1
|
Flores-Roco A, Lago BM, Villa-Bellosta R. Elevated glucose levels increase vascular calcification risk by disrupting extracellular pyrophosphate metabolism. Cardiovasc Diabetol 2024; 23:405. [PMID: 39529124 PMCID: PMC11555999 DOI: 10.1186/s12933-024-02502-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Vascular calcification is a major contributor to cardiovascular disease, especially diabetes, where it exacerbates morbidity and mortality. Although pyrophosphate is a recognized natural inhibitor of vascular calcification, there have been no prior studies examining its specific deficiency in diabetic conditions. This study is the first to analyze the direct link between elevated glucose levels and disruptions in extracellular pyrophosphate metabolism. METHODS Rat aortic smooth muscle cells, streptozotocin (STZ)-induced diabetic rats, and diabetic human aortic smooth muscle cells were used to assess the effects of elevated glucose levels on pyrophosphate metabolism and vascular calcification. The techniques used include extracellular pyrophosphate metabolism assays, thin-layer chromatography, phosphate-induced calcification assays, BrdU incorporation for DNA synthesis, aortic smooth muscle cell viability and proliferation assays, and quantitative PCR for enzyme expression analysis. Additionally, extracellular pyrophosphate metabolism was examined through the use of radiolabeled isotopes to track ATP and pyrophosphate transformations. RESULTS Elevated glucose led to a significant reduction in extracellular pyrophosphate across all diabetic models. This metabolic disruption was marked by notable downregulation of both the expression and activity of ectonucleotide pyrophosphatase/phosphodiesterase 1, a key enzyme that converts ATP to pyrophosphate. We also observed an upregulation of ectonucleoside triphosphate diphosphohydrolase 1, which preferentially hydrolyzes ATP to inorganic phosphate rather than pyrophosphate. Moreover, tissue-nonspecific alkaline phosphatase activity was markedly elevated across all diabetic models. This shift in enzyme activity significantly reduced the pyrophosphate/phosphate ratio. In addition, we noted a marked downregulation of matrix Gla protein, another inhibitor of vascular calcification. The impaired pyrophosphate metabolism was further corroborated by calcification experiments across all three diabetic models, which demonstrated an increased propensity for vascular calcification. CONCLUSIONS This study demonstrated that diabetes-induced high glucose disrupts extracellular pyrophosphate metabolism, compromising its protective role against vascular calcification. These findings identify pyrophosphate deficiency as a potential mechanism in diabetic vascular calcification, highlighting a new therapeutic target. Strategies aimed at restoring or enhancing pyrophosphate levels may offer significant potential in mitigating cardiovascular complications in diabetic patients, meriting further investigation.
Collapse
MESH Headings
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Diphosphates/metabolism
- Animals
- Humans
- Phosphoric Diester Hydrolases/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Pyrophosphatases/metabolism
- Pyrophosphatases/genetics
- Cells, Cultured
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Rats, Sprague-Dawley
- Alkaline Phosphatase/metabolism
- Alkaline Phosphatase/blood
- Extracellular Matrix Proteins/metabolism
- Extracellular Matrix Proteins/genetics
- Blood Glucose/metabolism
- Matrix Gla Protein
- Calcium-Binding Proteins/metabolism
- Calcium-Binding Proteins/genetics
- Cell Proliferation/drug effects
- GPI-Linked Proteins/metabolism
- Rats
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/prevention & control
- Aorta/metabolism
- Aorta/pathology
- Aorta/drug effects
- 5'-Nucleotidase
Collapse
Affiliation(s)
- Alicia Flores-Roco
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Belinda M Lago
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Ricardo Villa-Bellosta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706, Santiago de Compostela, Spain.
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Campus Vida, University of Santiago de Compostela, Avenida de Barcelona S/N, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
2
|
Babou Kammoe RB, Sévigny J. Extracellular nucleotides in smooth muscle contraction. Biochem Pharmacol 2024; 220:116005. [PMID: 38142836 DOI: 10.1016/j.bcp.2023.116005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Extracellular nucleotides and nucleosides are crucial signalling molecules, eliciting diverse biological responses in almost all organs and tissues. These molecules exert their effects by activating specific nucleotide receptors, which are finely regulated by ectonucleotidases that break down their ligands. In this comprehensive review, we aim to elucidate the relevance of extracellular nucleotides as signalling molecules in the context of smooth muscle contraction, considering the modulatory influence of ectonucleotidases on this intricate process. Specifically, we provide a detailed examination of the involvement of extracellular nucleotides in the contraction of non-vascular smooth muscles, including those found in the urinary bladder, the airways, the reproductive system, and the gastrointestinal tract. Furthermore, we present a broader overview of the role of extracellular nucleotides in vascular smooth muscle contraction.
Collapse
Affiliation(s)
- Romuald Brice Babou Kammoe
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada.
| |
Collapse
|
3
|
Cao H, Li L, Liu S, Wang Y, Liu X, Yang F, Dong W. The multifaceted role of extracellular ATP in sperm function: From spermatogenesis to fertilization. Theriogenology 2024; 214:98-106. [PMID: 37865020 DOI: 10.1016/j.theriogenology.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Extracellular adenosine 5'-triphosphate (ATP) is a vital signaling molecule involved in various physiological processes within the body. In recent years, studies have revealed its significant role in male reproduction, particularly in sperm function. This review explores the multifaceted role of extracellular ATP in sperm function, from spermatogenesis to fertilization. We discuss the impact of extracellular ATP on spermatogenesis, sperm maturation and sperm-egg fusion, highlighting the complex regulatory mechanisms and potential clinical applications in the context of male infertility. By examining the latest research, we emphasize the crucial role of extracellular ATP in sperm function and propose future research directions to further.
Collapse
Affiliation(s)
- Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xianglin Liu
- College of Forestry, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Fangxia Yang
- College of Forestry, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
4
|
Li X, Hu J, Yin P, Liu L, Chen Y. Mechanotransduction in the urothelium: ATP signalling and mechanoreceptors. Heliyon 2023; 9:e19427. [PMID: 37674847 PMCID: PMC10477517 DOI: 10.1016/j.heliyon.2023.e19427] [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: 04/23/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
The urothelium, which covers the inner surface of the bladder, is continuously exposed to a complex physical environment where it is stimulated by, and responds to, a wide range of mechanical cues. Mechanically activated ion channels endow the urothelium with functioning in the conversion of mechanical stimuli into biochemical events that influence the surface of the urothelium itself as well as suburothelial tissues, including afferent nerve fibres, interstitial cells of Cajal and detrusor smooth muscle cells, to ensure normal urinary function during the cycle of filling and voiding. However, under prolonged and abnormal loading conditions, the urothelial sensory system can become maladaptive, leading to the development of bladder dysfunction. In this review, we summarize developments in the understanding of urothelial mechanotransduction from two perspectives: first, with regard to the functions of urothelial mechanotransduction, particularly stretch-mediated ATP signalling and the regulation of urothelial surface area; and secondly, with regard to the mechanoreceptors present in the urothelium, primarily transient receptor potential channels and mechanosensitive Piezo channels, and the potential pathophysiological role of these channels in the bladder. A more thorough understanding of urothelial mechanotransduction function may inspire the development of new therapeutic strategies for lower urinary tract diseases.
