1
|
Jeong K, Je J, Dusabimana T, Karekezi J, Nugroho TA, Ndahigwa EN, Yun SP, Kim HJ, Kim H, Park SW. Activation of Purinergic P2Y2 Receptor Protects the Kidney Against Renal Ischemia and Reperfusion Injury in Mice. Int J Mol Sci 2024; 25:12563. [PMID: 39684275 DOI: 10.3390/ijms252312563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Extracellular ATP plays an important role in renal physiology as well as the pathogenesis of acute kidney injury induced by renal ischemia and reperfusion (IR). Expression of the purinergic P2Y2 receptor has been shown on inflammatory and structural cells of the kidney, and P2Y2R is preferably activated by ATP (or UTP). Here, we investigated the molecular mechanism of P2Y2R during IR injury by using P2Y2R knockout (KO) mice and a selective P2Y2R agonist, MRS2768. After renal IR, P2Y2R KO mice showed greater increases in plasma creatinine, tubular damage and neutrophil infiltration, and significant induction of proinflammatory cytokines and apoptotic markers than wild-type (WT) mice. In contrast, treatment with MRS2768 reduced plasma creatinine levels, tubular damage and inflammation, and renal apoptosis in mice subjected to renal IR. In cultured human proximal tubular HK-2 cells, MRS2768 upregulated P2Y2R mRNA levels and decreased TNF-α/cycloheximide-induced apoptosis and inflammation. Importantly, P2Y2R activation by MRS2768 increased the phosphorylation of protein kinase C (PKC), Src, and phosphatidylinositol 3-kinase (PI3K)/Akt. In addition, the inhibition of PI3K/Akt abolished the protective effects of MRS2768 against TNF-α/cycloheximide-induced apoptosis and inflammation in HK-2 cells. In conclusion, activation of P2Y2R protects against tubular apoptosis and inflammation during renal IR via the PKC/Src/Akt pathway, suggesting P2Y2R is a promising therapeutic target for acute kidney injury.
Collapse
Affiliation(s)
- Kyuho Jeong
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Biochemistry, College of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Jihyun Je
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Biochemistry, College of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Theodomir Dusabimana
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jacques Karekezi
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Tatang Aldi Nugroho
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Edvard Ntambara Ndahigwa
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Hwajin Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University Graduate School, Jinju 52727, Republic of Korea
| |
Collapse
|
2
|
Mounieb F, Abdel-Sattar SA, Balah A, Akool ES. P2 X 7 receptor is a critical regulator of extracellular ATP-induced profibrotic genes expression in rat kidney: implication of transforming growth factor-β/Smad signaling pathway. Purinergic Signal 2024; 20:421-430. [PMID: 37934321 PMCID: PMC11303607 DOI: 10.1007/s11302-023-09977-4] [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: 07/06/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
This study was designed to investigate the potential of extracellular adenosine 5'-triphosphate (ATP) via the P2 X 7 receptor to activate the renal fibrotic processes in rats. The present study demonstrates that administration of ATP rapidly activated transforming growth factor-β (TGF-β) to induce phosphorylation of Smad-2/3. Renal connective tissue growth factor (CTGF) and tissue inhibitor of metalloproteinase-1 (TIMP-1) mRNA and protein expressions were also increased following ATP administration. A decrease in TGF-β amount in serum as well as renal Smad-2/3 phosphorylation was noticed in animals pre-treated with the specific antagonist of P2 X 7 receptor, A 438,079. In addition, a significant reduction in mRNA and protein expression of CTGF and TIMP-1were also observed in the kidneys of those animals. Collectively, the current findings demonstrate that ATP has the ability to augment TGF-β-mediated Smad-2/3 phosphorylation and enhance the expression of the pro-fibrotic genes, CTGF and TIMP-1, an effect that is largely mediated via P2 X 7 receptor.
Collapse
Affiliation(s)
- Fatma Mounieb
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt
| | - Somaia A Abdel-Sattar
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt
| | - Amany Balah
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt.
| | - El-Sayed Akool
- Pharmacology and Toxicology Department, Faculty of Pharmacy (boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
3
|
Nespoux J, Monaghan MLT, Jones NK, Stewart K, Denby L, Czopek A, Mullins JJ, Menzies RI, Baker AH, Bailey MA. P2X7 receptor knockout does not alter renal function or prevent angiotensin II-induced kidney injury in F344 rats. Sci Rep 2024; 14:9573. [PMID: 38670993 PMCID: PMC11053004 DOI: 10.1038/s41598-024-59635-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
P2X7 receptors mediate immune and endothelial cell responses to extracellular ATP. Acute pharmacological blockade increases renal blood flow and filtration rate, suggesting that receptor activation promotes tonic vasoconstriction. P2X7 expression is increased in kidney disease and blockade/knockout is renoprotective. We generated a P2X7 knockout rat on F344 background, hypothesising enhanced renal blood flow and protection from angiotensin-II-induced renal injury. CRISPR/Cas9 introduced an early stop codon into exon 2 of P2rx7, abolishing P2X7 protein in kidney and reducing P2rx7 mRNA abundance by ~ 60% in bone-marrow derived macrophages. The M1 polarisation response to lipopolysaccharide was unaffected but P2X7 receptor knockout suppressed ATP-induced IL-1β release. In male knockout rats, acetylcholine-induced dilation of the renal artery ex vivo was diminished but not the response to nitroprusside. Renal function in male and female knockout rats was not different from wild-type. Finally, in male rats infused with angiotensin-II for 6 weeks, P2X7 knockout did not reduce albuminuria, tubular injury, renal macrophage accrual, and renal perivascular fibrosis. Contrary to our hypothesis, global P2X7 knockout had no impact on in vivo renal hemodynamics. Our study does not indicate a major role for P2X7 receptor activation in renal vascular injury.
Collapse
Affiliation(s)
- Josselin Nespoux
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Marie-Louise T Monaghan
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Natalie K Jones
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Kevin Stewart
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Laura Denby
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Alicja Czopek
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - John J Mullins
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Robert I Menzies
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Matthew A Bailey
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
4
|
Borselle D, Kaczorowski M, Gogolok B, Patkowski D, Polok M, Hałoń A, Apoznański W. Interstitial Cells of Cajal and P 2X 3 Receptors at Ureteropelvic Junction Obstruction and Their Relationship with Pain Response. J Clin Med 2024; 13:2109. [PMID: 38610874 PMCID: PMC11012584 DOI: 10.3390/jcm13072109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Introduction: Etiopathogenesis and the symptomatology of ureteropelvic junction obstruction (UPJO) in the pediatric population has not yet been definitely clarified, suggesting a multifactorial nature of the condition. The aim was to analyze the association between the number of Interstitial Cells of Cajal (ICCs), as well as P2X3 receptors in ureteropelvic junction (UPJ) and the pain response in pediatric patients with hydronephrosis. Methods: 50 patients with congenital hydronephrosis underwent open or laparoscopic pyeloplasty at one of two departments of pediatric surgery and urology in Poland. Patients were divided into two groups according to the pain symptoms before surgery. A total of 50 samples of UPJ were obtained intraoperatively and underwent histopathological and immunohistochemical (IHC) analysis. Quantitative assessment of ICCs was based on the number of CD117(+) cells of adequate morphology in the subepithelial layer and the muscularis propria. Expression of P2X3 receptors was evaluated as the intensity of IHC staining. Results: Patients with hydronephrosis and accompanying pain were on average 60 months older (77 vs. 17 months) than children with asymptomatic hydronephrosis (p = 0.017). Symptomatic children revealed higher numbers of ICCs in both the subepithelial layer and in the lamina muscularis propria. In particular, symptomatic patients aged 2 years or more exhibited significantly higher numbers of ICCs in the subepithelial layer. Significant differences in the distribution of ICCs between the subepithelial layer and the lamina muscularis propria were observed in both groups. Expression of P2X3 receptors was limited to the urothelium and the muscle layer and correlated between these structures. There was no relationship between pain response and the expression of P2X3 receptors. Conclusions: ICCs and P2X3 receptors may participate in the pathogenesis of UPJO and in the modulation of pain response to a dilatation of the pyelocaliceal system. Explanation of the role of ICCs and P2X3 receptors in propagation of ureteral peristaltic wave and the modulation of pain stimuli requires further studies.
