1
|
Akila AA, Gad RA, Ewees MGED, Abdul-Hamid M, Abdel-Reheim ES. Clopidogrel protects against gentamicin-induced nephrotoxicity through targeting oxidative stress, apoptosis, and coagulation pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2609-2625. [PMID: 39235475 PMCID: PMC11920383 DOI: 10.1007/s00210-024-03380-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
Gentamicin (Genta)-induced nephrotoxicity poses a significant clinical challenge due to its detrimental effects on kidney function. Clopidogrel (Clop), an antiplatelet drug known for its ability to prevent blood clots by inhibiting platelet aggregation, also has potential effects on oxidative stress and cell death. This study investigates Clop's protective role against Genta-induced nephrotoxicity, emphasizing the importance of the coagulation cascade. The 32 adult male albino rats were randomly assigned to four groups of eight (n = 8). The first group received only the vehicle. Genta was injected intraperitoneally at 100 mg/kg/day for 8 days in the second group. Groups 3 and 4 received oral Clop at 10 and 20 mg/kg/day for 1 week before Genta delivery and throughout the experiment. Renal tissue showed renal function tests, oxidative stress, pro-inflammatory cytokines, apoptotic markers, coagulation profile, and fibrin expression. Clop improved Genta-induced kidney function and histopathology. Clop substantially reduced pro-inflammatory cytokines, oxidative stress indicators, pro-apoptotic proteins, and fibrin protein. Clop also significantly boosted renal tissue anti-inflammatory and anti-apoptotic protein expression. Genta-induced nephrotoxicity involves oxidative stress, apoptosis, and coagulation system activation, according to studies. This study underscores that Genta-induced nephrotoxicity is associated with oxidative stress, apoptosis, and activation of the coagulation system. Clop's protective effects on nephrons are attributed to its anticoagulant, antioxidant, anti-inflammatory, and anti-apoptotic properties, presenting it as a promising therapeutic strategy against Genta-induced kidney damage.
Collapse
Affiliation(s)
- Asmaa A Akila
- Molecular Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Rania A Gad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, 62511, Egypt
| | - Mohamed Gamal El-Din Ewees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, 62511, Egypt.
| | - Manal Abdul-Hamid
- Cell Biology and Histology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Eman S Abdel-Reheim
- Molecular Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt
| |
Collapse
|
2
|
Lv L, Liu Y, Xiong J, Wang S, Li Y, Zhang B, Huang Y, Zhao J. Role of G protein coupled receptors in acute kidney injury. Cell Commun Signal 2024; 22:423. [PMID: 39223553 PMCID: PMC11367933 DOI: 10.1186/s12964-024-01802-8] [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: 06/11/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Acute kidney injury (AKI) is a clinical condition characterized by a rapid decline in kidney function, which is associated with local inflammation and programmed cell death in the kidney. The G protein-coupled receptors (GPCRs) represent the largest family of signaling transduction proteins in the body, and approximately 40% of drugs on the market target GPCRs. The expressions of various GPCRs, prostaglandin receptors and purinergic receptors, to name a few, are significantly altered in AKI models. And the role of GPCRs in AKI is catching the eyes of researchers due to their distinctive biological functions, such as regulation of hemodynamics, metabolic reprogramming, and inflammation. Therefore, in this review, we aim to discuss the role of GPCRs in the pathogenesis of AKI and summarize the relevant clinical trials involving GPCRs to assess the potential of GPCRs and their ligands as therapeutic targets in AKI and the transition to AKI-CKD.
Collapse
Affiliation(s)
- Liangjing Lv
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Yong Liu
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Jiachuan Xiong
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Shaobo Wang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Yan Li
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Bo Zhang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Yinghui Huang
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China
| | - Jinghong Zhao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
3
|
Farr E, Dimitrov D, Schmidt C, Turei D, Lobentanzer S, Dugourd A, Saez-Rodriguez J. MetalinksDB: a flexible and contextualizable resource of metabolite-protein interactions. Brief Bioinform 2024; 25:bbae347. [PMID: 39038934 PMCID: PMC11262834 DOI: 10.1093/bib/bbae347] [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: 02/06/2024] [Revised: 05/29/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
From the catalytic breakdown of nutrients to signaling, interactions between metabolites and proteins play an essential role in cellular function. An important case is cell-cell communication, where metabolites, secreted into the microenvironment, initiate signaling cascades by binding to intra- or extracellular receptors of neighboring cells. Protein-protein cell-cell communication interactions are routinely predicted from transcriptomic data. However, inferring metabolite-mediated intercellular signaling remains challenging, partially due to the limited size of intercellular prior knowledge resources focused on metabolites. Here, we leverage knowledge-graph infrastructure to integrate generalistic metabolite-protein with curated metabolite-receptor resources to create MetalinksDB. MetalinksDB is an order of magnitude larger than existing metabolite-receptor resources and can be tailored to specific biological contexts, such as diseases, pathways, or tissue/cellular locations. We demonstrate MetalinksDB's utility in identifying deregulated processes in renal cancer using multi-omics bulk data. Furthermore, we infer metabolite-driven intercellular signaling in acute kidney injury using spatial transcriptomics data. MetalinksDB is a comprehensive and customizable database of intercellular metabolite-protein interactions, accessible via a web interface (https://metalinks.omnipathdb.org/) and programmatically as a knowledge graph (https://github.com/biocypher/metalinks). We anticipate that by enabling diverse analyses tailored to specific biological contexts, MetalinksDB will facilitate the discovery of disease-relevant metabolite-mediated intercellular signaling processes.
Collapse
Affiliation(s)
- Elias Farr
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, United Kingdom
| | - Daniel Dimitrov
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Christina Schmidt
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Denes Turei
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Sebastian Lobentanzer
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- EMBL European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, United Kingdom
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- EMBL European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
4
|
van Megen WH, van Houtert TJ, Bos C, Peters DJM, de Baaij JHF, Hoenderop JGJ. Inhibition of pannexin-1 does not restore electrolyte balance in precystic Pkd1 knockout mice. Physiol Rep 2024; 12:e15956. [PMID: 38561249 PMCID: PMC10984814 DOI: 10.14814/phy2.15956] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/12/2024] [Accepted: 02/13/2024] [Indexed: 04/04/2024] Open
Abstract
Mutations in PKD1 and PKD2 cause autosomal dominant polycystic kidney disease (ADPKD), which is characterized by the formation of fluid-filled cysts in the kidney. In a subset of ADPKD patients, reduced blood calcium (Ca2+) and magnesium (Mg2+) concentrations are observed. As cystic fluid contains increased ATP concentrations and purinergic signaling reduces electrolyte reabsorption, we hypothesized that inhibiting ATP release could normalize blood Ca2+ and Mg2+ levels in ADPKD. Inducible kidney-specific Pkd1 knockout mice (iKsp-Pkd1-/-) exhibit hypocalcemia and hypomagnesemia in a precystic stage and show increased expression of the ATP-release channel pannexin-1. Therefore, we administered the pannexin-1 inhibitor brilliant blue-FCF (BB-FCF) every other day from Day 3 to 28 post-induction of Pkd1 gene inactivation. On Day 29, both serum Ca2+ and Mg2+ concentrations were reduced in iKsp-Pkd1-/- mice, while urinary Ca2+ and Mg2+ excretion was similar between the genotypes. However, serum and urinary levels of Ca2+ and Mg2+ were unaltered by BB-FCF treatment, regardless of genotype. BB-FCF did significantly decrease gene expression of the ion channels Trpm6 and Trpv5 in both control and iKsp-Pkd1-/- mice. Finally, no renoprotective effects of BB-FCF treatment were observed in iKsp-Pkd1-/- mice. Thus, administration of BB-FCF failed to normalize serum Ca2+ and Mg2+ levels.
Collapse
Affiliation(s)
- Wouter H. van Megen
- Department of Medical BiosciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Teun J. van Houtert
- Department of Medical BiosciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Caro Bos
- Department of Medical BiosciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Dorien J. M. Peters
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Jeroen H. F. de Baaij
- Department of Medical BiosciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Joost G. J. Hoenderop
- Department of Medical BiosciencesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
5
|
Cao H, Li L, Liu S, Wang Y, Liu X, Yang F, Dong W. The multifaceted role of extracellular ATP in sperm function: From spermatogenesis to fertilization. Theriogenology 2024; 214:98-106. [PMID: 37865020 DOI: 10.1016/j.theriogenology.2023.10.019] [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: 05/04/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Extracellular adenosine 5'-triphosphate (ATP) is a vital signaling molecule involved in various physiological processes within the body. In recent years, studies have revealed its significant role in male reproduction, particularly in sperm function. This review explores the multifaceted role of extracellular ATP in sperm function, from spermatogenesis to fertilization. We discuss the impact of extracellular ATP on spermatogenesis, sperm maturation and sperm-egg fusion, highlighting the complex regulatory mechanisms and potential clinical applications in the context of male infertility. By examining the latest research, we emphasize the crucial role of extracellular ATP in sperm function and propose future research directions to further.
Collapse
Affiliation(s)
- Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xianglin Liu
- College of Forestry, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Fangxia Yang
- College of Forestry, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
6
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
7
|
Grasselli C, Bombelli S, D'Esposito V, Di Tolla MF, L'Imperio V, Rocchio F, Miscione MS, Formisano P, Pagni F, Novelli R, Ruffini PA, Aramini A, Allegretti M, Perego R, De Filippis L. The therapeutic potential of an allosteric non-competitive CXCR1/2 antagonist for diabetic nephropathy. Diabetes Metab Res Rev 2023; 39:e3694. [PMID: 37470287 DOI: 10.1002/dmrr.3694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 07/21/2023]
Abstract
AIMS Diabetic nephropathy is a major consequence of inflammation developing in type 1 diabetes, with interleukin-8 (IL-8)-CXCR1/2 axis playing a key role in kidney disease progression. In this study, we investigated the therapeutic potential of a CXCR1/2 non-competitive allosteric antagonist (Ladarixin) in preventing high glucose-mediated injury in human podocytes and epithelial cells differentiated from renal stem/progenitor cells (RSC) cultured as nephrospheres. MATERIALS AND METHODS We used human RSCs cultured as nephrospheres through a sphere-forming functional assay to investigate hyperglycemia-mediated effects on IL-8 signalling in human podocytes and tubular epithelial cells. RESULTS High glucose impairs RSC self-renewal, induces an increase in IL-8 transcript expression and protein secretion and induces DNA damage in RSC-differentiated podocytes, while exerting no effect on RSC-differentiated epithelial cells. Accordingly, the supernatant from epithelial cells or podocytes cultured in high glucose was able to differentially activate leucocyte-mediated secretion of pro-inflammatory cytokines, suggesting that the crosstalk between immune and non-immune cells may be involved in disease progression in vivo. CONCLUSIONS Treatment with Ladarixin during RSC differentiation prevented high glucose-mediated effects on podocytes and modulated either podocyte or epithelial cell-dependent leucocyte secretion of pro-inflammatory cytokines, suggesting CXCR1/2 antagonists as possible pharmacological approaches for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Chiara Grasselli
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Silvia Bombelli
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Vittoria D'Esposito
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | - Vincenzo L'Imperio
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pathology Department, IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
| | | | | | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pathology Department, IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
| | - Rubina Novelli
- Research and Development, Dompé Farmaceutici S.p.A., Milano, Italy
| | | | | | | | - Roberto Perego
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | |
Collapse
|
8
|
Wang W, Wu S, Wang AY, Wu T, Luo H, Zhao JW, Chen J, Li Y, Ding H. Thrombomodulin activation driven by LXR agonist attenuates renal injury in diabetic nephropathy. Front Med (Lausanne) 2023; 9:916620. [PMID: 36698821 PMCID: PMC9870310 DOI: 10.3389/fmed.2022.916620] [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: 04/09/2022] [Accepted: 08/15/2022] [Indexed: 01/11/2023] Open
Abstract
Objective Inflammation and thrombosis are recognized as interrelated biological processes. Both thrombomodulin (TM) and factor XIII-A (FXIII-A) are involved in inflammation and coagulation process. However, their role in the pathogenesis of diabetic nephropathy (DN) remains unclear. In vitro study, the liver X receptor (LXR) agonist T0901317 can up-regulate the expression of TM in glomerular endothelial cells. Now we evaluated the interaction between TM activation and FXIII-A and their effects against renal injury. Methods We first evaluated the serum levels of FXIII-A and TM and the expression of TM, LXR-α and FXIII-A in renal tissues of patients with biopsy-proven DN. We then analyzed the expression of TM, LXR-α and FXIII-A in renal tissues of db/db DN mice after upregulating TM expression via T0901317 or downregulating its expression via transfection of TM shRNA-loaded adenovirus. We also investigated the serum levels of Tumor necrosis factor (TNF)-α, Interleukin (IL)-6, creatinine, and urinary microalbumin level in db/db mice. Results Our study showed that elevations in serum levels of FXIII-A positively correlated to the serum levels of TM and were also associated with end-stage kidney disease in patients with DN. The number of TM+ cells in the renal tissues of patients with DN negatively correlated with the number of FXIII-A+ cells and positively correlated with the number of LXR-α+ cells and estimated glomerular filtration rate (eGFR), whereas the number of FXIII-A+ cells negatively correlated with the eGFR. Conclusion Thrombomodulin activation with T0901317 downregulated FXIII-A expression in the kidney tissue and alleviated renal injury in db/db mice.