Collapse
Affiliation(s)
| | | | - Ping Yin
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Lumin Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yuelai Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| |
Collapse
|
5
|
Urinary ATP Levels Are Controlled by Nucleotidases Released from the Urothelium in a Regulated Manner. Metabolites 2022; 13:metabo13010030. [PMID: 36676954 PMCID: PMC9862892 DOI: 10.3390/metabo13010030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Adenosine 5′-triphosphate (ATP) is released in the bladder lumen during filling. Urothelial ATP is presumed to regulate bladder excitability. Urinary ATP is suggested as a urinary biomarker of bladder dysfunctions since ATP is increased in the urine of patients with overactive bladder, interstitial cystitis or bladder pain syndrome. Altered urinary ATP might also be associated with voiding dysfunctions linked to disease states associated with metabolic syndrome. Extracellular ATP levels are determined by ATP release and ATP hydrolysis by membrane-bound and soluble nucleotidases (s-NTDs). It is currently unknown whether s-NTDs regulate urinary ATP. Using etheno-ATP substrate and HPLC-FLD detection techniques, we found that s-NTDs are released in the lumen of ex vivo mouse detrusor-free bladders. Capillary immunoelectrophoresis by ProteinSimple Wes determined that intraluminal solutions (ILS) collected at the end of filling contain ENTPD3 > ENPP1 > ENPP3 ≥ ENTPD2 = NT5E = ALPL/TNAP. Activation of adenylyl cyclase with forskolin increased luminal s-NTDs release whereas the AC inhibitor SQ22536 had no effect. In contrast, forskolin reduced and SQ22536 increased s-NTDs release in the lamina propria. Adenosine enhanced s-NTDs release and accelerated ATP hydrolysis in ILS and lamina propria. Therefore, there is a regulated release of s-NTDs in the bladder lumen during filling. Aberrant release or functions of urothelial s-NTDs might cause elevated urinary ATP in conditions with abnormal bladder excitability.
Collapse
|
6
|
Maynard JP, Sfanos KS. P2 purinergic receptor dysregulation in urologic disease. Purinergic Signal 2022; 18:267-287. [PMID: 35687210 PMCID: PMC9184359 DOI: 10.1007/s11302-022-09875-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022] Open
Abstract
P2 purinergic receptors are involved in the normal function of the kidney, bladder, and prostate via signaling that occurs in response to extracellular nucleotides. Dysregulation of these receptors is common in pathological states and often associated with disease initiation, progression, or aggressiveness. Indeed, P2 purinergic receptor expression is altered across multiple urologic disorders including chronic kidney disease, polycystic kidney disease, interstitial cystitis, urinary incontinence, overactive bladder syndrome, prostatitis, and benign prostatic hyperplasia. P2 purinergic receptors are likewise indirectly associated with these disorders via receptor-mediated inflammation and pain, a common characteristic across most urologic disorders. Furthermore, select P2 purinergic receptors are overexpressed in urologic cancer including renal cell carcinoma, urothelial carcinoma, and prostate adenocarcinoma, and pre-clinical studies depict P2 purinergic receptors as potential therapeutic targets. Herein, we highlight the compelling evidence for the exploration of P2 purinergic receptors as biomarkers and therapeutic targets in urologic cancers and other urologic disease. Likewise, there is currently optimism for P2 purinergic receptor-targeted therapeutics for the treatment of inflammation and pain associated with urologic diseases. Further exploration of the common pathways linking P2 purinergic receptor dysregulation to urologic disease might ultimately help in gaining new mechanistic insight into disease processes and therapeutic targeting.
Collapse
Affiliation(s)
- Janielle P Maynard
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA.
| | - Karen S Sfanos
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Fleck D, Kenzler L, Mundt N, Strauch M, Uesaka N, Moosmann R, Bruentgens F, Missel A, Mayerhofer A, Merhof D, Spehr J, Spehr M. ATP activation of peritubular cells drives testicular sperm transport. eLife 2021; 10:e62885. [PMID: 33502316 PMCID: PMC7840184 DOI: 10.7554/elife.62885] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Spermatogenesis, the complex process of male germ cell proliferation, differentiation, and maturation, is the basis of male fertility. In the seminiferous tubules of the testes, spermatozoa are constantly generated from spermatogonial stem cells through a stereotyped sequence of mitotic and meiotic divisions. The basic physiological principles, however, that control both maturation and luminal transport of the still immotile spermatozoa within the seminiferous tubules remain poorly, if at all, defined. Here, we show that coordinated contractions of smooth muscle-like testicular peritubular cells provide the propulsive force for luminal sperm transport toward the rete testis. Using a mouse model for in vivo imaging, we describe and quantify spontaneous tubular contractions and show a causal relationship between peritubular Ca2+ waves and peristaltic transport. Moreover, we identify P2 receptor-dependent purinergic signaling pathways as physiological triggers of tubular contractions both in vitro and in vivo. When challenged with extracellular ATP, transport of luminal content inside the seminiferous tubules displays stage-dependent directionality. We thus suggest that paracrine purinergic signaling coordinates peristaltic recurrent contractions of the mouse seminiferous tubules to propel immotile spermatozoa to the rete testis.
