Finland.,Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Teemu Tolonen
- Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland.,Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Johanna U Mäenpää
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O.box 2000, 33521, Tampere, Finland.,Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tapio Visakorpi
- Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Annika Auranen
- Department of Obstetrics and Gynecology, Tampere University Hospital, P.O.box 2000, 33521, Tampere, Finland.,Tays Cancer Centre, Tampere University Hospital and Tampere University, Tampere, Finland
| |
Collapse
|
28
|
Ketz J, Saxena V, Arregui S, Jackson A, Schwartz GJ, Yagisawa T, Fairchild RL, Hains DS, Schwaderer AL. Developmental loss, but not pharmacological suppression, of renal carbonic anhydrase 2 results in pyelonephritis susceptibility. Am J Physiol Renal Physiol 2020; 318:F1441-F1453. [PMID: 32390512 DOI: 10.1152/ajprenal.00583.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Carbonic anhydrase II knockout (Car2-/-) mice have depleted numbers of renal intercalated cells, which are increasingly recognized to be innate immune effectors. We compared pyelonephritis susceptibility following reciprocal renal transplantations between Car2-/- and wild-type mice. We examined the effect of pharmacological CA suppression using acetazolamide in an experimental murine model of urinary tract infection. Car2-/- versus wild-type mice were compared for differences in renal innate immunity. In our transplant scheme, mice lacking CA-II in the kidney had increased pyelonephritis risk. Mice treated with acetazolamide had lower kidney bacterial burdens at 6 h postinfection, which appeared to be due to tubular flow from diuresis because comparable results were obtained when furosemide was substituted for acetazolamide. Isolated Car2-/- kidney cells enriched for intercalated cells demonstrated altered intercalated cell innate immune gene expression, notably increased calgizzarin and insulin receptor expression. Intercalated cell number and function along with renal tubular flow are determinants of pyelonephritis risk.
Collapse
Affiliation(s)
- John Ketz
- Center for Clinical and Translational Medicine, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Vijay Saxena
- Division of Pediatric Nephrology, Department of Pediatrics, Indiana University, Indianapolis, Indiana
| | - Samuel Arregui
- Division of Pediatric Nephrology, Department of Pediatrics, Indiana University, Indianapolis, Indiana
| | - Ashley Jackson
- Center for Clinical and Translational Medicine, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - George J Schwartz
- Division of Nephrology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Takafumi Yagisawa
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Robert L Fairchild
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio
| | - David S Hains
- Division of Pediatric Nephrology, Department of Pediatrics, Indiana University, Indianapolis, Indiana
| | - Andrew L Schwaderer
- Division of Pediatric Nephrology, Department of Pediatrics, Indiana University, Indianapolis, Indiana
| |
Collapse
|
29
|
Prasad SV, Fiedoruk K, Daniluk T, Piktel E, Bucki R. Expression and Function of Host Defense Peptides at Inflammation Sites. Int J Mol Sci 2019; 21:ijms21010104. [PMID: 31877866 PMCID: PMC6982121 DOI: 10.3390/ijms21010104] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/09/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023] Open
Abstract
There is a growing interest in the complex role of host defense peptides (HDPs) in the pathophysiology of several immune-mediated inflammatory diseases. The physicochemical properties and selective interaction of HDPs with various receptors define their immunomodulatory effects. However, it is quite challenging to understand their function because some HDPs play opposing pro-inflammatory and anti-inflammatory roles, depending on their expression level within the site of inflammation. While it is known that HDPs maintain constitutive host protection against invading microorganisms, the inducible nature of HDPs in various cells and tissues is an important aspect of the molecular events of inflammation. This review outlines the biological functions and emerging roles of HDPs in different inflammatory conditions. We further discuss the current data on the clinical relevance of impaired HDPs expression in inflammation and selected diseases.
Collapse
|
30
|
Becknell B, Ching C, Spencer JD. The Responses of the Ribonuclease A Superfamily to Urinary Tract Infection. Front Immunol 2019; 10:2786. [PMID: 31849967 PMCID: PMC6901906 DOI: 10.3389/fimmu.2019.02786] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022] Open
Abstract
The lower urinary tract is routinely exposed to microbes residing in the gastrointestinal tract, yet the urothelium resists invasive infections by gut microorganisms. This infection resistance is attributed to innate defenses in the bladder urothelium, kidney epithelium, and resident or circulating immune cells. In recent years, surmounting evidence suggests that these cell types produce and secrete soluble host defense peptides, including members of the Ribonuclease (RNase) A Superfamily, to combat invasive bacterial challenge. While some of these peptides, including RNase 4 and RNase 7, are abundantly produced by epithelial cells, the expression of others, like RNase 3 and RNase 6, increase at infection sites with immune cell recruitment. The objective of this mini-review is to highlight recent evidence showing the biological importance and responses of RNase A Superfamily members to infection in the kidney and bladder.