Collapse
Affiliation(s)
- Dominika Borselle
- Department of Pediatric Surgery and Urology, Wroclaw Medical University and Hospital, Borowska 213, 50-556 Wroclaw, Poland
| | - Maciej Kaczorowski
- Department of Clinical and Experimental Pathology, Wroclaw Medical University and Hospital, Borowska 213, 50-556 Wroclaw, Poland
| | - Bartosz Gogolok
- Department of Pediatric Surgery and Urology, Wroclaw Medical University and Hospital, Borowska 213, 50-556 Wroclaw, Poland
| | - Dariusz Patkowski
- Department of Pediatric Surgery and Urology, Wroclaw Medical University and Hospital, Borowska 213, 50-556 Wroclaw, Poland
| | - Marcin Polok
- Department of Pediatric Surgery and Urology, Collegium Medicum-University of Zielona Gora, 65-417 Zielona Gora, Poland
| | - Agnieszka Hałoń
- Department of Clinical and Experimental Pathology, Wroclaw Medical University and Hospital, Borowska 213, 50-556 Wroclaw, Poland
| | - Wojciech Apoznański
- Department of Pediatric Surgery and Urology, Wroclaw Medical University and Hospital, Borowska 213, 50-556 Wroclaw, Poland
| |
Collapse
|
5
|
Akasaka H, Naora H. Revisiting the Use of Normal Saline for Peritoneal Washing in Ovarian Cancer. Int J Mol Sci 2023; 24:16449. [PMID: 38003636 PMCID: PMC10671679 DOI: 10.3390/ijms242216449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
The omentum is the predominant site of ovarian cancer metastasis, but it is difficult to remove the omentum in its entirety. There is a critical need for effective approaches that minimize the risk of colonization of preserved omental tissues by occult cancer cells. Normal saline (0.9% sodium chloride) is commonly used to wash the peritoneal cavity during ovarian cancer surgery. The omentum has a prodigious ability to absorb fluid in the peritoneal cavity, but the impact of normal saline on the omentum is poorly understood. In this review article, we discuss why normal saline is not a biocompatible solution, drawing insights from clinical investigations of normal saline in fluid resuscitation and from the cytopathologic evaluation of peritoneal washings. We integrate these insights with the unique biology of the omentum and omental metastasis, highlighting the importance of considering the absorptive ability of the omentum when administering agents into the peritoneal cavity. Furthermore, we describe insights from preclinical studies regarding the mechanisms by which normal saline might render the omentum conducive for colonization by cancer cells. Importantly, we discuss the possibility that the risk of colonization of preserved omental tissues might be minimized by using balanced crystalloid solutions for peritoneal washing.
Collapse
Affiliation(s)
| | - Honami Naora
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
6
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
7
|
Wakefield B, Penuela S. Potential Implications of Exercise Training on Pannexin Expression and Function. J Vasc Res 2022; 60:114-124. [PMID: 36366809 DOI: 10.1159/000527240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/14/2022] [Indexed: 09/05/2023] Open
Abstract
Pannexins (PANX1, 2, 3) are channel-forming glycoproteins that are expressed throughout the cardiovascular and musculoskeletal system. The canonical function of these proteins is to release nucleotides that act as purinergic signalling at the cell membrane or Ca2+ channels at the endoplasmic reticulum membrane. These two forms of signalling are essential for autocrine and paracrine signalling in health, and alterations in this signalling have been implicated in the pathogenesis of many diseases. Many musculoskeletal and cardiovascular diseases are largely the result of a lack of physical activity which causes altered gene expression. Considering exercise training has been shown to alter a wide array of gene expression in musculoskeletal tissues, understanding the interaction between exercise training, gene function and expression in relevant diseases is warranted. With regards to pannexins, multiple publications have shown that exercise training can influence pannexin expression and may influence the significance of its function in certain diseases. This review further discusses the potential interaction between exercise training and pannexin biology in relevant tissues and disease models. We propose that exercise training in relevant animal and human models will provide a more comprehensive understanding of the implications of pannexin biology in disease.
Collapse
Affiliation(s)
- Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
8
|
Jasmer KJ, Muñoz Forti K, Woods LT, Cha S, Weisman GA. Therapeutic potential for P2Y 2 receptor antagonism. Purinergic Signal 2022:10.1007/s11302-022-09900-3. [PMID: 36219327 DOI: 10.1007/s11302-022-09900-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 10/17/2022] Open
Abstract
G protein-coupled receptors are the target of more than 30% of all FDA-approved drug therapies. Though the purinergic P2 receptors have been an attractive target for therapeutic intervention with successes such as the P2Y12 receptor antagonist, clopidogrel, P2Y2 receptor (P2Y2R) antagonism remains relatively unexplored as a therapeutic strategy. Due to a lack of selective antagonists to modify P2Y2R activity, studies using primarily genetic manipulation have revealed roles for P2Y2R in a multitude of diseases. These include inflammatory and autoimmune diseases, fibrotic diseases, renal diseases, cancer, and pathogenic infections. With the advent of AR-C118925, a selective and potent P2Y2R antagonist that became commercially available only a few years ago, new opportunities exist to gain a more robust understanding of P2Y2R function and assess therapeutic effects of P2Y2R antagonism. This review discusses the characteristics of P2Y2R that make it unique among P2 receptors, namely its involvement in five distinct signaling pathways including canonical Gαq protein signaling. We also discuss the effects of other P2Y2R antagonists and the pivotal development of AR-C118925. The remainder of this review concerns the mounting evidence implicating P2Y2Rs in disease pathogenesis, focusing on those studies that have evaluated AR-C118925 in pre-clinical disease models.
Collapse
Affiliation(s)
- Kimberly J Jasmer
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Kevin Muñoz Forti
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Lucas T Woods
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Gary A Weisman
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA. .,Department of Biochemistry, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
9
|
Hill DB, Button B, Rubinstein M, Boucher RC. Physiology and pathophysiology of human airway mucus. Physiol Rev 2022; 102:1757-1836. [PMID: 35001665 PMCID: PMC9665957 DOI: 10.1152/physrev.00004.2021] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 01/27/2023] Open
Abstract
The mucus clearance system is the dominant mechanical host defense system of the human lung. Mucus is cleared from the lung by cilia and airflow, including both two-phase gas-liquid pumping and cough-dependent mechanisms, and mucus transport rates are heavily dependent on mucus concentration. Importantly, mucus transport rates are accurately predicted by the gel-on-brush model of the mucociliary apparatus from the relative osmotic moduli of the mucus and periciliary-glycocalyceal (PCL-G) layers. The fluid available to hydrate mucus is generated by transepithelial fluid transport. Feedback interactions between mucus concentrations and cilia beating, via purinergic signaling, coordinate Na+ absorptive vs Cl- secretory rates to maintain mucus hydration in health. In disease, mucus becomes hyperconcentrated (dehydrated). Multiple mechanisms derange the ion transport pathways that normally hydrate mucus in muco-obstructive lung diseases, e.g., cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), non-CF bronchiectasis (NCFB), and primary ciliary dyskinesia (PCD). A key step in muco-obstructive disease pathogenesis is the osmotic compression of the mucus layer onto the airway surface with the formation of adherent mucus plaques and plugs, particularly in distal airways. Mucus plaques create locally hypoxic conditions and produce airflow obstruction, inflammation, infection, and, ultimately, airway wall damage. Therapies to clear adherent mucus with hydrating and mucolytic agents are rational, and strategies to develop these agents are reviewed.
Collapse
Affiliation(s)
- David B Hill
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael Rubinstein
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Mechanical Engineering and Materials Science, Biomedical Engineering, Physics, and Chemistry, Duke University, Durham, North Carolina
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
10
|
Sikka P, Behl T, Chandel P, Sehgal A, Singh S, Makeen HA, Albratty M, Alhazmi HA, Meraya AM. Scrutinizing the Therapeutic Promise of Purinergic Receptors Targeting Depression. Neurotox Res 2022; 40:1570-1585. [PMID: 35930172 DOI: 10.1007/s12640-022-00550-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
Abstract
Antidepressant use has resulted in a variety of negative consequences, including permanent brain damage and erectile dysfunction. So, the purpose lies in developing something more productive with minimal side effects and consequently improved efficacy. A growing body of evidences indicated a remarkable purinergic signalling system, which helped in dealing with this complication. This has been found to be a powerful formula in dealing with psychiatric disorders. P1 (adenosine), P2X, and P2Y (ATP) are the receptors, involved in the pathology as well as exhibiting the therapeutic action by triggering the purinergic pathway. It was found that A2A and P2X7 receptors specifically were involved and recognized as possible targets for treating depression. Further, the development of biomarkers for the diagnosis of depression has also been attributed to accelerate the process. One such biomarker includes serum uric acid. Many clinical studies reveal the importance of antagonizing P2X7 and A2A receptors, for promising research in understanding the molecular premises of depression. However, further investigations are still needed to be done to open several unfolded mysteries for a better and safe upshot. The selective antagonists for A2A and P2X7 receptors may have antidepressant effects showing positive results, in agreement with non-clinical testing. In this review, efforts are being devoted to the targeted receptors in bringing out antidepressant effects with a possible link involving depression and defined purinergic signalling. Additionally, the overview of various receptors, including their functions and distribution, is being explored in a representative way along with the biomarkers involved.