Collapse
Affiliation(s)
- David Fleck
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Lina Kenzler
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Nadine Mundt
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Research Training Group 2416 MultiSenses – MultiScales, RWTH Aachen UniversityAachenGermany
| | - Martin Strauch
- Institute of Imaging and Computer Vision, RWTH Aachen UniversityAachenGermany
| | - Naofumi Uesaka
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Department of Cognitive Neurobiology, Tokyo Medical and Dental UniversityTokyoJapan
| | - Robert Moosmann
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Felicitas Bruentgens
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Annika Missel
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Artur Mayerhofer
- Biomedical Center Munich (BMC), Cell Biology, Anatomy III, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Dorit Merhof
- Institute of Imaging and Computer Vision, RWTH Aachen UniversityAachenGermany
| | - Jennifer Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
| | - Marc Spehr
- Department of Chemosensation, Institute for Biology II, RWTH Aachen UniversityAachenGermany
- Research Training Group 2416 MultiSenses – MultiScales, RWTH Aachen UniversityAachenGermany
| |
Collapse
|
8
|
Babou Kammoe RB, Kauffenstein G, Pelletier J, Robaye B, Sévigny J. NTPDase1 Modulates Smooth Muscle Contraction in Mice Bladder by Regulating Nucleotide Receptor Activation Distinctly in Male and Female. Biomolecules 2021; 11:biom11020147. [PMID: 33498759 PMCID: PMC7911947 DOI: 10.3390/biom11020147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022] Open
Abstract
Nucleotides released by smooth muscle cells (SMCs) and by innervating nerve terminals activate specific P2 receptors and modulate bladder contraction. We hypothesized that cell surface enzymes regulate SMC contraction in mice bladder by controlling the concentration of nucleotides. We showed by immunohistochemistry, enzymatic histochemistry, and biochemical activities that nucleoside triphosphate diphosphohydrolase-1 (NTPDase1) and ecto-5′-nucleotidase were the major ectonucleotidases expressed by SMCs in the bladder. RT-qPCR revealed that, among the nucleotide receptors, there was higher expression of P2X1, P2Y1, and P2Y6 receptors. Ex vivo, nucleotides induced a more potent contraction of bladder strips isolated from NTPDase1 deficient (Entpd1−/−) mice compared to wild type controls. The strongest responses were obtained with uridine 5′-triphosphate (UTP) and uridine 5′-diphosphate (UDP), suggesting the involvement of P2Y6 receptors, which was confirmed with P2ry6−/− bladder strips. Interestingly, this response was reduced in female bladders. Our results also suggest the participation of P2X1, P2Y2 and/or P2Y4, and P2Y12 in these contractions. A reduced response to the thromboxane analogue U46619 was also observed in wild type, Entpd1−/−, and P2ry6−/− female bladders showing another difference due to sex. In summary, NTPDase1 modulates the activation of nucleotide receptors in mouse bladder SMCs, and contractions induced by P2Y6 receptor activation were weaker in female bladders.
Collapse
Affiliation(s)
- Romuald Brice Babou Kammoe
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada; (R.B.B.K.); (G.K.); (J.P.)
- Département de Microbiologie-Infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Gilles Kauffenstein
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada; (R.B.B.K.); (G.K.); (J.P.)
- Département de Microbiologie-Infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
- UMR INSERM 1260, Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, 67084 Strasbourg, France
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada; (R.B.B.K.); (G.K.); (J.P.)
| | - Bernard Robaye
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, 10 rue Adrienne Bolland, 6041 Gosselies, Belgium;
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada; (R.B.B.K.); (G.K.); (J.P.)
- Département de Microbiologie-Infectiologie et d’immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-525-4444 (ext. 46319); Fax: +1-418-654-2765
| |
Collapse
|
9
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Firouzmand S, Ajori L, Towse J, Allameh F, Najafi S, Javed S, John B, Langley SEM, Fry CH, Young JS. Investigating the associations of mucosal P2Y6 receptor expression and urinary ATP and ADP concentrations, with symptoms of overactive bladder. Neurourol Urodyn 2020; 39:926-934. [PMID: 32049380 DOI: 10.1002/nau.24322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/28/2020] [Indexed: 12/19/2022]
Abstract
AIM To characterize purinergic signaling in overactive bladder (OAB). METHODS Mucosal biopsies were taken by flexible cystoscopy from patients with storage symptoms referred to Urology Departments of collaborating hospitals. Immunohistochemistry (n = 12) and Western blot analysis (n = 28) were used to establish the qualitative and quantitative expression profile of P2Y6 in human mucosa. Participants from the general population provided a mid-stream urine sample. Bioluminescent assays were used to quantify adenosine triphosphate (ATP; n = 66) and adenosine diphosphate (ADP; n = 60) concentrations, which were normalized to creatinine (Cr) concentration. All participants completed a questionnaire (International Consultation on Incontinence Questionnaire - Overactive Bladder) to score urinary symptoms of OAB. RESULTS P2Y6 immunoreactivity, more prominent in the urothelium (colocalized with the uroepithelial marker pan-cytokeratin), was more greatly expressed in OAB compared to age- and sex-matched controls (benign prostatic hyperplasia) without OAB symptoms. Mucosal P2Y6 was positively correlated only with incontinence (P = .009). Both urinary ATP and its hydrolysis product, ADP, an agonist to P2Y6, were positively correlated with total OAB symptom score (P = .010 and P = .042, respectively). CONCLUSIONS The positive correlation of P2Y6 only with incontinence may indicate a different phenotype in OAB wet and warrants further investigation. Positive correlations of ATP and ADP with total OAB symptom score demonstrate upregulation in purinergic signaling in OAB; shown previously only in animal models. Further research is required to validate whether purinoceptors are indeed new therapeutic targets for this highly prevalent symptom complex.
Collapse
Affiliation(s)
- Sepinoud Firouzmand
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Ladan Ajori
- Department of Obstetrics and Gynecology, Preventive Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Urology and Gynecology, Shohada-e-Tajrish Hospital, Tehran, Iran
| | - Jonathan Towse
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Farzad Allameh
- Department of Obstetrics and Gynecology, Preventive Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Urology and Gynecology, Shohada-e-Tajrish Hospital, Tehran, Iran
| | - Saman Najafi
- Department of Obstetrics and Gynecology, Preventive Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saqib Javed
- Stokes Centre for Urology, Royal Surrey County Hospital, Guildford, United Kingdom
| | - Babbin John
- Stokes Centre for Urology, Royal Surrey County Hospital, Guildford, United Kingdom
| | - Stephen E M Langley
- Stokes Centre for Urology, Royal Surrey County Hospital, Guildford, United Kingdom
| | - Christopher H Fry
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, United Kingdom
| | - John S Young
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
11
|
Silva I, Magalhães-Cardoso MT, Ferreirinha F, Moreira S, Costa AF, Silva D, Vieira C, Silva-Ramos M, Correia-de-Sá P. β 3 Adrenoceptor-induced cholinergic inhibition in human and rat urinary bladders involves the exchange protein directly activated by cyclic AMP 1 favoring adenosine release. Br J Pharmacol 2020; 177:1589-1608. [PMID: 31721163 DOI: 10.1111/bph.14921] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE The mechanism by which β3 receptor agonists (e.g. mirabegron) control bladder overactivity may involve adenosine release from human and rat detrusor smooth muscle. Retrograde activation of adenosine A1 receptors reduces ACh release from cholinergic bladder nerves. β3 -Adrenoceptors usually couple to adenylyl cyclase. Here we investigated, which of the cAMP targets, protein kinase A or the exchange protein directly activated by cAMP (EPAC) could be involved in this cholinergic inhibition of the bladder. EXPERIMENTAL APPROACH [3 H]ACh and adenosine release from urothelium-denuded detrusor strips of cadaveric human organ donors and rats were measured by liquid scintillation spectrometry and HPLC, respectively. In vivo cystometry was also performed in urethane-anaesthetized rats. KEY RESULTS The exchange protein directly activated by cAMP (EPAC) inhibitor, ESI-09, prevented mirabegron- and isoprenaline-induced adenosine release from human and rat detrusor strips respectively. ESI-09, but not the PKA inhibitor, H-89, attenuated inhibition of [3 H]ACh release from stimulated (10 Hz) detrusor strips caused by activating β3 -adrenoceptors, AC (forskolin) and EPAC1 (8-CTP-2Me-cAMP). Isoprenaline-induced inhibition of [3 H]ACh release was also prevented by inhibitors of PKC (chelerythrine and Go6976) and of the equilibrative nucleoside transporter 1 (ENT1; dipyridamole and NBTI), but not by PLC inhibition with U73122. Pretreatment with ESI-09, but not with H-89, prevented the reduction of the voiding frequency caused by isoprenaline and forskolin in vivo. CONCLUSION AND IMPLICATIONS Data suggest that β3 -adrenoceptor-induced inhibition of cholinergic neurotransmission in human and rat urinary bladders involves activation of an EPAC1/PKC pathway downstream cAMP production resulting in adenosine outflow via ENT1.