Collapse
Affiliation(s)
- Wei Wang
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Song Wu
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Amanda Y. Wang
- Renal and Metabolic Division, The George Institute for Global Health, University of New South Wales Australia, Newtown, NSW, Australia,Department of Renal Medicine, Concord Repatriation General Hospital, Concord Clinical School, University of Sydney, Camperdown, NSW, Australia,Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia,*Correspondence: Amanda Y. Wang ✉
| | - Tao Wu
- Internal Medicine, Louisiana State University Health Science at Shreveport, Shreveport, LA, United States
| | - Haojun Luo
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jia Wei Zhao
- The Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Jin Chen
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yi Li
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hanlu Ding
- Renal Division and Institute of Nephrology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China,Hanlu Ding ✉
| |
Collapse
|
9
|
Wang L, Li J, Liao R, Li Y, Jiang L, Zhang Z, Geng J, Fu P, Su B, Zhao Y. Resolvin D1 attenuates sepsis induced acute kidney injury targeting mitochondria and NF-κB signaling pathway. Heliyon 2022; 8:e12269. [PMID: 36578378 PMCID: PMC9791840 DOI: 10.1016/j.heliyon.2022.e12269] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/13/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Background Acute kidney injury is a highly common and multifactorial renal disease resulting in significant morbidity and mortality, especially sepsis-induced acute kidney injury. There is no effective therapy available to treat or prevent sepsis-induced acute kidney injury. One of the specialized pro-resolving mediators, Resolvin D1 exhibits special anti-inflammatory effects in several inflammatory disease models, but there is little evidence about the effect and mechanism of Resolvin D1 in sepsis-induced acute kidney injury. Methods We conducted experiments to explore the effect and mechanism of Resolvin D1 in sepsis-induced acute kidney injury. In vitro, human proximal tubular epithelial cells were used to test the apoptosis ratio, cell viability and reactive oxygen species level. In vivo, C57BL/6 mice were injected with lipopolysaccharide to establish a sepsis-induced acute kidney injury model. Renal function and structure, apoptosis ratio of kidney cells, mitochondrial structure and function and related protein and gene levels were assessed. Results In vitro, the resolvin D1-treated group showed higher cell viability and lower reactive oxygen species levels and apoptosis ratios than the LPS group. In vivo, Resolvin D1 can not only improve renal function and mitochondrial function but also reduce the apoptosis ratio, while mediating mitochondrial dynamics and inhibiting NF-κB pathway. Conclusions Resolvin D1 has a good renoprotective effect by maintaining mitochondrial dynamics and inhibiting the NF-κB pathway.
Collapse
Affiliation(s)
- Liya Wang
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiameng Li
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruoxi Liao
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yupei Li
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu 610207, China,Disaster Medicine Center, Sichuan University, Chengdu, 610041, China
| | - Luojia Jiang
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuyun Zhang
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiwen Geng
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ping Fu
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China,Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Baihai Su
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China,The First People's Hospital of Shuangliu District, Chengdu, 610200, China,Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu 610207, China,Disaster Medicine Center, Sichuan University, Chengdu, 610041, China,Med-X Center for Materials, Sichuan University, Chengdu 610041, China,Corresponding author.
| | - Yuliang Zhao
- Department of Nephrology, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu 610041, China,Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China,Corresponding author.
| |
Collapse
|
10
|
Doan Ngoc TM, Tilly G, Danger R, Bonizec O, Masset C, Guérif P, Bruneau S, Glemain A, Harb J, Cadoux M, Vivet A, Mai HL, Garcia A, Laplaud D, Liblau R, Giral M, Blandin S, Feyeux M, Dubreuil L, Pecqueur C, Cyr M, Ni W, Brouard S, Degauque N. Effector Memory-Expressing CD45RA (TEMRA) CD8 + T Cells from Kidney Transplant Recipients Exhibit Enhanced Purinergic P2X4 Receptor-Dependent Proinflammatory and Migratory Responses. J Am Soc Nephrol 2022; 33:2211-2231. [PMID: 36280286 PMCID: PMC9731633 DOI: 10.1681/asn.2022030286] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/22/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The mechanisms regulating CD8+ T cell migration to nonlymphoid tissue during inflammation have not been fully elucidated, and the migratory properties of effector memory CD8+ T cells that re-express CD45RA (TEMRA CD8+ T cells) remain unclear, despite their roles in autoimmune diseases and allotransplant rejection. METHODS We used single-cell proteomic profiling and functional testing of CD8+ T cell subsets to characterize their effector functions and migratory properties in healthy volunteers and kidney transplant recipients with stable or humoral rejection. RESULTS We showed that humoral rejection of a kidney allograft is associated with an accumulation of cytolytic TEMRA CD8+ T cells in blood and kidney graft biopsies. TEMRA CD8+ T cells from kidney transplant recipients exhibited enhanced migratory properties compared with effector memory (EM) CD8+ T cells, with enhanced adhesion to activated endothelium and transmigration in response to the chemokine CXCL12. CXCL12 directly triggers a purinergic P2×4 receptor-dependent proinflammatory response of TEMRA CD8+ T cells from transplant recipients. The stimulation with IL-15 promotes the CXCL12-induced migration of TEMRA and EM CD8+ T cells and promotes the generation of functional PSGL1, which interacts with the cell adhesion molecule P-selectin and adhesion of these cells to activated endothelium. Although disruption of the interaction between functional PSGL1 and P-selectin prevents the adhesion and transmigration of both TEMRA and EM CD8+ T cells, targeting VLA-4 or LFA-1 (integrins involved in T cell migration) specifically inhibited the migration of TEMRA CD8+ T cells from kidney transplant recipients. CONCLUSIONS Our findings highlight the active role of TEMRA CD8+ T cells in humoral transplant rejection and suggest that kidney transplant recipients may benefit from therapeutics targeting these cells.
Collapse
Affiliation(s)
- Tra-My Doan Ngoc
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Gaëlle Tilly
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Richard Danger
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Orianne Bonizec
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Christophe Masset
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Pierrick Guérif
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Sarah Bruneau
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Alexandre Glemain
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jean Harb
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Marion Cadoux
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Anaïs Vivet
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Hoa Le Mai
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Alexandra Garcia
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - David Laplaud
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Roland Liblau
- CNRS, Institut National de la Santé et de la Recherche Médicale, UPS, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, Toulouse, France
- Department of Immunology, Toulouse University Hospital, Toulouse, France
| | - Magali Giral
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Stéphanie Blandin
- CHU Nantes, CNRS, Institut National de la Santé et de la Recherche Médicale, BioCore, US16, SFR Bonamy, Nantes Université, Nantes, France
| | - Magalie Feyeux
- CHU Nantes, CNRS, Institut National de la Santé et de la Recherche Médicale, BioCore, US16, SFR Bonamy, Nantes Université, Nantes, France
| | | | - Claire Pecqueur
- Université d’Angers, Institut National de la Santé et de la Recherche Médicale, CNRS, CRCI2NA, Nantes Université, Nantes, France
| | - Matthew Cyr
- IsoPlexis Corporation, Branford, Connecticut
| | - Weiming Ni
- IsoPlexis Corporation, Branford, Connecticut
| | - Sophie Brouard
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- CHU Nantes, Nantes Université, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Nicolas Degauque
- Institut National de la Santé et de la Recherche Médicale, Nantes Université, CHU Nantes, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
11
|
Giuliani KTK, Grivei A, Nag P, Wang X, Rist M, Kildey K, Law B, Ng MS, Wilkinson R, Ungerer J, Forbes JM, Healy H, Kassianos AJ. Hypoxic human proximal tubular epithelial cells undergo ferroptosis and elicit an NLRP3 inflammasome response in CD1c + dendritic cells. Cell Death Dis 2022; 13:739. [PMID: 36030251 PMCID: PMC9420140 DOI: 10.1038/s41419-022-05191-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023]
Abstract
Inflammasomes are multiprotein platforms responsible for the release of pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Mouse studies have identified inflammasome activation within dendritic cells (DC) as pivotal for driving tubulointerstitial fibrosis and inflammation, the hallmarks of chronic kidney disease (CKD). However, translation of this work to human CKD remains limited. Here, we examined the complex tubular cell death pathways mediating inflammasome activation in human kidney DC and, thus, CKD progression. Ex vivo patient-derived proximal tubular epithelial cells (PTEC) cultured under hypoxic (1% O2) conditions modelling the CKD microenvironment showed characteristics of ferroptotic cell death, including mitochondrial dysfunction, reductions in the lipid repair enzyme glutathione peroxidase 4 (GPX4) and increases in lipid peroxidation by-product 4-hydroxynonenal (4-HNE) compared with normoxic PTEC. The addition of ferroptosis inhibitor, ferrostatin-1, significantly reduced hypoxic PTEC death. Human CD1c+ DC activated in the presence of hypoxic PTEC displayed significantly increased production of inflammasome-dependent cytokines IL-1β and IL-18. Treatment of co-cultures with VX-765 (caspase-1/4 inhibitor) and MCC950 (NLRP3 inflammasome inhibitor) significantly attenuated IL-1β/IL-18 levels, supporting an NLRP3 inflammasome-dependent DC response. In line with these in vitro findings, in situ immunolabelling of human fibrotic kidney tissue revealed a significant accumulation of tubulointerstitial CD1c+ DC containing active inflammasome (ASC) specks adjacent to ferroptotic PTEC. These data establish ferroptosis as the primary pattern of PTEC necrosis under the hypoxic conditions of CKD. Moreover, this study identifies NLRP3 inflammasome signalling driven by complex tubulointerstitial PTEC-DC interactions as a key checkpoint for therapeutic targeting in human CKD.
Collapse
Affiliation(s)
- Kurt T. K. Giuliani
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Anca Grivei
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Purba Nag
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Xiangju Wang
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Melissa Rist
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Katrina Kildey
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia
| | - Becker Law
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1024.70000000089150953Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD Australia
| | - Monica S. Ng
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Institute of Molecular Biosciences, University of Queensland, Brisbane, QLD Australia ,grid.412744.00000 0004 0380 2017Department of Nephrology, Princess Alexandra Hospital, Brisbane, QLD Australia
| | - Ray Wilkinson
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1024.70000000089150953Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD Australia
| | - Jacobus Ungerer
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Josephine M. Forbes
- grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Mater Research Institute, University of Queensland, Brisbane, QLD Australia
| | - Helen Healy
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia
| | - Andrew J. Kassianos
- grid.415606.00000 0004 0380 0804Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD Australia ,grid.416100.20000 0001 0688 4634Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Faculty of Medicine, University of Queensland, Brisbane, QLD Australia ,grid.1024.70000000089150953Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD Australia
| |
Collapse
|
12
|
Lazzaroni E, Lunati ME, Montefusco L, Pastore I, Chebat E, Cimino V, Morpurgo PS, Muratori M, Plebani L, Bolla A, Rossi A, Vallone L, Gandolfi A, Tinari C, D'Addio F, Nasr MB, Loretelli C, Scaranna C, Bellante R, Manfrini R, Muratori F, Franzetti I, Orsi E, Gazzaruso C, Ghelardi R, Desenzani P, Genovese S, Girelli A, Folli F, Berra C, Fiorina P. Dapagliflozin acutely improves kidney function in type 2 diabetes mellitus. The PRECARE study. Pharmacol Res 2022; 183:106374. [PMID: 35908663 DOI: 10.1016/j.phrs.2022.106374] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/10/2023]
Abstract
Dapagliflozin has been demonstrated to improve glycemic control, blood pressure, and body weight in type 2 diabetes mellitus (T2D); indeed, it can also reduce the risk of progression to renal failure, of hospitalization for heart failure and of cardiovascular death. We aim to investigate the acute effect of Dapagliflozin on kidney function in the common clinical practice in T2D. This is a study including 1402 patients with T2D recruited from 11 centers in Lombardia, Italy, who were evaluated at baseline and after 6 months of treatment with Dapagliflozin 10 mg per day. The primary outcome of the study was the change in HbA1c, while the secondary outcomes were modification of weight, BMI, systolic and diastolic pressure, creatinine, eGFR and albuminuria status. After 24 weeks of treatment with Dapagliflozin, a reduction in Hb1Ac was observed (-0.6 ± 1.8%) as well as in BMI (-1.5 ± 5.2 kg/m2). Statistically significant changes were also found for systolic and diastolic blood pressure, cholesterol and triglycerides. Interestingly, a statistically significant acute improvement of kidney function was evident. Our analyses confirm the beneficial effects of dapagliflozin after 6 months of therapy, with improvements of glycemic and lipid profiles, blood pressure, BMI. Finally, an acute positive effect on albuminuria and KIDGO classes was observed during a 6 months treatment with dapagliflozin in patients with T2D.