Collapse
Affiliation(s)
- Priyanshi Sikka
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Parteek Chandel
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia.,Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Abdulkarim M Meraya
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
11
|
Carotti V, Rigalli JP, van Asbeck-van der Wijst J, G J Hoenderop J. Interplay between purinergic signalling and extracellular vesicles in health and disease. Biochem Pharmacol 2022; 203:115192. [PMID: 35905971 DOI: 10.1016/j.bcp.2022.115192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/25/2022]
Abstract
Purinergic signalling is a receptor-mediated process characterized by the binding of extracellular nucleotides and nucleosides to purinergic receptors, which results in the activation intracellular signalling pathways, and, ultimately, leads to changes in cell physiology. Purinergic signalling has been related to the regulation of important physiological processes (e.g., renal electrolyte reabsorption; platelet aggregation; immune response). In addition, it has been associated with pathophysiological situations such as cancer and inflammation. Extracellular vesicles (EVs) are nanoparticles released by all cells of the organism, which play a key role in cell-cell communication. In this regard, EVs can mediate effects on target cells located at distant locations. Within their cargo, EVs contain molecules with the potential to affect purinergic signalling at the target cells and tissues. Here, we review the studies addressing the regulation of purinergic signalling by EVs based on the cell type or tissue where the regulation takes place. In this regard, EVs are found to play a major role in modulating the extracellular ATP levels and, specially, adenosine. This has a clear impact on, for instance, the inflammatory and immune response against cancer cells. Furthermore, we discuss the data available on the regulation of EV secretion and its cargo by purinergic signalling. Here, a major role of the purinergic receptor P2X7 and again, an impact on processes such as inflammation, immune response and cancer pathogenesis has been established. Finally, we highlight uninvestigated aspects of these two regulatory networks and address their potential as therapeutic targets.
Collapse
Affiliation(s)
- Valentina Carotti
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, the Netherlands
| | - Juan P Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jenny van Asbeck-van der Wijst
- 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
|
12
|
Szrejder M, Rogacka D, Piwkowska A. Purinergic P2 receptors: Involvement and therapeutic implications in diabetes-related glomerular injury. Arch Biochem Biophys 2021; 714:109078. [PMID: 34742673 DOI: 10.1016/j.abb.2021.109078] [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: 09/08/2021] [Revised: 10/15/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023]
Abstract
The purinergic activation of P2 receptors initiates a powerful and rapid signaling cascade that contributes to the regulation of an array of physiological and pathophysiological processes in many organs, including the kidney. P2 receptors are broadly distributed in both epithelial and vascular renal cells. Disturbances of purinergic signaling can lead to impairments in renal function. A growing body of evidence indicates changes in P2 receptor expression and nucleotide metabolism in chronic renal injury and inflammatory diseases. Increasing attention has focused on purinergic P2X7 receptors, which are not normally expressed in healthy kidney tissue but are highly expressed at sites of tissue damage and inflammation. Under hyperglycemic conditions, several mechanisms that are linked to purinergic signaling and involve nucleotide release and degradation are disrupted, resulting in the accumulation of adenosine 5'-triphosphate in the bloodstream in diabetes. Dysfunction of the purinergic system might be associated with serious vascular complications in diabetes, including diabetic nephropathy. This review summarizes our current knowledge of the role of P2 receptors in diabetes-related glomerular injury and its implications for new therapeutics for diabetic nephropathy.
Collapse
Affiliation(s)
- Maria Szrejder
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland.
| | - Dorota Rogacka
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; Department of Molecular Biotechnology, University of Gdańsk, Faculty of Chemistry, Gdańsk, Poland
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Gdańsk, Poland; Department of Molecular Biotechnology, University of Gdańsk, Faculty of Chemistry, Gdańsk, Poland
| |
Collapse
|
13
|
Agarwal S, Sudhini YR, Polat OK, Reiser J, Altintas MM. Renal cell markers: lighthouses for managing renal diseases. Am J Physiol Renal Physiol 2021; 321:F715-F739. [PMID: 34632812 DOI: 10.1152/ajprenal.00182.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidneys, one of the vital organs in our body, are responsible for maintaining whole body homeostasis. The complexity of renal function (e.g., filtration, reabsorption, fluid and electrolyte regulation, and urine production) demands diversity not only at the level of cell types but also in their overall distribution and structural framework within the kidney. To gain an in depth molecular-level understanding of the renal system, it is imperative to discern the components of kidney and the types of cells residing in each of the subregions. Recent developments in labeling, tracing, and imaging techniques have enabled us to mark, monitor, and identify these cells in vivo with high efficiency in a minimally invasive manner. In this review, we summarize different cell types, specific markers that are uniquely associated with those cell types, and their distribution in the kidney, which altogether make kidneys so special and different. Cellular sorting based on the presence of certain proteins on the cell surface allowed for the assignment of multiple markers for each cell type. However, different studies using different techniques have found contradictions in cell type-specific markers. Thus, the term "cell marker" might be imprecise and suboptimal, leading to uncertainty when interpreting the data. Therefore, we strongly believe that there is an unmet need to define the best cell markers for a cell type. Although the compendium of renal-selective marker proteins presented in this review is a resource that may be useful to researchers, we acknowledge that the list may not be necessarily exhaustive.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | - Onur K Polat
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | |
Collapse
|
14
|
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] [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. Massé and colleagues identify enpp4 as a key regulator in the development of the kidney in Xenopus. The gene signalling pathways regulated by this ectonucleotidase are described and lipidic signalling regulatory mechanisms are explored.
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
|
15
|
Geis L, Boudriot FF, Wagner C. Connexin mRNA distribution in adult mouse kidneys. Pflugers Arch 2021; 473:1737-1747. [PMID: 34365513 PMCID: PMC8528753 DOI: 10.1007/s00424-021-02608-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 11/25/2022]
Abstract
Kidneys are thought to express eight different connexin isoforms (i.e., Cx 26, 30, 32, 37, 40, 43, 45, and 46), which form either hemichannels or gap junctions serving to intercellular communication and functional synchronization. Proper function of connexins has already been shown to be crucial for regulation of renal hemodynamics and renin secretion, and there is also growing evidence for connexins to play a role in pathologic conditions such as renal fibrosis or diabetic nephropathy. Therefore, exact intrarenal localization of the different connexin isoforms gains particular interest. Until now intrarenal expression of connexins has mainly been examined by immunohistochemistry, which in part generated conflicting results depending on antibodies and fixation protocols used. In this work, we used fluorescent RNAscope as an alternative technical approach to localize renal connexin mRNAs in healthy mouse kidneys. Addition of RNAscope probes for cell type specific mRNAs was used to assign connexin mRNA signals to specific cell types. We hereby found Cx26 mRNA strongly expressed in proximal tubules, Cx30 mRNA was selectively detected in the urothelium, and Cx32 mRNA was found in proximal tubules and to a lesser extent also in collecting ducts. Cx37 mRNA was mainly associated with vascular endothelium, Cx40 mRNA was largely found in glomerular mesangial and less in vascular endothelial cells, Cx43 mRNA was sparsely expressed by interstitial cells of all kidney zones, and Cx45 mRNA was predominantly found in smooth muscle cell layers of both blood vessels and ureter as well as in mesangial and interstitial (fibroblastic) cells. Cx46 mRNA could not be detected. In summary our results essentially confirm previous data on connexin expression in the renal vasculature and in glomeruli. In addition, they demonstrate strong connexin gene expression in proximal tubules, and they suggest significant connexin expression in resident tubulointerstitial cells.
Collapse
Affiliation(s)
- Lisa Geis
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany.
| | | | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
16
|
Jabalameli MR, Fitzpatrick FM, Colombo R, Howles SA, Leggatt G, Walker V, Wiberg A, Kunji ERS, Ennis S. Exome sequencing identifies a disease variant of the mitochondrial ATP-Mg/Pi carrier SLC25A25 in two families with kidney stones. Mol Genet Genomic Med 2021; 9:e1749. [PMID: 34346195 PMCID: PMC8683635 DOI: 10.1002/mgg3.1749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 05/13/2021] [Accepted: 07/01/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Calcium kidney stones are common and recurrences are often not preventable by available empiric remedies. Their etiology is multifactorial and polygenic, and an increasing number of genes are implicated. Their identification will enable improved management. METHODS DNA from three stone-formers in a Southampton family (UK) and two from an Italian family were analyzed independently by whole exome sequencing and selected variants were genotyped across all available members of both pedigrees. A disease variant of SLC25A25 (OMIM 608745), encoding the mitochondrial ATP-Mg/Pi carrier 3 (APC3) was identified, and analyzed structurally and functionally with respect to its calcium-regulated transport activity. RESULTS All five patients had a heterozygous dominant SLC25A25 variant (rs140777921; GRCh37.p13: chr 9 130868670 G>C; p.Gln349His; Reference Sequence NM_001006641.3). Non-stone formers also carried the variant indicating incomplete penetrance. Modeling suggests that the variant lacks a conserved polar interaction, which may cause structural instability. Calcium-regulated ATP transport was reduced to ~20% of the wild type, showing a large reduction in function. CONCLUSION The transporter is important in regulating mitochondrial ATP production. This rare variant may increase urine lithogenicity through impaired provision of ATP for solute transport processes in the kidney, and/or for purinergic signaling. Variants found in other genes may compound this abnormality.