Collapse
Affiliation(s)
- Brian Becknell
- Nephrology and Urology Research Affinity Group, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Center of Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Nephrology, Nationwide Children's Hospital, Columbus, OH, United States
| | - Christina Ching
- Nephrology and Urology Research Affinity Group, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Center of Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Urology, Nationwide Children's Hospital, Columbus, OH, United States
| | - John David Spencer
- Nephrology and Urology Research Affinity Group, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Center of Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Nephrology, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
31
|
Rademacher F, Dreyer S, Kopfnagel V, Gläser R, Werfel T, Harder J. The Antimicrobial and Immunomodulatory Function of RNase 7 in Skin. Front Immunol 2019; 10:2553. [PMID: 31749808 PMCID: PMC6848066 DOI: 10.3389/fimmu.2019.02553] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/15/2019] [Indexed: 11/13/2022] Open
Abstract
The human ribonuclease RNase 7 has been originally isolated from human skin and is a member of the human RNase A superfamily. RNase 7 is constantly released by keratinocytes and accumulates on the skin surface. The expression of RNase 7 in keratinocytes can be induced by diverse stimuli such as cytokines, growth factors, and microbial factors. RNase 7 exhibits a potent broad spectrum of antimicrobial activity against various microorganisms and contributes to control bacterial growth on the skin surface. The ribonuclease and antimicrobial activity of RNase 7 can be blocked by the endogenous ribonuclease inhibitor. There is also increasing evidence that RNase 7 exerts immunomodulatory activities and may participate in antiviral defense. In this review, we discuss how these characteristics of RNase 7 contribute to innate cutaneous defense and highlight its role in skin infection and inflammation. We also speculate how a potential dysregulation of RNase 7 promotes inflammatory skin diseases and if RNase 7 may have therapeutic potential.
Collapse
Affiliation(s)
| | - Sylvia Dreyer
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hanover, Germany
| | - Verena Kopfnagel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hanover, Germany
- Hannover Unified Biobank, Hannover Medical School, Hanover, Germany
| | - Regine Gläser
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hanover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hanover, Germany
| | - Jürgen Harder
- Department of Dermatology, University of Kiel, Kiel, Germany
| |
Collapse
|
32
|
Desloges I, Taylor JA, Leclerc JM, Brannon JR, Portt A, Spencer JD, Dewar K, Marczynski GT, Manges A, Gruenheid S, Le Moual H, Thomassin JL. Identification and characterization of OmpT-like proteases in uropathogenic Escherichia coli clinical isolates. Microbiologyopen 2019; 8:e915. [PMID: 31496120 PMCID: PMC6854850 DOI: 10.1002/mbo3.915] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/01/2019] [Accepted: 07/06/2019] [Indexed: 01/01/2023] Open
Abstract
Bacterial colonization of the urogenital tract is limited by innate defenses, including the production of antimicrobial peptides (AMPs). Uropathogenic Escherichia coli (UPEC) resist AMP‐killing to cause a range of urinary tract infections (UTIs) including asymptomatic bacteriuria, cystitis, pyelonephritis, and sepsis. UPEC strains have high genomic diversity and encode numerous virulence factors that differentiate them from non‐UTI‐causing strains, including ompT. As OmpT homologs cleave and inactivate AMPs, we hypothesized that UPEC strains from patients with symptomatic UTIs have high OmpT protease activity. Therefore, we measured OmpT activity in 58 clinical E. coli isolates. While heterogeneous OmpT activities were observed, OmpT activity was significantly greater in UPEC strains isolated from patients with symptomatic infections. Unexpectedly, UPEC strains exhibiting the greatest protease activities harbored an additional ompT‐like gene called arlC (ompTp). The presence of two OmpT‐like proteases in some UPEC isolates led us to compare the substrate specificities of OmpT‐like proteases found in E. coli. While all three cleaved AMPs, cleavage efficiency varied on the basis of AMP size and secondary structure. Our findings suggest the presence of ArlC and OmpT in the same UPEC isolate may confer a fitness advantage by expanding the range of target substrates.
Collapse
Affiliation(s)
- Isabelle Desloges
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - James A Taylor
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Jean-Mathieu Leclerc
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - John R Brannon
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Andrea Portt
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - John D Spencer
- Division of Nephrology, Nationwide Children's Hospital, Columbus, Ohio
| | - Ken Dewar
- Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Gregory T Marczynski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada
| | - Amee Manges
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada
| | - Hervé Le Moual
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada.,Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Jenny-Lee Thomassin
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| |
Collapse
|
33
|
Shen T, Levitman A, Li Y, Jacobs M, Xu K, Barasch J. Urinary defense begins in the kidney. Kidney Int 2019; 96:537-539. [PMID: 31445577 DOI: 10.1016/j.kint.2019.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/22/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Tian Shen
- Columbia University, Department of Medicine, New York, New York, USA
| | - Abraham Levitman
- Columbia University, Department of Medicine, New York, New York, USA
| | - Yuanji Li
- Columbia University, Department of Medicine, New York, New York, USA
| | - Mendel Jacobs
- Columbia University, Department of Medicine, New York, New York, USA
| | - Katherine Xu
- Columbia University, Department of Medicine, New York, New York, USA
| | - Jonathan Barasch
- Columbia University, Department of Medicine, New York, New York, USA.
| |
Collapse
|
34
|
Catinean A, Neag MA, Mitre AO, Bocsan CI, Buzoianu AD. Microbiota and Immune-Mediated Skin Diseases-An Overview. Microorganisms 2019; 7:microorganisms7090279. [PMID: 31438634 PMCID: PMC6781142 DOI: 10.3390/microorganisms7090279] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022] Open
Abstract
In recent years, increased attention has been paid to the relationship between microbiota and various diseases, especially immune-mediated diseases. Because conventional therapy for many autoimmune diseases is limited both in efficacy and safety, there is an increased interest in identifying nutraceuticals, particularly probiotics, able to modulate the microbiota and ameliorate these diseases. In this review, we analyzed the research focused on the role of gut microbiota and skin in immunity, their role in immune-mediated skin diseases (IMSDs), and the beneficial effect of probiotics in patients with this pathology. We selected articles published between 2009 and 2019 in PubMed and ScienceDirect that provided information regarding microbiota, IMSDs and the role of probiotics in these diseases. We included results from different types of studies including observational and interventional clinical trials or in vivo and in vitro experimental studies. Our results showed that probiotics have a beneficial effect in changing the microbiota of patients with IMSDs; they also influence disease progression. Further studies are needed to better understand the impact of new therapies on intestinal microbiota. It is also important to determine whether the microbiota of patients with autoimmune diseases can be manipulated in order to restore homeostasis of the microbiota.