Collapse
Affiliation(s)
- Elisa Lazzaroni
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy; Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università Degli Studi Milano, Milan, Italy
| | | | - Laura Montefusco
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Enrica Chebat
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Vincenzo Cimino
- Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università Degli Studi Milano, Milan, Italy; Endocrinology and Diabetology, Pio Albergo Trivulzio, Milan, Italy
| | | | - Milena Muratori
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Laura Plebani
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Andrea Bolla
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Antonio Rossi
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Luciana Vallone
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Camilla Tinari
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Francesca D'Addio
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy; Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università Degli Studi Milano, Milan, Italy
| | - Moufida Ben Nasr
- Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università Degli Studi Milano, Milan, Italy
| | - Cristian Loretelli
- Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università Degli Studi Milano, Milan, Italy
| | - Cristiana Scaranna
- Division of Endocrinology and Diabetology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Rosalia Bellante
- Division of Endocrinology and Diabetology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Roberto Manfrini
- Endocrinology and Metabolism, Department of Health Science, Università degli Studi di Milano, Milan, Italy; Departmental Unit of Diabetes and Metabolism, San Paolo Hospital, ASST Santi Paolo e Carlo, Italy
| | - Fabrizio Muratori
- Division of Endocrinology and Diabetology, Sant'Anna Hospital - ASST Lariana, Como, Italy
| | - Ivano Franzetti
- Division of Endocrinology and Diabetology, S. Antonio Abate Hospital, Gallarate, Italy
| | - Emanuela Orsi
- Diabetes Service, Endocrinology and Metabolic Diseases Unit, IRCCS "Cà Granda - Ospedale Maggiore Policlinico" Foundation, Italy
| | - Carmine Gazzaruso
- Diabetes and Endocrine, Metabolic and Vascular Diseases Unit and the Centre for Applied Clinical Research (Ce.R.C.A.) Clinical Institute "Beato Matteo" (Hospital Group San Donato), Vigevano, Italy
| | - Renata Ghelardi
- Unit of Diabetology, ASST Melegnano-Martesana, San Giuliano Milanese, Italy
| | - Paolo Desenzani
- Unit of Diabetology, ASST Spedali Civili, Montichiari, Brescia, Italy
| | | | | | - Franco Folli
- Endocrinology and Metabolism, Department of Health Science, Università degli Studi di Milano, Milan, Italy; Departmental Unit of Diabetes and Metabolism, San Paolo Hospital, ASST Santi Paolo e Carlo, Italy
| | - Cesare Berra
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS Multimedica, Milan, Italy
| | - Paolo Fiorina
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy; Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università Degli Studi Milano, Milan, Italy; Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Bao S, Wang X, Ma Q, Wei C, Nan J, Ao W. Mongolian medicine in treating type 2 diabetes mellitus combined with nonalcoholic fatty liver disease via FXR/LXR-mediated P2X7R/NLRP3/NF-κB pathway activation. CHINESE HERBAL MEDICINES 2022; 14:367-375. [PMID: 36118003 PMCID: PMC9476729 DOI: 10.1016/j.chmed.2022.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/17/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD) are the most problematic metabolic diseases in the world. NAFLD encompasses a spectrum of severity, ranging from simple steatosis to non-alcoholic steatohepatitis (NASH) and fibrosis, increasing the risk of cirrhosis and hepatocellular carcinoma. Importantly, NAFLD is closely linked to obesity and tightly interrelated with insulin resistance and T2DM. T2DM and NAFLD (T2DM-NAFLD) are called as the Xike Rixijing Disease and Tonglaga Indigestion Disease respectively, in Mongolian medicine. Xike Rixijing Disease maybe develop into Tonglaga Indigestion Disease. Forturnately many Mongolian medicines show efficient treatment of T2DM-NAFLD, such as Agriophyllum squarrosum, Haliyasu (dried powder of camel placenta), Digeda-4 (herbs of Lomatogonium carinthiacum, rhizomata of Coptis chinensis, ripe fruits of Gardenia jasminoides, herbs of Dianthus superbus), Guangmingyan Siwei Decoction Powder (Halite, ripe fruits of Terminalia chebula, rhizomata of Zingiber officinale, fruit clusters of Piper longum), Tonglaga-5 (ripe fruits of Punica granatum, barks of Cinnamomum cassia, ripe fruits of Amomum kravanh, fruit clusters of Piper longum, flowers of Carthamus tinctorius), Tegexidegeqi (rhizomata of Inula helenium, ripe fruits of Gardenia jasminoides, rhizomata of Platycodon grandiflorum, rhizomata of Coptis chinensis, heartwood of Caesalpinia sappan), Ligan Shiliu Bawei San (ripe fruits of Punica granatum, barks of Cinnamomum cassia, ripe fruits of Amomum kravanh, fruit clusters of Piper longum, flowers of Carthamus tinctorius, ripe fruits of Amomum tsao-ko, rhizomata of Zingiber officinale), etc. Principles of Mongolian medicine in treating diseases: by balancing “three essences or roots” and “seven elements”, strengthening liver and kidney function, transporting nutrients to enhance physical strength and disease resistance, and combined with drugs for comprehensive conditioning treatment. However, their molecular mechanisms remain unclear. In this review, we prospect that Mongolian medicines might be a promising treatment for T2DM-NAFLD by activating P2X7R/NLRP3/NF-κB inflammatory pathway via lipid-sensitive nuclear receptors (i.e., FXR and LXR).
Collapse
Affiliation(s)
- Shuyin Bao
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao 028000, China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
- Jilin Key Laboratory for Traditional Chinese Korean Medicine, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Xiuzhi Wang
- Department of Medicines and Foods, Tongliao Vocational College, Tongliao 028000, China
| | - Qianqian Ma
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao 028000, China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Chengxi Wei
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao 028000, China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
- Corresponding authors.
| | - Jixing Nan
- Jilin Key Laboratory for Traditional Chinese Korean Medicine, College of Pharmacy, Yanbian University, Yanji 133002, China
- Corresponding authors.
| | - Wuliji Ao
- Research and development center, Inner Mongolia Research Institute of Traditional Mongolian Medicine Engineering Technology, Tongliao 028000, China
- Mongolian Medicine R&D National Local Union Engineering Research Center, Inner Mongolia Minzu University, Tongliao 028000, China
- Corresponding authors.
| |
Collapse
|
14
|
Blocking connexin 43 and its promotion of ATP release from renal tubular epithelial cells ameliorates renal fibrosis. Cell Death Dis 2022; 13:511. [PMID: 35641484 PMCID: PMC9156700 DOI: 10.1038/s41419-022-04910-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023]
Abstract
Whether metabolites derived from injured renal tubular epithelial cells (TECs) participate in renal fibrosis is poorly explored. After TEC injury, various metabolites are released and among the most potent is adenosine triphosphate (ATP), which is released via ATP-permeable channels. In these hemichannels, connexin 43 (Cx43) is the most common member. However, its role in renal interstitial fibrosis (RIF) has not been fully examined. We analyzed renal samples from patients with obstructive nephropathy and mice with unilateral ureteral obstruction (UUO). Cx43-KSP mice were generated to deplete Cx43 in TECs. Through transcriptomics, metabolomics, and single-cell sequencing multi-omics analysis, the relationship among tubular Cx43, ATP, and macrophages in renal fibrosis was explored. The expression of Cx43 in TECs was upregulated in both patients and mice with obstructive nephropathy. Knockdown of Cx43 in TECs or using Cx43-specific inhibitors reduced UUO-induced inflammation and fibrosis in mice. Single-cell RNA sequencing showed that ATP specific receptors, including P2rx4 and P2rx7, were distributed mainly on macrophages. We found that P2rx4- or P2rx7-positive macrophages underwent pyroptosis after UUO, and in vitro ATP directly induced pyroptosis by macrophages. The administration of P2 receptor or P2X7 receptor blockers to UUO mice inhibited macrophage pyroptosis and demonstrated a similar degree of renoprotection as Cx43 genetic depletion. Further, we found that GAP 26 (a Cx43 hemichannel inhibitor) and A-839977 (an inhibitor of the pyroptosis receptor) alleviated UUO-induced fibrosis, while BzATP (the agonist of pyroptosis receptor) exacerbated fibrosis. Single-cell sequencing demonstrated that the pyroptotic macrophages upregulated the release of CXCL10, which activated intrarenal fibroblasts. Cx43 mediates the release of ATP from TECs during renal injury, inducing peritubular macrophage pyroptosis, which subsequently leads to the release of CXCL10 and activation of intrarenal fibroblasts and acceleration of renal fibrosis.
Collapse
|
15
|
Liu X, Du H, Sun Y, Shao L. Role of abnormal energy metabolism in the progression of chronic kidney disease and drug intervention. Ren Fail 2022; 44:790-805. [PMID: 35535500 PMCID: PMC9103584 DOI: 10.1080/0886022x.2022.2072743] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) is a severe clinical syndrome with significant socioeconomic impact worldwide. Orderly energy metabolism is essential for normal kidney function and energy metabolism disorders are increasingly recognized as an important player in CKD. Energy metabolism disorders are characterized by ATP deficits and reactive oxygen species increase. Oxygen and mitochondria are essential for ATP production, hypoxia and mitochondrial dysfunction both affect the energy production process. Renin-angiotensin and adenine signaling pathway also play important regulatory roles in energy metabolism. In addition, disturbance of energy metabolism is a key factor in the development of hereditary nephropathy such as autosomal dominant polycystic kidney disease. Currently, drugs with clinically clear renal function protection, such as Angiotensin II Type 1 receptor blockers and fenofibrate, have been proven to improve energy metabolism disorders. The sodium-glucose co-transporter inhibitors 2 that can mediate glucose metabolism disorders not only delay the progress of diabetic nephropathy, but also have significant protective effects in non-diabetic nephropathy. Hypoxia-inducible factor enhances ATP production to the kidney by improving renal oxygen supply and increasing glycolysis, and the mitochondria targeted peptides (SS-31) plays a protective role by stabilizing the mitochondrial inner membrane. Moreover, several drugs are being studied and are predicted to have potential renal protective properties. We propose that the regulation of energy metabolism represents a promising strategy to delay the progression of CKD.
Collapse
Affiliation(s)
- Xuyan Liu
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Huasheng Du
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Yan Sun
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| | - Leping Shao
- Department of Nephrology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
16
|
Ben-Nissan G, Katzir N, Füzesi-Levi MG, Sharon M. Biology of the Extracellular Proteasome. Biomolecules 2022; 12:619. [PMID: 35625547 PMCID: PMC9139032 DOI: 10.3390/biom12050619] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
Proteasomes are traditionally considered intracellular complexes that play a critical role in maintaining proteostasis by degrading short-lived regulatory proteins and removing damaged proteins. Remarkably, in addition to these well-studied intracellular roles, accumulating data indicate that proteasomes are also present in extracellular body fluids. Not much is known about the origin, biological role, mode(s) of regulation or mechanisms of extracellular transport of these complexes. Nevertheless, emerging evidence indicates that the presence of proteasomes in the extracellular milieu is not a random phenomenon, but rather a regulated, coordinated physiological process. In this review, we provide an overview of the current understanding of extracellular proteasomes. To this end, we examine 143 proteomic datasets, leading us to the realization that 20S proteasome subunits are present in at least 25 different body fluids. Our analysis also indicates that while 19S subunits exist in some of those fluids, the dominant proteasome activator in these compartments is the PA28α/β complex. We also elaborate on the positive correlations that have been identified in plasma and extracellular vesicles, between 20S proteasome and activity levels to disease severity and treatment efficacy, suggesting the involvement of this understudied complex in pathophysiology. In addition, we address the considerations and practical experimental methods that should be taken when investigating extracellular proteasomes. Overall, we hope this review will stimulate new opportunities for investigation and thoughtful discussions on this exciting topic that will contribute to the maturation of the field.
Collapse
Affiliation(s)
| | | | | | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel; (G.B.-N.); (N.K.); (M.G.F.-L.)
| |
Collapse
|
17
|
Hossain S, Chow CWK, Cook D, Sawade E, Hewa GA. Review of Nitrification Monitoring and Control Strategies in Drinking Water System. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19074003. [PMID: 35409686 PMCID: PMC8997939 DOI: 10.3390/ijerph19074003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 12/29/2022]
Abstract
Nitrification is a major challenge in chloraminated drinking water systems, resulting in undesirable loss of disinfectant residual. Consequently, heterotrophic bacteria growth is increased, which adversely affects the water quality, causing taste, odour, and health issues. Regular monitoring of various water quality parameters at susceptible areas of the water distribution system (WDS) helps to detect nitrification at an earlier stage and allows sufficient time to take corrective actions to control it. Strategies to monitor nitrification in a WDS require conducting various microbiological tests or assessing surrogate parameters that are affected by microbiological activities. Additionally, microbial decay factor (Fm) is used by water utilities to monitor the status of nitrification. In contrast, approaches to manage nitrification in a WDS include controlling various factors that affect monochloramine decay rate and ammonium substrate availability, and that can inhibit nitrification. However, some of these control strategies may increase the regulated disinfection-by-products level, which may be a potential health concern. In this paper, various strategies to monitor and control nitrification in a WDS are critically examined. The key findings are: (i) the applicability of some methods require further validation using real WDS, as the original studies were conducted on laboratory or pilot systems; (ii) there is no linkage/formula found to relate the surrogate parameters to the concentration of nitrifying bacteria, which possibly improve nitrification monitoring performance; (iii) improved methods/monitoring tools are required to detect nitrification at an earlier stage; (iv) further studies are required to understand the effect of soluble microbial products on the change of surrogate parameters. Based on the current review, we recommend that the successful outcome using many of these methods is often site-specific, hence, water utilities should decide based on their regular experiences when considering economic and sustainability aspects.
