1
|
Díaz-García E, García-Sánchez A, Alfaro E, López-Fernández C, Mañas E, Casitas R, Vega S, Cano-Pumarega I, García-Río F, Cubillos-Zapata C. Dysregulation in CD39/CD73 Axis May Trigger the Upsurge of the Immune Suppressive Agent Adenosine in OSA Patients. Arch Bronconeumol 2024; 60:207-214. [PMID: 38485582 DOI: 10.1016/j.arbres.2024.02.013] [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: 01/11/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Although higher incidence of cancer represents a major burden for obstructive sleep apnea (OSA) patients, the molecular pathways driving this association are not completely understood. Interestingly, adenosinergic signaling has emerged as a powerful immune checkpoint driving tumor development and progression. METHODS Here, we explored the expression of the adenosinergic ecto-enzymes CD39 and CD73 in T-lymphocytes of OSA patients without any evidence of cancer, as well as their soluble forms in plasma (sCD39 and sCD73), along with adenosine. In addition, we explored the role of intermittent hypoxia (IH) in this context by in vitro models. RESULTS Our results showed that CD39 is upregulated while CD73 is downregulated in OSA T-cells' membrane. Moreover, our findings suggest that IH, through HIF-1, mediates the upregulation of both CD39 and CD73; and that CD73 downregulation could be mediated by a higher release of sCD73 by OSA T-lymphocytes. Importantly, we found that both sCD39 and sCD73 are upregulated in OSA plasma, suggesting T-lymphocytes as a potential source for plasmatic sCD73. Finally, our data propose the alterations in CD39/CD73 axis could underlie the upsurge of adenosine levels in the plasma of OSA patients. CONCLUSION Our study reveals a hypoxia-mediated alteration of the CD39/CD73 axis in OSA patients, which could trigger ADO upregulation, thus potentially contributing to the immune suppressive environment and ultimately facilitating tumor development and progression. Therefore, our data highlights the need for new longitudinal studies evaluating CD39 and/or CD73 as potential cancer-risk prognostic biomarkers in OSA patients.
Collapse
Affiliation(s)
- Elena Díaz-García
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Aldara García-Sánchez
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Servicio de Neumología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Enrique Alfaro
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain; Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | - Cristina López-Fernández
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Eva Mañas
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Raquel Casitas
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Sara Vega
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | | | - Francisco García-Río
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain; Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain.
| | - Carolina Cubillos-Zapata
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain; Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, Madrid, Spain.
| |
Collapse
|
2
|
Aresta Branco MSL, Gutierrez Cruz A, Peri LE, Mutafova-Yambolieva VN. The Pannexin 1 Channel and the P2X7 Receptor Are in Complex Interplay to Regulate the Release of Soluble Ectonucleotidases in the Murine Bladder Lamina Propria. Int J Mol Sci 2023; 24:9964. [PMID: 37373111 PMCID: PMC10298213 DOI: 10.3390/ijms24129964] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The bladder urothelium releases ATP into the lamina propria (LP) during filling, which can activate P2X receptors on afferent neurons and trigger the micturition reflex. Effective ATP concentrations are largely dependent on metabolism by membrane-bound and soluble ectonucleotidases (s-ENTDs), and the latter are released in the LP in a mechanosensitive manner. Pannexin 1 (PANX1) channel and P2X7 receptor (P2X7R) participate in urothelial ATP release and are physically and functionally coupled, hence we investigated whether they modulate s-ENTDs release. Using ultrasensitive HPLC-FLD, we evaluated the degradation of 1,N6-etheno-ATP (eATP, substrate) to eADP, eAMP, and e-adenosine (e-ADO) in extraluminal solutions that were in contact with the LP of mouse detrusor-free bladders during filling prior to substrate addition, as an indirect measure of s-ENDTS release. Deletion of Panx1 increased the distention-induced, but not the spontaneous, release of s-ENTDs, whereas activation of P2X7R by BzATP or high concentration of ATP in WT bladders increased both. In Panx1-/- bladders or WT bladders treated with the PANX1 inhibitory peptide 10Panx, however, BzATP had no effect on s-ENTDS release, suggesting that P2X7R activity depends on PANX1 channel opening. We concluded, therefore, that P2X7R and PANX1 are in complex interaction to regulate s-ENTDs release and maintain suitable ATP concentrations in the LP. Thus, while stretch-activated PANX1 hinders s-ENTDS release possibly to preserve effective ATP concentration at the end of bladder filling, P2X7R activation, presumably in cystitis, would facilitate s-ENTDs-mediated ATP degradation to counteract excessive bladder excitability.
Collapse
Affiliation(s)
| | | | | | - Violeta N. Mutafova-Yambolieva
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada Reno, Reno, NV 89557, USA; (M.S.L.A.B.); (A.G.C.); (L.E.P.)
| |
Collapse
|
3
|
Kroll RG, Powell C, Chen J, Snider NT, St. Hilaire C, Reddy A, Kim J, Pinsky DJ, Murthy VL, Sutton NR. Circulating Ectonucleotidases Signal Impaired Myocardial Perfusion at Rest and Stress. J Am Heart Assoc 2023; 12:e027920. [PMID: 37119076 PMCID: PMC10227209 DOI: 10.1161/jaha.122.027920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/20/2023] [Indexed: 04/30/2023]
Abstract
Background Ectonucleotidases maintain vascular homeostasis by metabolizing extracellular nucleotides, modulating inflammation and thrombosis, and potentially, myocardial flow through adenosine generation. Evidence implicates dysfunction or deficiency of ectonucleotidases CD39 or CD73 in human disease; the utility of measuring levels of circulating ectonucleotidases as plasma biomarkers of coronary artery dysfunction or disease has not been previously reported. Methods and Results A total of 529 individuals undergoing clinically indicated positron emission tomography stress testing between 2015 and 2019 were enrolled in this single-center retrospective analysis. Baseline demographics, clinical data, nuclear stress test, and coronary artery calcium score variables were collected, as well as a blood sample. CD39 and CD73 levels were assessed as binary (detectable, undetectable) or continuous variables using ELISAs. Plasma CD39 was detectable in 24% of White and 8% of Black study participants (P=0.02). Of the clinical history variables examined, ectonucleotidase levels were most strongly associated with underlying liver disease and not other traditional coronary artery disease risk factors. Intriguingly, detection of circulating ectonucleotidase was inversely associated with stress myocardial blood flow (2.3±0.8 mL/min per g versus 2.7 mL/min per g±1.1 for detectable versus undetectable CD39 levels, P<0.001) and global myocardial flow reserve (Pearson correlation between myocardial flow reserve and log(CD73) -0.19, P<0.001). A subanalysis showed these differences held true independent of liver disease. Conclusions Vasodilatory adenosine is the expected product of local ectonucleotidase activity, yet these data support an inverse relationship between plasma ectonucleotidases, stress myocardial blood flow (CD39), and myocardial flow reserve (CD73). These findings support the conclusion that plasma levels of ectonucleotidases, which may be shed from the endothelial surface, contribute to reduced stress myocardial blood flow and myocardial flow reserve.
Collapse
Affiliation(s)
- Rachel G. Kroll
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Corey Powell
- Consulting for Statistics, Computing, and Analytics ResearchUniversity of MichiganAnn ArborMI
| | - Jun Chen
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Natasha T. Snider
- Department of Cell Biology and PhysiologyUniversity of North Carolina at Chapel HillChapel HillNC
| | - Cynthia St. Hilaire
- Division of Cardiology, Departments of Medicine and BioengineeringVascular Medicine Institute, University of PittsburghPittsburghPAUSA
| | - Akshay Reddy
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Judy Kim
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - David J. Pinsky
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMI
| | - Venkatesh L. Murthy
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
| | - Nadia R. Sutton
- Division of Cardiovascular Medicine, Department of MedicineMichigan MedicineAnn ArborMI
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTN
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN
| |
Collapse
|
4
|
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: 4.0] [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
|
5
|
Pilard M, Ollivier EL, Gourdou-Latyszenok V, Couturaud F, Lemarié CA. Endothelial Cell Phenotype, a Major Determinant of Venous Thrombo-Inflammation. Front Cardiovasc Med 2022; 9:864735. [PMID: 35528838 PMCID: PMC9068971 DOI: 10.3389/fcvm.2022.864735] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023] Open
Abstract
Reduced blood flow velocity in the vein triggers inflammation and is associated with the release into the extracellular space of alarmins or damage-associated molecular patterns (DAMPs). These molecules include extracellular nucleic acids, extracellular purinergic nucleotides (ATP, ADP), cytokines and extracellular HMGB1. They are recognized as a danger signal by immune cells, platelets and endothelial cells. Hence, endothelial cells are capable of sensing environmental cues through a wide variety of receptors expressed at the plasma membrane. The endothelium is then responding by expressing pro-coagulant proteins, including tissue factor, and inflammatory molecules such as cytokines and chemokines involved in the recruitment and activation of platelets and leukocytes. This ultimately leads to thrombosis, which is an active pro-inflammatory process, tightly regulated, that needs to be properly resolved to avoid further vascular damages. These mechanisms are often dysregulated, which promote fibrinolysis defects, activation of the immune system and irreversible vascular damages further contributing to thrombotic and inflammatory processes. The concept of thrombo-inflammation is now widely used to describe the complex interactions between the coagulation and inflammation in various cardiovascular diseases. In endothelial cells, activating signals converge to multiple intracellular pathways leading to phenotypical changes turning them into inflammatory-like cells. Accumulating evidence suggest that endothelial to mesenchymal transition (EndMT) may be a major mechanism of endothelial dysfunction induced during inflammation and thrombosis. EndMT is a biological process where endothelial cells lose their endothelial characteristics and acquire mesenchymal markers and functions. Endothelial dysfunction might play a central role in orchestrating and amplifying thrombo-inflammation thought induction of EndMT processes. Mechanisms regulating endothelial dysfunction have been only partially uncovered in the context of thrombotic diseases. In the present review, we focus on the importance of the endothelial phenotype and discuss how endothelial plasticity may regulate the interplay between thrombosis and inflammation. We discuss how the endothelial cells are sensing and responding to environmental cues and contribute to thrombo-inflammation with a particular focus on venous thromboembolism (VTE). A better understanding of the precise mechanisms involved and the specific role of endothelial cells is needed to characterize VTE incidence and address the risk of recurrent VTE and its sequelae.