Collapse
Affiliation(s)
- Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Maria Adriana Neag
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
| | - Andrei Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Corina Ioana Bocsan
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca Dana Buzoianu
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
35
|
Eichler T, Bender K, Murtha MJ, Schwartz L, Metheny J, Solden L, Jaggers RM, Bailey MT, Gupta S, Mosquera C, Ching C, La Perle K, Li B, Becknell B, Spencer JD. Ribonuclease 7 Shields the Kidney and Bladder from Invasive Uropathogenic Escherichia coli Infection. J Am Soc Nephrol 2019; 30:1385-1397. [PMID: 31239387 PMCID: PMC6683711 DOI: 10.1681/asn.2018090929] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 04/17/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Evidence suggests that antimicrobial peptides, components of the innate immune response, protect the kidneys and bladder from bacterial challenge. We previously identified ribonuclease 7 (RNase 7) as a human antimicrobial peptide that has bactericidal activity against uropathogenic Escherichia coli (UPEC). Functional studies assessing RNase 7's contributions to urinary tract defense are limited. METHODS To investigate RNase 7's role in preventing urinary tract infection (UTI), we quantified urinary RNase 7 concentrations in 29 girls and adolescents with a UTI history and 29 healthy female human controls. To assess RNase 7's antimicrobial activity in vitro in human urothelial cells, we used siRNA to silence urothelial RNase 7 production and retroviral constructs to stably overexpress RNase 7; we then evaluated UPEC's ability to bind and invade these cells. For RNase 7 in vivo studies, we developed humanized RNase 7 transgenic mice, subjected them to experimental UTI, and enumerated UPEC burden in the urine, bladder, and kidneys. RESULTS Compared with controls, study participants with a UTI history had 1.5-fold lower urinary RNase 7 concentrations. When RNase 7 was silenced in vitro, the percentage of UPEC binding or invading human urothelial cells increased; when cells overexpressed RNase 7, UPEC attachment and invasion decreased. In the transgenic mice, we detected RNase 7 expression in the kidney's intercalated cells and bladder urothelium. RNase 7 humanized mice exhibited marked protection from UPEC. CONCLUSIONS These findings provide evidence that RNase 7 has a role in kidney and bladder host defense against UPEC and establish a foundation for investigating RNase 7 as a UTI prognostic marker or nonantibiotic-based therapy.
Collapse
Affiliation(s)
- Tad Eichler
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
| | - Kristin Bender
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
| | - Matthew J Murtha
- Centers for Clinical and Translational Research and
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio; and
| | - Laura Schwartz
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
| | | | - Lindsey Solden
- Microbial Pathogenesis, The Research Institute at Nationwide Children's, Columbus, Ohio
| | - Robert M Jaggers
- Microbial Pathogenesis, The Research Institute at Nationwide Children's, Columbus, Ohio
| | - Michael T Bailey
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio; and
- Microbial Pathogenesis, The Research Institute at Nationwide Children's, Columbus, Ohio
| | - Sudipti Gupta
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
| | | | - Christina Ching
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio; and
- Divisions of Urology and
- Departments of Pediatric Surgery and
| | - Krista La Perle
- Comparative Pathology and Mouse Phenotyping Shared Resource, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Birong Li
- Centers for Clinical and Translational Research and
| | - Brian Becknell
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio; and
- Nephrology, and
- Pediatrics, Nationwide Children's, Columbus, Ohio
| | - John David Spencer
- Centers for Clinical and Translational Research and
- Nephrology and Urology Research Affinity Group
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio; and
- Nephrology, and
- Pediatrics, Nationwide Children's, Columbus, Ohio
| |
Collapse
|
36
|
An Exosomal Urinary miRNA Signature for Early Diagnosis of Renal Fibrosis in Lupus Nephritis. Cells 2019; 8:cells8080773. [PMID: 31349698 PMCID: PMC6721515 DOI: 10.3390/cells8080773] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
For lupus nephritis (LN) management, it is very important to detect fibrosis at an early stage. Urinary exosomal miRNAs profiling can be used as a potential multi-marker phenotyping tool to identify early fibrosis. We isolated and characterised urinary exosomes and cellular pellets from patients with biopsy-proven LN (n = 45) and healthy controls (n = 20). LN chronicity index (CI) correlated with urinary exosomal miR-21, miR-150, and miR-29c (r = 0.565, 0.840, −0.559, respectively). This miRNA profile distinguished low CI from moderate-high CI in LN patients with a high sensitivity and specificity (94.4% and 99.8%). Furthermore, this multimarker panel predicted an increased risk of progression to end-stage renal disease (ESRD). Pathway analysis identified VEGFA and SP1 as common target genes for the three miRNAs. Immunohistochemistry in LN renal biopsies revealed a significant increase of COL1A1 and COL4A1 correlated with renal chronicity. SP1 decreased significantly in the high-CI group (p = 0.002). VEGFA levels showed no differences. In vitro experiments suggest that these miRNA combinations promote renal fibrosis by increasing profibrotic molecules through SP1 and Smad3/TGFβ pathways. In conclusion, a urinary exosomal multimarker panel composed of miR-21, miR-150, and miR-29c provides a non-invasive method to detect early renal fibrosis and predict disease progression in LN.