Collapse
Affiliation(s)
- M Reza Jabalameli
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| | - Fiona M Fitzpatrick
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Roberto Colombo
- Faculty of Medicine 'Agostino Gemelli', Catholic University of the Sacred Heart, Rome, Italy.,Center for the Study of Rare Inherited Diseases, Niguarda Ca´Granda Metropolitan Hospital, Milan, Italy
| | - Sarah A Howles
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Gary Leggatt
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK.,Wessex Kidney Centre, Queen Alexandra Hospital, Portsmouth, UK
| | - Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton, Southampton, UK
| | - Akira Wiberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Edmund R S Kunji
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Sarah Ennis
- Department of Human Genetics and Genomic Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
17
|
Barros Lamus ER, Carotti V, de Vries CRS, Witsel F, Arntz OJ, van de Loo FAJ, Carvajal CA, Bindels RJM, Hoenderop JGJ, Rigalli JP. Extracellular vesicles regulate purinergic signaling and epithelial sodium channel expression in renal collecting duct cells. FASEB J 2021; 35:e21506. [PMID: 33811695 DOI: 10.1096/fj.202002559r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
Purinergic signaling regulates several renal physiological and pathophysiological processes. Extracellular vesicles (EVs) are nanoparticles released by most cell types, which, in non-renal tissues, modulate purinergic signaling. The aim of this study was to investigate the effect of EVs from renal proximal tubule (HK2) and collecting duct cells (HCD) on intra- and intersegment modulation of extracellular ATP levels, the underlying molecular mechanisms, and the impact on the expression of the alpha subunit of the epithelial sodium channel (αENaC). HK2 cells were exposed to HK2 EVs, while HCD cells were exposed to HK2 and HCD EVs. Extracellular ATP levels and αENaC expression were measured by chemiluminescence and qRT-PCR, respectively. ATPases in EV populations were identified by mass spectrometry. The effect of aldosterone was assessed using EVs from aldosterone-treated cells and urinary EVs (uEVs) from primary aldosteronism (PA) patients. HK2 EVs downregulated ectonucleoside-triphosphate-diphosphohydrolase-1 (ENTPD1) expression, increased extracellular ATP and downregulated αENaC expression in HCD cells. ENTPD1 downregulation could be attributed to increased miR-205-3p and miR-505 levels. Conversely, HCD EVs decreased extracellular ATP levels and upregulated αENaC expression in HCD cells, probably due to enrichment of 14-3-3 isoforms with ATPase activity. Pretreatment of donor cells with aldosterone or exposure to uEVs from PA patients enhanced the effects on extracellular ATP and αENaC expression. We demonstrated inter- and intrasegment modulation of renal purinergic signaling by EVs. Our findings postulate EVs as carriers of information along the renal tubules, whereby processes affecting EV release and/or cargo may impact on purinergically regulated processes.
Collapse
Affiliation(s)
- Eric R Barros Lamus
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina Carotti
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Christine R S de Vries
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Femke Witsel
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Onno J Arntz
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fons A J van de Loo
- Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cristian A Carvajal
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - René J M Bindels
- 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
| | - Juan P Rigalli
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Himmel NJ, Rogers RT, Redd SK, Wang Y, Blount MA. Purinergic signaling is enhanced in the absence of UT-A1 and UT-A3. Physiol Rep 2021; 9:e14636. [PMID: 33369887 PMCID: PMC7769175 DOI: 10.14814/phy2.14636] [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: 02/26/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 11/24/2022] Open
Abstract
ATP is an important paracrine regulator of renal tubular water and urea transport. The activity of P2Y2 , the predominant P2Y receptor of the medullary collecting duct, is mediated by ATP, and modulates urinary concentration. To investigate the role of purinergic signaling in the absence of urea transport in the collecting duct, we studied wild-type (WT) and UT-A1/A3 null (UT-A1/A3 KO) mice in metabolic cages to monitor urine output, and collected tissue samples for analysis. We confirmed that UT-A1/A3 KO mice are polyuric, and concurrently observed lower levels of urinary cAMP as compared to WT, despite elevated serum vasopressin (AVP) levels. Because P2Y2 inhibits AVP-stimulated transport by dampening cAMP synthesis, we suspected that, similar to other models of AVP-resistant polyuria, purinergic signaling is increased in UT-A1/A3 KO mice. In fact, we observed that both urinary ATP and purinergic-mediated prostanoid (PGE2 ) levels were elevated. Collectively, our data suggest that the reduction of medullary osmolality due to the lack of UT-A1 and UT-A3 induces an AVP-resistant polyuria that is possibly exacerbated by, or at least correlated with, enhanced purinergic signaling.
Collapse
Affiliation(s)
- Nathaniel J. Himmel
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Richard T. Rogers
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Sara K. Redd
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Yirong Wang
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
| | - Mitsi A. Blount
- Renal DivisionDepartment of MedicineEmory University School of MedicineAtlantaGAUSA
- Department of PhysiologyEmory University School of MedicineAtlantaGAUSA
| |
Collapse
|
19
|
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
|
20
|
Extracellular ATP modulates podocyte function through P2Y purinergic receptors and pleiotropic effects on AMPK and cAMP/PKA signaling pathways. Arch Biochem Biophys 2020; 695:108649. [PMID: 33122160 DOI: 10.1016/j.abb.2020.108649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/25/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Podocytes and their foot processes interlinked by slit diaphragms, constitute a continuous outermost layer of the glomerular capillary and seem to be crucial for maintaining the integrity of the glomerular filtration barrier. Purinergic signaling is involved in a wide range of physiological processes in the renal system, including regulating glomerular filtration. We evaluated the role of nucleotide receptors in cultured rat podocytes using non-selective P2 receptor agonists and agonists specific for the P2Y1, P2Y2, and P2Y4 receptors. The results showed that extracellular ATP evokes cAMP-dependent pathways through P2 receptors and influences remodeling of the podocyte cytoskeleton and podocyte permeability to albumin via coupling with RhoA signaling. Our findings highlight the relevance of the P2Y4 receptor in protein kinase A-mediated signal transduction to the actin cytoskeleton. We observed increased cAMP concentration and decreased RhoA activity after treatment with a P2Y4 agonist. Moreover, protein kinase A inhibitors reversed P2Y4-induced changes in RhoA activity and intracellular F-actin staining. P2Y4 stimulation resulted in enhanced AMPK phosphorylation and reduced reactive oxygen species generation. Our findings identify P2Y-PKA-RhoA signaling as the regulatory mechanism of the podocyte contractile apparatus and glomerular filtration. We describe a protection mechanism for the glomerular barrier linked to reduced oxidative stress and reestablished energy balance.
Collapse
|
21
|
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: 27] [Impact Index Per Article: 5.4] [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
|
22
|
Gyimesi G, Pujol-Giménez J, Kanai Y, Hediger MA. Sodium-coupled glucose transport, the SLC5 family, and therapeutically relevant inhibitors: from molecular discovery to clinical application. Pflugers Arch 2020; 472:1177-1206. [PMID: 32767111 PMCID: PMC7462921 DOI: 10.1007/s00424-020-02433-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Sodium glucose transporters (SGLTs) belong to the mammalian solute carrier family SLC5. This family includes 12 different members in human that mediate the transport of sugars, vitamins, amino acids, or smaller organic ions such as choline. The SLC5 family belongs to the sodium symporter family (SSS), which encompasses transporters from all kingdoms of life. It furthermore shares similarity to the structural fold of the APC (amino acid-polyamine-organocation) transporter family. Three decades after the first molecular identification of the intestinal Na+-glucose cotransporter SGLT1 by expression cloning, many new discoveries have evolved, from mechanistic analysis to molecular genetics, structural biology, drug discovery, and clinical applications. All of these advances have greatly influenced physiology and medicine. While SGLT1 is essential for fast absorption of glucose and galactose in the intestine, the expression of SGLT2 is largely confined to the early part of the kidney proximal tubules, where it reabsorbs the bulk part of filtered glucose. SGLT2 has been successfully exploited by the pharmaceutical industry to develop effective new drugs for the treatment of diabetic patients. These SGLT2 inhibitors, termed gliflozins, also exhibit favorable nephroprotective effects and likely also cardioprotective effects. In addition, given the recent finding that SGLT2 is also expressed in tumors of pancreas and prostate and in glioblastoma, this opens the door to potential new therapeutic strategies for cancer treatment by specifically targeting SGLT2. Likewise, further discoveries related to the functional association of other SGLTs of the SLC5 family to human pathologies will open the door to potential new therapeutic strategies. We furthermore hope that the herein summarized information about the physiological roles of SGLTs and the therapeutic benefits of the gliflozins will be useful for our readers to better understand the molecular basis of the beneficial effects of these inhibitors, also in the context of the tubuloglomerular feedback (TGF), and the renin-angiotensin system (RAS). The detailed mechanisms underlying the clinical benefits of SGLT2 inhibition by gliflozins still warrant further investigation that may serve as a basis for future drug development.