Collapse
Affiliation(s)
- Sharif Hossain
- Scarce Resources and Circular Economy (ScaRCE), UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia; (C.W.K.C.); (G.A.H.)
- Correspondence:
| | - Christopher W. K. Chow
- Scarce Resources and Circular Economy (ScaRCE), UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia; (C.W.K.C.); (G.A.H.)
- Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - David Cook
- South Australian Water Corporation, Adelaide, SA 5000, Australia; (D.C.); (E.S.)
| | - Emma Sawade
- South Australian Water Corporation, Adelaide, SA 5000, Australia; (D.C.); (E.S.)
| | - Guna A. Hewa
- Scarce Resources and Circular Economy (ScaRCE), UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia; (C.W.K.C.); (G.A.H.)
| |
Collapse
|
18
|
de Oliveira AA, Mendoza VO, Rastogi S, Nunes KP. New insights into the role and therapeutic potential of HSP70 in diabetes. Pharmacol Res 2022; 178:106173. [PMID: 35278625 DOI: 10.1016/j.phrs.2022.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
Abstract
Emerging evidence indicates that HSP70 represents a key mechanism in the pathophysiology of β-cell dysfunction, insulin resistance, and various diabetic complications, including micro- and macro-vascular alterations, as well as impaired hemostasis. Hyperglycemia, a hallmark of both types of diabetes, increases the circulating levels of HSP70 (eHSP70), but there is still divergence about whether diabetes up- or down-regulates the intracellular fraction of this protein (iHSP70). Here, we consider that iHSP70 levels reduce in diabetic arterial structures and that the vascular system is in direct contact with all other systems in the body suggesting that a systemic response might also be happening for iHSP70, which is characterized by decreased levels of HSP70 in the vasculature. Furthermore, although many pathways have been proposed to explain HSP70's functions in diabetes, and organs/tissues/cells-specific variations occur, the membrane-bound receptor of the innate immune system, Toll-like receptor 4, and its downstream signal transduction pathways appear to be a constant, not only when we explore the actions of eHSP70, but also when we assess the contributions of iHSP70. In this review, we focus on discussing the multiple roles of HSP70 across organs/tissues/cells affected by hyperglycemia to further explore the possibility of targeting this protein with pharmacological and non-pharmacological approaches in the context of diabetes.
Collapse
Affiliation(s)
- Amanda Almeida de Oliveira
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Valentina Ochoa Mendoza
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Swasti Rastogi
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Kenia Pedrosa Nunes
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States.
| |
Collapse
|
19
|
Xie Y, Hu X, Li S, Qiu Y, Cao R, Xu C, Lu C, Wang Z, Yang J. Pharmacological targeting macrophage phenotype via gut-kidney axis ameliorates renal fibrosis in mice. Pharmacol Res 2022; 178:106161. [DOI: 10.1016/j.phrs.2022.106161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/06/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022]
|
20
|
Novel Soluble Mediators of Innate Immune System Activation in Solid Allograft Rejection. Transplantation 2022; 106:500-509. [PMID: 34049364 DOI: 10.1097/tp.0000000000003834] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During the past years, solid allograft rejection has been considered the consequence of either cellular- or antibody-mediated reaction both being part of the adaptive immune response, whereas the role of innate immunity has been mostly considered less relevant. Recently, a large body of evidence suggested that the innate immune response and its soluble mediators may play a more important role during solid allograft rejection than originally thought. This review will highlight the role of novel soluble mediators that are involved in the activation of innate immunity during alloimmune response and solid allograft rejection. We will also discuss emerging strategies to alleviate the aforementioned events. Hence, novel, feasible, and safe clinical therapies are needed to prevent allograft loss in solid organ transplantation. Fully understanding the role of soluble mediators of innate immune system activation may help to mitigate solid allograft rejection and improve transplanted recipients' outcomes.
Collapse
|
21
|
Kulesza T, Typiak M, Rachubik P, Audzeyenka I, Rogacka D, Angielski S, Saleem MA, Piwkowska A. Hyperglycemic environment disrupts phosphate transporter function and promotes calcification processes in podocytes and isolated glomeruli. J Cell Physiol 2022; 237:2478-2491. [PMID: 35150131 DOI: 10.1002/jcp.30700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/20/2022] [Accepted: 02/02/2022] [Indexed: 11/10/2022]
Abstract
Soft tissue calcification is a pathological phenomenon that often occurs in end-stage chronic kidney disease (CKD), which is caused by diabetic nephropathy, among other factors. Hyperphosphatemia present during course of CKD contributes to impairments in kidney function, particularly damages in the glomerular filtration barrier (GFB). Essential elements of the GFB include glomerular epithelial cells, called podocytes. In the present study, we found that human immortalized podocytes express messenger RNA and protein of phosphate transporters, including NaPi 2c (SLC34A3), Pit 1 (SLC20A1), and Pit 2 (SLC20A2), which are sodium-dependent and mediate intracellular phosphate (Pi) transport, and XPR1, which is responsible for extracellular Pi transport. We found that cells that were grown in a medium with a high glucose (HG) concentration (30 mM) expressed less Pit 1 and Pit 2 protein than podocytes that were cultured in a standard glucose medium (11 mM). We found that exposure of the analyzed transporters in the cell membrane of the podocyte is altered by HG conditions. We also found that the activity of tissue nonspecific alkaline phosphatase increased in HG, causing a rise in Pi generation. Additionally, HG led to a reduction of the amount of ectonucleotide pyrophosphatase/phosphodiesterase 1 in the cell membrane of podocytes. The extracellular concentration of pyrophosphate also decreased under HG conditions. These data suggest that a hyperglycemic environment enhances the production of Pi in podocytes and its retention in the extracellular space, which may induce glomerular calcification.
Collapse
Affiliation(s)
- Tomasz Kulesza
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Marlena Typiak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland.,Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Patrycja Rachubik
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Irena Audzeyenka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland.,Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Dorota Rogacka
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland.,Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Stefan Angielski
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | | | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland.,Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| |
Collapse
|
22
|
Massé K, Bhamra S, Paroissin C, Maneta-Peyret L, Boué-Grabot E, Jones EA. The enpp4 ectonucleotidase regulates kidney patterning signalling networks in Xenopus embryos. Commun Biol 2021; 4:1158. [PMID: 34620987 PMCID: PMC8497618 DOI: 10.1038/s42003-021-02688-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/17/2021] [Indexed: 11/30/2022] Open
Abstract
The enpp ectonucleotidases regulate lipidic and purinergic signalling pathways by controlling the extracellular concentrations of purines and bioactive lipids. Although both pathways are key regulators of kidney physiology and linked to human renal pathologies, their roles during nephrogenesis remain poorly understood. We previously showed that the pronephros was a major site of enpp expression and now demonstrate an unsuspected role for the conserved vertebrate enpp4 protein during kidney formation in Xenopus. Enpp4 over-expression results in ectopic renal tissues and, on rare occasion, complete mini-duplication of the entire kidney. Enpp4 is required and sufficient for pronephric markers expression and regulates the expression of RA, Notch and Wnt pathway members. Enpp4 is a membrane protein that binds, without hydrolyzing, phosphatidylserine and its effects are mediated by the receptor s1pr5, although not via the generation of S1P. Finally, we propose a novel and non-catalytic mechanism by which lipidic signalling regulates nephrogenesis.
Collapse
Affiliation(s)
- Karine Massé
- School of Life Sciences, Warwick University, Coventry, CV47AL, UK.
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000, Bordeaux, France.
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000, Bordeaux, France.
| | - Surinder Bhamra
- School of Life Sciences, Warwick University, Coventry, CV47AL, UK
| | - Christian Paroissin
- Université de Pau et des Pays de l'Adour, Laboratoire de Mathématiques et de leurs Applications-UMR CNRS 5142, 64013, Pau cedex, France
| | - Lilly Maneta-Peyret
- Université de Bordeaux, CNRS, Laboratoire de Biogenèse Membranaire UMR 5200, F-33800, Villenave d'Ornon, France
| | - Eric Boué-Grabot
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000, Bordeaux, France
| | | |
Collapse
|
23
|
Kerkman PF, Dernstedt A, Tadala L, Mittler E, Dannborg M, Sundling C, Maleki KT, Tauriainen J, Tuiskunen‐Bäck A, Wigren Byström J, Ocaya P, Thunberg T, Jangra RK, Román‐Sosa G, Guardado‐Calvo P, Rey FA, Klingström J, Chandran K, Puhar A, Ahlm C, Forsell MNE. Generation of plasma cells and CD27 -IgD - B cells during hantavirus infection is associated with distinct pathological findings. Clin Transl Immunology 2021; 10:e1313. [PMID: 34277007 PMCID: PMC8275445 DOI: 10.1002/cti2.1313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 04/19/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Human hantavirus infections can cause haemorrhagic fever with renal syndrome (HFRS). The pathogenic mechanisms are not fully understood, nor if they affect the humoral immune system. The objective of this study was to investigate humoral immune responses to hantavirus infection and to correlate them to the typical features of HFRS: thrombocytopenia and transient kidney dysfunction. METHODS We performed a comprehensive characterisation of longitudinal antiviral B-cell responses of 26 hantavirus patients and combined this with paired clinical data. In addition, we measured extracellular adenosine triphosphate (ATP) and its breakdown products in circulation and performed in vitro stimulations to address its effect on B cells. RESULTS We found that thrombocytopenia was correlated to an elevated frequency of plasmablasts in circulation. In contrast, kidney dysfunction was indicative of an accumulation of CD27-IgD- B cells and CD27-/low plasmablasts. Finally, we provide evidence that high levels of extracellular ATP and matrix metalloproteinase 8 can contribute to shedding of CD27 during human hantavirus infection. CONCLUSION Our findings demonstrate that thrombocytopenia and kidney dysfunction associate with distinctly different effects on the humoral immune system. Moreover, hantavirus-infected individuals have significantly elevated levels of extracellular ATP in circulation.