Collapse
Affiliation(s)
- Isabel Silva
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - M Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Sílvia Moreira
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana Filipa Costa
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Diogo Silva
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cátia Vieira
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Miguel Silva-Ramos
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
12
|
Silva-Ramos M, Silva I, Faria M, Ferreirinha F, Correia-de-Sá P. Activation of Prejunctional P2x2/3 Heterotrimers by ATP Enhances the Cholinergic Tone in Obstructed Human Urinary Bladders. J Pharmacol Exp Ther 2019; 372:63-72. [PMID: 31636173 DOI: 10.1124/jpet.119.261610] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
The objective of this study was to investigate the role of ATP in cholinergic neurotransmission in the urinary bladder of control men and of patients obstructed as a result of benign prostatic hyperplasia (BPH). Human detrusor samples were collected from 41 patients who submitted to transvesical prostatectomy resulting from BPH and 26 male organ donors. The release of [3H]acetylcholine ([3H]ACh) was evoked by electrical field stimulation (10 Hz, 200 pulses) in urothelium-denuded detrusor strips. Myographic recordings were performed to test detrusor strip sensitivity to ACh and ATP. Nerve-evoked [3H]ACh release was 1.5-fold higher in detrusor strips from BPH patients compared with controls. This difference was abolished after desensitization of ionotropic P2X1-3 receptors with an ATP analog, α,β-methylene ATP (30 μM, applied for 15 minutes). TNP-ATP (10 nM, a preferential P2X2/3 antagonist) and A317491 (100 nM, a selective P2X3 antagonist) were about equipotent in decreasing nerve-evoked [3H]ACh release in control detrusor strips, but the selective P2X1 receptor antagonist NF023 (3 μM) was devoid of effect. The inhibitory effect of TNP-ATP (10 nM) increased from 27% ± 9% to 43% ± 6% in detrusor strips of BPH patients, but the effect of A317491 (100 nM) [3H]ACh release unaltered (20% ± 2% vs. 24% ± 4%). The amplitude of ACh (0.1-100 μM)-induced myographic recordings decreased, whereas sensitivity to ATP (0.01-3 mM) increased in detrusor strips from BPH patients. Besides the well characterized P2X1 receptor-mediated contractile activity of ATP in pathologic human bladders, we show here for the first time that cholinergic hyperactivity in the detrusor of BPH patients is facilitated by activation of ATP-sensitive P2X2/3 heterotrimers. SIGNIFICANCE STATEMENT: Bladder outlet obstruction often leads to detrusor overactivity and reduced bladder compliance in parallel to atropine-resistant increased purinergic tone. Our data show that P2X1 purinoceptors are overexpressed in the detrusor of patients with benign prostatic hyperplasia. Besides the P2X1 receptor-mediated detrusor contractions, ATP favors nerve-evoked acetylcholine release via the activation of prejunctional P2X2/3 excitatory receptors in these patients Thus, our hypothesis is that manipulation of the purinergic tone may be therapeutically useful to counteract cholinergic overstimulation in obstructed patients.
Collapse
Affiliation(s)
- M Silva-Ramos
- Laboratório de Farmacologia e Neurobiologia and Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal (M.S.-R., I.S., M.F., F.F., P.C.-S.); and Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal (M.S.-R.)
| | - I Silva
- Laboratório de Farmacologia e Neurobiologia and Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal (M.S.-R., I.S., M.F., F.F., P.C.-S.); and Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal (M.S.-R.)
| | - M Faria
- Laboratório de Farmacologia e Neurobiologia and Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal (M.S.-R., I.S., M.F., F.F., P.C.-S.); and Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal (M.S.-R.)
| | - F Ferreirinha
- Laboratório de Farmacologia e Neurobiologia and Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal (M.S.-R., I.S., M.F., F.F., P.C.-S.); and Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal (M.S.-R.)
| | - P Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia and Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal (M.S.-R., I.S., M.F., F.F., P.C.-S.); and Serviço de Urologia, Centro Hospitalar Universitário do Porto (CHUP), Porto, Portugal (M.S.-R.)
| |
Collapse
|
13
|
Taidi Z, Mansfield KJ, Bates L, Sana-Ur-Rehman H, Liu L. Purinergic P2X7 receptors as therapeutic targets in interstitial cystitis/bladder pain syndrome; key role of ATP signaling in inflammation. Bladder (San Franc) 2019; 6:e38. [PMID: 32775480 PMCID: PMC7401983 DOI: 10.14440/bladder.2019.789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/06/2018] [Accepted: 12/17/2018] [Indexed: 12/23/2022] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic lower urinary tract condition. Patients with IC/BPS suffer from debilitating pain and urinary urgency. The underlying etiology of IC/BPS is unknown and as such current treatments are mostly symptomatic with no real cure. Many theories have been proposed to describe the etiology of IC/BPS, but this review focuses on the role of inflammation. In IC/BPS patients, the permeability of the urothelium barrier is compromised and inflammatory cells infiltrate the bladder wall. There are increased levels of many inflammatory mediators in patients with IC/BPS and symptoms such as pain and urgency that have been associated with the degree of inflammation. Recent evidence has highlighted the role of purinergic receptors, specifically the P2X7 receptor, in the process of inflammation. The results from studies in animals including cyclophosphamide-induced hemorrhagic cystitis strongly support the role of P2X7 receptors in inflammation. Furthermore, the deletion of the P2X7 receptor or antagonism of this receptor significantly reduces inflammatory mediator release from the bladder and improves symptoms. Research results from IC/BPS patients and animal models of IC/BPS strongly support the crucial role of inflammation in the pathophysiology of this painful disease. Purinergic signaling and purinergic receptors, especially the P2X7 receptor, play an undisputed role in inflammation. Purinergic receptor antagonists show positive results in treating different symptoms of IC/BPS.