Collapse
Affiliation(s)
- Gergely Gyimesi
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension, and Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Office D845, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Jonai Pujol-Giménez
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension, and Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Office D845, Freiburgstrasse 15, CH-3010, Bern, Switzerland
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Matthias A Hediger
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension, and Department of Biomedical Research, Inselspital, University of Bern, Kinderklinik, Office D845, Freiburgstrasse 15, CH-3010, Bern, Switzerland.
| |
Collapse
|
23
|
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
|
24
|
Pizzoni A, Bazzi Z, Di Giusto G, Alvarez CL, Rivarola V, Capurro C, Schwarzbaum PJ, Ford P. Release of ATP by TRPV4 activation is dependent upon the expression of AQP2 in renal cells. J Cell Physiol 2020; 236:2559-2571. [PMID: 33094506 DOI: 10.1002/jcp.30013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/18/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022]
Abstract
Increasing evidence indicates that aquaporins (AQPs) exert an influence in cell signaling by the interplay with the transient receptor potential vanilloid 4 (TRPV4) channel. We previously found that TRPV4 physically and functionally interacts with AQP2 in cortical collecting ducts (CCD) cells, favoring cell volume regulation and cell migration. Because TRPV4 was implicated in ATP release in several tissues, we investigated the possibility that TRPV4/AQP2 interaction influences ATP release in CCD cells. Using two CCD cell lines expressing or not AQP2, we measured extracellular ATP (ATPe) under TRPV4 activation and intracellular Ca2+ under ATP addition. We found that AQP2 is critical for the release of ATP induced by TRPV4 activation. This ATP release occurs by an exocytic and a conductive route. ATPe, in turn, stimulates purinergic receptors leading to ATPe-induced ATP release by a Ca2+ -dependent mechanism. We propose that AQP2 by modulating Ca2+ and ATP differently could explain AQP2-increased cell migration.
Collapse
Affiliation(s)
- Alejandro Pizzoni
- Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Facultad de Medicina, Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-HOUSSAY), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Zaher Bazzi
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisico-Química Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini," Consejo Nacional de Investigaciones Científicas y Técnicas, Cátedra de Química Biológica Superior, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gisela Di Giusto
- Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Facultad de Medicina, Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-HOUSSAY), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cora L Alvarez
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisico-Química Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini," Consejo Nacional de Investigaciones Científicas y Técnicas, Cátedra de Química Biológica Superior, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Valeria Rivarola
- Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Facultad de Medicina, Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-HOUSSAY), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Claudia Capurro
- Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Facultad de Medicina, Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-HOUSSAY), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo J Schwarzbaum
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisico-Química Biológicas (IQUIFIB) "Prof. Alejandro C. Paladini," Consejo Nacional de Investigaciones Científicas y Técnicas, Cátedra de Química Biológica Superior, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paula Ford
- Departamento de Ciencias Fisiológicas, Laboratorio de Biomembranas, Facultad de Medicina, Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-HOUSSAY), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
25
|
Resolving the Ionotropic P2X4 Receptor Mystery Points Towards a New Therapeutic Target for Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21145005. [PMID: 32679900 PMCID: PMC7404342 DOI: 10.3390/ijms21145005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022] Open
Abstract
Adenosine triphosphate (ATP) is a primordial versatile autacoid that changes its role from an intracellular energy saver to a signaling molecule once released to the extracellular milieu. Extracellular ATP and its adenosine metabolite are the main activators of the P2 and P1 purinoceptor families, respectively. Mounting evidence suggests that the ionotropic P2X4 receptor (P2X4R) plays pivotal roles in the regulation of the cardiovascular system, yet further therapeutic advances have been hampered by the lack of selective P2X4R agonists. In this review, we provide the state of the art of the P2X4R activity in the cardiovascular system. We also discuss the role of P2X4R activation in kidney and lungs vis a vis their interplay to control cardiovascular functions and dysfunctions, including putative adverse effects emerging from P2X4R activation. Gathering this information may prompt further development of selective P2X4R agonists and its translation to the clinical practice.
Collapse
|
26
|
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: 70] [Impact Index Per Article: 14.0] [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
|
27
|
Gerbino A, De Zio R, Russo D, Milella L, Milano S, Procino G, Pusch M, Svelto M, Carmosino M. Role of PKC in the Regulation of the Human Kidney Chloride Channel ClC-Ka. Sci Rep 2020; 10:10268. [PMID: 32581267 PMCID: PMC7314819 DOI: 10.1038/s41598-020-67219-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/02/2020] [Indexed: 12/03/2022] Open
Abstract
The physiological role of the renal ClC-Ka/ClC-K1 channels is to confer a high Cl- permeability to the thin Ascending Limb of Henle (tAL), which in turn is essential for establishing the high osmolarity of the renal medulla that drives water reabsorption from collecting ducts. Here, we investigated by whole-cell patch-clamp measurements on HEK293 cells co-expressing ClC-Ka (tagged with GFP) and the accessory subunit barttin (tagged with m-Cherry) the effect of a natural diuretic extract from roots of Dandelion (DRE), and other compounds activating PKC, such as ATP, on ClC-Ka activity and its membrane localization. Treatment with 400 µg/ml DRE significantly inhibited Cl- currents time-dependently within several minutes. Of note, the same effect on Cl- currents was obtained upon treatment with 100 µM ATP. Pretreatment of cells with either the intracellular Ca2+ chelator BAPTA-AM (30 μM) or the PKC inhibitor Calphostin C (100 nM) reduced the inhibitory effect of DRE. Conversely, 1 µM of phorbol meristate acetate (PMA), a specific PKC activator, mimicked the inhibitory effect of DRE on ClC-Ka. Finally, we found that pretreatment with 30 µM Heclin, an E3 ubiquitin ligase inhibitor, did not revert DRE-induced Cl- current inhibition. In agreement with this, live-cell confocal analysis showed that DRE treatment did not induce ClC-Ka internalization. In conclusion, we demonstrate for the first time that the activity of ClC-Ka in renal cells could be significantly inhibited by the activation of PKC elicited by classical maneuvers, such as activation of purinergic receptors, or by exposure to herbal extracts that activates a PKC-dependent pathway. Overall, we provide both new information regarding the regulation of ClC-Ka and a proof-of-concept study for the use of DRE as new diuretic.
Collapse
Affiliation(s)
- Andrea Gerbino
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Roberta De Zio
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Daniela Russo
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Luigi Milella
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Michael Pusch
- National Research Council, Institute of Biophysics, Genova, IT, Italy
| | - Maria Svelto
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, IT, Italy. .,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy.
| |
Collapse
|
28
|
Jackson EK, Gillespie DG, Cheng D, Mi Z, Menshikova EV. Characterization of the N 6-etheno-bridge method to assess extracellular metabolism of adenine nucleotides: detection of a possible role for purine nucleoside phosphorylase in adenosine metabolism. Purinergic Signal 2020; 16:187-211. [PMID: 32367441 PMCID: PMC7367995 DOI: 10.1007/s11302-020-09699-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
The goal of this study was to determine the validity of using N6-etheno-bridged adenine nucleotides to evaluate ecto-nucleotidase activity. We observed that the metabolism of N6-etheno-ATP versus ATP was quantitatively similar when incubated with recombinant CD39, ENTPD2, ENTPD3, or ENPP-1, and the quantitative metabolism of N6-etheno-AMP versus AMP was similar when incubated with recombinant CD73. This suggests that ecto-nucleotidases process N6-etheno-bridged adenine nucleotides similarly to endogenous adenine nucleotides. Four cell types rapidly (t1/2, 0.21 to 0.66 h) metabolized N6-etheno-ATP. Applied N6-etheno-ATP was recovered in the medium as N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and surprisingly N6-etheno-adenine; intracellular N6-etheno compounds were undetectable. This suggests minimal cellular uptake, intracellular metabolism, or deamination of these compounds. N6-etheno-ATP, N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and N6-etheno-adenine had little affinity for recombinant A1, A2A, or A2B receptors, for a subset of P2X receptors (3H-α,β-methylene-ATP binding to rat bladder membranes), or for a subset of P2Y receptors (35S-ATP-αS binding to rat brain membranes), suggesting minimal pharmacological activity. N6-etheno-adenosine was partially converted to N6-etheno-adenine in four different cell types; this was blocked by purine nucleoside phosphorylase (PNPase) inhibition. Intravenous N6-etheno-ATP was quickly metabolized, with N6-etheno-adenine being the main product in naïve rats, but not in rats pretreated with a PNPase inhibitor. PNPase inhibition reduced the urinary excretion of endogenous adenine and attenuated the conversion of exogenous adenosine to adenine in the renal cortex. The N6-etheno-bridge method is a valid technique to assess extracellular metabolism of adenine nucleotides by ecto-nucleotidases. Also, rats express an enzyme with PNPase-like activity that metabolizes N6-etheno-adenosine to N6-etheno-adenine.