Collapse
Affiliation(s)
- Priscilla F Kerkman
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Andy Dernstedt
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Lalitha Tadala
- The Laboratory for Molecular Infection Medicine Sweden (MIMS)Umeå Centre for Microbial Research (UCMR)UmeaSweden
- Department of Molecular BiologyUmeå UniversityUmeaSweden
| | - Eva Mittler
- Department of Microbiology & ImmunologyAlbert Einstein College of MedicineBronxNYUSA
| | - Mirjam Dannborg
- The Laboratory for Molecular Infection Medicine Sweden (MIMS)Umeå Centre for Microbial Research (UCMR)UmeaSweden
- Department of Molecular BiologyUmeå UniversityUmeaSweden
| | - Christopher Sundling
- Department of MedicineKarolinska InstitutetSolnaSweden
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
| | - Kimia T Maleki
- Department of MedicineKarolinska InstitutetHuddingeSweden
| | | | - Anne Tuiskunen‐Bäck
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Julia Wigren Byström
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Pauline Ocaya
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Therese Thunberg
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Rohit K Jangra
- Department of Microbiology & ImmunologyAlbert Einstein College of MedicineBronxNYUSA
| | - Gleyder Román‐Sosa
- Structural Virology UnitVirology DepartmentInstitut PasteurCNRS UMR 3569ParisFrance
| | - Pablo Guardado‐Calvo
- Structural Virology UnitVirology DepartmentInstitut PasteurCNRS UMR 3569ParisFrance
| | - Felix A Rey
- Structural Virology UnitVirology DepartmentInstitut PasteurCNRS UMR 3569ParisFrance
| | | | - Kartik Chandran
- Department of Microbiology & ImmunologyAlbert Einstein College of MedicineBronxNYUSA
| | - Andrea Puhar
- The Laboratory for Molecular Infection Medicine Sweden (MIMS)Umeå Centre for Microbial Research (UCMR)UmeaSweden
- Department of Molecular BiologyUmeå UniversityUmeaSweden
| | - Clas Ahlm
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| | - Mattias NE Forsell
- Department of Clinical MicrobiologyUmeå Centre for Microbial Research (UCMR)UmeaSweden
| |
Collapse
|
24
|
Zhao L, Zhang J, Lei S, Ren H, Zou Y, Bai L, Zhang R, Xu H, Li L, Zhao Y, Cooper ME, Tong N, Zhang J, Liu F. Combining glomerular basement membrane and tubular basement membrane assessment improves the prediction of diabetic end-stage renal disease. J Diabetes 2021; 13:572-584. [PMID: 33352010 PMCID: PMC8246816 DOI: 10.1111/1753-0407.13150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/17/2020] [Accepted: 12/20/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To address the prognostic value of combining tubular basement membrane (TBM) and glomerular basement membrane (GBM) thickness in diabetic nephropathy (DN). METHODS This retrospective study enrolled 110 patients with type 2 diabetes and biopsy-proven DN from 2011 to 2018. The pathological findings were confirmed according to the Renal Pathology Society classifications. GBM and TBM thicknesses were determined using the Haas' direct measurement/arithmetic mean method and orthogonal intercept method, respectively. Cox proportional hazard models were used to investigate the hazard ratios (HRs) for the influence of combined GBM and TBM thickness for predicting end-stage renal disease (ESRD). RESULTS Patients were assigned to three groups according to the median GBM and TBM thickness: GBMlo TBMlo (GBM < 681 nm and TBM < 1200 nm), GBMhi TBMlo /GBMlo TBMhi (GBM ≥ 681 nm and TBM < 1200 nm, or GBM < 681 nm and TBM ≥ 1200 nm), and GBMhi TBMhi (GBM ≥ 681 nm and TBM ≥ 1200 nm). The GBMhi TBMlo /GBMlo TBMhi and GBMhi TBMhi groups displayed poorer renal function, a more severe glomerular classification and interstitial inflammation, and poorer renal survival rates than the GBMlo TBMlo group The GBMhi TBMlo /GBMlo TBMhi and GBMhi TBMhi groups had adjusted HRs of 1.49 (95% confidence interval [CI], 1.21-9.75) and 3.07 (95% CI, 2.87-12.78), respectively, compared with the GBMlo TBMlo group. CONCLUSIONS TBM thickness enhanced GBM thickness for renal prognosis in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Lijun Zhao
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
- Division of General PracticeWest China Hospital of Sichuan UniversityChengduChina
| | - Junlin Zhang
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Song Lei
- Division of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Honghong Ren
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Yutong Zou
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Lin Bai
- Histology and Imaging platform, Core Facility of West China HospitalChengduChina
| | - Rui Zhang
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Huan Xu
- Division of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Lin Li
- Division of PathologyWest China Hospital of Sichuan UniversityChengduChina
| | - Yuancheng Zhao
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| | - Mark E. Cooper
- Division of DiabetesCentral Clinical School, Monash UniversityMelbourneMelbourneAustralia
| | - Nanwei Tong
- Division of EndocrinologyWest China Hospital of Sichuan UniversityChengduChina
| | - Jie Zhang
- Histology and Imaging platform, Core Facility of West China HospitalChengduChina
| | - Fang Liu
- Division of NephrologyWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
25
|
Squires PE, Price GW, Mouritzen U, Potter JA, Williams BM, Hills CE. Danegaptide Prevents TGFβ1-Induced Damage in Human Proximal Tubule Epithelial Cells of the Kidney. Int J Mol Sci 2021; 22:2809. [PMID: 33802083 PMCID: PMC7999212 DOI: 10.3390/ijms22062809] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is a global health problem associated with a number of comorbidities. Recent evidence implicates increased hemichannel-mediated release of adenosine triphosphate (ATP) in the progression of tubulointerstitial fibrosis, the main underlying pathology of CKD. Here, we evaluate the effect of danegaptide on blocking hemichannel-mediated changes in the expression and function of proteins associated with disease progression in tubular epithelial kidney cells. Primary human proximal tubule epithelial cells (hPTECs) were treated with the beta1 isoform of the pro-fibrotic cytokine transforming growth factor (TGFβ1) ± danegaptide. qRT-PCR and immunoblotting confirmed mRNA and protein expression, whilst a cytokine antibody array assessed the expression/secretion of proinflammatory and profibrotic cytokines. Carboxyfluorescein dye uptake and ATP biosensing measured hemichannel activity and ATP release, whilst transepithelial electrical resistance was used to assess paracellular permeability. Danegaptide negated carboxyfluorescein dye uptake and ATP release and protected against protein changes associated with tubular injury. Blocking Cx43-mediated ATP release was paralleled by partial restoration of the expression of cell cycle inhibitors, adherens and tight junction proteins and decreased paracellular permeability. Furthermore, danegaptide inhibited TGFβ1-induced changes in the expression and secretion of key adipokines, cytokines, chemokines, growth factors and interleukins. The data suggest that as a gap junction modulator and hemichannel blocker, danegaptide has potential in the future treatment of CKD.
Collapse
Affiliation(s)
- Paul E. Squires
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK; (P.E.S.); (G.W.P.); (J.A.P.); (B.M.W.)
| | - Gareth W. Price
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK; (P.E.S.); (G.W.P.); (J.A.P.); (B.M.W.)
| | - Ulrik Mouritzen
- Ciana Therapeutics, Ved Hegnet 2, 2960 Rungsted Kyst, Copenhagen, Denmark;
| | - Joe A. Potter
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK; (P.E.S.); (G.W.P.); (J.A.P.); (B.M.W.)
| | - Bethany M. Williams
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK; (P.E.S.); (G.W.P.); (J.A.P.); (B.M.W.)
| | - Claire E. Hills
- School of Life Sciences, Joseph Banks Laboratories, University of Lincoln, Lincoln LN6 7DL, UK; (P.E.S.); (G.W.P.); (J.A.P.); (B.M.W.)
| |
Collapse
|
26
|
Potter JA, Price GW, Cliff CL, Williams BM, Hills CE, Squires PE. Carboxyfluorescein Dye Uptake to Measure Connexin-mediated Hemichannel Activity in Cultured Cells. Bio Protoc 2021; 11:e3901. [PMID: 33732788 DOI: 10.21769/bioprotoc.3901] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Connexins are membrane bound proteins that facilitate direct and local paracrine mediated cell-to-cell communication through their ability to oligomerise into hexameric hemichannels. When neighbouring channels align, they form gap-junctions that provide a direct route for information transfer between cells. In contrast to intact gap junctions, which typically open under physiological conditions, undocked hemichannels have a low open probability and mainly open in response to injury. Hemichannels permit the release of small molecules and ions (approximately 1kDa) into the local intercellular environment, and excessive expression/activity has been linked to a number of disease conditions. Carboxyfluorescein dye uptake measures functional expression of hemichannels, where increased hemichannel activity/function reflects increased loading. The technique relies on the uptake of a membrane-impermeable fluorescent tracer through open hemichannels, and can be used to compare channel activity between cell monolayers cultured under different conditions, e.g. control versus disease. Other techniques, such as biotinylation and electrophysiology can measure cell surface expression and hemichannel open probability respectively, however, carboxyfluorescein uptake provides a simple, rapid and cost-effective method to determine hemichannel activity in vitro in multiple cell types. Graphic abstract: Using dye uptake to measure hemichannel activity.
Collapse
Affiliation(s)
- Joe A Potter
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Gareth W Price
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Chelsy L Cliff
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Bethany M Williams
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Claire E Hills
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Paul E Squires
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
27
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
28
|
Liu Z, Lu J, Zhang D, Niu L, Shi B. Decreased serum C1Q/TNF-related protein 4 concentrations are associated with type 2 diabetes mellitus. Ther Adv Endocrinol Metab 2021; 12:20420188211059884. [PMID: 34868546 PMCID: PMC8637797 DOI: 10.1177/20420188211059884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/25/2021] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE To detect serum C1Q/TNF-related protein 4 (CTRP4) concentrations in patients with newly diagnosed type 2 diabetes mellitus (T2DM) and evaluate the correlation between CTRP4 and other variables in T2DM. METHOD Sixty-five patients with newly diagnosed T2DM and eighty-nine healthy volunteers were enrolled in this study. Anthropometric and biochemical data of the study participants was collected, and serum CTRP4 concentrations were detected by enzyme-linked immunosorbent assay (ELISA) kit. The correlation between serum CTRP4 and other indexes was analyzed by Spearman correlation analysis. Trend χ2 test and binary multivariate stepwise logistic regression were performed to assess the correlation between CTRP4 and the risk of T2DM. RESULTS Serum CTRP4 concentrations in the T2DM group were significantly lower than those in the control group (P < .01). Spearman correlation analysis showed that CTRP4 concentrations were negatively correlated with BMI, hs-CRP, HOMA-IR, FBG and TG (r = - 0.430, - 0.453, - 0.371, - 0.361, - 0.506, P < .05), and positively correlated with HDL-c (r = 0.303, P < .05). Trend χ2 test indicated that with the increase of CTRP4 levels in the population, the risk of T2DM presented a general downward trend (P < .01). Binary multivariate stepwise logistic regression suggested that serum CTRP4 was an independent impact factor for T2DM and high serum CTRP4 levels were related to the decreased risk of T2DM (P < .05). CONCLUSIONS Serum CTRP4 concentrations decrease in patients with newly diagnosed T2DM. Serum CTRP4 levels are negatively associated with the risk of T2DM.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Endocrinology, The First
Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinhua Lu
- Department of Geriatrics, The First Affiliated
Hospital of Soochow University, Suzhou, China
| | - Daiyi Zhang
- Medical Center, The First Affiliated Hospital
of Soochow University, Suzhou, China
| | | | - Bimin Shi
- Department of Endocrinology, The First
Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
29
|
Monaghan MLT, Bailey MA, Unwin RJ. Purinergic signalling in the kidney: In physiology and disease. Biochem Pharmacol 2020; 187:114389. [PMID: 33359067 DOI: 10.1016/j.bcp.2020.114389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
Historically, the control of renal vascular and tubular function has, for the most part, concentrated on neural and endocrine regulation. However, in addition to these extrinsic factors, it is now appreciated that several complex humoral control systems exist within the kidney that can act in an autocrine and/or paracrine fashion. These paracrine systems complement neuroendocrine regulation by dynamically fine-tuning renal vascular and tubular function to buffer rapid changes in nephron perfusion and flow rate of tubular fluid. One of the most pervasive is the extracellular nucleotide/P2 receptor system, which is central to many of the intrinsic regulatory feedback loops within the kidney such as renal haemodynamic autoregulation and tubuloglomerular feedback (TGF). Although physiological actions of extracellular adenine nucleotides were reported almost 100 years ago, the conceptual framework for purinergic regulation of renal function owes much to the work of Geoffrey Burnstock. In this review, we reflect on our >20-year collaboration with Professor Burnstock and highlight the research that is still unlocking the potential of the renal purinergic system to understand and treat kidney disease.
Collapse
Affiliation(s)
- Marie-Louise T Monaghan
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom
| | - Matthew A Bailey
- British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, United Kingdom
| | - Robert J Unwin
- The Department of Renal Medicine, University College London, United Kingdom.
| |
Collapse
|
30
|
Hering L, Rahman M, Potthoff SA, Rump LC, Stegbauer J. Role of α2-Adrenoceptors in Hypertension: Focus on Renal Sympathetic Neurotransmitter Release, Inflammation, and Sodium Homeostasis. Front Physiol 2020; 11:566871. [PMID: 33240096 PMCID: PMC7680782 DOI: 10.3389/fphys.2020.566871] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
The kidney is extensively innervated by sympathetic nerves playing an important role in the regulation of blood pressure homeostasis. Sympathetic nerve activity is ultimately controlled by the central nervous system (CNS). Norepinephrine, the main sympathetic neurotransmitter, is released at prejunctional neuroeffector junctions in the kidney and modulates renin release, renal vascular resistance, sodium and water handling, and immune cell response. Under physiological conditions, renal sympathetic nerve activity (RSNA) is modulated by peripheral mechanisms such as the renorenal reflex, a complex interaction between efferent sympathetic nerves, central mechanism, and afferent sensory nerves. RSNA is increased in hypertension and, therefore, critical for the perpetuation of hypertension and the development of hypertensive kidney disease. Renal sympathetic neurotransmission is not only regulated by RSNA but also by prejunctional α2-adrenoceptors. Prejunctional α2-adrenoceptors serve as autoreceptors which, when activated by norepinephrine, inhibit the subsequent release of norepinephrine induced by a sympathetic nerve impulse. Deletion of α2-adrenoceptors aggravates hypertension ultimately by modulating renal pressor response and sodium handling. α2-adrenoceptors are also expressed in the vasculature, renal tubules, and immune cells and exert thereby effects related to vascular tone, sodium excretion, and inflammation. In the present review, we highlight the role of α2-adrenoceptors on renal sympathetic neurotransmission and its impact on hypertension. Moreover, we focus on physiological and pathophysiological functions mediated by non-adrenergic α2-adrenoceptors. In detail, we discuss the effects of sympathetic norepinephrine release and α2-adrenoceptor activation on renal sodium transporters, on renal vascular tone, and on immune cells in the context of hypertension and kidney disease.