Collapse
|
6
|
Thrombo-Inflammation: A Focus on NTPDase1/CD39. Cells 2021; 10:cells10092223. [PMID: 34571872 PMCID: PMC8469976 DOI: 10.3390/cells10092223] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
There is increasing evidence for a link between inflammation and thrombosis. Following tissue injury, vascular endothelium becomes activated, losing its antithrombotic properties whereas inflammatory mediators build up a prothrombotic environment. Platelets are the first elements to be activated following endothelial damage; they participate in physiological haemostasis, but also in inflammatory and thrombotic events occurring in an injured tissue. While physiological haemostasis develops rapidly to prevent excessive blood loss in the endothelium activated by inflammation, hypoxia or by altered blood flow, thrombosis develops slowly. Activated platelets release the content of their granules, including ATP and ADP released from their dense granules. Ectonucleoside triphosphate diphosphohydrolase-1 (NTPDase1)/CD39 dephosphorylates ATP to ADP and to AMP, which in turn, is hydrolysed to adenosine by ecto-5'-nucleotidase (CD73). NTPDase1/CD39 has emerged has an important molecule in the vasculature and on platelet surfaces; it limits thrombotic events and contributes to maintain the antithrombotic properties of endothelium. The aim of the present review is to provide an overview of platelets as cellular elements interfacing haemostasis and inflammation, with a particular focus on the emerging role of NTPDase1/CD39 in controlling both processes.
Collapse
|
7
|
Role of Purinergic Signalling in Endothelial Dysfunction and Thrombo-Inflammation in Ischaemic Stroke and Cerebral Small Vessel Disease. Biomolecules 2021; 11:biom11070994. [PMID: 34356618 PMCID: PMC8301873 DOI: 10.3390/biom11070994] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/20/2022] Open
Abstract
The cerebral endothelium is an active interface between blood and the central nervous system. In addition to being a physical barrier between the blood and the brain, the endothelium also actively regulates metabolic homeostasis, vascular tone and permeability, coagulation, and movement of immune cells. Being part of the blood–brain barrier, endothelial cells of the brain have specialized morphology, physiology, and phenotypes due to their unique microenvironment. Known cardiovascular risk factors facilitate cerebral endothelial dysfunction, leading to impaired vasodilation, an aggravated inflammatory response, as well as increased oxidative stress and vascular proliferation. This culminates in the thrombo-inflammatory response, an underlying cause of ischemic stroke and cerebral small vessel disease (CSVD). These events are further exacerbated when blood flow is returned to the brain after a period of ischemia, a phenomenon termed ischemia-reperfusion injury. Purinergic signaling is an endogenous molecular pathway in which the enzymes CD39 and CD73 catabolize extracellular adenosine triphosphate (eATP) to adenosine. After ischemia and CSVD, eATP is released from dying neurons as a damage molecule, triggering thrombosis and inflammation. In contrast, adenosine is anti-thrombotic, protects against oxidative stress, and suppresses the immune response. Evidently, therapies that promote adenosine generation or boost CD39 activity at the site of endothelial injury have promising benefits in the context of atherothrombotic stroke and can be extended to current CSVD known pathomechanisms. Here, we have reviewed the rationale and benefits of CD39 and CD39 therapies to treat endothelial dysfunction in the brain.
Collapse
|
8
|
Giuliani AL, Sarti AC, Di Virgilio F. Ectonucleotidases in Acute and Chronic Inflammation. Front Pharmacol 2021; 11:619458. [PMID: 33613285 PMCID: PMC7887318 DOI: 10.3389/fphar.2020.619458] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Ectonucleotidases are extracellular enzymes with a pivotal role in inflammation that hydrolyse extracellular purine and pyrimidine nucleotides, e.g., ATP, UTP, ADP, UDP, AMP and NAD+. Ectonucleotidases, expressed by virtually all cell types, immune cells included, either as plasma membrane-associated or secreted enzymes, are classified into four main families: 1) nucleoside triphosphate diphosphohydrolases (NTPDases), 2) nicotinamide adenine dinucleotide glycohydrolase (NAD glycohydrolase/ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1), 3) ecto-5′-nucleotidase (NT5E), and 4) ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs). Concentration of ATP, UTP and NAD+ can be increased in the extracellular space thanks to un-regulated, e.g., cell damage or cell death, or regulated processes. Regulated processes include secretory exocytosis, connexin or pannexin hemichannels, ATP binding cassette (ABC) transporters, calcium homeostasis modulator (CALMH) channels, the ATP-gated P2X7 receptor, maxi-anion channels (MACs) and volume regulated ion channels (VRACs). Hydrolysis of extracellular purine nucleotides generates adenosine, an important immunosuppressant. Extracellular nucleotides and nucleosides initiate or dampen inflammation via P2 and P1 receptors, respectively. All these agents, depending on their level of expression or activation and on the agonist concentration, are potent modulators of inflammation and key promoters of host defences, immune cells activation, pathogen clearance, tissue repair and regeneration. Thus, their knowledge is of great importance for a full understanding of the pathophysiology of acute and chronic inflammatory diseases. A selection of these pathologies will be briefly discussed here.
Collapse
Affiliation(s)
- Anna Lisa Giuliani
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alba Clara Sarti
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Di Virgilio
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
9
|
Yegutkin GG. Adenosine metabolism in the vascular system. Biochem Pharmacol 2020; 187:114373. [PMID: 33340515 DOI: 10.1016/j.bcp.2020.114373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
The concept of extracellular purinergic signaling was first proposed by Geoffrey Burnstock in the early 1970s. Since then, extracellular ATP and its metabolites ADP and adenosine have attracted an enormous amount of attention in terms of their involvement in a wide range of immunomodulatory, thromboregulatory, angiogenic, vasoactive and other pathophysiological activities in different organs and tissues, including the vascular system. In addition to significant progress in understanding the properties of nucleotide- and adenosine-selective receptors, recent studies have begun to uncover the complexity of regulatory mechanisms governing the duration and magnitude of the purinergic signaling cascade. This knowledge has led to the development of new paradigms in understanding the entire purinome by taking into account the multitude of signaling and metabolic pathways involved in biological effects of ATP and adenosine and compartmentalization of the adenosine system. Along with the "canonical route" of ATP breakdown to adenosine via sequential ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39) and ecto-5'-nucleotidase/CD73 activities, it has now become clear that purine metabolism is the result of concerted effort between ATP release, its metabolism through redundant nucleotide-inactivating and counteracting ATP-regenerating ectoenzymatic pathways, as well as cellular nucleoside uptake and phosphorylation of adenosine to ATP through complex phosphotransfer reactions. In this review I provide an overview of key enzymes involved in adenosine metabolic network, with special emphasis on the emerging roles of purine-converting ectoenzymes as novel targets for cancer and vascular therapies.
Collapse
|
10
|
Zeng J, Ning Z, Wang Y, Xiong H. Implications of CD39 in immune-related diseases. Int Immunopharmacol 2020; 89:107055. [PMID: 33045579 DOI: 10.1016/j.intimp.2020.107055] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/10/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
Abstract
Extracellular adenosine triphosphate (eATP) mediates pro-inflammatory responses by recruiting and activating inflammatory cells. CD39 can hydrolyze eATP into adenosine monophosphate (AMP), while CD73 can convert AMP into the immunosuppressive nucleoside adenosine (ADO). CD39 is a rate-limiting enzyme in this cascade, which is regarded as an immunological switch shifting the ATP-mediated pro-inflammatory environment to the ADO- mediated anti-inflammatory status. The CD39 expression can be detected in a wide spectrum of immunocytes, which is under the influence of environmental and genetic factors. It is increasingly suggested that, CD39 participates in some pathophysiological processes, like inflammatory bowel disease (IBD), sepsis, multiple sclerosis (MS), allergic diseases, ischemia-reperfusion (I/R) injury, systemic lupus erythematosus (SLE), diabetes and cancer. Here, we focus on the current understanding of CD39 in immunity, and comprehensively illustrate the diverse CD39 functions within a variety of disorders.
Collapse
Affiliation(s)
- Jianrui Zeng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Yuzhong Wang
- Department of Neurology and Central Laboratory, Affiliated Hospital of Jining Medical University, Shandong 272000, China.
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China.
| |
Collapse
|
11
|
Chaurasia SN, Kushwaha G, Pandey A, Dash D. Human platelets express functional ectonucleotidases that restrict platelet activation signaling. Biochem Biophys Res Commun 2020; 527:104-109. [PMID: 32446352 DOI: 10.1016/j.bbrc.2020.04.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Platelets play central role in thrombosis and haemostasis. Platelets store adenine nucleotides in their dense granules, which are released upon agonist-stimulation. Level of these nucleotides in extracellular fluid is regulated by activities of ectonucleotidases such as ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and ecto-5'-nucleotidase (CD73) expressed on platelet surface. Here we demonstrate that, expression of surface-bound ectonucleotidases rose significantly in platelets, concomitant with upregulation of their enzymatic activities, when cells were stimulated with thrombin. Interestingly, inhibition of CD73 in thrombin-treated platelets led to enhanced tyrosine phosphorylation of proteins and rise in intracellular free calcium, [Ca2+]i, thus signifying the inhibitory role of the ectonucleotidase on agonist-mediated platelet signaling.
Collapse
Affiliation(s)
- Susheel N Chaurasia
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Geeta Kushwaha
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Abhishek Pandey
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Debabrata Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
12
|
Vuerich M, Robson SC, Longhi MS. Ectonucleotidases in Intestinal and Hepatic Inflammation. Front Immunol 2019; 10:507. [PMID: 30941139 PMCID: PMC6433995 DOI: 10.3389/fimmu.2019.00507] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 02/25/2019] [Indexed: 12/21/2022] Open
Abstract
Purinergic signaling modulates systemic and local inflammatory responses. Extracellular nucleotides, including eATP, promote inflammation, at least in part via the inflammasome upon engagement of P2 purinergic receptors. In contrast, adenosine generated during eATP phosphohydrolysis by ectonucleotidases, triggers immunosuppressive/anti-inflammatory pathways. Mounting evidence supports the role of ectonucleotidases, especially ENTPD1/CD39 and CD73, in the control of several inflammatory conditions, ranging from infectious disease, organ fibrosis to oncogenesis. Our experimental data generated over the years have indicated both CD39 and CD73 serve as pivotal regulators of intestinal and hepatic inflammation. In this context, immune cell responses are regulated by the balance between eATP and adenosine, potentially impacting disease outcomes as in gastrointestinal infection, inflammatory bowel disease, ischemia reperfusion injury of the bowel and liver, autoimmune or viral hepatitis and other inflammatory conditions, such as cancer. In this review, we report the most recent discoveries on the role of ENTPD1/CD39, CD73, and other ectonucleotidases in the regulation of intestinal and hepatic inflammation. We discuss the present knowledge, highlight the most intriguing and promising experimental data and comment on important aspects that still need to be addressed to develop purinergic-based therapies for these important illnesses.