Collapse
|
37
|
Abstract
The epithelium of the kidney collecting duct (CD) is composed mainly of two different types of cells with distinct and complementary functions. CD principal cells traditionally have been considered to have a major role in Na+ and water regulation, while intercalated cells (ICs) were thought to largely modulate acid-base homeostasis. In recent years, our understanding of IC function has improved significantly owing to new research findings. Thus, we now have a new model for CD transport that integrates mechanisms of salt and water reabsorption, K+ homeostasis, and acid-base status between principal cells and ICs. There are three main types of ICs (type A, type B, and non-A, non-B), which first appear in the late distal convoluted tubule or in the connecting segment in a species-dependent manner. ICs can be detected in CD from cortex to the initial part of the inner medulla, although some transport proteins that are key components of ICs also are present in medullary CD, cells considered inner medullary. Of the three types of ICs, each has a distinct morphology and expresses different complements of membrane transport proteins that translate into very different functions in homeostasis and contributions to CD luminal pro-urine composition. This review includes recent discoveries in IC intracellular and paracrine signaling that contributes to acid-base regulation as well as Na+, Cl-, K+, and Ca2+ homeostasis. Thus, these new findings highlight the potential role of ICs as targets for potential hypertension treatments.
Collapse
Affiliation(s)
- Renee Rao
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Núria M Pastor-Soler
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA.
| |
Collapse
|
38
|
Pottanat ND, Brook AC, Bartosova M, Cortado H, Gupta S, Li B, Jackson AR, Vonau M, Cohen S, Ferrara M, Ching CB, Spencer JD, Brauner A, Fraser DJ, Schmitt CP, Eberl M, Ayoob R, Becknell B. Analysis of the Ribonuclease A Superfamily of Antimicrobial Peptides in Patients Undergoing Chronic Peritoneal Dialysis. Sci Rep 2019; 9:7753. [PMID: 31123272 PMCID: PMC6533318 DOI: 10.1038/s41598-019-44219-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/09/2019] [Indexed: 02/03/2023] Open
Abstract
Infectious peritonitis is a common complication in patients undergoing chronic peritoneal dialysis (PD), limiting the duration of PD as a modality for renal replacement therapy and increasing patient morbidity and mortality. Antimicrobial peptides (AMPs) serve critical roles in mucosal defense, but their expression and activity during peritonitis are poorly understood. We hypothesized that AMPs belonging to the Ribonuclease (RNase) A Superfamily are present in peritoneal fluid and increase during peritonitis in patients undergoing chronic PD. In the absence of peritonitis, we detected RNase 3, RNase 6, and RNase 7 in cell-free supernatants and viable cells obtained from peritoneal fluid of chronic PD patients. The cellular sources of these RNases were eosinophils (RNase 3), macrophages (RNase 6), and mesothelial cells (RNase 7). During peritonitis, RNase 3 increased 55-fold and RNase 7 levels increased 3-fold on average, whereas RNase 6 levels were unchanged. The areas under the receiver-operating characteristic curves for RNase 3 and RNase 7 were 0.99 (95% confidence interval (CI): 0.96-1.0) and 0.79 (95% CI: 0.64-0.93), respectively, indicating their potential as biomarkers of peritonitis. Discrete omental reservoirs of these RNases were evident in patients with end stage kidney disease prior to PD initiation, and omental RNase 3 reactive cells increased in patients undergoing PD with a history of peritonitis. We propose that constitutive and inducible pools of antimicrobial RNases form a network to shield the peritoneal cavity from microbial invasion in patients undergoing chronic PD.
Collapse
Affiliation(s)
- Neha Dhingra Pottanat
- Division of Nephrology, Department of Pediatrics, Riley Children's Hospital and Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amy C Brook
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, United Kingdom
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Hanna Cortado
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Sudipti Gupta
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Birong Li
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ashley R Jackson
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Martin Vonau
- Department of Pediatrics and Internal Medicine, Ohio State University College of Medicine, Columbus, OH, USA
| | - Shira Cohen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Maria Ferrara
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christina B Ching
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Pediatric Urology, Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - John David Spencer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| | - Annelie Brauner
- Department of Microbiology, Tumor and Cell Biology, Division of Clinical Microbiology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Donald J Fraser
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, United Kingdom
- Wales Kidney Research Unit, Cardiff University, Cardiff, United Kingdom
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, United Kingdom
| | - Rose Ayoob
- Division of Nephrology, Department of Pediatrics, Charleston, WV, USA.