Collapse
Affiliation(s)
- Lydia Hering
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Masudur Rahman
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sebastian A Potthoff
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Lars C Rump
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
31
|
AZD-9056, a P2X7 receptor inhibitor, suppresses ATP-induced melanogenesis. J Dermatol Sci 2020; 100:227-229. [PMID: 33051087 DOI: 10.1016/j.jdermsci.2020.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/16/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022]
|
32
|
Dusabimana T, Kim SR, Park EJ, Je J, Jeong K, Yun SP, Kim HJ, Kim H, Park SW. P2Y2R contributes to the development of diabetic nephropathy by inhibiting autophagy response. Mol Metab 2020; 42:101089. [PMID: 32987187 PMCID: PMC7568185 DOI: 10.1016/j.molmet.2020.101089] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/14/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Diabetic nephropathy (DN) is one of the most common complications of diabetes and a critical risk factor for developing end-stage renal disease. Activation of purinergic receptors, including P2Y2R has been associated with the pathogenesis of renal diseases, such as polycystic kidney and glomerulonephritis. However, the role of P2Y2R and its precise mechanisms in DN remain unknown. We hypothesised that P2Y2R deficiency may play a protective role in DN by modulating the autophagy signalling pathway. METHODS We used a mouse model of DN by combining a treatment of high-fat diet and streptozotocin after unilateral nephrectomy in wild-type or P2Y2R knockout mice. We measured renal functional parameter in plasma, examined renal histology, and analysed expression of autophagy regulatory proteins. RESULTS Hyperglycaemia and ATP release were induced in wild type-DN mice and positively correlated with renal dysfunction. Conversely, P2Y2R knockout markedly attenuates albuminuria, podocyte loss, development of glomerulopathy, renal tubular injury, apoptosis and interstitial fibrosis induced by DN. These protective effects were associated with inhibition of AKT-mediated FOXO3a (forkhead box O3a) phosphorylation and induction of FOXO3a-induced autophagy gene transcription. Furthermore, inhibitory phosphorylation of ULK-1 was decreased, and the downstream Beclin-1 autophagy signalling was activated in P2Y2R deficiency. Increased SIRT-1 (sirtuin-1) and FOXO3a expression in P2Y2R deficiency also enhanced autophagy response, thereby ameliorating renal dysfunction in DN. CONCLUSIONS P2Y2R contributes to the pathogenesis of DN by impairing autophagy and serves as a therapeutic target for treating DN.
Collapse
Affiliation(s)
- Theodomir Dusabimana
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea; Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - So Ra Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
| | - Eun Jung Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
| | - Jihyun Je
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
| | - Kyuho Jeong
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea; Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea; Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Hwajin Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea.
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Republic of Korea; Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea.
| |
Collapse
|
33
|
DeLalio LJ, Masati E, Mendu S, Ruddiman CA, Yang Y, Johnstone SR, Milstein JA, Keller TCS, Weaver RB, Guagliardo NA, Best AK, Ravichandran KS, Bayliss DA, Sequeira-Lopez MLS, Sonkusare SN, Shu XH, Desai B, Barrett PQ, Le TH, Gomez RA, Isakson BE. Pannexin 1 channels in renin-expressing cells influence renin secretion and blood pressure homeostasis. Kidney Int 2020; 98:630-644. [PMID: 32446934 PMCID: PMC7483468 DOI: 10.1016/j.kint.2020.04.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Kidney function and blood pressure homeostasis are regulated by purinergic signaling mechanisms. These autocrine/paracrine signaling pathways are initiated by the release of cellular ATP, which influences kidney hemodynamics and steady-state renin secretion from juxtaglomerular cells. However, the mechanism responsible for ATP release that supports tonic inputs to juxtaglomerular cells and regulates renin secretion remains unclear. Pannexin 1 (Panx1) channels localize to both afferent arterioles and juxtaglomerular cells and provide a transmembrane conduit for ATP release and ion permeability in the kidney and the vasculature. We hypothesized that Panx1 channels in renin-expressing cells regulate renin secretion in vivo. Using a renin cell-specific Panx1 knockout model, we found that male Panx1 deficient mice exhibiting a heightened activation of the renin-angiotensin-aldosterone system have markedly increased plasma renin and aldosterone concentrations, and elevated mean arterial pressure with altered peripheral hemodynamics. Following ovariectomy, female mice mirrored the male phenotype. Furthermore, constitutive Panx1 channel activity was observed in As4.1 renin-secreting cells, whereby Panx1 knockdown reduced extracellular ATP accumulation, lowered basal intracellular calcium concentrations and recapitulated a hyper-secretory renin phenotype. Moreover, in response to stress stimuli that lower blood pressure, Panx1-deficient mice exhibited aberrant "renin recruitment" as evidenced by reactivation of renin expression in pre-glomerular arteriolar smooth muscle cells. Thus, renin-cell Panx1 channels suppress renin secretion and influence adaptive renin responses when blood pressure homeostasis is threatened.
Collapse
Affiliation(s)
- Leon J DeLalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Ester Masati
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Suresh Mendu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Yang Yang
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Scott R Johnstone
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jenna A Milstein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Rachel B Weaver
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nick A Guagliardo
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Angela K Best
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Maria Luisa S Sequeira-Lopez
- Pediatric Center of Excellence in Nephrology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Swapnil N Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Xiaohong H Shu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Bimal Desai
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Paula Q Barrett
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Thu H Le
- Department of Medicine, Division of Nephrology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - R Ariel Gomez
- Pediatric Center of Excellence in Nephrology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| |
Collapse
|
34
|
Sabapathy V, Venkatadri R, Dogan M, Sharma R. The Yin and Yang of Alarmins in Regulation of Acute Kidney Injury. Front Med (Lausanne) 2020; 7:441. [PMID: 32974364 PMCID: PMC7472534 DOI: 10.3389/fmed.2020.00441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is a major clinical burden affecting 20 to 50% of hospitalized and intensive care patients. Irrespective of the initiating factors, the immune system plays a major role in amplifying the disease pathogenesis with certain immune cells contributing to renal damage, whereas others offer protection and facilitate recovery. Alarmins are small molecules and proteins that include granulysins, high-mobility group box 1 protein, interleukin (IL)-1α, IL-16, IL-33, heat shock proteins, the Ca++ binding S100 proteins, adenosine triphosphate, and uric acid. Alarmins are mostly intracellular molecules, and their release to the extracellular milieu signals cellular stress or damage, generally leading to the recruitment of the cells of the immune system. Early studies indicated a pro-inflammatory role for the alarmins by contributing to immune-system dysregulation and worsening of AKI. However, recent developments demonstrate anti-inflammatory mechanisms of certain alarmins or alarmin-sensing receptors, which may participate in the prevention, resolution, and repair of AKI. This dual function of alarmins is intriguing and has confounded the role of alarmins in AKI. In this study, we review the contribution of various alarmins to the pathogenesis of AKI in experimental and clinical studies. We also analyze the approaches for the therapeutic utilization of alarmins for AKI.
Collapse
Affiliation(s)
| | | | | | - Rahul Sharma
- Division of Nephrology, Department of Medicine, Center for Immunity, Inflammation, and Regenerative Medicine (CIIR), University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
35
|
Conversion of extracellular ATP into adenosine: a master switch in renal health and disease. Nat Rev Nephrol 2020; 16:509-524. [PMID: 32641760 DOI: 10.1038/s41581-020-0304-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
ATP and its ultimate degradation product adenosine are potent extracellular signalling molecules that elicit a variety of pathophysiological functions in the kidney through the activation of P2 and P1 purinergic receptors, respectively. Extracellular purines can modulate immune responses, balancing inflammatory processes and immunosuppression; indeed, alterations in extracellular nucleotide and adenosine signalling determine outcomes of inflammation and healing processes. The functional activities of ectonucleotidases such as CD39 and CD73, which hydrolyse pro-inflammatory ATP to generate immunosuppressive adenosine, are therefore pivotal in acute inflammation. Protracted inflammation may result in aberrant adenosinergic signalling, which serves to sustain inflammasome activation and worsen fibrotic reactions. Alterations in the expression of ectonucleotidases on various immune cells, such as regulatory T cells and macrophages, as well as components of the renal vasculature, control purinergic receptor-mediated effects on target tissues within the kidney. The role of CD39 as a rheostat that can have an impact on purinergic signalling in both acute and chronic inflammation is increasingly supported by the literature, as detailed in this Review. Better understanding of these purinergic processes and development of novel drugs targeting these pathways could lead to effective therapies for the management of acute and chronic kidney disease.
Collapse
|
36
|
Price GW, Chadjichristos CE, Kavvadas P, Tang SCW, Yiu WH, Green CR, Potter JA, Siamantouras E, Squires PE, Hills CE. Blocking Connexin-43 mediated hemichannel activity protects against early tubular injury in experimental chronic kidney disease. Cell Commun Signal 2020; 18:79. [PMID: 32450899 PMCID: PMC7249671 DOI: 10.1186/s12964-020-00558-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Tubulointerstitial fibrosis represents the key underlying pathology of Chronic Kidney Disease (CKD), yet treatment options remain limited. In this study, we investigated the role of connexin43 (Cx43) hemichannel-mediated adenosine triphosphate (ATP) release in purinergic-mediated disassembly of adherens and tight junction complexes in early tubular injury. METHODS Human primary proximal tubule epithelial cells (hPTECs) and clonal tubular epithelial cells (HK2) were treated with Transforming Growth Factor Beta1 (TGF-β1) ± apyrase, or ATPγS for 48 h. For inhibitor studies, cells were co-incubated with Cx43 mimetic Peptide 5, or purinergic receptor antagonists Suramin, A438079 or A804598. Immunoblotting, single-cell force spectroscopy and trans-epithelial electrical resistance assessed protein expression, cell-cell adhesion and paracellular permeability. Carboxyfluorescein uptake and biosensing measured hemichannel activity and real-time ATP release, whilst a heterozygous Cx43+/- mouse model with unilateral ureteral obstruction (UUO) assessed the role of Cx43 in vivo. RESULTS Immunohistochemistry of biopsy material from patients with diabetic nephropathy confirmed increased expression of purinergic receptor P2X7. TGF-β1 increased Cx43 mediated hemichannel activity and ATP release in hPTECs and HK2 cells. The cytokine reduced maximum unbinding forces and reduced cell-cell adhesion, which translated to increased paracellular permeability. Changes were reversed when cells were co-incubated with either Peptide 5 or P2-purinoceptor inhibitors. Cx43+/- mice did not exhibit protein changes associated with early tubular injury in a UUO model of fibrosis. CONCLUSION Data suggest that Cx43 mediated ATP release represents an initial trigger in early tubular injury via its actions on the adherens and tight junction complex. Since Cx43 is highly expressed in nephropathy, it represents a novel target for intervention of tubulointerstitial fibrosis in CKD. Video Abstract In proximal tubular epithelial cells (PTECs), tight junction proteins, including zona occuludens-1 (ZO-1), contribute to epithelial integrity, whilst the adherens junction protein epithelial (E)-cadherin (ECAD) maintains cell-cell coupling, facilitating connexin 43 (Cx43) gap junction-mediated intercellular communication (GJIC) and the direct transfer of small molecules and ions between cells. In disease, such as diabetic nephropathy, the pro-fibrotic cytokine transforming growth factor beta1 (TGF-β1) binds to its receptor and recruits SMAD2/3 signalling ahead of changes in gene transcription and up-regulation of Cx43-mediated hemichannels (HC). Uncoupled hemichannels permit the release of adenosine triphosphate (ATP) in to the extracellular space (↑[ATP]e), where ATP binds to the P2X7 purinoreceptor and activates the nucleotide-binding domain and leucine-rich repeat containing (NLR) protein-3 (NLRP3) inflammasome. Inflammation results in epithelial-to-mesenchymal transition (EMT), fibrosis and tubular injury. A major consequence is further loss of ECAD and reduced stickiness between cells, which can be functionally measured as a decrease in the maximum unbinding force needed to uncouple two adherent cells (Fmax). Loss of ECAD feeds forward to further lessen cell-cell coupling exacerbating the switch from GJIC to HC-mediated release of ATP. Reduction in ZO-1 impedes tight junction effectiveness and decreases trans-epithelial resistance (↓TER), resulting in increased paracellular permeability.
Collapse
Affiliation(s)
- Gareth W. Price
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Christos E. Chadjichristos
- National Institutes for Health and Medical Research Unite Mixte de Recherche S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
| | - Panagiotis Kavvadas
- National Institutes for Health and Medical Research Unite Mixte de Recherche S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
| | - Sydney C. W. Tang
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wai Han Yiu
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Colin R. Green
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Joe A. Potter
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Eleftherios Siamantouras
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Paul E. Squires
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Claire E. Hills
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| |
Collapse
|
37
|
Serralha RS, Rodrigues IF, Bertolini A, Lima DY, Nascimento M, Mouro MG, Punaro GR, Visoná I, Rodrigues AM, Higa EMS. Esculin reduces P2X7 and reverses mitochondrial dysfunction in the renal cortex of diabetic rats. Life Sci 2020; 254:117787. [PMID: 32417372 DOI: 10.1016/j.lfs.2020.117787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
AIMS To evaluate the effects of esculin treatment on P2X7 receptor and mitochondrial dysfunction in the renal cortex of diabetic rats. MAIN METHODS Male Wistar rats, 7 weeks old, were unilaterally nephrectomized. Part of these animals were induced to diabetes using streptozotocin (60 mg/kg). Diabetes was confirmed 48 h after induction, with blood glucose levels ≥200 mg/dL. Part of control and diabetic animals were selected to receive daily doses of esculin (50 mg/kg), during 8 weeks. The animals were placed in metabolic cages at the eighth week of protocol for 24 h urine collection and a small aliquot of blood was collected for biochemical analysis. After this procedure, the animals were euthanized and the remaining kidney was stored for histopathological analysis, Western blotting and mitochondrial high-resolution respirometry. KEY FINDINGS Although esculin did not change metabolic parameters, renal biochemical function, neither TBARS in DM rats, esculin reduced P2X7 levels in these animals and restored mitochondrial function via glycolysis substrates and β-oxidation. Besides, at the histological analysis, we observed that esculin reduced inflammatory infiltrates and collagen IV deposits as compared to diabetic group. SIGNIFICANCE Esculin attenuated the development of renal injuries caused by hyperglycemia, proinflammatory and oxidative mechanisms mediated by P2X7 receptor, as seen by histological findings and improved mitochondrial function in diabetic animals. This suggests that esculin could be used as an adjuvant therapy to prevent the diabetic nephropathy.