Collapse
Affiliation(s)
- Zhinoos Taidi
- School of Medical Sciences, The University of New South Wales, Sydney NSW 2052, Australia
| | - Kylie J Mansfield
- School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Lucy Bates
- Westmead Hospital, Westmead, NSW 2145, Australia
| | - Hafiz Sana-Ur-Rehman
- School of Medical Sciences, The University of New South Wales, Sydney NSW 2052, Australia
| | - Lu Liu
- School of Medical Sciences, The University of New South Wales, Sydney NSW 2052, Australia
| |
Collapse
|
14
|
Modulation of lower urinary tract smooth muscle contraction and relaxation by the urothelium. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:675-694. [DOI: 10.1007/s00210-018-1510-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/08/2018] [Indexed: 10/14/2022]
|
15
|
Guan NN, Sharma N, Hallén-Grufman K, Jager EWH, Svennersten K. The role of ATP signalling in response to mechanical stimulation studied in T24 cells using new microphysiological tools. J Cell Mol Med 2018; 22:2319-2328. [PMID: 29392898 PMCID: PMC5867107 DOI: 10.1111/jcmm.13520] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 12/01/2017] [Indexed: 01/06/2023] Open
Abstract
The capacity to store urine and initiate voiding is a valued characteristic of the human urinary bladder. To maintain this feature, it is necessary that the bladder can sense when it is full and when it is time to void. The bladder has a specialized epithelium called urothelium that is believed to be important for its sensory function. It has been suggested that autocrine ATP signalling contributes to this sensory function of the urothelium. There is well‐established evidence that ATP is released via vesicular exocytosis as well as by pannexin hemichannels upon mechanical stimulation. However, there are still many details that need elucidation and therefore there is a need for the development of new tools to further explore this fascinating field. In this work, we use new microphysiological systems to study mechanostimulation at a cellular level: a mechanostimulation microchip and a silicone‐based cell stretcher. Using these tools, we show that ATP is released upon cell stretching and that extracellular ATP contributes to a major part of Ca2+ signalling induced by stretching in T24 cells. These results contribute to the increasing body of evidence for ATP signalling as an important component for the sensory function of urothelial cells. This encourages the development of drugs targeting P2 receptors to relieve suffering from overactive bladder disorder and incontinence.
Collapse
Affiliation(s)
- Na N Guan
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institutet, Stockholm, Sweden.,Department of Urology, Karolinska University Hospital, Stockholm, Sweden.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nimish Sharma
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institutet, Stockholm, Sweden.,Department of Urology, Karolinska University Hospital, Stockholm, Sweden
| | - Katarina Hallén-Grufman
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institutet, Stockholm, Sweden.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Edwin W H Jager
- Department of Physics, Chemistry and Biology (IFM), Linköping University, Linköping, Sweden
| | - Karl Svennersten
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institutet, Stockholm, Sweden.,Department of Urology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Vieira C, Ferreirinha F, Magalhães-Cardoso MT, Silva I, Marques P, Correia-de-Sá P. Post-inflammatory Ileitis Induces Non-neuronal Purinergic Signaling Adjustments of Cholinergic Neurotransmission in the Myenteric Plexus. Front Pharmacol 2017; 8:811. [PMID: 29167643 PMCID: PMC5682326 DOI: 10.3389/fphar.2017.00811] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/26/2017] [Indexed: 12/11/2022] Open
Abstract
Uncoupling between ATP overflow and extracellular adenosine formation changes purinergic signaling in post-inflammatory ileitis. Adenosine neuromodulation deficits were ascribed to feed-forward inhibition of ecto-5′-nucleotidase/CD73 by high extracellular adenine nucleotides in the inflamed ileum. Here, we hypothesized that inflammation-induced changes in cellular density may also account to unbalance the release of purines and their influence on [3H]acetylcholine release from longitudinal muscle-myenteric plexus preparations of the ileum of 2,4,6-trinitrobenzenesulfonic acid (TNBS)-treated rats. The population of S100β-positive glial cells increase, whereas Ano-1-positive interstitial cells of Cajal (ICCs) diminished, in the ileum 7-days after the inflammatory insult. In the absence of changes in the density of VAChT-positive cholinergic nerves detected by immunofluorescence confocal microscopy, the inflamed myenteric plexus released smaller amounts of [3H]acetylcholine which also became less sensitive to neuronal blockade by tetrodotoxin (1 μM). Instead, [3H]acetylcholine release was attenuated by sodium fluoroacetate (5 mM), carbenoxolone (10 μM) and A438079 (3 μM), which prevent activation of glial cells, pannexin-1 hemichannels and P2X7 receptors, respectively. Sodium fluoroacetate also decreased ATP overflow without significantly affecting the extracellular adenosine levels, thus indicating that surplus ATP release parallels reactive gliosis in post-inflammatory ileitis. Conversely, loss of ICCs may explain the lower amounts of adenosine detected in TNBS-treated preparations, since blockade of Cav3 (T-type) channels existing in ICCs with mibefradil (3 μM) or inhibition of the equilibrative nucleoside transporter 1 with dipyridamole (0.5 μM), both decreased extracellular adenosine. Data indicate that post-inflammatory ileitis operates a shift on purinergic neuromodulation reflecting the upregulation of ATP-releasing enteric glial cells and the depletion of ICCs accounting for decreased adenosine overflow via equilibrative nucleoside transporters.
Collapse
Affiliation(s)
- Cátia Vieira
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria T Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Isabel Silva
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Patrícia Marques
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
17
|
Silva I, Costa AF, Moreira S, Ferreirinha F, Magalhães-Cardoso MT, Calejo I, Silva-Ramos M, Correia-de-Sá P. Inhibition of cholinergic neurotransmission by β 3-adrenoceptors depends on adenosine release and A 1-receptor activation in human and rat urinary bladders. Am J Physiol Renal Physiol 2017; 313:F388-F403. [PMID: 28446460 DOI: 10.1152/ajprenal.00392.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 12/12/2022] Open
Abstract
The direct detrusor relaxant effect of β3-adrenoceptor agonists as a primary mechanism to improve overactive bladder symptoms has been questioned. Among other targets, activation of β3-adrenoceptors downmodulate nerve-evoked acetylcholine (ACh) release, but there is insufficient evidence for the presence of these receptors on bladder cholinergic nerve terminals. Our hypothesis is that adenosine formed from the catabolism of cyclic AMP in the detrusor may act as a retrograde messenger via prejunctional A1 receptors to explain inhibition of cholinergic activity by β3-adrenoceptors. Isoprenaline (1 µM) decreased [3H]ACh release from stimulated (10 Hz, 200 pulses) human (-47 ± 5%) and rat (-38 ± 1%) detrusor strips. Mirabegron (0.1 µM, -53 ± 8%) and CL316,243 (1 µM, -37 ± 7%) mimicked isoprenaline (1 µM) inhibition, and their effects were prevented by blocking β3-adrenoceptors with L748,337 (30 nM) and SR59230A (100 nM), respectively, in human and rat detrusor. Mirabegron and isoprenaline increased extracellular adenosine in the detrusor. Blockage of A1 receptors with 1,3-dipropyl-8-cyclopentylxanthine (DPCPX, 100 nM) or the equilibrative nucleoside transporters (ENT) with dipyridamole (0.5 µM) prevented mirabegron and isoprenaline inhibitory effects. Dipyridamole prevented isoprenaline-induced adenosine outflow from the rat detrusor, and this effect was mimicked by the ENT1 inhibitor, S-(4-nitrobenzyl)-6-thioinosine (NBTI, 30 µM). Cystometry recordings in anesthetized rats demonstrated that SR59230A, DPCPX, dipyridamole, and NBTI reversed the decrease in the voiding frequency caused by isoprenaline (0.1-1,000 nM). Data suggest that inhibition of cholinergic neurotransmission by β3-adrenoceptors results from adenosine release via equilibrative nucleoside transporters and prejunctional A1-receptor stimulation in human and rat urinary bladder.