Collapse
Affiliation(s)
- Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Delbert G. Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Elizabeth V. Menshikova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| |
Collapse
|
29
|
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
|
30
|
Feliu C, Peyret H, Vautier D, Djerada Z. Simultaneous quantification of 8 nucleotides and adenosine in cells and their medium using UHPLC-HRMS. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1148:122156. [PMID: 32446186 DOI: 10.1016/j.jchromb.2020.122156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Purinergic signalling is involved in physiological processes, particularly during ischemia-reperfusion injuries for which it has a protective effect. The purpose of this work was to develop a method for simultaneous quantification of eight nucleotides and adenosine in biological matrices by liquid chromatography coupled with high-resolution mass spectrometry. A method was developed that was sufficiently robust to quantify the targeted analytes in 20 min with good sensitivity. Analysis of extracellular media from cultured endothelial cells detected the release of nucleotides and adenosine during 2 h of hypoxia. The quantification of cylic adenosine monophosphate (cAMP) allowed to establish a dose-response curve after receptor stimulation. Therefore, our method allows us to study the involvement of nucleotides in various processes in both the intracellular and extracellular compartment.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Hélène Peyret
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Damien Vautier
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Zoubir Djerada
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France.
| |
Collapse
|
31
|
Guan Z, Makled MN, Inscho EW. Purinoceptors, renal microvascular function and hypertension. Physiol Res 2020; 69:353-369. [PMID: 32301620 DOI: 10.33549/physiolres.934463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Proper renal blood flow (RBF) and glomerular filtration rate (GFR) are critical for maintaining normal blood pressure, kidney function and water and electrolyte homeostasis. The renal microvasculature expresses a multitude of receptors mediating vasodilation and vasoconstriction, which can influence glomerular blood flow and capillary pressure. Despite this, RBF and GFR remain quite stable when arterial pressure fluctuates because of the autoregulatory mechanism. ATP and adenosine participate in autoregulatory control of RBF and GFR via activation of two different purinoceptor families (P1 and P2). Purinoceptors are widely expressed in renal microvasculature and tubules. Emerging data show altered purinoceptor signaling in hypertension-associated kidney injury, diabetic nephropathy, sepsis, ischemia-reperfusion induced acute kidney injury and polycystic kidney disease. In this brief review, we highlight recent studies and new insights on purinoceptors regulating renal microvascular function and renal hemodynamics. We also address the mechanisms underlying renal microvascular injury and impaired renal autoregulation, focusing on purinoceptor signaling and hypertension-induced renal microvascular dysfunction. Interested readers are directed to several excellent and comprehensive reviews that recently covered the topics of renal autoregulation, and nucleotides in kidney function under physiological and pathophysiological conditions (Inscho 2009, Navar et al. 2008, Carlstrom et al. 2015, Vallon et al. 2020).
Collapse
Affiliation(s)
- Z Guan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, South Birmingham, USA.
| | | | | |
Collapse
|
32
|
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
|
33
|
Analysis of purine receptor expression and functionality in alveolar epithelial cells. Purinergic Signal 2020; 16:213-229. [PMID: 32236789 DOI: 10.1007/s11302-020-09696-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023] Open
Abstract
Despite its fundamental role in providing an extensive surface for gas exchange, the alveolar epithelium (AE) serves as an immunological barrier through, e.g., the release of proinflammatory cytokines and secretion of surfactant to prevent alveolar collapse. Thus, AE is important for sustaining lung homeostasis. Extracellular ATP secreted by alveolar epithelial cells (AECs) is involved in physiological and pathological conditions and acts mainly through the activation of purine receptors (P2Rs). When studying P2R-mediated processes, primary isolated type II AECs (piAECs) still represent the gold standard in in vitro research, although their preparation is time-consuming and requires the sacrifice of many animals. Hence, cultivated immortalized and tumor-derived AEC lines may constitute a valuable alternative. In this work, we examined P2R expression and functionality in piAECs, in immortalized and tumor-derived AEC lines with the purpose of gaining a better understanding of purinergic signaling in different cell systems and assisting researchers in the choice of a suitable cell line with a certain P2R in demand. We combined mRNA and protein analysis to evaluate the expression of P2R. For pharmacological testing, we conducted calcium ([Ca2+]) measurements and siRNA receptor knockdown. Interestingly, the mRNA and protein levels of P2Y2, P2Y6, and P2X4 were detected on all cell lines. Concerning functionality, P2XR could be narrowed to L2 and piAECs while P2YR were active in all cell lines.
Collapse
|
34
|
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
|
35
|
Verschuren EHJ, Castenmiller C, Peters DJM, Arjona FJ, Bindels RJM, Hoenderop JGJ. Sensing of tubular flow and renal electrolyte transport. Nat Rev Nephrol 2020; 16:337-351. [DOI: 10.1038/s41581-020-0259-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2020] [Indexed: 02/06/2023]
|
36
|
Price GW, Potter JA, Williams BM, Cliff CL, Squires PE, Hills CE. Connexin-mediated cell communication in the kidney: A potential therapeutic target for future intervention of diabetic kidney disease?: Joan Mott Prize Lecture. Exp Physiol 2020; 105:219-229. [PMID: 31785013 DOI: 10.1113/ep087770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
Abstract
The ability of cells to communicate and synchronise their activity is essential for the maintenance of tissue structure, integrity and function. A family of membrane-bound proteins called connexins are largely responsible for mediating the local transfer of information between cells. Assembled in the cell membrane as a hexameric connexon, they either function as a conduit for paracrine signalling, forming a transmembrane hemi-channel, or, if aligned with connexons on neighbouring cells, form a continuous aqueous pore or gap junction, which allows for the direct transmission of metabolic and electrical signals. Regulation of connexin synthesis and activity is critical to cellular function, and a number of diseases are attributed to changes in the expression and/or function of these important proteins. A link between hyperglycaemia, connexin expression, altered nucleotide concentrations and impaired function highlights a potential role for connexin-mediated cell communication in complications of diabetes. In the diabetic kidney, glycaemic injury is the leading cause of end-stage renal failure, reflecting multiple aetiologies including glomerular hyperfiltration, albuminuria, increased deposition of extracellular matrix and tubulointerstitial fibrosis. Loss of connexin-mediated cell-to-cell communication in diabetic nephropathy may represent an early sign of disease progression, but our understanding of the process remains severely limited. This review focuses on recent evidence demonstrating that glucose-evoked changes in connexin-mediated cell communication and associated purinergic signalling may contribute to the pathogenesis of kidney disease in diabetes, highlighting the tantalising potential of targeting these proteins as a novel therapeutic intervention.
Collapse
Affiliation(s)
- Gareth W Price
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Joe A Potter
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Bethany M Williams
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Chelsy L Cliff
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Paul E Squires
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Claire E Hills
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| |
Collapse
|
37
|
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: 58] [Impact Index Per Article: 9.7] [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
|
38
|
Blanchard C, Boué-Grabot E, Massé K. Comparative Embryonic Spatio-Temporal Expression Profile Map of the Xenopus P2X Receptor Family. Front Cell Neurosci 2019; 13:340. [PMID: 31402854 PMCID: PMC6676501 DOI: 10.3389/fncel.2019.00340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/10/2019] [Indexed: 01/27/2023] Open
Abstract
P2X receptors are ATP-gated cations channels formed by the homo or hetero-trimeric association from the seven cloned subunits (P2X1-7). P2X receptors are widely distributed in different organs and cell types throughout the body including the nervous system and are involved in a large variety of physiological but also pathological processes in adult mammals. However, their expression and function during embryogenesis remain poorly understood. Here, we report the cloning and the comparative expression map establishment of the entire P2X subunit family in the clawed frog Xenopus. Orthologous sequences for 6 mammalian P2X subunits were identified in both X. laevis and X. tropicalis, but not for P2X3 subunit, suggesting a potential loss of this subunit in the Pipidae family. Three of these genes (p2rx1, p2rx2, and p2rx5) exist as homeologs in the pseudoallotetraploid X. laevis, making a total of 9 subunits in this species. Phylogenetic analyses demonstrate the high level of conservation of these receptors between amphibian and other vertebrate species. RT-PCR revealed that all subunits are expressed during the development although zygotic p2rx6 and p2rx7 transcripts are mainly detected at late organogenesis stages. Whole mount in situ hybridization shows that each subunit displays a specific spatio-temporal expression profile and that these subunits can therefore be grouped into two groups, based on their expression or not in the developing nervous system. Overlapping expression in the central and peripheral nervous system and in the sensory organs suggests potential heteromerization and/or redundant functions of P2X subunits in Xenopus embryos. The developmental expression of the p2rx subunit family during early phases of embryogenesis indicates that these subunits may have distinct roles during vertebrate development, especially embryonic neurogenesis.