Collapse
Affiliation(s)
- Marta Vuerich
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Simon C Robson
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Maria Serena Longhi
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
13
|
Di Virgilio F, Sarti AC, Falzoni S, De Marchi E, Adinolfi E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer 2018; 18:601-618. [PMID: 30006588 DOI: 10.1038/s41568-018-0037-0] [Citation(s) in RCA: 470] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Modulation of the biochemical composition of the tumour microenvironment is a new frontier of cancer therapy. Several immunosuppressive mechanisms operate in the milieu of most tumours, a condition that makes antitumour immunity ineffective. One of the most potent immunosuppressive factors is adenosine, which is generated in the tumour microenvironment owing to degradation of extracellular ATP. Accruing evidence over the past few years shows that ATP is one of the major biochemical constituents of the tumour microenvironment, where it acts at P2 purinergic receptors expressed on both tumour and host cells. Stimulation of P2 receptors has different effects depending on the extracellular ATP concentration, the P2 receptor subtype engaged and the target cell type. Among P2 receptors, the P2X purinergic receptor 7 (P2X7R) subtype appears to be a main player in host-tumour cell interactions. Preclinical studies in several tumour models have shown that P2X7R targeting is potentially a very effective anticancer treatment, and many pharmaceutical companies have now developed potent and selective small molecule inhibitors of P2X7R. In this Review, we report on the multiple mechanisms by which extracellular ATP shapes the tumour microenvironment and how its stimulation of host and tumour cell P2 receptors contributes to determining tumour fate.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Elena Adinolfi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
14
|
Mononuclear-cell-derived microparticles attenuate endothelial inflammation by transfer of miR-142-3p in a CD39 dependent manner. Purinergic Signal 2018; 14:423-432. [PMID: 30244433 DOI: 10.1007/s11302-018-9624-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/29/2018] [Indexed: 12/22/2022] Open
Abstract
Plasma microparticles (MP) bear functional active ectonucleotidases of the CD39 family with implications in vascular inflammation. MP appear to be able to fuse with cells and transfer genetic information. Here, we tested whether levels of different immunomodulatory microRNAs (miRs) in plasma MP are modulated by CD39 after experimental hepatectomy. We further investigated whether horizontal transfer of miR-142-3p between mononuclear (MNC) and endothelial cells via MP is regulated by purinergic signaling. Partial hepatectomy was performed in C57BL/6 wild type and Cd39 null mice. MP were collected via ultracentrifugation. MNC were stimulated with nucleotides and nucleosides, in vitro, and tested for miR-142-3p levels. Fusion of MNC-derived MP and endothelial cells with subsequent transfer of miR-142-3p was imaged by flow cytometry and confocal microscopy. Endothelial inflammation and apoptosis were quantified after transfection with miR-142-3p. Significantly lower miR-142-3p levels were observed in plasma MP of Cd39 null mice after partial hepatectomy, when compared to C57BL/6 wild types (p < 0.05). In contrast to extracellular nucleotides, anti-inflammatory adenosine significantly increased miR-142-3p levels in MNC-derived MP, in vitro (p < 0.05). MNC-derived MP are able to transfer miR-142-3p to endothelial cells by fusion. Transfection of endothelial cells with miR-142-3p decreased TNF-α levels (p < 0.05) and endothelial apoptosis (p < 0.05). MiR-142-3p levels in MNC-derived MP are modulated by nucleoside signaling and might reflect compensatory responses in vascular inflammation. Our data suggest the transfer of genetic information via shed MP as a putative mechanism of intercellular communication-with implications in organ regeneration.
Collapse
|
15
|
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: 4.3] [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
|
16
|
Meyer J, Balaphas A, Fontana P, Sadoul K, Morel P, Gonelle-Gispert C, Bühler L. Platelets in liver regeneration. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/voxs.12382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- J. Meyer
- Division of Digestive and Transplantation Surgery; University Hospitals of Geneva; Genève Switzerland
- Unit of Surgical Research; University of Geneva; Genève Switzerland
| | - A. Balaphas
- Division of Digestive and Transplantation Surgery; University Hospitals of Geneva; Genève Switzerland
- Unit of Surgical Research; University of Geneva; Genève Switzerland
| | - P. Fontana
- Division of Angiology and Haemostasis; University Hospitals of Geneva; Genève Switzerland
- Geneva Platelet Group; University of Geneva; Genève Switzerland
| | - K. Sadoul
- Regulation and pharmacology of the cytoskeleton; Institute for Advanced Biosciences; Université Grenoble Alpes; Grenoble France
| | - P. Morel
- Division of Digestive and Transplantation Surgery; University Hospitals of Geneva; Genève Switzerland
- Unit of Surgical Research; University of Geneva; Genève Switzerland
| | | | - L. Bühler
- Division of Digestive and Transplantation Surgery; University Hospitals of Geneva; Genève Switzerland
- Unit of Surgical Research; University of Geneva; Genève Switzerland
| |
Collapse
|
17
|
Boopathy GTK, Kulkarni M, Ho SY, Boey A, Chua EWM, Barathi VA, Carney TJ, Wang X, Hong W. Cavin-2 regulates the activity and stability of endothelial nitric-oxide synthase (eNOS) in angiogenesis. J Biol Chem 2017; 292:17760-17776. [PMID: 28912276 PMCID: PMC5663877 DOI: 10.1074/jbc.m117.794743] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/04/2017] [Indexed: 01/26/2023] Open
Abstract
Angiogenesis is a highly regulated process for formation of new blood vessels from pre-existing ones. Angiogenesis is dysregulated in various pathologies, including age-related macular degeneration, arthritis, and cancer. Inhibiting pathological angiogenesis therefore represents a promising therapeutic strategy for treating these disorders, highlighting the need to study angiogenesis in more detail. To this end, identifying the genes essential for blood vessel formation and elucidating their function are crucial for a complete understanding of angiogenesis. Here, focusing on potential candidate genes for angiogenesis, we performed a morpholino-based genetic screen in zebrafish and identified Cavin-2, a membrane-bound phosphatidylserine-binding protein and critical organizer of caveolae (small microdomains in the plasma membrane), as a regulator of angiogenesis. Using endothelial cells, we show that Cavin-2 is required for in vitro angiogenesis and also for endothelial cell proliferation, migration, and invasion. We noted a high level of Cavin-2 expression in the neovascular tufts in the mouse model of oxygen-induced retinopathy, suggesting a role for Cavin-2 in pathogenic angiogenesis. Interestingly, we also found that Cavin-2 regulates the production of nitric oxide (NO) in endothelial cells by controlling the stability and activity of the endothelial nitric-oxide synthase (eNOS) and that Cavin-2 knockdown cells produce much less NO than WT cells. Also, mass spectrometry, flow cytometry, and electron microscopy analyses indicated that Cavin-2 is secreted in endothelial microparticles (EMPs) and is required for EMP biogenesis. Taken together, our results indicate that in addition to its function in caveolae biogenesis, Cavin-2 plays a critical role in endothelial cell maintenance and function by regulating eNOS activity.
Collapse
Affiliation(s)
- Gandhi T K Boopathy
- From the Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, .,the SERI-IMCB Programme in Retinal Angiogenic Diseases (SIPRAD), SERI-IMCB, Singapore
| | - Madhura Kulkarni
- the Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sze Yuan Ho
- the Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Adrian Boey
- From the Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore
| | - Edmond Wei Min Chua
- From the Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore
| | - Veluchamy A Barathi
- the SERI-IMCB Programme in Retinal Angiogenic Diseases (SIPRAD), SERI-IMCB, Singapore.,the Singapore Eye Research Institute (SERI), 20 College Road, 169856 Singapore.,the Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Graduate Medical School, 8 College Rd., 169857 Singapore.,the Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, and
| | - Tom J Carney
- From the Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore.,the SERI-IMCB Programme in Retinal Angiogenic Diseases (SIPRAD), SERI-IMCB, Singapore.,the Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Xiaomeng Wang
- From the Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore.,the SERI-IMCB Programme in Retinal Angiogenic Diseases (SIPRAD), SERI-IMCB, Singapore.,the Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,the Singapore Eye Research Institute (SERI), 20 College Road, 169856 Singapore
| | - Wanjin Hong
- From the Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, .,the SERI-IMCB Programme in Retinal Angiogenic Diseases (SIPRAD), SERI-IMCB, Singapore
| |
Collapse
|
18
|
Feldbrügge L, Moss AC, Yee EU, Csizmadia E, Mitsuhashi S, Longhi MS, Sandhu B, Stephan H, Wu Y, Cheifetz AS, Müller CE, Sévigny J, Robson SC, Jiang ZG. Expression of Ecto-nucleoside Triphosphate Diphosphohydrolases-2 and -3 in the Enteric Nervous System Affects Inflammation in Experimental Colitis and Crohn's Disease. J Crohns Colitis 2017; 11:1113-1123. [PMID: 28472257 PMCID: PMC5881706 DOI: 10.1093/ecco-jcc/jjx058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 04/03/2017] [Accepted: 04/24/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Recent studies have suggested that the enteric nervous system can modulate gut immunity. Ecto-nucleoside triphosphate diphosphohydrolases [E-NTPDases] regulate purinergic signalling by sequential phosphohydrolysis of pro-inflammatory extracellular adenosine 5'-triphosphate [ATP]. Herein, we test the hypothesis that E-NTPDases modulate gut inflammation via neuro-immune crosstalk. DESIGN We determined expression patterns of NTPDase2 and NTPDase3 in murine and human colon. Experimental colitis was induced by dextran sodium sulphate [DSS] in genetically engineered mice deficient in NTPDase2 or NTPDase3. We compared plasma adenosine diphosphatase [ADPase] activity from Crohn's patients and healthy controls, and linked the enzyme activity to Crohn's disease activity. RESULTS NTPDase2 and -3 were chiefly expressed in cells of the enteric nervous system in both murine and human colon. When compared with wild type, DSS-induced colitis was exacerbated in Entpd2, and to a lesser extent, Entpd3 null mice as measured by disease activity score and histology, and marked anaemia was seen in both. Colonic macrophages isolated from Entpd2 null mice displayed a pro-inflammatory phenotype compared with wild type. In human plasma, Crohn's patients had decreases in ADPase activity when compared with healthy controls. The drop in ADPase activity was likely associated with changes in NTPDase2 and -3, as suggested by inhibitor studies, and were correlated with Crohn's disease activity. CONCLUSIONS NTPDase2 and -3 are ecto-enzymes expressed in the enteric nervous system. Both enzymes confer protection against gut inflammation in experimental colitis and exhibit alterations in Crohn's disease. These observations suggest that purinergic signalling modulated by E-NTPDases governs neuro-immune interactions that are relevant in Crohn's disease.