Collapse
Affiliation(s)
- Isabel Silva
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| | - Ana Filipa Costa
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| | - Sílvia Moreira
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| | - Maria Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| | - Isabel Calejo
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| | - Miguel Silva-Ramos
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal.,Serviço de Urologia, Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Universidade do Porto, Porto, Portugal; .,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal; and
| |
Collapse
|
18
|
P2Y 6-deficiency increases micturition frequency and attenuates sustained contractility of the urinary bladder in mice. Sci Rep 2017; 7:771. [PMID: 28396595 PMCID: PMC5429706 DOI: 10.1038/s41598-017-00824-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/14/2017] [Indexed: 12/30/2022] Open
Abstract
The role of the P2Y6 receptor in bladder function has recently attracted a great deal of attention in lower urinary tract research. We conducted this study to determine contributions of the P2Y6 receptor in lower urinary tract function of normal phenotypes by comparing P2Y6-deficient mice and wild-type mice. In in vivo experiments, P2Y6-deficient mice had more frequent micturition with smaller bladder capacity compared to wild-type mice; however, there was no difference between these groups in bladder-filling pressure/volume relationships during cystometry under decerebrate, unanaesthetized conditions. Analysis of in vivo bladder contraction revealed significant difference between the 2 groups, with P2Y6-deficient mice presenting markedly shorter bladder contraction duration but no difference in peak contraction pressure. However, analysis of in vitro experiments showed no P2Y6 involvements in contraction and relaxation of bladder muscle strips and in ATP release by mechanical stimulation of primary-cultured urothelial cells. These results suggest that the P2Y6 receptor in the central nervous system, dorsal root ganglion, or both is involved in inhibition of bladder afferent signalling or sensitivity in the pontine micturition centre and that the receptor in the detrusor may be implicated in facilitation to sustain bladder contraction force.
Collapse
|
19
|
Chen BY, Chen D, Lyu JX, Li KQ, Jiang MM, Zeng JJ, He XJ, Hao K, Tao HQ, Mou XZ, Ying YM, Zhang W, Zhu MH, Wang Z. Marsdeniae tenacissimae extract (MTE) suppresses cell proliferation by attenuating VEGF/VEGFR2 interactions and promotes apoptosis through regulating PKC pathway in human umbilical vein endothelial cells. Chin J Nat Med 2017; 14:922-930. [PMID: 28262119 DOI: 10.1016/s1875-5364(17)30017-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Indexed: 01/13/2023]
Abstract
Marsdeniae tenacissimae extract (MTE), commonly known as Xiao-Ai-Ping in China, is a traditional Chinese herb medicine capable of inhibiting proliferation and metastasis and boosting apoptosis in various cancer cells. However, little is known about the contribution of MTE towards tumor angiogenesis and the underlying mechanism. The present study aimed to evaluate the effects of MTE on the proliferation and apoptosis of human umbilical vein endothelial cells (HUVECs) and the molecular mechanism. 3-(4,5-dimethylthiazol-2-yl)-5(3-carboxymethoxyphenyl)-2-(4-sulfopheny)-2H-tetrazolium, inner salt (MTS) and PI-stained flow cytometry assays revealed that MTE dose-dependently reduced the proliferation of HUVECs by arresting cell cycle at S phase (P < 0.05). Annexin V-FITC/PI-stained flow cytometry confirmed that MTE (160 μL·L-1) enhanced the apoptosis of HUVECs significantly (P < 0.001). Real-time quantitative RT-PCR and Western blot analyses showed an increase in Bax expression and a sharply decline in Bcl-2 expression; caspase-3 was activated simultaneously in a dose-dependent manner (P < 0.05). Further study observed the dose-dependent down-regulation of vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2), P2Y6 receptor (P2Y6R), and chemokine (C-C motif) ligand 2 (CCL-2), along with the activation of PKC Δ and up-regulation of p53 in a dose-dependent manner in MTE-treated selected cells (P < 0.05). Collectively, the results from the present study suggested that MTE suppressed the proliferation by attenuating CCL-2-mediated VEGF/VEGFR2 interactions and promoted the apoptosis through PKCΔ-induced p53-dependent mitochondrial pathway in HUVECs, supporting that MTE may be developed as a potent anti-cancer medicine.
Collapse
Affiliation(s)
- Bing-Yu Chen
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Dong Chen
- Wenzhou Center for Disease Control and Prevention, Wenzhou 325001, China
| | - Jian-Xin Lyu
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Kai-Qiang Li
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Meng-Meng Jiang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Jing-Jing Zeng
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Xu-Jun He
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Ke Hao
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Hou-Quan Tao
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - You-Min Ying
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wei Zhang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Meng-Hua Zhu
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China.
| | - Zhen Wang
- Research Center of Blood Transfusion Medicine, Education Ministry Key Laboratory of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, 310014, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
20
|
Silva-Ramos M, Silva I, Faria M, Magalhães-Cardoso MT, Correia J, Ferreirinha F, Correia-de-Sá P. Impairment of ATP hydrolysis decreases adenosine A1 receptor tonus favoring cholinergic nerve hyperactivity in the obstructed human urinary bladder. Purinergic Signal 2015; 11:595-606. [PMID: 26521170 DOI: 10.1007/s11302-015-9478-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022] Open
Abstract
This study was designed to investigate whether reduced adenosine formation linked to deficits in extracellular ATP hydrolysis by NTPDases contributes to detrusor neuromodulatory changes associated with bladder outlet obstruction in men with benign prostatic hyperplasia (BPH). The kinetics of ATP catabolism and adenosine formation as well as the role of P1 receptor agonists on muscle tension and nerve-evoked [(3)H]ACh release were evaluated in mucosal-denuded detrusor strips from BPH patients (n = 31) and control organ donors (n = 23). The neurogenic release of ATP and [(3)H]ACh was higher (P < 0.05) in detrusor strips from BPH patients. The extracellular hydrolysis of ATP and, subsequent, adenosine formation was slower (t (1/2) 73 vs. 36 min, P < 0.05) in BPH detrusor strips. The A(1) receptor-mediated inhibition of evoked [(3)H]ACh release by adenosine (100 μM), NECA (1 μM), and R-PIA (0.3 μM) was enhanced in BPH bladders. Relaxation of detrusor contractions induced by acetylcholine required 30-fold higher concentrations of adenosine. Despite VAChT-positive cholinergic nerves exhibiting higher A(1) immunoreactivity in BPH bladders, the endogenous adenosine tonus revealed by adenosine deaminase is missing. Restoration of A1 inhibition was achieved by favoring (1) ATP hydrolysis with apyrase (2 U mL(-1)) or (2) extracellular adenosine accumulation with dipyridamole or EHNA, as these drugs inhibit adenosine uptake and deamination, respectively. In conclusion, reduced ATP hydrolysis leads to deficient adenosine formation and A(1) receptor-mediated inhibition of cholinergic nerve activity in the obstructed human bladder. Thus, we propose that pharmacological manipulation of endogenous adenosine levels and/or A(1) receptor activation might be useful to control bladder overactivity in BPH patients.