Collapse
Affiliation(s)
- R S Serralha
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil.
| | - I F Rodrigues
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - A Bertolini
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - D Y Lima
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - M Nascimento
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology, Universidade Federal de Sao Paulo, Brazil
| | - M G Mouro
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - G R Punaro
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - I Visoná
- Pathology Department, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - A M Rodrigues
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - E M S Higa
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Emergency Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| |
Collapse
|
38
|
Nascimento M, Punaro GR, Serralha RS, Lima DY, Mouro MG, Oliveira LCG, Casarini DE, Rodrigues AM, Higa EMS. Inhibition of the P2X 7 receptor improves renal function via renin-angiotensin system and nitric oxide on diabetic nephropathy in rats. Life Sci 2020; 251:117640. [PMID: 32259603 DOI: 10.1016/j.lfs.2020.117640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/27/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023]
Abstract
AIM To evaluate the effects of P2X7 receptor blockade on renin-angiotensin system (RAS) in rats with diabetic nephropathy (DN). MAIN METHODS Wistar rats were unilaterally nephrectomized and received streptozotocin for diabetes mellitus (DM) induction; control animals (CTL) received the drug vehicle. The animals were submitted to P2X7 receptor silencing, forming the group (DM + siRNA). The animals were placed in metabolic cages for data collection and evaluation of renal function; at the end of the protocol, the kidney was removed for analysis of P2X7, renin, angiotensin-converting enzyme (ACE), ACE2, angiotensin, thiobarbituric acid reactive substance levels (TBARS), nitric oxide (NO) and qualitative histological. KEY FINDINGS The metabolic profile was attenuated in DM + siRNA vs. DM and there was a significant improvement in creatinine, urea and proteinuria levels in the same group. Renin expression was significantly decreased in DM + siRNA vs. DM. ACE and ACE2 were significantly reduced in DM + siRNA vs. DM. TBARS levels were decreased and NO showed an increase in DM + siRNA vs. DM, both significant. All histological alterations were improved in DM + siRNA vs. DM. SIGNIFICANCE Data have shown that although silencing of the P2X7 receptor did not decrease fasting glucose, it promoted an improvement in the metabolic profile and a significant recovery of renal function, revealing a protective action by the inhibition of this receptor. This effect must have occurred due to the inhibition of RAS and the increase of NO, suggesting that the use of P2X7 receptors inhibitors could be used as adjuvant therapy against DN progression.
Collapse
Affiliation(s)
- M Nascimento
- Nephrology, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil
| | - G R Punaro
- Nephrology, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil.
| | - R S Serralha
- Translational Medicine, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil
| | - D Y Lima
- Nephrology, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil
| | - M G Mouro
- Translational Medicine, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil
| | | | - D E Casarini
- Nephrology, Universidade Federal de Sao Paulo, Brazil; Translational Medicine, Universidade Federal de Sao Paulo, Brazil
| | - A M Rodrigues
- Translational Medicine, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil
| | - E M S Higa
- Nephrology, Universidade Federal de Sao Paulo, Brazil; Translational Medicine, Universidade Federal de Sao Paulo, Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo, Brazil; Emergency Division, Universidade Federal de Sao Paulo, Brazil
| |
Collapse
|
39
|
Han SJ, Lovaszi M, Kim M, D’Agati V, Haskó G, Lee HT. P2X4 receptor exacerbates ischemic AKI and induces renal proximal tubular NLRP3 inflammasome signaling. FASEB J 2020; 34:5465-5482. [PMID: 32086866 PMCID: PMC7136150 DOI: 10.1096/fj.201903287r] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 12/24/2022]
Abstract
We tested the hypothesis that the P2X4 purinergic receptor (P2X4) exacerbates ischemic acute kidney injury (AKI) by promoting renal tubular inflammation after ischemia and reperfusion (IR). Supporting this, P2X4-deficient (KO) mice were protected against ischemic AKI with significantly attenuated renal tubular necrosis, inflammation, and apoptosis when compared to P2X4 wild-type (WT) mice subjected to renal IR. Furthermore, WT mice treated with P2X4 allosteric agonist ivermectin had exacerbated renal IR injury whereas P2X4 WT mice treated with a selective P2X4 antagonist (5-BDBD) were protected against ischemic AKI. Mechanistically, induction of kidney NLRP3 inflammasome signaling after renal IR was significantly attenuated in P2X4 KO mice. A P2 agonist ATPγS increased NLRP3 inflammasome signaling (NLRP3 and caspase 1 induction and IL-1β processing) in isolated renal proximal tubule cells from WT mice whereas these increases were absent in renal proximal tubules isolated from P2X4 KO mice. Moreover, 5-BDBD attenuated ATPγS induced NLRP3 inflammasome induction in renal proximal tubules from WT mice. Finally, P2X4 agonist ivermectin induced NLRP3 inflammasome and pro-inflammatory cytokines in cultured human proximal tubule cells. Taken together, our studies suggest that renal proximal tubular P2X4 activation exacerbates ischemic AKI and promotes NLRP3 inflammasome signaling.
Collapse
Affiliation(s)
- Sang Jun Han
- Department of Anesthesiology,College of Physicians and Surgeons of Columbia University, New York, NY
| | - Marianna Lovaszi
- Department of Anesthesiology,College of Physicians and Surgeons of Columbia University, New York, NY
| | - Mihwa Kim
- Department of Anesthesiology,College of Physicians and Surgeons of Columbia University, New York, NY
| | - Vivette D’Agati
- Department of Pathology, College of Physicians and Surgeons of Columbia University, New York, NY
| | - György Haskó
- Department of Anesthesiology,College of Physicians and Surgeons of Columbia University, New York, NY
| | - H. Thomas Lee
- Department of Anesthesiology,College of Physicians and Surgeons of Columbia University, New York, NY
| |
Collapse
|
40
|
Verschuren EHJ, Rigalli JP, Castenmiller C, Rohrbach MU, Bindels RJM, Peters DJM, Arjona FJ, Hoenderop JGJ. Pannexin-1 mediates fluid shear stress-sensitive purinergic signaling and cyst growth in polycystic kidney disease. FASEB J 2020; 34:6382-6398. [PMID: 32159259 DOI: 10.1096/fj.201902901r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/07/2020] [Accepted: 03/01/2020] [Indexed: 12/16/2022]
Abstract
Tubular ATP release is regulated by mechanosensation of fluid shear stress (FSS). Polycystin-1/polycystin-2 (PC1/PC2) functions as a mechanosensory complex in the kidney. Extracellular ATP is implicated in polycystic kidney disease (PKD), where PC1/PC2 is dysfunctional. This study aims to provide new insights into the ATP signaling under physiological conditions and PKD. Microfluidics, pharmacologic inhibition, and loss-of-function approaches were combined to assess the ATP release in mouse distal convoluted tubule 15 (mDCT15) cells. Kidney-specific Pkd1 knockout mice (iKsp-Pkd1-/- ) and zebrafish pkd2 morphants (pkd2-MO) were as models for PKD. FSS-exposed mDCT15 cells displayed increased ATP release. Pannexin-1 inhibition and knockout decreased FSS-modulated ATP release. In iKsp-Pkd1-/- mice, elevated renal pannexin-1 mRNA expression and urinary ATP were observed. In Pkd1-/- mDCT15 cells, elevated ATP release was observed upon the FSS mechanosensation. In these cells, increased pannexin-1 mRNA expression was observed. Importantly, pannexin-1 inhibition in pkd2-MO decreased the renal cyst growth. Our results demonstrate that pannexin-1 channels mediate ATP release into the tubular lumen due to pro-urinary flow. We present pannexin-1 as novel therapeutic target to prevent the renal cyst growth in PKD.
Collapse
Affiliation(s)
- Eric H J Verschuren
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Juan P Rigalli
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Charlotte Castenmiller
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Meike U Rohrbach
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Francisco J Arjona
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
41
|
Thongnak L, Pongchaidecha A, Lungkaphin A. Renal Lipid Metabolism and Lipotoxicity in Diabetes. Am J Med Sci 2019; 359:84-99. [PMID: 32039770 DOI: 10.1016/j.amjms.2019.11.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/13/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022]
Abstract
The pathogenesis of diabetic kidney disease is a complex process caused by both glucotoxicity and lipotoxicity due to lipid accumulation. In cases of diabetic animals, lipid deposition is found in both tubular and glomerular portions of the kidneys, which are the major sites of diabetic nephropathy lesions. The aim of this review was to provide insights into the mechanisms that lead to the development of renal lipid accumulation and the effects of renal lipotoxicity in the diabetic condition. An increased number of lipogenic genes and a decreased number of lipid oxidation genes are also detected in diabetic kidneys, both of which lead to lipid accumulation. The induction of oxidative stress, inflammation, fibrosis and apoptosis caused by lipid accumulation and lipid metabolites is called lipotoxicity. Renal lipotoxicity due to derangement in lipid metabolism may be a pathogenic mechanism leading to diabetic nephropathy and renal dysfunction.
Collapse
Affiliation(s)
- Laongdao Thongnak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anchalee Pongchaidecha
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Functional Food Research Center for Well-Being, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
42
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
43
|
Arkhipov SN, Pavlov TS. ATP release into ADPKD cysts via pannexin-1/P2X7 channels decreases ENaC activity. Biochem Biophys Res Commun 2019; 513:166-171. [PMID: 30952430 PMCID: PMC6475605 DOI: 10.1016/j.bbrc.2019.03.177] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/30/2022]
Abstract
Genetic predisposition is necessary for polycystic kidney disease (PKD) initiation, although there are other, incompletely identified downstream processes that are required for cyst growth. Their characterization may provide a unique opportunity for clinical interventions. One of the poorly studied phenomena in PKD is high ATP content in cysts. Unfortunately, neither origins of uncontrolled ATP release, nor consequences of abnormal purinergic signaling in relation to epithelial transport are well explored in the polycystic kidney. We tested the distribution of pannexin-1 (Panx1) and P2X7, two proteins potentially involved in ATP release, in the kidneys of the Pkd1RC/RC mice, a model of autosomal dominant PKD (ADPKD). Abundances of both proteins were abnormally increased in the cyst lining cells compared to non-dilated collecting ducts. To establish if pannexin-1 contributes to ATP release in the collecting ducts (CD), we measured luminal accumulation of ATP in M1 cell renal CD monolayers, and found that treatment with probenecid, a Panx1 blocker, prevents ATP release. Single channel patch clamp analysis of polarized M1 cells revealed that apical stimulation of P2X receptors with αβ-MeATP acutely reduces ENaC activity. We conclude that in ADPKD progression, an abnormal hyperexpression of both PANX1 and P2RX7 occurs in the cyst lining epithelial cells. High abundance of both proteins is not typical for non-dilated CDs but, when it happens in cysts, pannexin1/P2X7 cooperation elevates ATP release into the luminal space. High ATP level is a pathogenic factor facilitating cystogenesis by reducing ENaC-mediated reabsorption from the lumen.
Collapse
Affiliation(s)
- Sergey N Arkhipov
- Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, MI, USA
| | - Tengis S Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Health System, Detroit, MI, USA.
| |
Collapse
|
44
|
Role of the P2X7 receptor in the pathogenesis of type 2 diabetes and its microvascular complications. Curr Opin Pharmacol 2019; 47:75-81. [PMID: 30954933 DOI: 10.1016/j.coph.2019.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 12/16/2022]
Abstract
P2X7 receptors can be found in many tissues and organs, where they mediate several biological functions. This review summarizes the current knowledge about the role of this receptor in the pathogenesis of type 2 diabetes, in which the key clinical features are impaired insulin secretion and sensitivity, hyperglycemia, coexistence of other cardiovascular risk factors such as dyslipidemia and hypertension, and subclinical inflammation. The receptor modulates crucial pathways in the pancreatic islets (where it can either exert a trophic or detrimental action on β cells), and in the liver, in the adipose tissue and in the skeletal muscle, which are main sites of insulin resistance. P2X7 receptors also modulate a series of inflammatory responses that participate in the development of the microvascular complications of the disease. Potent and selective P2X7R blockers are available to be tested in Phase I/II clinical studies for the treatment of several chronic diseases, and it might be worthwhile to consider inclusion of patients with type 2 diabetes and its complications.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Purine nucleosides and nucleotides are released in the extracellular space following cell injury and act as paracrine mediators through a number of dedicated membrane receptors. In particular, extracellular ATP (eATP) significantly influences T-lymphocyte activation and phenotype. The purpose of this review is to discuss the role of ATP signaling in the T-cell-mediated alloimmune response. RECENT FINDINGS In various animal models of solid transplantation, the purinergic axis has been targeted to prevent acute rejection and to promote long-term graft tolerance. The inhibition of ATP-gated P2X receptors has been shown to halt lymphocyte activation, to downregulate both Th1 and Th17 responses and to promote T-regulatory (Treg) cell differentiation. Similarly, the inhibition of ATP signaling attenuated graft-versus-host disease in mice undergoing hematopoietic cell transplantation. Significantly, different drugs targeting the purinergic system have been recently approved for human use and may be a viable therapeutic option for transplant patients. SUMMARY The inhibition of eATP signaling downregulates the alloimmune response, expands Treg cells and promotes graft survival. This robust preclinical evidence and the recent advances in pharmacological research may lead to intriguing clinical applications.