Collapse
Affiliation(s)
- Linda Feldbrügge
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Surgery, Charité Universitätsmedizin, Berlin, Germany
| | - Alan C Moss
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Eric U Yee
- Department of Pathology, OU Medical Center, Oklahoma City, USA
| | - Eva Csizmadia
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shuji Mitsuhashi
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria Serena Longhi
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Bynvant Sandhu
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Holger Stephan
- Helmholtz-Zentrum Dresden–Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Yan Wu
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Adam S Cheifetz
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Jean Sévigny
- Département de Microbiologie-infectiologie et d’Immunologie, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Simon C Robson
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Z Gordon Jiang
- Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Neudecker V, Colgan SP, Eltzschig HK. Novel therapeutic concepts for inflammatory bowel disease-from bench to bedside. J Mol Med (Berl) 2017; 95:899-903. [PMID: 28780611 PMCID: PMC5800411 DOI: 10.1007/s00109-017-1574-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Viola Neudecker
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany.
| | - Sean P Colgan
- Department of Medicine and the Mucosal Inflammation Program, University of Colorado Denver, Aurora, CO, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
20
|
Purinergic signaling during intestinal inflammation. J Mol Med (Berl) 2017; 95:915-925. [PMID: 28547076 DOI: 10.1007/s00109-017-1545-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease (IBD) is a devastating disease that is associated with excessive inflammation in the intestinal tract in genetically susceptible individuals and potentially triggered by microbial dysbiosis. This illness markedly predisposes patients to thrombophilia and chronic debility as well as bowel, lymphatic, and liver cancers. Development of new therapies is needed to re-establish long-term immune tolerance in IBD patients without increasing the risk of opportunistic infections and cancer. Aberrant purinergic signaling pathways have been implicated in disordered thromboregulation and immune dysregulation, as noted in the pathogenesis of IBD and other gastrointestinal/hepatic autoimmune diseases. Expression of CD39 on endothelial or immune cells allows for homeostatic integration of hemostasis and immunity, which are disrupted in IBD. Our focus in this review is on novel aspects of the functions of CD39 and related NTPDases in IBD. Regulated CD39 activity allows for scavenging of extracellular nucleotides, the maintenance of P2-receptor integrity and coordination of adenosinergic signaling responses. CD39 together with CD73, serves as an integral component of the immunosuppressive machinery of dendritic cells, myeloid cells, T and B cells. Genetic inheritance and environental factors closely regulate the levels of expression and phosphohydrolytic activity of CD39, both on immune cells and released microparticles. Purinergic mechanisms associated with T regulatory and supressor T helper type 17 cells modulate disease activity in IBD, as can be modeled in experimental colitis. As a recent example, upregulation of CD39 is dependent upon ligation of the aryl hydrocarbon receptor (AHR), as with natural ligands such as bilirubin and 2-(1' H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE). Decreased expression of CD39 and/or dysfunctional AHR signaling, however, abrogates the protective effects of immunosuppressive AHR ligands. These factors could also serve as biomarkers of disease activity in IBD. Heightened thrombosis, inflammation, and immune disturbances as seen in IBD appear to be associated with aberrant purinergic signaling. Ongoing development of therapeutic strategies augmenting CD39 ectonucleotidase bioactivity via cytokines or AHR ligands offers promise for management of thrombophilia, disordered inflammation, and aberrant immune reactivity in IBD.
Collapse
|
21
|
Silkin YA, Silkina EN. The role of ecto-ATPases of erythrocyte plasma membrane in hemodynamics of fishes. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017010094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Covarrubias R, Chepurko E, Reynolds A, Huttinger ZM, Huttinger R, Stanfill K, Wheeler DG, Novitskaya T, Robson SC, Dwyer KM, Cowan PJ, Gumina RJ. Role of the CD39/CD73 Purinergic Pathway in Modulating Arterial Thrombosis in Mice. Arterioscler Thromb Vasc Biol 2016; 36:1809-20. [PMID: 27417582 DOI: 10.1161/atvbaha.116.307374] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/29/2016] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Circulating blood cells and endothelial cells express ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and ecto-5'-nucleotidase (CD73). CD39 hydrolyzes extracellular ATP or ADP to AMP. CD73 hydrolyzes AMP to adenosine. The goal of this study was to examine the interplay between CD39 and CD73 cascade in arterial thrombosis. APPROACH AND RESULTS To determine how CD73 activity influences in vivo thrombosis, the time to ferric chloride-induced arterial thrombosis was measured in CD73-null mice. In response to 5% FeCl3, but not to 10% FeCl3, there was a significant decrease in the time to thrombosis in CD73-null mice compared with wild-type mice. In mice overexpressing CD39, ablation of CD73 did not inhibit the prolongation in the time to thrombosis conveyed by CD39 overexpression. However, the CD73 inhibitor α-β-methylene-ADP nullified the prolongation in the time to thrombosis in human CD39 transgenic (hC39-Tg)/CD73-null mice. To determine whether hematopoietic-derived cells or endothelial cell CD39 activity regulates in vivo arterial thrombus, bone marrow transplant studies were conducted. FeCl3-induced arterial thrombosis in chimeric mice revealed a significant prolongation in the time to thrombosis in hCD39-Tg reconstituted wild-type mice, but not on wild-type reconstituted hCD39-Tg mice. Monocyte depletion with clodronate-loaded liposomes normalized the time to thrombosis in hCD39-Tg mice compared with hCD39-Tg mice treated with control liposomes, demonstrating that increased CD39 expression on monocytes protects against thrombosis. CONCLUSIONS These data demonstrate that ablation of CD73 minimally effects in vivo thrombosis, but increased CD39 expression on hematopoietic-derived cells, especially monocytes, attenuates in vivo arterial thrombosis.
Collapse
Affiliation(s)
- Roman Covarrubias
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Elena Chepurko
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Adam Reynolds
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Zachary M Huttinger
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Ryan Huttinger
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Katherine Stanfill
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Debra G Wheeler
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Tatiana Novitskaya
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Simon C Robson
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Karen M Dwyer
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Peter J Cowan
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Richard J Gumina
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.).
| |
Collapse
|
23
|
Mahaut-Smith MP, Taylor KA, Evans RJ. Calcium Signalling through Ligand-Gated Ion Channels such as P2X1 Receptors in the Platelet and other Non-Excitable Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:305-29. [PMID: 27161234 DOI: 10.1007/978-3-319-26974-0_13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ligand-gated ion channels on the cell surface are directly activated by the binding of an agonist to their extracellular domain and often referred to as ionotropic receptors. P2X receptors are ligand-gated non-selective cation channels with significant permeability to Ca(2+) whose principal physiological agonist is ATP. This chapter focuses on the mechanisms by which P2X1 receptors, a ubiquitously expressed member of the family of ATP-gated channels, can contribute to cellular responses in non-excitable cells. Much of the detailed information on the contribution of P2X1 to Ca(2+) signalling and downstream functional events has been derived from the platelet. The underlying primary P2X1-generated signalling event in non-excitable cells is principally due to Ca(2+) influx, although Na(+) entry will also occur along with membrane depolarization. P2X1 receptor stimulation can lead to additional Ca(2+) mobilization via a range of routes such as amplification of G-protein-coupled receptor-dependent Ca(2+) responses. This chapter also considers the mechanism by which cells generate extracellular ATP for autocrine or paracrine activation of P2X1 receptors. For example cytosolic ATP efflux can result from opening of pannexin anion-permeable channels or following damage to the cell membrane. Alternatively, ATP stored in specialised secretory vesicles can undergo quantal release via the process of exocytosis. Examples of physiological or pathophysiological roles of P2X1-dependent signalling in non-excitable cells are also discussed, such as thrombosis and immune responses.
Collapse
Affiliation(s)
- Martyn P Mahaut-Smith
- Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK.
| | - Kirk A Taylor
- Department of Biomedical and Forensic Sciences, Anglia Ruskin University, Cambridge, UK
| | - Richard J Evans
- Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester, LE1 9HN, UK
| |
Collapse
|
24
|
Phillips CD, Baker RJ. Secretory Gene Recruitments in Vampire Bat Salivary Adaptation and Potential Convergences With Sanguivorous Leeches. Front Ecol Evol 2015. [DOI: 10.3389/fevo.2015.00122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
25
|
Banz Y, Item GM, Vogt A, Rieben R, Candinas D, Beldi G. Endothelial- and Platelet-Derived Microparticles Are Generated During Liver Resection in Humans. J INVEST SURG 2015; 29:20-31. [DOI: 10.3109/08941939.2015.1047540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Hohmann JD, Peter K. Activated-platelet targeting of CD39 as a potential way forward. The quest for efficient antithrombotic therapy without associated bleeding complications. Hamostaseologie 2015; 36:17-25. [PMID: 26328528 DOI: 10.5482/hamo-14-12-0085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 08/27/2015] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED Antiplatelet therapy is given to millions of patients and has saved numerous lives. However, it is also associated with complications including fatal bleedings. Clinically used antiplatelet drugs seem to follow the rule of an inherent link of improved anti-thrombotic potency with increased risk of bleeding complications. Therefore, there is an ongoing quest to develop drugs that are able to break this link that has prevented many patients from receiving antiplatelet protection and has resulted in substantial mortality and morbidity. We describe a new antiplatelet approach that is based on an recombinant antibody protein, a drug format that has recently attracted major interest. Two unique components are genetically combined in this molecule: 1) The ecto-nucleoside triphosphate diphosphohydrolase NTPDase CD39, which enzymatically degrades ATP and ADP to AMP, which is then further degraded to adenosine by the endothelially expressed CD73. Thereby, the platelet activating ADP is reduced and replaced by the platelet inhibiting adenosine resulting in a strong antiplatelet effect. 2) A single-chain antibody (scFv) that specifically binds to the activated GPIIb/IIIa receptor and thus allows targeting to activated platelets. The described fusion protein results in strong enrichment of CD39's antiplatelet effect, resulting in potent inhibition of platelet adhesion and aggregation and thrombosis in mice. The activated platelet targeting allows using a low systemic concentration that does not interfere with normal haemostasis and thus does not cause bleeding time prolongation in mice. CONCLUSION We describe a new antiplatelet approach that promises to deliver strong localized antithrombotic effects without associated bleeding problems.