Collapse
Affiliation(s)
- M Silva-Ramos
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,Serviço de Urologia, Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - I Silva
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - M Faria
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - M T Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - J Correia
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - F Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - P Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal. .,Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
21
|
Calcium signaling and the novel anti-proliferative effect of the UTP-sensitive P2Y11 receptor in rat cardiac myofibroblasts. Cell Calcium 2015; 58:518-33. [PMID: 26324417 DOI: 10.1016/j.ceca.2015.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/06/2015] [Accepted: 08/14/2015] [Indexed: 12/16/2022]
Abstract
During myocardial ischemia and reperfusion both purines and pyrimidines are released into the extracellular milieu, thus creating a signaling wave that propagates to neighboring cells via membrane-bound P2 purinoceptors activation. Cardiac fibroblasts (CF) are important players in heart remodeling, electrophysiological changes and hemodynamic alterations following myocardial infarction. Here, we investigated the role UTP on calcium signaling and proliferation of CF cultured from ventricles of adult rats. Co-expression of discoidin domain receptor 2 and α-smooth muscle actin indicate that cultured CF are activated myofibroblasts. Intracellular calcium ([Ca(2+)]i) signals were monitored in cells loaded with Fluo-4 NW. CF proliferation was evaluated by the MTT assay. UTP and the selective P2Y4 agonist, MRS4062, caused a fast desensitizing [Ca(2+)]i rise originated from thapsigargin-sensitive internal stores, which partially declined to a plateau providing the existence of Ca(2+) in the extracellular fluid. The biphasic [Ca(2+)]i response to UTP was attenuated respectively by P2Y4 blockers, like reactive blue-2 and suramin, and by the P2Y11 antagonist, NF340. UTP and the P2Y2 receptor agonist MRS2768 increased, whereas the selective P2Y11 agonist NF546 decreased, CF growth; MRS4062 was ineffective. Blockage of the P2Y11 receptor or its coupling to adenylate cyclase boosted UTP-induced CF proliferation. Confocal microscopy and Western blot analysis confirmed the presence of P2Y2, P2Y4 and P2Y11 receptors. Data indicate that besides P2Y4 and P2Y2 receptors which are responsible for UTP-induced [Ca(2+)]i transients and growth of CF, respectively, synchronous activation of the previously unrecognized P2Y11 receptor may represent an important target for anti-fibrotic intervention in cardiac remodeling.
Collapse
|
22
|
Silva I, Ferreirinha F, Magalhães-Cardoso MT, Silva-Ramos M, Correia-de-Sá P. Activation of P2Y6 Receptors Facilitates Nonneuronal Adenosine Triphosphate and Acetylcholine Release from Urothelium with the Lamina Propria of Men with Bladder Outlet Obstruction. J Urol 2015; 194:1146-54. [PMID: 26004864 DOI: 10.1016/j.juro.2015.05.080] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2015] [Indexed: 12/22/2022]
Abstract
PURPOSE Deregulation of purinergic bladder signaling may contribute to persistent detrusor overactivity in patients with bladder outlet obstruction. Activation of uridine diphosphate sensitive P2Y6 receptors increases voiding frequency in rats indirectly by releasing adenosine triphosphate from the urothelium. To our knowledge this mechanism has never been tested in the human bladder. MATERIALS AND METHODS We examined the role of the uridine diphosphate sensitive P2Y6 receptor on tetrodotoxin insensitive nonneuronal adenosine triphosphate and [(3)H]acetylcholine release from the human urothelium with the lamina propria of control organ donors and patients with benign prostatic hyperplasia. RESULTS The adenosine triphosphate-to-[(3)H]acetylcholine ratio was fivefold higher in mucosal urothelium/lamina propria strips from benign prostatic hyperplasia patients than control men. The selective P2Y6 receptor agonist PSB0474 (100 nM) augmented by a similar amount adenosine triphosphate and [(3)H]acetylcholine release from mucosal urothelium/lamina propria strips from both groups of individuals. The facilitatory effect of PSB0474 was prevented by MRS2578 (50 nM) and by carbenoxolone (10 μM), which block P2Y6 receptor and pannexin-1 hemichannels, respectively. Blockade of P2X3 (and/or P2X2/3) receptors with A317491 (100 nM) also attenuated release facilitation by PSB0474 in control men but not in patients with benign prostatic hyperplasia. Immunolocalization studies showed that P2Y6, P2X2 and P2X3 receptors were present in choline acetyltransferase positive urothelial cells. In contrast to P2Y6 staining, choline acetyltransferase, P2X2 and P2X3 immunoreactivity decreased in the urothelium of benign prostatic hyperplasia patients. CONCLUSIONS Activation of P2Y6 receptor amplifies mucosal adenosine triphosphate release underlying bladder overactivity in patients with benign prostatic hyperplasia. Therefore, we propose selective P2Y6 receptor blockade as a novel therapeutic strategy to control persistent storage symptoms in obstructed patients.