Collapse
|
46
|
Rennert L, Zschiedrich S, Sandner L, Hartleben B, Cicko S, Ayata CK, Meyer C, Zech A, Zeiser R, Huber TB, Idzko M, Grahammer F. P2Y2R Signaling Is Involved in the Onset of Glomerulonephritis. Front Immunol 2018; 9:1589. [PMID: 30061884 PMCID: PMC6054981 DOI: 10.3389/fimmu.2018.01589] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022] Open
Abstract
Endogenously released adenosine-5’-triphosphate (ATP) is a key regulator of physiological function and inflammatory responses in the kidney. Genetic or pharmacological inhibition of purinergic receptors has been linked to attenuation of inflammatory disorders and hence constitutes promising new avenues for halting and reverting inflammatory renal diseases. However, the involvement of purinergic receptors in glomerulonephritis (GN) has only been incompletely mapped. Here, we demonstrate that induction of GN in an experimental antibody-mediated GN model results in a significant increase of urinary ATP-levels and an upregulation of P2Y2R expression in resident kidney cells as well as infiltrating leukocytes pointing toward a possible role of the ATP/P2Y2R-axis in glomerular disease initiation. In agreement, decreasing extracellular ATP-levels or inhibition of P2R during induction of antibody-mediated GN leads to a reduction in all cardinal features of GN such as proteinuria, glomerulosclerosis, and renal failure. The specific involvement of P2Y2R could be further substantiated by demonstrating the protective effect of the lack of P2Y2R in antibody-mediated GN. To systematically differentiate between the function of P2Y2R on resident renal cells versus infiltrating leukocytes, we performed bone marrow-chimera experiments revealing that P2Y2R on hematopoietic cells is the main driver of the ATP/P2Y2R-mediated disease progression in antibody-mediated GN. Thus, these data unravel an important pro-inflammatory role for P2Y2R in the pathogenesis of GN.
Collapse
Affiliation(s)
- Laura Rennert
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Zschiedrich
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Sandner
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Björn Hartleben
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sanja Cicko
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Cemil Korcan Ayata
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Charlotte Meyer
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Andreas Zech
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany
| | - Tobias B Huber
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,BIOSS Center for Biological Signalling Studies, Albert-Ludwigs-University, Freiburg, Germany
| | - Marco Idzko
- Department of Pneumology, University Medical Center Freiburg, Freiburg, Germany.,Division of Pulmonology, Department of Medicine II, Medical University Vienna, Vienna, Austria
| | - Florian Grahammer
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
47
|
Jankowski J, Perry HM, Medina CB, Huang L, Yao J, Bajwa A, Lorenz UM, Rosin DL, Ravichandran KS, Isakson BE, Okusa MD. Epithelial and Endothelial Pannexin1 Channels Mediate AKI. J Am Soc Nephrol 2018; 29:1887-1899. [PMID: 29866797 DOI: 10.1681/asn.2017121306] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/11/2018] [Indexed: 12/24/2022] Open
Abstract
Background Pannexin1 (Panx1), an ATP release channel, is present in most mammalian tissues, but the role of Panx1 in health and disease is not fully understood. Panx1 may serve to modulate AKI; ATP is a precursor to adenosine and may function to block inflammation, or ATP may act as a danger-associated molecular pattern and initiate inflammation.Methods We used pharmacologic and genetic approaches to evaluate the effect of Panx1 on kidney ischemia-reperfusion injury (IRI), a mouse model of AKI.Results Pharmacologic inhibition of gap junctions, including Panx1, by administration of carbenoxolone protected mice from IRI. Furthermore, global deletion of Panx1 preserved kidney function and morphology and diminished the expression of proinflammatory molecules after IRI. Analysis of bone marrow chimeric mice revealed that Panx1 expressed on parenchymal cells is necessary for ischemic injury, and both proximal tubule and vascular endothelial Panx1 tissue-specific knockout mice were protected from IRI. In vitro, Panx1-deficient proximal tubule cells released less and retained more ATP under hypoxic stress.Conclusions Panx1 is involved in regulating ATP release from hypoxic cells, and reducing this ATP release may protect kidneys from AKI.
Collapse
Affiliation(s)
- Jakub Jankowski
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Departments of
| | - Heather M Perry
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Departments of
| | - Christopher B Medina
- Pharmacology.,Microbiology, Immunology, and Cancer Biology, and.,Beirne Carter Center for Immunology.,Center for Cell Clearance, University of Virginia, Charlottesville, Virginia
| | - Liping Huang
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Departments of
| | - Junlan Yao
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Departments of
| | - Amandeep Bajwa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Departments of
| | - Ulrike M Lorenz
- Microbiology, Immunology, and Cancer Biology, and.,Beirne Carter Center for Immunology
| | | | - Kodi S Ravichandran
- Microbiology, Immunology, and Cancer Biology, and.,Beirne Carter Center for Immunology.,Center for Cell Clearance, University of Virginia, Charlottesville, Virginia
| | - Brant E Isakson
- Molecular Physiology and Biological Physics.,Robert M. Berne Cardiovascular Research Center, and
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, Departments of
| |
Collapse
|
48
|
Kishore BK, Robson SC, Dwyer KM. CD39-adenosinergic axis in renal pathophysiology and therapeutics. Purinergic Signal 2018; 14:109-120. [PMID: 29332180 PMCID: PMC5940625 DOI: 10.1007/s11302-017-9596-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022] Open
Abstract
Extracellular ATP interacts with purinergic type 2 (P2) receptors and elicits many crucial biological functions. Extracellular ATP is sequentially hydrolyzed to ADP and AMP by the actions of defined nucleotidases, such as CD39, and AMP is converted to adenosine, largely by CD73, an ecto-5'-nucleotidase. Extracellular adenosine interacts with P1 receptors and often opposes the effects of P2 receptor activation. The balance between extracellular ATP and adenosine in the blood and extracellular fluid is regulated chiefly by the activities of CD39 and CD73, which constitute the CD39-adenosinergic axis. In recent years, several studies have shown this axis to play critical roles in transport of water/sodium, tubuloglomerular feedback, renin secretion, ischemia reperfusion injury, renal fibrosis, hypertension, diabetic nephropathy, transplantation, inflammation, and macrophage transformation. Important developments include global and targeted gene knockout and/or transgenic mouse models of CD39 or CD73, biological or small molecule inhibitors, and soluble engineered ectonucleotidases to directly impact the CD39-adenosinergic axis. This review presents a comprehensive picture of the multiple roles of CD39-adenosinergic axis in renal physiology, pathophysiology, and therapeutics. Scientific advances and greater understanding of the role of this axis in the kidney, in both health and illness, will direct development of innovative therapies for renal diseases.
Collapse
Affiliation(s)
- Bellamkonda K. Kishore
- Departments of Internal Medicine and Nutrition & Integrative Physiology, and Center on Aging, University of Utah Health, Salt Lake City, UT USA
- Nephrology Research, VA Salt Lake City Health Care System, 500 Foothill Drive (151M), Salt Lake City, UT 84148 USA
| | - Simon C. Robson
- Division of Gastroenterology/Hepatology and Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215 USA
| | - Karen M. Dwyer
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3220 Australia
| |
Collapse
|
49
|
Li H, Han SJ, Kim M, Cho A, Choi Y, D'Agati V, Lee HT. Divergent roles for kidney proximal tubule and granulocyte PAD4 in ischemic AKI. Am J Physiol Renal Physiol 2018; 314:F809-F819. [PMID: 29357426 PMCID: PMC6031910 DOI: 10.1152/ajprenal.00569.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/05/2017] [Accepted: 12/05/2017] [Indexed: 02/06/2023] Open
Abstract
We previously demonstrated that kidney peptidylarginine deiminase-4 (PAD4) plays a critical role in ischemic acute kidney injury (AKI) in mice by promoting renal tubular inflammation and neutrophil infiltration (Ham A, Rabadi M, Kim M, Brown KM, Ma Z, D'Agati V, Lee HT. Am J Physiol Renal Physiol 307: F1052-F1062, 2014). Although the role of PAD4 in granulocytes including neutrophils is well known, we surprisingly observed profound renal proximal tubular PAD4 induction after renal ischemia-reperfusion (I/R) injury. Here we tested the hypothesis that renal proximal tubular PAD4 rather than myeloid-cell lineage PAD4 plays a critical role in exacerbating ischemic AKI by utilizing mice lacking PAD4 in renal proximal tubules (PAD4ff PEPCK Cre mice) or in granulocytes (PAD4ff LysM Cre mice). Mice lacking renal proximal tubular PAD4 were significantly protected against ischemic AKI compared with wild-type (PAD4ff) mice. Surprisingly, mice lacking PAD4 in myeloid cells were also protected against renal I/R injury although this protection was less compared with renal proximal tubular PAD4-deficient mice. Renal proximal tubular PAD4-deficient mice had profoundly reduced renal tubular apoptosis, whereas myeloid-cell PAD4-deficient mice showed markedly reduced renal neutrophil infiltration. Taken together, our studies suggest that both renal proximal tubular PAD4 as well as myeloid-cell lineage PAD4 play a critical role in exacerbating ischemic AKI. Renal proximal tubular PAD4 appears to contribute to ischemic AKI by promoting renal tubular apoptosis, whereas myeloid-cell PAD4 is preferentially involved in promoting neutrophil infiltration to the kidney and inflammation after renal I/R.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University , New York, New York
| | - Sang Jun Han
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University , New York, New York
| | - Mihwa Kim
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University , New York, New York
| | - Ahyeon Cho
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University , New York, New York
| | - Yewoon Choi
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University , New York, New York
| | - Vivette D'Agati
- Department of Pathology, College of Physicians and Surgeons of Columbia University , New York, New York
| | - H Thomas Lee
- Department of Anesthesiology, College of Physicians and Surgeons of Columbia University , New York, New York
| |
Collapse
|
50
|
Hansson SF, Zhou AX, Vachet P, Eriksson JW, Pereira MJ, Skrtic S, Jongsma Wallin H, Ericsson-Dahlstrand A, Karlsson D, Ahnmark A, Sörhede Winzell M, Magnone MC, Davidsson P. Secretagogin is increased in plasma from type 2 diabetes patients and potentially reflects stress and islet dysfunction. PLoS One 2018; 13:e0196601. [PMID: 29702679 PMCID: PMC5922551 DOI: 10.1371/journal.pone.0196601] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/16/2018] [Indexed: 01/09/2023] Open
Abstract
Beta cell dysfunction accompanies and drives the progression of type 2 diabetes mellitus (T2D), but there are few clinical biomarkers available to assess islet cell stress in humans. Secretagogin, a protein enriched in pancreatic islets, demonstrates protective effects on beta cell function in animals. However, its potential as a circulating biomarker released from human beta cells and islets has not been studied. In this study primary human islets, beta cells and plasma samples were used to explore secretion and expression of secretagogin in relation to the T2D pathology. Secretagogin was abundantly and specifically expressed and secreted from human islets. Furthermore, T2D patients had an elevated plasma level of secretagogin compared with matched healthy controls, which was confirmed in plasma of diabetic mice transplanted with human islets. Additionally, the plasma secretagogin level of the human cohort had an inverse correlation to clinical assessments of beta cell function. To explore the mechanism of secretagogin release in vitro, human beta cells (EndoC-βH1) were exposed to elevated glucose or cellular stress-inducing agents. Secretagogin was not released in parallel with glucose stimulated insulin release, but was markedly elevated in response to endoplasmic reticulum stressors and cytokines. These findings indicate that secretagogin is a potential novel biomarker, reflecting stress and islet cell dysfunction in T2D patients.
Collapse
Affiliation(s)
- Sara F. Hansson
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
- * E-mail:
| | - Alex-Xianghua Zhou
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Paulina Vachet
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Jan W. Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J. Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Stanko Skrtic
- Translational Medicine Unit CVRM, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Daniel Karlsson
- Bioscience, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Andrea Ahnmark
- Bioscience, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Maria Sörhede Winzell
- Bioscience, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Maria Chiara Magnone
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Pia Davidsson
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|