| | - Brian Becknell
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
- Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
39
|
Lee HH, Wang YN, Hung MC. Functional roles of the human ribonuclease A superfamily in RNA metabolism and membrane receptor biology. Mol Aspects Med 2019; 70:106-116. [PMID: 30902663 DOI: 10.1016/j.mam.2019.03.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/17/2019] [Indexed: 02/08/2023]
Abstract
The human ribonuclease A (hRNase A) superfamily is comprised of 13 members of secretory RNases, most of which are recognized as catabolic enzymes for their ribonucleolytic activity to degrade ribonucleic acids (RNAs) in the extracellular space, where they play a role in innate host defense and physiological homeostasis. Interestingly, human RNases 9-13, which belong to a non-canonical subgroup of the hRNase A superfamily, are ribonucleolytic activity-deficient proteins with unclear biological functions. Moreover, accumulating evidence indicates that secretory RNases, such as human RNase 5, can be internalized into cells facilitated by membrane receptors like the epidermal growth factor receptor to regulate intracellular RNA species, in particular non-coding RNAs, and signaling pathways by either a ribonucleolytic activity-dependent or -independent manner. In this review, we summarize the classical role of hRNase A superfamily in the metabolism of extracellular and intracellular RNAs and update its non-classical function as a cognate ligand of membrane receptors. We further discuss the biological significance and translational potential of using secretory RNases as predictive biomarkers or therapeutic agents in certain human diseases and the pathological settings for future investigations.
Collapse
Affiliation(s)
- Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Unit 108, 1515 Holcombe Boulevard, Houston, TX, 77030, USA; Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
40
|
Saxena V, Fitch J, Ketz J, White P, Wetzel A, Chanley MA, Spencer JD, Becknell B, Pierce KR, Arregui SW, Nelson RD, Schwartz GJ, Velazquez V, Walker LA, Chen X, Yan P, Hains DS, Schwaderer AL. Whole Transcriptome Analysis of Renal Intercalated Cells Predicts Lipopolysaccharide Mediated Inhibition of Retinoid X Receptor alpha Function. Sci Rep 2019; 9:545. [PMID: 30679625 PMCID: PMC6345901 DOI: 10.1038/s41598-018-36921-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 11/23/2018] [Indexed: 01/14/2023] Open
Abstract
The renal collecting duct consists of intercalated cells (ICs) and principal cells (PCs). We have previously demonstrated that collecting ducts have a role in the innate immune defense of the kidney. Transcriptomics is an important tool used to enhance systems-level understanding of cell biology. However, transcriptomics performed on whole kidneys provides limited insight of collecting duct cell gene expression, because these cells comprise a small fraction of total kidney cells. Recently we generated reporter mouse models to enrich collecting duct specific PC and ICs and reported targeted gene expression of anti-microbial peptide genes. Here we report transcriptomics on enriched ICs and PCs and performed a pilot study sequencing four single ICs. We identified 3,645 genes with increased relative expression in ICs compared to non-ICs. In comparison to non-PCs, 2,088 genes had higher relative expression in PCs. IC associated genes included the innate interleukin 1 receptor, type 1 and the antimicrobial peptide(AMP) adrenomedullin. The top predicted canonical pathway for enriched ICs was lipopolysaccharide/Interleukin 1 mediated inhibition of Retinoid X Receptor alpha function and decreased Retinoid X Receptor expression was confirmed to occur 1-hour post experimental murine UTI in ICs but not in non-ICs.
Collapse
Affiliation(s)
- Vijay Saxena
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| | - James Fitch
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - John Ketz
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
| | - Peter White
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Amy Wetzel
- The Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Melinda A Chanley
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
| | - John D Spencer
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Brian Becknell
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University, Columbus, Ohio, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Keith R Pierce
- Innate Immunity Translational Research Center, Children's Foundation Research Institute at Le Bonheur Children's Hospital, Memphis, Tennessee, United States
| | - Sam W Arregui
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States
| | - Raoul D Nelson
- Division of Nephrology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - George J Schwartz
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, United States
| | - Victoria Velazquez
- Research Institute at Nationwide Children's Hospital Flow Cytometry Core Laboratory, Columbus, Ohio, United States
| | - Logan A Walker
- Department of Physics, College of Arts and Sciences, The Ohio State University, Columbus, Ohio, United States
| | - Xi Chen
- Genomics Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Pearlly Yan
- Genomics Shared Resource, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - David S Hains
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| | - Andrew L Schwaderer
- Indiana University School of Medicine, Riley Children's Hospital, Indianapolis, Indiana, United States.
| |
Collapse
|
41
|
Abstract
People with diabetes mellitus are at higher risk of developing serious ascending infections of the urinary tract. The traditional explanation has focused on the role of glycosuria in promoting bacterial growth. Using mouse models, Murtha et al. demonstrate that when the intracellular insulin signaling pathway is compromised, antimicrobial defenses are compromised too, and the mice are unable to effectively handle uropathogenic E. coli introduced experimentally into the urinary tract. These observations strongly support the hypothesis that the antimicrobial defenses of the kidney are dependent on insulin, and the urinary tract infections associated with diabetes occur due to reduced expression of these key effectors of innate immunity.
Collapse
|
42
|
The Immunomodulatory and Antimicrobial Properties of the Vertebrate Ribonuclease A Superfamily. Vaccines (Basel) 2018; 6:vaccines6040076. [PMID: 30463297 PMCID: PMC6313885 DOI: 10.3390/vaccines6040076] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/31/2018] [Accepted: 11/16/2018] [Indexed: 02/08/2023] Open
Abstract
The Ribonuclease A Superfamily is composed of cationic peptides that are secreted by immune cells and epithelial tissues. Although their physiological roles are unclear, several members of the vertebrate Ribonuclease A Superfamily demonstrate antimicrobial and immune modulation activities. The objective of this review is to provide an overview of the published literature on the Ribonuclease A Superfamily with an emphasis on each peptide’s regulation, antimicrobial properties, and immunomodulatory functions. As additional insights emerge regarding the mechanisms in which these ribonucleases eradicate invading pathogens and modulate immune function, these ribonucleases may have the potential to be developed as a novel class of therapeutics for some human diseases.