Collapse
Affiliation(s)
- Isabel Silva
- Laboratório de Farmacologia e Neurobiologia, Porto, Portugal; Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Porto, Portugal; Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Maria Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Porto, Portugal; Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Miguel Silva-Ramos
- Laboratório de Farmacologia e Neurobiologia, Porto, Portugal; Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto and Serviço de Urologia, Centro Hospitalar do Porto, Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Porto, Portugal; Center for Drug Discovery and Innovative Medicines, Porto, Portugal.
| |
Collapse
|
23
|
Cardoso AM, Schetinger MRC, Correia-de-Sá P, Sévigny J. Impact of ectonucleotidases in autonomic nervous functions. Auton Neurosci 2015; 191:25-38. [PMID: 26008223 DOI: 10.1016/j.autneu.2015.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 01/05/2023]
Abstract
Adenine and uracil nucleotides play key functions in the autonomic nervous system (ANS). For instance, ATP acts as a neurotransmitter, co-transmitter and neuromodulator in the ANS. The purinergic system encompasses (1) receptors that respond to extracellular purines, which are designated as P1 and P2 purinoceptors, (2) purine release and uptake, and (3) a cascade of enzymes that regulate the concentration of purines near the cell surface. Ectonucleotidases and adenosine deaminase (ADA) are enzymes responsible for the hydrolysis of ATP (and other nucleotides such as ADP, UTP, UDP, AMP) and adenosine, respectively. Accordingly, these enzymes are expected to play an important role in the control of neuro-effector transmission in tissues innervated by both the sympathetic and parasympathetic divisions of the ANS. Indeed, ectonucleotidases have the ability to either terminate P2 receptor responses initiated by nucleoside triphosphates (ATP and UTP), and/or to favor the activation of ADP (e.g. P2Y1,12,13) and UDP (e.g. P2Y6) and/or adenosine (P1) specific receptors. In addition, ectonucleotidases can also importantly protect some P2 receptors from desensitization (e.g. P2X1, P2Y1). In this review, we present the (putative) roles of ectonucleotidases and ADA in the ANS with a focus on their regulatory activity at neuro-effector junctions in the following tissues: heart, vas deferens, urinary bladder, salivary glands, blood vessels and the intestine. We also present their implication in nociceptive transmission.
Collapse
Affiliation(s)
- Andréia Machado Cardoso
- Post-Graduation Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology of the Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria Rio Grande do Sul, Brazil; Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec G1V 0A6, Canada; Centre de Recherche du CHU de Québec - Université Laval, Québec G1V 4G2, Canada.
| | - Maria Rosa Chitolina Schetinger
- Post-Graduation Program in Toxicological Biochemistry, Department of Biochemistry and Molecular Biology of the Center of Natural and Exact Sciences of the Federal University of Santa Maria, Santa Maria Rio Grande do Sul, Brazil
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, MedInUP, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), 4050-313 Porto, Portugal
| | - Jean Sévigny
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec G1V 0A6, Canada; Centre de Recherche du CHU de Québec - Université Laval, Québec G1V 4G2, Canada.
| |
Collapse
|
24
|
Abstract
It is well established that in most species, exocytotic vesicular release of ATP from parasympathetic neurons contributes to contraction of the bladder. However, ATP is released not only from parasympathetic nerves, but also from the urothelium. During bladder filling, the urothelium is stretched and ATP is released from the umbrella cells thereby activating mechanotransduction pathways. ATP release can also be induced by various mediators present in the urine and and/or released from nerves or other components of the lamina propria. Urothelial release of ATP is mainly attributable to vesicular transport or exocytosis and, to a smaller extent, to pannexin hemichannel conductive efflux. After release, ATP acts on P2X3 and P2X2/3 receptors on suburothelial sensory nerves to initiate the voiding reflex and to mediate the sensation of bladder filling and urgency. ATP also acts on suburothelial interstitial cells/myofibroblasts generating an inward Ca(2+) transient that via gap junctions could provide a mechanism for long-distance spread of signals from the urothelium to the detrusor muscle. ATP release can be affected by urological diseases, e.g., interstitial cystitis and both the mechanisms of release and the receptors activated by ATP may be targets for future drugs for treatment of lower urinary tract disorders.
Collapse
|
25
|
Moreira-Rodrigues M, Graça AL, Ferreira M, Afonso J, Serrão P, Morato M, Ferreirinha F, Correia-de-Sá P, Ebert SN, Moura D. Attenuated aortic vasodilation and sympathetic prejunctional facilitation in epinephrine-deficient mice: selective impairment of β2-adrenoceptor responses. J Pharmacol Exp Ther 2014; 351:243-9. [PMID: 25161169 DOI: 10.1124/jpet.114.217281] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It has been suggested that there is a link between epinephrine synthesis and the development of β2-adrenoceptor-mediated effects, but it remains to be determined whether this development is triggered by epinephrine. The aim of this study was to characterize β-adrenoceptor-mediated relaxation and facilitation of norepinephrine release in the aorta of phenylethanolamine-N-methyltransferase-knockout (Pnmt-KO) mice. Catecholamines were quantified by reverse-phase high-performance liquid chromatography-electrochemical detection. Aortic rings were mounted in a myograph to determine concentration-response curves to selective β1- or β2-adrenoceptor agonists in the absence or presence of selective β1- or β2-adrenoceptor antagonists. Aortic rings were also preincubated with [(3)H]norepinephrine to measure tritium overflow elicited by electrical stimulation in the presence of increasing concentrations of nonselective β- or selective β2-adrenoceptor agonists. β2-Adrenoceptor protein density was evaluated by Western blotting and β2-adrenoceptor localization by immunohistochemistry. Epinephrine is absent in Pnmt-KO mice. The potency and the maximal effect of the β2-adrenoceptor agonist terbutaline were lower in Pnmt-KO than in wild-type (WT) mice. The selective β2-adrenoceptor antagonist ICI 118,551 [(±)-erythro-(S*,S*)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol hydrochloride] antagonized the relaxation caused by terbutaline in WT but not in Pnmt-KO mice. Isoproterenol and terbutaline induced concentration-dependent increases in tritium overflow in WT mice only. β2-Adrenoceptor protein density was decreased in membrane aorta homogenates of Pnmt-KO mice, and this finding was supported by immunofluorescence confocal microscopy. In conclusion, epinephrine is crucial for β2-adrenoceptor-mediated vasodilation and facilitation of norepinephrine release. In the absence of epinephrine, β2-adrenoceptor protein density was decreased in aorta cell membranes, thus potentially hindering its functional activity.
Collapse
Affiliation(s)
- Mónica Moreira-Rodrigues
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.).
| | - Ana L Graça
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Marlene Ferreira
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.).
| | - Joana Afonso
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Paula Serrão
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Manuela Morato
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Fátima Ferreirinha
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Paulo Correia-de-Sá
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Steven N Ebert
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| | - Daniel Moura
- Laboratory of General Physiology (M.M.-R.) and Laboratory of Pharmacology and Neurobiology (F.F., P.C.), Unit for Multidisciplinary Investigation in Biomedicine, Institute of Biomedical Sciences Abel Salazar, University of Porto; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto (A.L.G., M.F., J.A., P.S., D.M.); Neuropharmacology, Institute of Molecular and Cellular Biology, University of Porto (M.M., D.M.); Center for Drug Discovery and Innovative Medicines, University of Porto (M.M.-R., A.L.G., M.F., J.A., P.S., M.M., F.F., P.C., D.M.); Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto and Rede de Química e Tecnologia (REQUIMTE), Porto, Portugal (M.M.); and Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida (S.N.E.)
| |
Collapse
|