Collapse
Affiliation(s)
- Camille Blanchard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Eric Boué-Grabot
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Karine Massé
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
39
|
Smaini M, Smaini I, Ennachete M, Laghlimi C, Saâdane H, Moutcine A, Chtaini A. Electrochemical determination of adenosine by natural phosphate modified carbon paste electrode: Analytical application in serum. SENSING AND BIO-SENSING RESEARCH 2019. [DOI: 10.1016/j.sbsr.2019.100272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
40
|
Craigie E, Menzies RI, Larsen CK, Jacquillet G, Carrel M, Wildman SS, Loffing J, Leipziger J, Shirley DG, Bailey MA, Unwin RJ. The renal and blood pressure response to low sodium diet in P2X4 receptor knockout mice. Physiol Rep 2018; 6:e13899. [PMID: 30350402 PMCID: PMC6198136 DOI: 10.14814/phy2.13899] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 01/02/2023] Open
Abstract
In the kidney, purinergic (P2) receptor-mediated ATP signaling has been shown to be an important local regulator of epithelial sodium transport. Appropriate sodium regulation is crucial for blood pressure (BP) control and disturbances in sodium balance can lead to hypo- or hypertension. Links have already been established between P2 receptor signaling and the development of hypertension, attributed mainly to vascular and/or inflammatory effects. A transgenic mouse model with deletion of the P2X4 receptor (P2X4-/- ) is known to have hypertension, which is thought to reflect endothelial dysfunction and impaired nitric oxide (NO) release. However, renal function in this model has not been characterized; moreover, studies in vitro have shown that the P2X4 receptor can regulate renal epithelial Na+ channel (ENaC) activity. Therefore, in the present study we investigated renal function and sodium handling in P2X4-/- mice, focusing on ENaC-mediated Na+ reabsorption. We confirmed an elevated BP in P2X4-/- mice compared with wild-type mice, but found that ENaC-mediated Na+ reabsorption is no different from wild-type and does not contribute to the raised BP observed in the knockout. However, when P2X4-/- mice were placed on a low sodium diet, BP normalized. Plasma aldosterone concentration tended to increase according to sodium restriction status in both genotypes; in contrast to wild-types, P2X4-/- mice did not show an increase in functional ENaC activity. Thus, although the increased BP in P2X4-/- mice has been attributed to endothelial dysfunction and impaired NO release, there is also a sodium-sensitive component.
Collapse
Affiliation(s)
- Eilidh Craigie
- Centre for NephrologyUniversity College London Medical SchoolLondonUnited Kingdom
- Institue for AnatomyUniversity of ZürichZürichSwitzerland
| | - Robert I. Menzies
- British Heart Foundation Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUnited Kingdom
| | - Casper K. Larsen
- Department of Biomedicine, PhysiologyAarhus UniversityAarhus CDenmark
| | - Grégory Jacquillet
- Centre for NephrologyUniversity College London Medical SchoolLondonUnited Kingdom
| | - Monique Carrel
- Institue for AnatomyUniversity of ZürichZürichSwitzerland
| | - Scott S. Wildman
- Urinary System Physiology UnitMedway School of PharmacyUniversity of KentKentUnited Kingdom
| | | | - Jens Leipziger
- Department of Biomedicine, PhysiologyAarhus UniversityAarhus CDenmark
| | - David G. Shirley
- Centre for NephrologyUniversity College London Medical SchoolLondonUnited Kingdom
| | - Matthew A. Bailey
- British Heart Foundation Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUnited Kingdom
| | - Robert J. Unwin
- Centre for NephrologyUniversity College London Medical SchoolLondonUnited Kingdom
- CVRM iMEDAstraZeneca GothenburgGothenburgSweden
| |
Collapse
|
41
|
Alicic RZ, Johnson EJ, Tuttle KR. SGLT2 Inhibition for the Prevention and Treatment of Diabetic Kidney Disease: A Review. Am J Kidney Dis 2018; 72:267-277. [DOI: 10.1053/j.ajkd.2018.03.022] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/06/2018] [Indexed: 02/06/2023]
|
42
|
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
|
43
|
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
|
44
|
Olabiyi AA, Carvalho FB, Bottari NB, Morsch VM, Morel AF, Oboh G, Schetinger MR. Tiger nut and walnut extracts modulate extracellular metabolism of ATP and adenosine through the NOS/cGMP/PKG signalling pathway in kidney slices. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 43:140-149. [PMID: 29747747 DOI: 10.1016/j.phymed.2018.04.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 02/19/2018] [Accepted: 04/15/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Tiger nut (Cyperus esculentus L.) and walnut (Tetracarpidium conophorum Müll. Arg.) have been reportedly used in the treatment of inflammatory diseases such as atherosclerosis, prevent heart attack and improve blood circulation, reduce serum cholesterol level as well as inhibit oxidation reactions. PURPOSE This study investigated the effect of tiger nut and walnut hydro-alcoholic extracts on extracellular metabolism of ATP through the NOS/cGMP/PKG signaling pathway induced by Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME) in kidney slices. METHODS The plants were extracted for 24 h in 10 ml of 70% ethanol and 30% distilled water per gram milled material on a mechanical shaker and filtered using Whatman filter paper. The effect of the extracts on ecto-nucleotidases (NTPDase and 5' nucleotidase) and adenosine deaminase activities, nitrites and nitrates levels (NO, markers of NO production) as well as lipid and protein oxidation reactions in kidney slices were evaluated. Also, the phenolic components of the nut samples were determined using High Performance Liquid Chromatography (HPLC). RESULTS The results revealed a protective effect of tiger nut and walnut on co-incubation with L-NAME of the enzyme activities, increased NO significantly (P < 0.05) when compared to the vehicle. L-NAME also increased the thiobabituric reactive substances but co-incubation with the extracts caused a significant reduction while protein oxidation across groups showed no significant difference when compared to the vehicle group. HPLC finger printing revealed the presence of quercetin and kaempferol as the most abundant phenolic compounds in tiger nut and walnut respectively. CONCLUSION Tiger nut and walnut extracts showed a protective effect on L-NAME induced kidney slices by reducing the activities of NTPDase (ATP as substrate) and adenosine deaminase, increased NO levels as well as prevent oxidative damage. The effect observed may be attributed to the phenolic compounds present in both nuts as depicted by HPLC finger printing.
Collapse
Affiliation(s)
- Ayodeji A Olabiyi
- Department of Biochemistry, Functional Foods and Nutraceuticals Unit, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Department of Medical Biochemistry, Afe Babalola University Ado-Ekiti, P.M.B. 5454, Nigeria; Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria/RS 97105-900, Brazil.
| | - Fabiano B Carvalho
- Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria/RS 97105-900, Brazil
| | - Nathieli B Bottari
- Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria/RS 97105-900, Brazil
| | - Vera M Morsch
- Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria/RS 97105-900, Brazil
| | - Ademir F Morel
- Department of Chemistry, Center of Natural and Exacts Sciences, Federal University of Santa Maria, RS 97105-900, Brazil
| | - Ganiyu Oboh
- Department of Biochemistry, Functional Foods and Nutraceuticals Unit, Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria.
| | - Maria Rosa Schetinger
- Department of Biochemistry and Molecular Biology, Center of Natural and Exacts Sciences, Federal University of Santa Maria, Santa Maria/RS 97105-900, Brazil.
| |
Collapse
|
45
|
Burnstock G, Knight GE. The potential of P2X7 receptors as a therapeutic target, including inflammation and tumour progression. Purinergic Signal 2018; 14:1-18. [PMID: 29164451 PMCID: PMC5842154 DOI: 10.1007/s11302-017-9593-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
Seven P2X ion channel nucleotide receptor subtypes have been cloned and characterised. P2X7 receptors (P2X7R) are unusual in that there are extra amino acids in the intracellular C terminus. Low concentrations of ATP open cation channels sometimes leading to cell proliferation, whereas high concentrations of ATP open large pores that release inflammatory cytokines and can lead to apoptotic cell death. Since many diseases involve inflammation and immune responses, and the P2X7R regulates inflammation, there has been recent interest in the pathophysiological roles of P2X7R and the potential of P2X7R antagonists to treat a variety of diseases. These include neurodegenerative diseases, psychiatric disorders, epilepsy and a number of diseases of peripheral organs, including the cardiovascular, airways, kidney, liver, bladder, skin and musculoskeletal. The potential of P2X7R drugs to treat tumour progression is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK.
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
- Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia.
| | - Gillian E Knight
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK
| |
Collapse
|
46
|
Savio LEB, de Andrade Mello P, da Silva CG, Coutinho-Silva R. The P2X7 Receptor in Inflammatory Diseases: Angel or Demon? Front Pharmacol 2018; 9:52. [PMID: 29467654 PMCID: PMC5808178 DOI: 10.3389/fphar.2018.00052] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Under physiological conditions, adenosine triphosphate (ATP) is present at low levels in the extracellular milieu, being massively released by stressed or dying cells. Once outside the cells, ATP and related nucleotides/nucleoside generated by ectonucleotidases mediate a high evolutionary conserved signaling system: the purinergic signaling, which is involved in a variety of pathological conditions, including inflammatory diseases. Extracellular ATP has been considered an endogenous adjuvant that can initiate inflammation by acting as a danger signal through the activation of purinergic type 2 receptors-P2 receptors (P2Y G-protein coupled receptors and P2X ligand-gated ion channels). Among the P2 receptors, the P2X7 receptor is the most extensively studied from an immunological perspective, being involved in both innate and adaptive immune responses. P2X7 receptor activation induces large-scale ATP release via its intrinsic ability to form a membrane pore or in association with pannexin hemichannels, boosting purinergic signaling. ATP acting via P2X7 receptor is the second signal to the inflammasome activation, inducing both maturation and release of pro-inflammatory cytokines, such as IL-1β and IL-18, and the production of reactive nitrogen and oxygen species. Furthermore, the P2X7 receptor is involved in caspases activation, as well as in apoptosis induction. During adaptive immune response, P2X7 receptor modulates the balance between the generation of T helper type 17 (Th17) and T regulatory (Treg) lymphocytes. Therefore, this receptor is involved in several inflammatory pathological conditions. In infectious diseases and cancer, P2X7 receptor can have different and contrasting effects, being an angel or a demon depending on its level of activation, cell studied, type of pathogen, and severity of infection. In neuroinflammatory and neurodegenerative diseases, P2X7 upregulation and function appears to contribute to disease progression. In this review, we deeply discuss P2X7 receptor dual function and its pharmacological modulation in the context of different pathologies, and we also highlight the P2X7 receptor as a potential target to treat inflammatory related diseases.