Collapse
|
43
|
Murtha MJ, Eichler T, Bender K, Metheny J, Li B, Schwaderer AL, Mosquera C, James C, Schwartz L, Becknell B, Spencer JD. Insulin receptor signaling regulates renal collecting duct and intercalated cell antibacterial defenses. J Clin Invest 2018; 128:5634-5646. [PMID: 30418175 DOI: 10.1172/jci98595] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
People with diabetes mellitus have increased infection risk. With diabetes, urinary tract infection (UTI) is more common and has worse outcomes. Here, we investigate how diabetes and insulin resistance impact the kidney's innate defenses and urine sterility. We report that type 2 diabetic mice have increased UTI risk. Moreover, insulin-resistant prediabetic mice have increased UTI susceptibility, independent of hyperglycemia or glucosuria. To identify how insulin resistance affects renal antimicrobial defenses, we genetically deleted the insulin receptor in the kidney's collecting tubules and intercalated cells. Intercalated cells, located within collecting tubules, contribute to epithelial defenses by acidifying the urine and secreting antimicrobial peptides (AMPs) into the urinary stream. Collecting duct and intercalated cell-specific insulin receptor deletion did not impact urine acidification, suppressed downstream insulin-mediated targets and AMP expression, and increased UTI susceptibility. Specifically, insulin receptor-mediated signaling regulates AMPs, including lipocalin 2 and ribonuclease 4, via phosphatidylinositol-3-kinase signaling. These data suggest that insulin signaling plays a critical role in renal antibacterial defenses.
Collapse
Affiliation(s)
- Matthew J Murtha
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Tad Eichler
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kristin Bender
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jackie Metheny
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Birong Li
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Andrew L Schwaderer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Claudia Mosquera
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Cindy James
- Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, Ohio, USA
| | - Laura Schwartz
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Brian Becknell
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - John David Spencer
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,The Ohio State University College of Medicine, Columbus, Ohio, USA.,Division of Nephrology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
44
|
Saxena V, Hains DS, Ketz J, Chanley M, Spencer JD, Becknell B, Pierce KR, Nelson RD, Purkerson JM, Schwartz GJ, Schwaderer AL. Cell-specific qRT-PCR of renal epithelial cells reveals a novel innate immune signature in murine collecting duct. Am J Physiol Renal Physiol 2018; 315:F812-F823. [PMID: 28468965 PMCID: PMC6230735 DOI: 10.1152/ajprenal.00512.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/23/2017] [Accepted: 04/25/2017] [Indexed: 01/17/2023] Open
Abstract
The urinary tract is usually culture negative despite its close proximity to microbial flora. The precise mechanism by which the kidneys and urinary tract defends against infection is not well understood. The initial kidney cells to encounter ascending pathogens are the collecting tubule cells that consist of principal cells (PCs) that express aquaporin 2 (AQP2) and intercalated cells (ICs) that express vacuolar H+-ATPase (V-ATPase, B1 subunit). We have previously shown that ICs are involved with the human renal innate immune defense. Here we generated two reporter mice, VATPase B1-cre+tdT+ mice to fluorescently label ICs and AQP2-cre+tdT+ mice to fluorescently label PCs, and then performed flow sorting to enrich PCs and ICs for analysis. Isolated ICs and PCs along with proximal tubular cells were used to measure antimicrobial peptide (AMP) mRNA expression. ICs and PCs were significantly enriched for AMPs. Isolated ICs responded to uropathogenic Escherichia coli (UPEC) challenge in vitro and had higher RNase4 gene expression than control while both ICs and PCs responded to UPEC challenge in vivo by upregulating Defb1 mRNA expression. To our knowledge, this is the first report of isolating murine collecting tubule cells and performing targeted analysis for multiple classes of AMPs.