Collapse
Affiliation(s)
| | - K Peter
- Prof. Karlheinz Peter, Baker IDI Heart and Diabetes Institute, PO Box 6492, St Kilda Road Central, Melbourne, Victoria 8008, Australia,
| |
Collapse
|
27
|
Yegutkin GG. Enzymes involved in metabolism of extracellular nucleotides and nucleosides: functional implications and measurement of activities. Crit Rev Biochem Mol Biol 2015; 49:473-97. [PMID: 25418535 DOI: 10.3109/10409238.2014.953627] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular nucleotides and nucleosides mediate diverse signaling effects in virtually all organs and tissues. Most models of purinergic signaling depend on functional interactions between distinct processes, including (i) the release of endogenous ATP and other nucleotides, (ii) triggering of signaling events via a series of nucleotide-selective ligand-gated P2X and metabotropic P2Y receptors as well as adenosine receptors and (iii) ectoenzymatic interconversion of purinergic agonists. The duration and magnitude of purinergic signaling is governed by a network of ectoenzymes, including the enzymes of the nucleoside triphosphate diphosphohydrolase (NTPDase) family, the nucleotide pyrophosphatase/phosphodiesterase (NPP) family, ecto-5'-nucleotidase/CD73, tissue-nonspecific alkaline phosphatase (TNAP), prostatic acid phosphatase (PAP) and other alkaline and acid phosphatases, adenosine deaminase (ADA) and purine nucleoside phosphorylase (PNP). Along with "classical" inactivating ectoenzymes, recent data provide evidence for the co-existence of a counteracting ATP-regenerating pathway comprising the enzymes of the adenylate kinase (AK) and nucleoside diphosphate kinase (NDPK/NME/NM23) families and ATP synthase. This review describes recent advances in this field, with special emphasis on purine-converting ectoenzymes as a complex and integrated network regulating purinergic signaling in such (patho)physiological states as immunomodulation, inflammation, tumorigenesis, arterial calcification and other diseases. The second part of this review provides a comprehensive overview and basic principles of major approaches employed for studying purinergic activities, including spectrophotometric Pi-liberating assays, high-performance liquid chromatographic (HPLC) and thin-layer chromatographic (TLC) analyses of purine substrates and metabolites, capillary electrophoresis, bioluminescent, fluorometric and electrochemical enzyme-coupled assays, histochemical staining, and further emphasizes their advantages, drawbacks and suitability for assaying a particular catalytic reaction.
Collapse
Affiliation(s)
- Gennady G Yegutkin
- Department of Medical Microbiology and Immunology, University of Turku , Turku , Finland
| |
Collapse
|
28
|
Abstract
There are nineteen different receptor proteins for adenosine, adenine and uridine nucleotides, and nucleotide sugars, belonging to three families of G protein-coupled adenosine and P2Y receptors, and ionotropic P2X receptors. The majority are functionally expressed in blood vessels, as purinergic receptors in perivascular nerves, smooth muscle and endothelial cells, and roles in regulation of vascular contractility, immune function and growth have been identified. The endogenous ligands for purine receptors, ATP, ADP, UTP, UDP and adenosine, can be released from different cell types within the vasculature, as well as from circulating blood cells, including erythrocytes and platelets. Many purine receptors can be activated by two or more of the endogenous ligands. Further complexity arises because of interconversion between ligands, notably adenosine formation from the metabolism of ATP, leading to complex integrated responses through activation of different subtypes of purine receptors. The enzymes responsible for this conversion, ectonucleotidases, are present on the surface of smooth muscle and endothelial cells, and may be coreleased with neurotransmitters from nerves. What selectivity there is for the actions of purines/pyrimidines comes from differential expression of their receptors within the vasculature. P2X1 receptors mediate the vasocontractile actions of ATP released as a neurotransmitter with noradrenaline (NA) from sympathetic perivascular nerves, and are located on the vascular smooth muscle adjacent to the nerve varicosities, the sites of neurotransmitter release. The relative contribution of ATP and NA as functional cotransmitters varies with species, type and size of blood vessel, neuronal firing pattern, the tone/pressure of the blood vessel, and in ageing and disease. ATP is also a neurotransmitter in non-adrenergic non-cholinergic perivascular nerves and mediates vasorelaxation via smooth muscle P2Y-like receptors. ATP and adenosine can act as neuromodulators, with the most robust evidence being for prejunctional inhibition of neurotransmission via A1 adenosine receptors, but also prejunctional excitation and inhibition of neurotransmission via P2X and P2Y receptors, respectively. P2Y2, P2Y4 and P2Y6 receptors expressed on the vascular smooth muscle are coupled to vasocontraction, and may have a role in pathophysiological conditions, when purines are released from damaged cells, or when there is damage to the protective barrier that is the endothelium. Adenosine is released during hypoxia to increase blood flow via vasodilator A2A and A2B receptors expressed on the endothelium and smooth muscle. ATP is released from endothelial cells during hypoxia and shear stress and can act at P2Y and P2X4 receptors expressed on the endothelium to increase local blood flow. Activation of endothelial purine receptors leads to the release of nitric oxide, hyperpolarising factors and prostacyclin, which inhibits platelet aggregation and thus ensures patent blood flow. Vascular purine receptors also regulate endothelial and smooth muscle growth, and inflammation, and thus are involved in the underlying processes of a number of cardiovascular diseases.
Collapse
Affiliation(s)
- Vera Ralevic
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| | - William R Dunn
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
29
|
Jalkanen J, Yegutkin GG, Hollmén M, Aalto K, Kiviniemi T, Salomaa V, Jalkanen S, Hakovirta H. Aberrant circulating levels of purinergic signaling markers are associated with several key aspects of peripheral atherosclerosis and thrombosis. Circ Res 2015; 116:1206-15. [PMID: 25645301 DOI: 10.1161/circresaha.116.305715] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE Purinergic signaling plays an important role in inflammation and vascular integrity, but little is known about purinergic mechanisms during the pathogenesis of atherosclerosis in humans. OBJECTIVE The objective of this study is to study markers of purinergic signaling in a cohort of patients with peripheral artery disease. METHODS AND RESULTS Plasma ATP and ADP levels and serum nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39) and ecto-5'-nucleotidase/CD73 activities were measured in 226 patients with stable peripheral artery disease admitted for nonurgent invasive imaging and treatment. The major findings were that ATP, ADP, and CD73 values were higher in atherosclerotic patients than in controls without clinically evident peripheral artery disease (P<0.0001). Low CD39 activity was associated with disease progression (P=0.01). In multivariable linear regression models, high CD73 activity was associated with chronic hypoxia (P=0.001). Statin use was associated with lower ADP (P=0.041) and tended to associate with higher CD73 (P=0.054), while lower ATP was associated with the use of angiotensin receptor blockers (P=0.015). CONCLUSIONS Purinergic signaling plays an important role in peripheral artery disease progression. Elevated levels of circulating ATP and ADP are especially associated with atherosclerotic diseases of younger age and smoking. The antithrombotic and anti-inflammatory effects of statins may partly be explained by their ability to lower ADP. We suggest that the prothrombotic nature of smoking could be a cause of elevated ADP, and this may explain why cardiovascular patients who smoke benefit from platelet P2Y12 receptor antagonists more than their nonsmoking peers.
Collapse
Affiliation(s)
- Juho Jalkanen
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| | - Gennady G Yegutkin
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| | - Maija Hollmén
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| | - Kristiina Aalto
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| | - Tuomas Kiviniemi
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| | - Veikko Salomaa
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| | - Sirpa Jalkanen
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.).
| | - Harri Hakovirta
- From the Department of Vascular Surgery (J.J., H.H.) and Heart Center (T.K.), Turku University Hospital, Turku, Finland; Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland (G.G.Y., M.H., K.A., S.J.); and National Institute for Health and Welfare, Helsinki, Finland (V.S.)
| |
Collapse
|
30
|
Abstract
Extracellular nucleotides play a critical role in vascular thrombosis and inflammation. Alterations in purinergic extracellular nucleotide concentrations activate pathways that result in platelet degranulation and aggregation, and endothelial and leukocyte activation and recruitment. CD39, the dominant vascular nucleotidase, hydrolyzes ATP and ADP to provide the substrate for generation of the anti-inflammatory and antithrombotic mediator adenosine. The purinergic signaling system, with CD39 at its center, plays an important role in modulating vascular homeostasis and the response to vascular injury, as seen in clinically relevant diseases such as stroke, ischemia-reperfusion injury, and pulmonary hypertension. A growing body of knowledge of the purinergic signaling pathway implicates CD39 as a critical modulator of vascular thrombosis and inflammation. Therapeutic strategies targeting CD39 offer promising opportunities in the management of vascular thromboinflammatory diseases.
Collapse
|
31
|
Characterization of circulating microparticle-associated CD39 family ecto-nucleotidases in human plasma. Purinergic Signal 2014; 10:611-8. [PMID: 25165006 DOI: 10.1007/s11302-014-9423-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 08/18/2014] [Indexed: 01/28/2023] Open
Abstract
Phosphohydrolysis of extracellular ATP and ADP is an essential step in purinergic signaling that regulates key pathophysiological processes, such as those linked to inflammation. Classically, this reaction has been known to occur in the pericellular milieu catalyzed by membrane bound cellular ecto-nucleotidases, which can be released in the form of both soluble ecto-enzymes as well as being associated with exosomes. Circulating ecto-nucleoside triphosphate diphosphohydrolase 1 (NTPDase 1/CD39) and adenylate kinase 1 (AK1) activities have been shown to be present in plasma. However, other ecto-nucleotidases have not been characterized in depth. An in vitro ADPase assay was developed to probe the ecto-enzymes responsible for the ecto-nucleotidase activity in human platelet-free plasma, in combination with various specific biochemical inhibitors. Identities of ecto-nucleotidases were further characterized by chromatography, immunoblotting, and flow cytometry of circulating exosomes. We noted that microparticle-bound E-NTPDases and soluble AK1 constitute the highest levels of ecto-nucleotidase activity in human plasma. All four cell membrane expressed E-NTPDases are also found in circulating microparticles in human plasma, inclusive of: CD39, NTPDase 2 (CD39L1), NTPDase 3 (CD39L3), and NTPDase 8. CD39 family members and other ecto-nucleotidases are found on distinct microparticle populations. A significant proportion of the microparticle-associated ecto-nucleotidase activity is sensitive to POM6, inferring the presence of NTPDases, either -2 or/and -3. We have refined ADPase assays of human plasma from healthy volunteers and have found that CD39, NTPDases 2, 3, and 8 to be associated with circulating microparticles, whereas soluble AK1 is present in human plasma. These ecto-enzymes constitute the bulk circulating ADPase activity, suggesting a broader implication of CD39 family and other ecto-enzymes in the regulation of extracellular nucleotide metabolism.