Collapse
Affiliation(s)
- Luiz E B Savio
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola de Andrade Mello
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Cleide Gonçalves da Silva
- Division of Vascular Surgery, Department of Surgery, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
47
|
Dou L, Chen YF, Cowan PJ, Chen XP. Extracellular ATP signaling and clinical relevance. Clin Immunol 2017; 188:67-73. [PMID: 29274390 DOI: 10.1016/j.clim.2017.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/12/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022]
Abstract
Since purinergic signaling was discovered in the early 1970s, it has been shown that extracellular nucleotides, and their derivative nucleosides, are released in a regulated or unregulated manner by cells in various challenging settings and then bind defined purinergic receptors to activate intricate signaling networks. Extracellular ATP plays a role based on different P2 receptor subtypes expressed on specific cell types. Sequential hydrolysis of extracellular ATP catalyzed by ectonucleotidases (e.g. CD39, CD73) is the main pathway for the generation of adenosine, which in turn activates P1 receptors. Many studies have demonstrated that extracellular ATP signaling functions as an important dynamic regulatory pathway to coordinate appropriate immune responses in various pathological processes, including intracellular infection, host-tumor interaction, pro-inflammation vascular injury, and transplant immunity. ATP receptors and CD39 also participate in related clinical settings. Here, we review the latest research in to the development of promising clinical treatment strategies.
Collapse
Affiliation(s)
- Lei Dou
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Fa Chen
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Peter J Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Australia.
| | - Xiao-Ping Chen
- Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
48
|
Battistone MA, Nair AV, Barton CR, Liberman RN, Peralta MA, Capen DE, Brown D, Breton S. Extracellular Adenosine Stimulates Vacuolar ATPase-Dependent Proton Secretion in Medullary Intercalated Cells. J Am Soc Nephrol 2017; 29:545-556. [PMID: 29222395 DOI: 10.1681/asn.2017060643] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 10/18/2017] [Indexed: 12/29/2022] Open
Abstract
Acidosis is an important complication of AKI and CKD. Renal intercalated cells (ICs) express the proton pumping vacuolar H+-ATPase (V-ATPase) and are extensively involved in acid-base homeostasis. H+ secretion in type A intercalated cells (A-ICs) is regulated by apical vesicle recycling and stimulated by cAMP. In other cell types, cAMP is increased by extracellular agonists, including adenosine, through purinergic receptors. Adenosine is a Food and Drug Administration-approved drug, but very little is known about the effect of adenosine on IC function. Therefore, we investigated the role of adenosine in the regulation of V-ATPase in ICs. Intravenous treatment of mice with adenosine or agonists of ADORA2A and ADORA2B purinergic P1 receptors induced V-ATPase apical membrane accumulation in medullary A-ICs but not in cortical A-ICs or other IC subtypes. Both receptors are located in A-IC apical membranes, and adenosine injection increased urine adenosine concentration and decreased urine pH. Cell fractionation showed that adenosine or an ADORA2A or ADORA2B agonist induced V-ATPase translocation from vesicles to the plasma membrane and increased protein kinase A (PKA)-dependent protein phosphorylation in purified medullary ICs that were isolated from mice. Either ADORA2A or ADORA2B antagonists or the PKA inhibitor mPKI blocked these effects. Finally, a fluorescence pH assay showed that adenosine activates V-ATPase in isolated medullary ICs. Our study shows that medullary A-ICs respond to luminal adenosine through ADORA2A and ADORA2B receptors in a cAMP/PKA pathway-dependent mechanism to induce V-ATPase-dependent H+ secretion.
Collapse
Affiliation(s)
- Maria A Battistone
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anil V Nair
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Claire R Barton
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rachel N Liberman
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria A Peralta
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Diane E Capen
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sylvie Breton
- Program in Membrane Biology, Center for Systems Biology, Nephrology Division, and Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
49
|
Plattner H, Verkhratsky A. Inseparable tandem: evolution chooses ATP and Ca2+ to control life, death and cellular signalling. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0419. [PMID: 27377729 DOI: 10.1098/rstb.2015.0419] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2016] [Indexed: 01/01/2023] Open
Abstract
From the very dawn of biological evolution, ATP was selected as a multipurpose energy-storing molecule. Metabolism of ATP required intracellular free Ca(2+) to be set at exceedingly low concentrations, which in turn provided the background for the role of Ca(2+) as a universal signalling molecule. The early-eukaryote life forms also evolved functional compartmentalization and vesicle trafficking, which used Ca(2+) as a universal signalling ion; similarly, Ca(2+) is needed for regulation of ciliary and flagellar beat, amoeboid movement, intracellular transport, as well as of numerous metabolic processes. Thus, during evolution, exploitation of atmospheric oxygen and increasingly efficient ATP production via oxidative phosphorylation by bacterial endosymbionts were a first step for the emergence of complex eukaryotic cells. Simultaneously, Ca(2+) started to be exploited for short-range signalling, despite restrictions by the preset phosphate-based energy metabolism, when both phosphates and Ca(2+) interfere with each other because of the low solubility of calcium phosphates. The need to keep cytosolic Ca(2+) low forced cells to restrict Ca(2+) signals in space and time and to develop energetically favourable Ca(2+) signalling and Ca(2+) microdomains. These steps in tandem dominated further evolution. The ATP molecule (often released by Ca(2+)-regulated exocytosis) rapidly grew to be the universal chemical messenger for intercellular communication; ATP effects are mediated by an extended family of purinoceptors often linked to Ca(2+) signalling. Similar to atmospheric oxygen, Ca(2+) must have been reverted from a deleterious agent to a most useful (intra- and extracellular) signalling molecule. Invention of intracellular trafficking further increased the role for Ca(2+) homeostasis that became critical for regulation of cell survival and cell death. Several mutually interdependent effects of Ca(2+) and ATP have been exploited in evolution, thus turning an originally unholy alliance into a fascinating success story.This article is part of the themed issue 'Evolution brings Ca(2+) and ATP together to control life and death'.
Collapse
Affiliation(s)
- Helmut Plattner
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Alexei Verkhratsky
- Faculty of Biological Sciences, University of Manchester, Manchester M13 9PT, UK Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
50
|
Burnstock G. Purinergic Signalling: Therapeutic Developments. Front Pharmacol 2017; 8:661. [PMID: 28993732 PMCID: PMC5622197 DOI: 10.3389/fphar.2017.00661] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
Abstract
Purinergic signalling, i.e., the role of nucleotides as extracellular signalling molecules, was proposed in 1972. However, this concept was not well accepted until the early 1990's when receptor subtypes for purines and pyrimidines were cloned and characterised, which includes four subtypes of the P1 (adenosine) receptor, seven subtypes of P2X ion channel receptors and 8 subtypes of the P2Y G protein-coupled receptor. Early studies were largely concerned with the physiology, pharmacology and biochemistry of purinergic signalling. More recently, the focus has been on the pathophysiology and therapeutic potential. There was early recognition of the use of P1 receptor agonists for the treatment of supraventricular tachycardia and A2A receptor antagonists are promising for the treatment of Parkinson's disease. Clopidogrel, a P2Y12 antagonist, is widely used for the treatment of thrombosis and stroke, blocking P2Y12 receptor-mediated platelet aggregation. Diquafosol, a long acting P2Y2 receptor agonist, is being used for the treatment of dry eye. P2X3 receptor antagonists have been developed that are orally bioavailable and stable in vivo and are currently in clinical trials for the treatment of chronic cough, bladder incontinence, visceral pain and hypertension. Antagonists to P2X7 receptors are being investigated for the treatment of inflammatory disorders, including neurodegenerative diseases. Other investigations are in progress for the use of purinergic agents for the treatment of osteoporosis, myocardial infarction, irritable bowel syndrome, epilepsy, atherosclerosis, depression, autism, diabetes, and cancer.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical SchoolLondon, United Kingdom
- Department of Pharmacology and Therapeutics, The University of Melbourne, MelbourneVIC, Australia
| |
Collapse
|