Collapse
Affiliation(s)
- Vijay Saxena
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University , Columbus, Ohio
| | - David S Hains
- Innate Immunity Translational Research Center, Children's Foundation Research Institute at Le Bonheur Children's Hospital , Memphis, Tennessee
| | - John Ketz
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University , Columbus, Ohio
| | - Melinda Chanley
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University , Columbus, Ohio
| | - John D Spencer
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University , Columbus, Ohio
| | - Brian Becknell
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University , Columbus, Ohio
| | - Keith R Pierce
- Innate Immunity Translational Research Center, Children's Foundation Research Institute at Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Raoul D Nelson
- Division of Nephrology, Department of Pediatrics, University of Utah , Salt Lake City, Utah
| | - Jeffrey M Purkerson
- University of Rochester Medical Center, School of Medicine and Dentistry , Rochester, New York
| | - George J Schwartz
- University of Rochester Medical Center, School of Medicine and Dentistry , Rochester, New York
| | - Andrew L Schwaderer
- The Research Institute at Nationwide Children's, Center for Clinical and Translational Research, Columbus, Ohio, and College of Medicine, Ohio State University , Columbus, Ohio
| |
Collapse
|
45
|
Schwab S, Jobin K, Kurts C. Urinary tract infection: recent insight into the evolutionary arms race between uropathogenic Escherichia coli and our immune system. Nephrol Dial Transplant 2018; 32:1977-1983. [PMID: 28340252 DOI: 10.1093/ndt/gfx022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/20/2016] [Indexed: 12/16/2022] Open
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections worldwide. Humans evolved various immune-dependent and independent defense mechanisms, while pathogens evolved multiple virulence factors to fight back. This article summarizes recent findings regarding the arms race between hosts and pathogens in UTIs. It was recently reported that macrophage subsets regulate neutrophil-mediated defense in primary UTIs but seem to subvert adaptive immunity upon re-infection. Moreover, some bacterial strains can survive inside macrophages, leading to recurrent infections. Inflammasome activation results in infected host cell death and pathogen release, facilitating the removal of intracellular bacteria. As a counteraction, some bacteria evolved mechanisms to disrupt inflammasome activation. Mucosal-associated invariant T cells are further effectors that can lyse infected epithelial cells and release intracellular bacteria. Once released, the bacteria are phagocytosed by neutrophils. However, some bacteria can inhibit neutrophil migration and deprive neutrophils of nutrients. Furthermore, the complement system, considered generally bactericidal, is exploited by the bacteria for cellular invasion. Another weapon against UTI is antimicrobial peptides, e.g. ribonuclease 7, but its production is inhibited by certain bacterial strains. Thus the arms race in UTI is ongoing, and knowing the enemy's methods can help in developing new drugs to win the race.
Collapse
Affiliation(s)
- Sebastian Schwab
- Institute of Molecular Medicine and Experimental Immunology, Friedrich-Wilhelms University, Bonn, Germany.,Medical Clinic I, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Katarzyna Jobin
- Institute of Molecular Medicine and Experimental Immunology, Friedrich-Wilhelms University, Bonn, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, Friedrich-Wilhelms University, Bonn, Germany
| |
Collapse
|
46
|
Urine: Waste product or biologically active tissue? Neurourol Urodyn 2018; 37:1162-1168. [PMID: 29464759 DOI: 10.1002/nau.23414] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/18/2017] [Indexed: 01/01/2023]
Abstract
AIMS Historically, urine has been viewed primarily as a waste product with little biological role in the overall health of an individual. Increasingly, data suggest that urine plays a role in human health beyond waste excretion. For example, urine might act as an irritant and contribute to symptoms through interaction with-and potential compromise of-the urothelium. METHODS To explore the concept that urine may be a vehicle for agents with potential or occult bioactivity and to discuss existing evidence and novel research questions that may yield insight into such a role, the National Institute of Diabetes and Digestive and Kidney Disease invited experts in the fields of comparative evolutionary physiology, basic science, nephrology, urology, pediatrics, metabolomics, and proteomics (among others) to a Urinology Think Tank meeting on February 9, 2015. RESULTS This report reflects ideas that evolved from this meeting and current literature, including the concept of urine quality, the biological, chemical, and physical characteristics of urine, including the microbiota, cells, exosomes, pH, metabolites, proteins, and specific gravity (among others). Additionally, the manuscript presents speculative, and hopefully testable, ideas about the functional roles of urine constituents in health and disease. CONCLUSION Moving forward, there are several questions that need further understanding and pursuit. There were suggestions to consider actively using various animal models and their biological specimens to elaborate on basic mechanistic information regarding human bladder dysfunction.
Collapse
|
47
|
Torres-Juarez F, Touqui L, Leon-Contreras J, Rivas-Santiago C, Enciso-Moreno JA, Hernández-Pando R, Rivas-Santiago B. RNase 7 but not psoriasin nor sPLA2-IIA associates with Mycobacterium tuberculosis during airway epithelial cell infection. Pathog Dis 2018; 76:4810750. [DOI: 10.1093/femspd/fty005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/15/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Flor Torres-Juarez
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
- Department of Immunology, School of Medicine, Universidad Autonoma de San Luis Potosi, San Luis Potosi, Mexico
| | - Lousseine Touqui
- Unité d’histopathologie humaine et modèles animaux, Institut Pasteur, Paris, France
| | - Juan Leon-Contreras
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition ‘Salvador Zubirán’, México City, Mexico
| | - Cesar Rivas-Santiago
- National Council of Science and Technology (CONACyT), Catedras-CONACyT, México
- Biological Sciences School, University Autonomous of Zacatecas, Zacatecas, Mexico
| | - Jose A Enciso-Moreno
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition ‘Salvador Zubirán’, México City, Mexico
| | - Bruno Rivas-Santiago
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security-IMSS, Zacatecas, Mexico
| |
Collapse
|
48
|
Peng H, Purkerson JM, Schwaderer AL, Schwartz GJ. Metabolic acidosis stimulates the production of the antimicrobial peptide cathelicidin in rabbit urine. Am J Physiol Renal Physiol 2017; 313:F1061-F1067. [PMID: 28747361 PMCID: PMC7276924 DOI: 10.1152/ajprenal.00701.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 11/22/2022] Open
Abstract
Intercalated cells of the collecting duct (CD) are critical for acid-base homeostasis and innate immune defense of the kidney. Little is known about the impact of acidosis on innate immune defense in the distal nephron. Urinary tract infections are mainly due to Escherichia coli and are an important risk factor for development of chronic kidney disease. While the effect of urinary pH on growth of E. coli is well established, in this study, we demonstrate that acidosis increases urine antimicrobial activity due, at least in part, to induction of cathelicidin expression within the CD. Acidosis was induced in rabbits by adding NH4Cl to the drinking water and reducing food intake over 3 days or by casein supplementation. Microdissected CDs were examined for cathelicidin mRNA expression and antimicrobial activity, and cathelicidin protein levels in rabbit urine were measured. Cathelicidin expression in CD cells was detected in kidney sections. CDs from acidotic rabbits expressed three times more cathelicidin mRNA than those isolated from normal rabbits. Urine from acidotic rabbits had significantly more antimicrobial activity (vs. E. coli) than normal urine, and most of this increased activity was blocked by cathelicidin antibody. The antibody had little effect on antimicrobial activity of normal urine. Urine from acidotic rabbits had at least twice the amount of cathelicidin protein as did normal urine. We conclude that metabolic acidosis not only stimulates CD acid secretion but also induces expression of cathelicidin and, thereby, enhances innate immune defense against urinary tract infections via induction of antimicrobial peptide expression.