Collapse
|
32
|
Hargett LA, Bauer NN. On the origin of microparticles: From "platelet dust" to mediators of intercellular communication. Pulm Circ 2014; 3:329-40. [PMID: 24015332 PMCID: PMC3757826 DOI: 10.4103/2045-8932.114760] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Microparticles are submicron vesicles shed from a variety of cells. Peter Wolf first identified microparticles in the midst of ongoing blood coagulation research in 1967 as a product of platelets. He termed them platelet dust. Although initially thought to be useless cellular trash, decades of research focused on the tiny vesicles have defined their roles as participators in coagulation, cellular signaling, vascular injury, and homeostasis. The purpose of this review is to highlight the science leading up to the discovery of microparticles, feature discoveries made by key contributors to the field of microparticle research, and discuss their positive and negative impact on the pulmonary circulation.
Collapse
Affiliation(s)
- Leslie A Hargett
- Department of Pharmacology, Center for Lung Biology and College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | | |
Collapse
|
33
|
Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev 2013; 66:102-92. [PMID: 24335194 DOI: 10.1124/pr.113.008029] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purinergic signaling plays important roles in control of vascular tone and remodeling. There is dual control of vascular tone by ATP released as a cotransmitter with noradrenaline from perivascular sympathetic nerves to cause vasoconstriction via P2X1 receptors, whereas ATP released from endothelial cells in response to changes in blood flow (producing shear stress) or hypoxia acts on P2X and P2Y receptors on endothelial cells to produce nitric oxide and endothelium-derived hyperpolarizing factor, which dilates vessels. ATP is also released from sensory-motor nerves during antidromic reflex activity to produce relaxation of some blood vessels. In this review, we stress the differences in neural and endothelial factors in purinergic control of different blood vessels. The long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides in promoting migration and proliferation of both vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis and vessel remodeling during restenosis after angioplasty are described. The pathophysiology of blood vessels and therapeutic potential of purinergic agents in diseases, including hypertension, atherosclerosis, ischemia, thrombosis and stroke, diabetes, and migraine, is discussed.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; and Department of Pharmacology, The University of Melbourne, Australia.
| | | |
Collapse
|
34
|
Tapper EB, Robson S, Malik R. Reply to: "Circulating platelet derived microparticles are not increased in patients with cirrhosis". J Hepatol 2013; 59:913. [PMID: 23816703 DOI: 10.1016/j.jhep.2013.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 06/20/2013] [Accepted: 06/23/2013] [Indexed: 12/04/2022]
Affiliation(s)
- Elliot B Tapper
- Division of Gastroenterology and Hepatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|
35
|
Pettengill M, Robson S, Tresenriter M, Millán JL, Usheva A, Bingham T, Belderbos M, Bergelson I, Burl S, Kampmann B, Gelinas L, Kollmann T, Bont L, Levy O. Soluble ecto-5'-nucleotidase (5'-NT), alkaline phosphatase, and adenosine deaminase (ADA1) activities in neonatal blood favor elevated extracellular adenosine. J Biol Chem 2013; 288:27315-27326. [PMID: 23897810 PMCID: PMC3779727 DOI: 10.1074/jbc.m113.484212] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Extracellular adenosine, a key regulator of physiology and immune cell function that is found at elevated levels in neonatal blood, is generated by phosphohydrolysis of adenine nucleotides released from cells and catabolized by deamination to inosine. Generation of adenosine monophosphate (AMP) in blood is driven by cell-associated enzymes, whereas conversion of AMP to adenosine is largely mediated by soluble enzymes. The identities of the enzymes responsible for these activities in whole blood of neonates have been defined in this study and contrasted to adult blood. We demonstrate that soluble 5′-nucleotidase (5′-NT) and alkaline phosphatase (AP) mediate conversion of AMP to adenosine, whereas soluble adenosine deaminase (ADA) catabolizes adenosine to inosine. Newborn blood plasma demonstrates substantially higher adenosine-generating 5′-NT and AP activity and lower adenosine-metabolizing ADA activity than adult plasma. In addition to a role in soluble purine metabolism, abundant AP expressed on the surface of circulating neonatal neutrophils is the dominant AMPase on these cells. Plasma samples from infant observational cohorts reveal a relative plasma ADA deficiency at birth, followed by a gradual maturation of plasma ADA through infancy. The robust adenosine-generating capacity of neonates appears functionally relevant because supplementation with AMP inhibited whereas selective pharmacologic inhibition of 5′-NT enhanced Toll-like receptor-mediated TNF-α production in neonatal whole blood. Overall, we have characterized previously unrecognized age-dependent expression patterns of plasma purine-metabolizing enzymes that result in elevated plasma concentrations of anti-inflammatory adenosine in newborns. Targeted manipulation of purine-metabolizing enzymes may benefit this vulnerable population.
Collapse
Affiliation(s)
- Matthew Pettengill
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115; Harvard Medical School, Boston, Massachusetts 02115
| | - Simon Robson
- Harvard Medical School, Boston, Massachusetts 02115; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
| | - Megan Tresenriter
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Anny Usheva
- Harvard Medical School, Boston, Massachusetts 02115; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
| | - Taiese Bingham
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115; Harvard Medical School, Boston, Massachusetts 02115
| | - Mirjam Belderbos
- Department of Pediatrics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ilana Bergelson
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115
| | - Sarah Burl
- Vaccinology Theme Group, Medical Research Council Unit, Fajara, The Gambia; Department of Pediatrics, Imperial College London, London W2 IPG, United Kingdom
| | - Beate Kampmann
- Vaccinology Theme Group, Medical Research Council Unit, Fajara, The Gambia; Department of Pediatrics, Imperial College London, London W2 IPG, United Kingdom
| | - Laura Gelinas
- Experimental Medicine Program, Department of Medicine
| | - Tobias Kollmann
- Division of Infectious and Immunologic Diseases, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Louis Bont
- Department of Pediatrics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115; Harvard Medical School, Boston, Massachusetts 02115.
| |
Collapse
|
36
|
Increased plasma levels of microparticles expressing CD39 and CD133 in acute liver injury. Transplantation 2013; 95:63-9. [PMID: 23232366 DOI: 10.1097/tp.0b013e318278d3cd] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We have previously demonstrated that CD133 and CD39 are expressed by hematopoietic stem cells (HSC), which are mobilized after liver injury and target sites of injury, limit vascular inflammation, and boost hepatic regeneration. Plasma microparticles (MP) expressing CD39 can block endothelial activation. Here, we tested whether CD133 MP might be shed in a CD39-dependent manner in a model of liver injury and could potentially serve as biomarkers of liver failure in the clinic. METHODS Wild-type and Cd39-null mice were subjected to acetaminophen-induced liver injury. Mice were sacrificed and plasma MP were isolated by ultracentrifugation. HSC and CD133 MP levels were analyzed by fluorescence-activated cell sorting. Patients were enrolled with acute (n=5) and acute on chronic (n=5) liver injury with matched controls (n=7). Blood was collected at admission and plasma CD133 and CD39 MP subsets were analyzed by fluorescence-activated cell sorting. RESULTS HSC and CD133 MP levels were significantly increased only in the plasma of wild-type mice with acetaminophen hepatotoxicity (P<0.05). No increases in CD133 MP were noted in Cd39-null mice. Plasma MP increases were observed in patients with liver injury. These MP were characterized by significantly higher levels of CD39 (P<0.05). CONCLUSIONS HSC and plasma CD133 MP levels increase in a CD39-dependent manner during experimental acute liver injury. Increased levels of CD39 MP are differentially noted in patients with liver injury. Further research is needed to determine whether MP fluxes are secondary to pathophysiologic insults to the liver or might reflect compensatory responses.
Collapse
|
37
|
Abstract
Membrane microparticles are submicron fragments of membrane shed into extracellular space from cells under conditions of stress/injury. They may be distinguished from other classes of extracellular vesicles (i.e. exosomes) on the basis of size, content and mechanism of formation. Microparticles are found in plasma and other biological fluids from healthy individuals and their levels are altered in various diseases, including diabetes, chronic kidney disease, pre-eclampsia and hypertension among others. Accordingly, they have been considered biomarkers of vascular injury and pro-thrombotic or pro-inflammatory conditions. In addition to this, emerging evidence suggests that microparticles are not simply a consequence of disease, but that they themselves may contribute to pathological processes. Thus microparticles appear to serve as both markers and mediators of pathology. The present review examines the evidence for microparticles as both biomarkers of, and contributors to, the progression of disease. Approaches for the detection of microparticles are summarized and novel concepts relating to the formation of microparticles and their biological effects are examined.
Collapse
|
38
|
Delayed targeting of CD39 to activated platelet GPIIb/IIIa via a single-chain antibody: breaking the link between antithrombotic potency and bleeding? Blood 2013; 121:3067-75. [PMID: 23380744 DOI: 10.1182/blood-2012-08-449694] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ecto-nucleoside triphosphate diphosphohydrolase CD39 represents a promising antithrombotic therapeutic. It degrades adenosine 5'-diphosphate (ADP), a main platelet activating/recruiting agent. We hypothesized that delayed enrichment of CD39 on developing thrombi will allow for a low and safe systemic concentration and thus avoid bleeding. We use a single-chain antibody (scFv, specific for activated GPIIb/IIIa) for targeting CD39. This should allow delayed enrichment on growing thrombi but not on the initial sealing layer of platelets, which do not yet express activated GPIIb/IIIa. CD39 was recombinantly fused to an activated GPIIb/IIIa-specific scFv (targ-CD39) and a nonfunctional scFv (non-targ-CD39). Targ-CD39 was more effective at preventing ADP-induced platelet activation than non-targ-CD39. In a mouse carotid artery thrombosis model, non-targ-CD39, although protective against vessel occlusion, was associated with significant bleeding on tail transection. In contrast, targ-CD39 concentrated at the thrombus site; hence, a dose ∼10 times less of CD39 prevented vessel occlusion to a similar extent as high-dose non-targ-CD39, without prolonged bleeding time. An equimolar dose of non-targ-CD39 at this low concentration was ineffective at preventing vessel occlusion. Thus, delayed targeting of CD39 via scFv to activated platelets provides strong antithrombotic potency and yet prevents bleeding and thereby promotes CD39 toward clinical use.
Collapse
|
39
|
Rozisky JR, Nonose Y, Laste G, Dos Santos VS, de Macedo IC, Battastini AMO, Caumo W, Torres IL. Morphine treatment alters nucleotidase activities in rat blood serum. J Exp Pharmacol 2012; 4:187-93. [PMID: 27186131 PMCID: PMC4863557 DOI: 10.2147/jep.s34033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Morphine has been widely used in neonatal pain management. However, this treatment may produce adaptive changes in several physiologic systems. Our laboratory has demonstrated that morphine treatment in neonate rats alters nucleoside triphosphate diphosphohydrolase (NTPDase) activity and gene expression in central nervous system structures. Considering the relationship between the opioid and purinergic systems, our aim was to verify whether treatment with morphine from postnatal days 8 (P8) through 14 (P14) at a dose of 5 μg per day alters NTPDase and 5′-nucleotidase activities in rat serum over the short, medium, and long terms. After the in vivo assay, the morphine group showed increased hydrolysis of all nucleotides at P30, and a decrease in adenosine 5′-diphosphate hydrolysis at P60. Moreover, we found that nucleotidase activities change with age; adenosine 5′-triphosphate hydrolysis activity was lower at P16, and adenosine 5′-monophosphate hydrolysis activity was higher at P60. These changes are very important because these enzymes are the main regulators of blood nucleotide levels and, consequently, nucleotide signaling. Our findings showed that in vivo morphine treatment alters nucleotide hydrolysis in rat blood serum, suggesting that purine homeostasis can be influenced by opioid treatment during the neonatal period.
Collapse
Affiliation(s)
- Joanna Ripoll Rozisky
- Laboratório de Farmacologia da Dor e Neuromodulação: Modelos Animais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Medicina, Ciências Médicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Yasmine Nonose
- Laboratório de Farmacologia da Dor e Neuromodulação: Modelos Animais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriela Laste
- Laboratório de Farmacologia da Dor e Neuromodulação: Modelos Animais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Medicina, Ciências Médicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Vinicius Souza Dos Santos
- Laboratório de Farmacologia da Dor e Neuromodulação: Modelos Animais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Isabel Cristina de Macedo
- Laboratório de Farmacologia da Dor e Neuromodulação: Modelos Animais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Medicina, Ciências Médicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação em Medicina, Ciências Médicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil
| | - Wolnei Caumo
- Programa de Pós-Graduação em Medicina, Ciências Médicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Iraci Ls Torres
- Laboratório de Farmacologia da Dor e Neuromodulação: Modelos Animais, Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Medicina, Ciências Médicas, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul, Brazil; Unidade de Experimentação Animal e Grupo de Pesquisa e Pós-Graduação, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
40
|
Roberts V, Lu B, Rajakumar S, Cowan PJ, Dwyer KM. The CD39-adenosinergic axis in the pathogenesis of renal ischemia-reperfusion injury. Purinergic Signal 2012. [PMID: 23188420 DOI: 10.1007/s11302-012-9342-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hypoxic injury occurs when the blood supply to an organ is interrupted; subsequent reperfusion halts ongoing ischemic damage but paradoxically leads to further inflammation. Together this is termed ischemia-reperfusion injury (IRI). IRI is inherent to organ transplantation and impacts both the short- and long-term outcomes of the transplanted organ. Activation of the purinergic signalling pathway is intrinsic to the pathogenesis of, and endogenous response to IRI. Therapies targeting the purinergic pathway in IRI are an attractive avenue for the improvement of transplant outcomes and the basis of ongoing research. This review aims to examine the role of adenosine receptor signalling and the ecto-nucleotidases, CD39 and CD73, in IRI, with a particular focus on renal IRI.
Collapse
Affiliation(s)
- Veena Roberts
- St. Vincent's Hospital Melbourne, Immunology Research Centre, Melbourne, Australia.
| | | | | | | | | |
Collapse
|
41
|
Zimmermann H, Zebisch M, Sträter N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal 2012; 8:437-502. [PMID: 22555564 PMCID: PMC3360096 DOI: 10.1007/s11302-012-9309-4] [Citation(s) in RCA: 778] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 02/01/2012] [Indexed: 12/12/2022] Open
Abstract
Ecto-nucleotidases play a pivotal role in purinergic signal transmission. They hydrolyze extracellular nucleotides and thus can control their availability at purinergic P2 receptors. They generate extracellular nucleosides for cellular reuptake and salvage via nucleoside transporters of the plasma membrane. The extracellular adenosine formed acts as an agonist of purinergic P1 receptors. They also can produce and hydrolyze extracellular inorganic pyrophosphate that is of major relevance in the control of bone mineralization. This review discusses and compares four major groups of ecto-nucleotidases: the ecto-nucleoside triphosphate diphosphohydrolases, ecto-5'-nucleotidase, ecto-nucleotide pyrophosphatase/phosphodiesterases, and alkaline phosphatases. Only recently and based on crystal structures, detailed information regarding the spatial structures and catalytic mechanisms has become available for members of these four ecto-nucleotidase families. This permits detailed predictions of their catalytic mechanisms and a comparison between the individual enzyme groups. The review focuses on the principal biochemical, cell biological, catalytic, and structural properties of the enzymes and provides brief reference to tissue distribution, and physiological and pathophysiological functions.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Institute of Cell Biology and Neuroscience, Molecular and Cellular Neurobiology, Biologicum, Goethe-University Frankfurt, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| | | | | |
Collapse
|
42
|
Visovatti SH, Hyman MC, Bouis D, Neubig R, McLaughlin VV, Pinsky DJ. Increased CD39 nucleotidase activity on microparticles from patients with idiopathic pulmonary arterial hypertension. PLoS One 2012; 7:e40829. [PMID: 22792409 PMCID: PMC3394716 DOI: 10.1371/journal.pone.0040829] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 06/17/2012] [Indexed: 12/02/2022] Open
Abstract
Background Idiopathic pulmonary arterial hypertension (IPAH) is a devastating disease characterized by increased pulmonary vascular resistance, smooth muscle and endothelial cell proliferation, perivascular inflammatory infiltrates, and in situ thrombosis. Circulating intravascular ATP, ADP, AMP and adenosine activate purinergic cell signaling pathways and appear to induce many of the same pathologic processes that underlie IPAH. Extracellular dephosphorylation of ATP to ADP and AMP occurs primarily via CD39 (ENTPD1), an ectonucleotidase found on the surface of leukocytes, platelets, and endothelial cells [1]. Microparticles are micron-sized phospholipid vesicles formed from the membranes of platelets and endothelial cells. Objectives: Studies here examine whether CD39 is an important microparticle surface nucleotidase, and whether patients with IPAH have altered microparticle-bound CD39 activity that may contribute to the pathophysiology of the disease. Methodology/ Principal Findings Kinetic parameters, inhibitor blocking experiments, and immunogold labeling with electron microscopy support the role of CD39 as a major nucleotidase on the surface of microparticles. Comparison of microparticle surface CD39 expression and nucleotidase activity in 10 patients with advanced IPAH and 10 healthy controls using flow cytometry and thin layer chromatograph demonstrate the following: 1) circulating platelet (CD39+CD31+CD42b+) and endothelial (CD39+CD31+CD42b−) microparticle subpopulations in patients with IPAH show increased CD39 expression; 2) microparticle ATPase and ADPase activity in patients with IPAH is increased. Conclusions/ Significance We demonstrate for the first time increased CD39 expression and function on circulating microparticles in patients with IPAH. Further research is needed to elucidate whether these findings identify an important trigger for the development of the disease, or reflect a physiologic response to IPAH.
Collapse
Affiliation(s)
- Scott H Visovatti
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America.
| | | | | | | | | | | |
Collapse
|
43
|
Yegutkin GG, Wieringa B, Robson SC, Jalkanen S. Metabolism of circulating ADP in the bloodstream is mediated via integrated actions of soluble adenylate kinase-1 and NTPDase1/CD39 activities. FASEB J 2012; 26:3875-83. [PMID: 22637533 DOI: 10.1096/fj.12-205658] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Extracellular ATP and ADP trigger inflammatory, vasodilatatory, and prothrombotic signaling events in the vasculature, and their turnover is governed by networks of membrane-associated enzymes. The contribution of soluble activities to intravascular nucleotide homeostasis remains controversial. By using thin-layer chromatographic assays, we revealed transphosphorylation of [γ-(32)P]ATP and AMP by human and murine sera, which was progressively inhibited by specific adenylate kinase (AK) inhibitor Ap(5)A. This phosphotransfer reaction was diminished markedly in serum from knockout mice lacking the major AK isoform, AK1, and in human serum immunodepleted of AK1. We also showed that ∼75% ADP in cell-free serum is metabolized via reversible AK1 reaction 2ADP ↔ ATP + AMP. The generated ATP and AMP are then metabolized through the coupled nucleotide pyrophosphatase/phosphodiesterase and 5'-nucleotidase/CD73 reactions, respectively. Constitutive presence of another nucleotide-converting enzyme, nucleoside triphosphate diphosphohydrolase-1 (NTPDase1, known as CD39), was ascertained by the relative deficiency of serum from CD39-null mice to dephosphorylate [(3)H]ADP and [γ-(32)P]ATP, and also by diminished [(3)H]ADP hydrolysis by human serum pretreated with NTPDase1 inhibitors, POM-1 and ARL-67156. In summary, we have identified hitherto unrecognized soluble forms of AK1 and NTPDase1/CD39 that contribute in the active cycling between the principal platelet-recruiting agent ADP and other circulating nucleotides.
Collapse
Affiliation(s)
- Gennady G Yegutkin
- MediCity Research Laboratory, University of Turku, Tykistökatu 6A, 20520 Turku, Finland.
| | | | | | | |
Collapse
|
44
|
Andriantsitohaina R, Gaceb A, Vergori L, Martínez MC. Microparticles as Regulators of Cardiovascular Inflammation. Trends Cardiovasc Med 2012; 22:88-92. [DOI: 10.1016/j.tcm.2012.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Schmelzle M, Cowan PJ, Robson SC. Which anti-platelet therapies might be beneficial in xenotransplantation? Xenotransplantation 2011; 18:79-87. [PMID: 21496115 DOI: 10.1111/j.1399-3089.2011.00628.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Xenotransplantation could provide an unlimited and elective supply of grafts, once mechanisms of graft loss and vascular injury are better understood. The development of α-1,3-galactosyltransferase gene-knockout (GalT-KO) swine with the removal of a dominant xeno-antigen has been an important advance; however, delayed xenograft and acute vascular reaction in GalT-KO animals persist. These occur, at least in part, because of humoral reactions that result in vascular injury. Intrinsic molecular incompatibilities in the regulation of blood clotting and extracellular nucleotide homeostasis between discordant species may also predispose to thrombophilia within the vasculature of xenografts. Although limited benefits have been achieved with currently available pharmacological anti-thrombotics and anti-coagulants, the highly complex mechanisms of platelet activation and thrombosis in xenograft rejection also require potent immunosuppressive interventions. We will focus on recent thromboregulatory approaches while elucidating appropriate anti-platelet mechanisms. We will discuss potential benefits of additional anti-thrombotic interventions that are possible in transgenic swine and review recent developments in pharmacological anti-platelet therapy.