Collapse
Affiliation(s)
- Hu Peng
- University of Rochester Medical Center, Rochester, New York; and
| | | | | | | |
Collapse
|
49
|
Cutaneous Burn Injury Modulates Urinary Antimicrobial Peptide Responses and the Urinary Microbiome. Crit Care Med 2017; 45:e543-e551. [PMID: 28333758 DOI: 10.1097/ccm.0000000000002304] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Characterization of urinary bacterial microbiome and antimicrobial peptides after burn injury to identify potential mechanisms leading to urinary tract infections and associated morbidities in burn patients. DESIGN Retrospective cohort study using human urine from control and burn subjects. SETTING University research laboratory. PATIENTS Burn patients. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Urine samples from catheterized burn patients were collected hourly for up to 40 hours. Control urine was collected from "healthy" volunteers. The urinary bacterial microbiome and antimicrobial peptide levels and activity were compared with patient outcomes. We observed a significant increase in urinary microbial diversity in burn patients versus controls, which positively correlated with a larger percent burn and with the development of urinary tract infection and sepsis postadmission, regardless of age or gender. Urinary psoriasin and β-defensin antimicrobial peptide levels were significantly reduced in burn patients at 1 and 40 hours postadmission. We observed a shift in antimicrobial peptide hydrophobicity and activity between control and burn patients when urinary fractions were tested against Escherichia coli and Enterococcus faecalis urinary tract infection isolates. Furthermore, the antimicrobial peptide activity in burn patients was more effective against E. coli than E. faecalis. Urinary tract infection-positive burn patients with altered urinary antimicrobial peptide activity developed either an E. faecalis or Pseudomonas aeruginosa urinary tract infection, suggesting a role for urinary antimicrobial peptides in susceptibility to select uropathogens. CONCLUSIONS Our data reveal potential links for urinary tract infection development and several morbidities in burn patients through alterations in the urinary microbiome and antimicrobial peptides. Overall, this study supports the concept that early assessment of urinary antimicrobial peptide responses and the bacterial microbiome may be used to predict susceptibility to urinary tract infections and sepsis in burn patients.
Collapse
|
50
|
Xu Y, Qin S, An T, Tang Y, Huang Y, Zheng L. MiR-145 detection in urinary extracellular vesicles increase diagnostic efficiency of prostate cancer based on hydrostatic filtration dialysis method. Prostate 2017; 77:1167-1175. [PMID: 28617988 DOI: 10.1002/pros.23376] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/18/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) can be detected in body fluids and may serve as disease biomarkers. Increasing evidence suggests that circulating miRNAs in serum and urine may be potential non-invasive biomarkers for prostate cancer (PCa). In the present study, we aimed to investigate whether hydrostatic filtration dialysis (HFD) is suitable for urinary EVs (UEVs) isolation and whether such reported PCa-related miRNAs can be detected in UEVs as PCa biomarkers. METHODS To analyze EVs miRNAs, we searched for an easy and economic method to enrich EVs from urine samples. We compared the efficiency of HFD method and conventional ultracentrifugation (UC) in isolating UEVs. Subsequently, UEVs were isolated from patients with PCa, patients with benign prostate hyperplasia (BPH) and healthy individuals. Differential expression of four PCa-related miRNAs (miR-572, miR-1290, miR-141, and miR-145) were measured in UEVs and paired serum EVs using SYBR Green-based quantitative reverse transcription-polymerase chain reaction (qRT-PCR). RESULTS The overall performance of HFD was similar to UC. In miRNA yield, both HFD and UC can meet the needs of further analysis. The level of miR-145 in UEVs was significantly increased in patients with PCa compared with the patients with BPH (P = 0.018). In addition, significant increase was observed in miR-145 levels when patients with Gleason score ≥8 tumors compared with Gleason score ≤7 (P = 0.020). Receiver-operating characteristic curve (ROC) revealed that miR-145 in UEVs combined with serum PSA could differentiate PCa from BPH better than PSA alone (AUC 0.863 and AUC 0.805, respectively). In serum EVs, four miRNAs were significantly higher in patients with PCa than with BPH. CONCLUSION HFD is appropriate for UEVs isolation and miRNA analysis when compared with conventional UC. miR-145 in UEVs is upregulated from PCa patients compared BPH patients and healthy controls. We suggest the potential use of UEVs miR-145 as a biomarker of PCa.
Collapse
Affiliation(s)
- Yong Xu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sihua Qin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Taixue An
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yueting Tang
- Department of Clinical Laboratory, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|