Collapse
Affiliation(s)
- Moritz Schmelzle
- Liver Center and Transplantation Institute, Department of Medicine and Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
46
|
Ceruti S, Colombo L, Magni G, Viganò F, Boccazzi M, Deli MA, Sperlágh B, Abbracchio MP, Kittel A. Oxygen-glucose deprivation increases the enzymatic activity and the microvesicle-mediated release of ectonucleotidases in the cells composing the blood-brain barrier. Neurochem Int 2011; 59:259-71. [PMID: 21672581 DOI: 10.1016/j.neuint.2011.05.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 05/19/2011] [Accepted: 05/26/2011] [Indexed: 01/25/2023]
Abstract
The blood-brain barrier (BBB), the dynamic interface between the nervous tissue and the blood, is composed by endothelial cells, pericytes and astrocytes. Extracellular nucleotides and nucleosides and their receptors (the purinergic system) constitute a widely diffused signaling system involved in many pathophysiological processes. However, the role of this system in controlling BBB functions is still largely unknown. By using cultures of these three cell types grown separately and a BBB in vitro model consisting of triple co-cultures, we studied for the first time the expression and distribution of the ecto-enzymes nucleoside triphosphate diphosphohydrolases (NTPDases, the enzymes which hydrolyze extracellular nucleotides) under control and ischemic (oxygen-glucose deprivation in vitro; OGD) conditions. NTPDase1 was detected in all three cell types, whereas NTPDase2 was expressed by astrocytes and pericytes and, to a lesser extent, by endothelial cells. Endothelial cells were extremely susceptible to cell death when OGD was applied to mimic in vitro the cytotoxicity induced by ischemia, whereas astrocytes and pericytes were more resistant. A semi-quantitative assay highlighted markedly increased e-ATPase activity following exposure to OGD in all three cell types, either when grown separately or when co-cultured together to resemble the composition of the BBB. Moreover, electron microscopy analysis showed that both endothelial cells and astrocytes shed microvesicles containing NTPDases from their membrane, which may suggest a novel mechanism to increase the breakdown of ATP released to toxic levels by damaged BBB cells. We hypothesize that this phenomenon could have a protective and/or modulatory effect for brain parenchymal cells. This in vitro model is therefore useful to study the role of extracellular nucleotides in modulating BBB responses to ischemic events, and to develop new effective purinergic-based approaches for brain ischemia.
Collapse
Affiliation(s)
- Stefania Ceruti
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological Sciences, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Straub A, Krajewski S, Hohmann JD, Westein E, Jia F, Bassler N, Selan C, Kurz J, Wendel HP, Dezfouli S, Yuan Y, Nandurkar H, Jackson S, Hickey MJ, Peter K. Evidence of platelet activation at medically used hypothermia and mechanistic data indicating ADP as a key mediator and therapeutic target. Arterioscler Thromb Vasc Biol 2011; 31:1607-16. [PMID: 21512161 DOI: 10.1161/atvbaha.111.226373] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Hypothermia is used in various clinical settings to inhibit ischemia-related organ damage. However, prothrombotic effects have been described as potential side effects. This study aimed to elucidate the mechanism of hypothermia-induced platelet activation and subsequent prothrombotic events and to develop preventative pharmacological strategies applicable during clinically used hypothermia. METHODS AND RESULTS Platelet function was investigated ex vivo and in vivo at clinically used hypothermia (28°C/18°C). Hypothermic mice demonstrated increased expression of platelet activation marker P-selectin, platelet-leukocyte aggregate formation, and thrombocytopenia. Intravital microscopy of FeCl(3)-injured murine mesenteric arteries revealed increased platelet thrombus formation with hypothermia. Ex vivo flow chamber experiments indicated increased platelet-fibrinogen adhesion under hypothermia. We show that hypothermia results in reduced ADP hydrolysis via reduction of CD39 (E-NTPDase1) activity, resulting in increased levels of ADP and subsequent augmented primary and secondary platelet activation. In vivo administration of ADP receptor P(2)Y(12) antagonists and recombinant soluble CD39 prevented hypothermia-induced thrombus formation and thrombocytopenia, respectively. CONCLUSIONS The platelet agonist ADP plays a key role in hypothermia-induced platelet activation. Inhibition of receptor binding or hydrolysis of ADP has the potential to protect platelets against hypothermia-induced activation. Our findings provide a rational basis for further evaluation of novel antithrombotic strategies in clinically applied hypothermia.
Collapse
Affiliation(s)
- Andreas Straub
- Atherothrombosis and Vascular Biology, Baker IDI Heart and Diabetes Institute, PO Box 6492 St Kilda Rd Central, Melbourne, Victoria 8008, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Jones S, Evans RJ, Mahaut-Smith MP. Extracellular Ca(2+) modulates ADP-evoked aggregation through altered agonist degradation: implications for conditions used to study P2Y receptor activation. Br J Haematol 2011; 153:83-91. [PMID: 21332705 PMCID: PMC3084511 DOI: 10.1111/j.1365-2141.2010.08499.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
ADP is considered a weak platelet agonist due to the limited aggregation responses it induces in vitro at physiological concentrations of extracellular Ca2+ [(Ca2+)o]. Lowering [Ca2+]o paradoxically enhances ADP-evoked aggregation, an effect that has been attributed to enhanced thromboxane A2 production. This study examined the role of ectonucleotidases in the [Ca2+]o-dependence of platelet activation. Reducing [Ca2+]o from millimolar to micromolar levels converted ADP (10 μmol/l)-evoked platelet aggregation from a transient to a sustained response in both platelet-rich plasma and washed suspensions. Blocking thromboxane A2 production with aspirin had no effect on this [Ca2+]o-dependence. Prevention of ADP degradation abolished the differences between low and physiological [Ca2+]o resulting in a robust and sustained aggregation in both conditions. Measurements of extracellular ADP revealed reduced degradation in both plasma and apyrase-containing saline at micromolar compared to millimolar [Ca2+]o. As reported previously, thromboxane A2 generation was enhanced at low [Ca2+]o, however this was independent of ectonucleotidase activity. P2Y receptor antagonists cangrelor and MRS2179 demonstrated the necessity of P2Y12 receptors for sustained ADP-evoked aggregation, with a minor role for P2Y1. In conclusion, Ca2+-dependent ectonucleotidase activity is a major factor determining the extent of platelet aggregation to ADP and must be controlled for in studies of P2Y receptor activation.
Collapse
Affiliation(s)
- Sarah Jones
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | | | | |
Collapse
|
49
|
Deaglio S, Robson SC. Ectonucleotidases as regulators of purinergic signaling in thrombosis, inflammation, and immunity. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2011; 61:301-32. [PMID: 21586363 PMCID: PMC5879773 DOI: 10.1016/b978-0-12-385526-8.00010-2] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Evolving studies in models of transplant rejection, inflammatory bowel disease, and cancer, among others, have implicated purinergic signaling in clinical manifestations of vascular injury and thrombophilia, inflammation, and immune disturbance. Within the vasculature, spatial and temporal expression of CD39 nucleoside triphosphate diphosphohydrolase (NTPDase) family members together with CD73 ecto-5'-nucleotidase control platelet activation, thrombus size, and stability. This is achieved by closely regulated phosphohydrolytic activities to scavenge extracellular nucleotides, maintain P2-receptor integrity, and coordinate adenosinergic signaling responses. The CD38/CD157 family of extracellular NADases degrades NAD(+) and generates Ca(2+)-active metabolites, including cyclic ADP ribose and ADP ribose. These mediators regulate leukocyte adhesion and chemotaxis. These mechanisms are crucial in vascular homeostasis, hemostasis, thrombogenesis, and during inflammation. There has been recent interest in ectonucleotidase expression by immune cells. CD39 expression identifies Langerhans-type dendritic cells and efficiently distinguishes T regulatory cells from other resting or activated T cells. CD39, together with CD73 in mice, serves as an integral component of the suppressive machinery of T cells. Purinergic responses also impact generation of T helper-type 17 cells. Further, CD38 and changes in NAD(+) availability modulate ADP ribosylation of the cytolytic P2X7 receptor that deletes T regulatory cells. Expression of CD39, CD73, and CD38 ectonucleotidases on either endothelial or immune cells allows for homeostatic integration and control of vascular inflammatory and immune cell reactions at sites of injury. Ongoing development of therapeutic strategies targeting these and other ectonucleotidases offers promise for the management of vascular thrombosis, disordered inflammation, and aberrant immune reactivity.
Collapse
Affiliation(s)
- Silvia Deaglio
- Department of Genetics, Biology, and Biochemistry, University of Turin & Human Genetics Foundation, Italy
| | | |
Collapse
|
50
|
Mause SF, Weber C. Microparticles: protagonists of a novel communication network for intercellular information exchange. Circ Res 2010; 107:1047-57. [PMID: 21030722 DOI: 10.1161/circresaha.110.226456] [Citation(s) in RCA: 609] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Microparticles represent a heterogeneous population of vesicles with a diameter of 100 to 1000 nm that are released by budding of the plasma membrane and express antigens specific of their parental cells. Although microparticle formation represents a physiological phenomenon, a multitude of pathologies are associated with a considerable increase in circulating microparticles, including inflammatory and autoimmune diseases, atherosclerosis, and malignancies. Microparticles display an broad spectrum of bioactive substances and receptors on their surface and harbor a concentrated set of cytokines, signaling proteins, mRNA, and microRNA. Recent studies provided evidence for the concept of microparticles as veritable vectors for the intercellular exchange of biological signals and information. Indeed, microparticles may transfer part of their components and content to selected target cells, thus mediating cell activation, phenotypic modification, and reprogramming of cell function. Because microparticles readily circulate in the vasculature, they may serve as shuttle modules and signaling transducers not only in their local environment but also at remarkable distance from their site of origin. Altogether, this transcellular delivery system may extend the confines of the limited transcriptome and proteome of recipient cells and establishes a communication network in which specific properties and information among cells can be efficiently shared. At least in same cases, the sequential steps of the transfer process underlie complex regulatory mechanisms, including selective sorting ("packaging") of microparticle components and content, specificity of interactions with target cells determined by surface receptors, and ultimately finely tuned and signal-dependent release and delivery of microparticle content.
Collapse
Affiliation(s)
- Sebastian F Mause
- Institut für Molekulare Herz-Kreislaufforschung, Universitätsklinikum Aachen, Pauwelsstrasse 30, Aachen, Germany
| | | |
Collapse
|