1
|
Gerasimovskaya E, Patil RS, Davies A, Maloney ME, Simon L, Mohamed B, Cherian-Shaw M, Verin AD. Extracellular purines in lung endothelial permeability and pulmonary diseases. Front Physiol 2024; 15:1450673. [PMID: 39234309 PMCID: PMC11372795 DOI: 10.3389/fphys.2024.1450673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
The purinergic signaling system is an evolutionarily conserved and critical regulatory circuit that maintains homeostatic balance across various organ systems and cell types by providing compensatory responses to diverse pathologies. Despite cardiovascular diseases taking a leading position in human morbidity and mortality worldwide, pulmonary diseases represent significant health concerns as well. The endothelium of both pulmonary and systemic circulation (bronchial vessels) plays a pivotal role in maintaining lung tissue homeostasis by providing an active barrier and modulating adhesion and infiltration of inflammatory cells. However, investigations into purinergic regulation of lung endothelium have remained limited, despite widespread recognition of the role of extracellular nucleotides and adenosine in hypoxic, inflammatory, and immune responses within the pulmonary microenvironment. In this review, we provide an overview of the basic aspects of purinergic signaling in vascular endothelium and highlight recent studies focusing on pulmonary microvascular endothelial cells and endothelial cells from the pulmonary artery vasa vasorum. Through this compilation of research findings, we aim to shed light on the emerging insights into the purinergic modulation of pulmonary endothelial function and its implications for lung health and disease.
Collapse
Affiliation(s)
| | - Rahul S Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Adrian Davies
- Department of Internal Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - McKenzie E Maloney
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Office of Academic Affairs, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liselle Simon
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Basmah Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
2
|
Kidder E, Gangopadhyay S, Francis S, Alfaidi M. "How to Release or Not Release, That Is the Question." A Review of Interleukin-1 Cellular Release Mechanisms in Vascular Inflammation. J Am Heart Assoc 2024; 13:e032987. [PMID: 38390810 PMCID: PMC10944040 DOI: 10.1161/jaha.123.032987] [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: 10/06/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide, characterized by atherosclerotic activity within large and medium-sized arteries. Inflammation has been shown to be a primary driver of atherosclerotic plaque formation, with interleukin-1 (IL-1) having a principal role. This review focuses on the current state of knowledge of molecular mechanisms of IL-1 release from cells in atherosclerotic plaques. A more in-depth understanding of the process of IL-1's release into the vascular environment is necessary for the treatment of inflammatory disease processes, as the current selection of medicines being used primarily target IL-1 after it has been released. IL-1 is secreted by several heterogenous mechanisms, some of which are cell type-specific and could provide further specialized targets for therapeutic intervention. A major unmet challenge is to understand the mechanism before and leading to IL-1 release, especially by cells in atherosclerotic plaques, including endothelial cells, vascular smooth muscle cells, and macrophages. Data so far indicate a heterogeneity of IL-1 release mechanisms that vary according to cell type and are stimulus-dependent. Unraveling this complexity may reveal new targets to block excess vascular inflammation.
Collapse
Affiliation(s)
- Evan Kidder
- Division of Cardiology, Department of Internal MedicineLouisiana State University Health Sciences CentreShreveportLAUSA
| | - Siddhartha Gangopadhyay
- Division of Cardiology, Department of Internal MedicineLouisiana State University Health Sciences CentreShreveportLAUSA
| | - Sheila Francis
- School of Medicine and Population HealthUniversity of SheffieldSheffieldUK
| | - Mabruka Alfaidi
- Division of Cardiology, Department of Internal MedicineLouisiana State University Health Sciences CentreShreveportLAUSA
| |
Collapse
|
3
|
Sudi S, Thomas FM, Daud SK, Ag Daud DM, Sunggip C. The Pleiotropic Role of Extracellular ATP in Myocardial Remodelling. Molecules 2023; 28:molecules28052102. [PMID: 36903347 PMCID: PMC10004151 DOI: 10.3390/molecules28052102] [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: 01/26/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/12/2023] Open
Abstract
Myocardial remodelling is a molecular, cellular, and interstitial adaptation of the heart in response to altered environmental demands. The heart undergoes reversible physiological remodelling in response to changes in mechanical loading or irreversible pathological remodelling induced by neurohumoral factors and chronic stress, leading to heart failure. Adenosine triphosphate (ATP) is one of the potent mediators in cardiovascular signalling that act on the ligand-gated (P2X) and G-protein-coupled (P2Y) purinoceptors via the autocrine or paracrine manners. These activations mediate numerous intracellular communications by modulating the production of other messengers, including calcium, growth factors, cytokines, and nitric oxide. ATP is known to play a pleiotropic role in cardiovascular pathophysiology, making it a reliable biomarker for cardiac protection. This review outlines the sources of ATP released under physiological and pathological stress and its cell-specific mechanism of action. We further highlight a series of cardiovascular cell-to-cell communications of extracellular ATP signalling cascades in cardiac remodelling, which can be seen in hypertension, ischemia/reperfusion injury, fibrosis, hypertrophy, and atrophy. Finally, we summarize current pharmacological intervention using the ATP network as a target for cardiac protection. A better understanding of ATP communication in myocardial remodelling could be worthwhile for future drug development and repurposing and the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Suhaini Sudi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Fiona Macniesia Thomas
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Siti Kadzirah Daud
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Dayang Maryama Ag Daud
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Health through Exercise and Active Living (HEAL) Research Unit, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Caroline Sunggip
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Correspondence:
| |
Collapse
|
4
|
Li R, Ma W, Lang H, He H, Lv L, Zhang Y, Chen X, Hou L. Hemostatic mechanism of the effect of Jianpi Yiqi Shexue decoction on vascular factors in immune thrombocytopenia model mice. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2022. [DOI: 10.1016/j.jtcms.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
5
|
Comish PB, Carlson D, Kang R, Tang D. Damage-Associated Molecular Patterns and the Systemic Immune Consequences of Severe Thermal Injury. THE JOURNAL OF IMMUNOLOGY 2021; 205:1189-1197. [PMID: 32839211 DOI: 10.4049/jimmunol.2000439] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/05/2020] [Indexed: 12/31/2022]
Abstract
Thermal injury is often associated with a proinflammatory state resulting in serious complications. After a burn, the innate immune system is activated with subsequent immune cell infiltration and cytokine production. Although the innate immune response is typically beneficial, an excessive activation leads to cytokine storms, multiple organ failure, and even death. This overwhelming immune response is regulated by damage-associated molecular patterns (DAMPs). DAMPs are endogenous molecules that are actively secreted by immune cells or passively released by dead or dying cells that can bind to pathogen recognition receptors in immune and nonimmune cells. Recent studies involving animal models along with human studies have drawn great attention to the possible pathological role of DAMPs as an immune consequence of thermal injury. In this review, we outline DAMPs and their function in thermal injury, shedding light on the mechanism of sterile inflammation during tissue injury and identifying new immune targets for treating thermal injury.
Collapse
Affiliation(s)
- Paul B Comish
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Deborah Carlson
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
6
|
Andrejew R, Glaser T, Oliveira-Giacomelli Á, Ribeiro D, Godoy M, Granato A, Ulrich H. Targeting Purinergic Signaling and Cell Therapy in Cardiovascular and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:275-353. [PMID: 31898792 DOI: 10.1007/978-3-030-31206-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular purines exert several functions in physiological and pathophysiological mechanisms. ATP acts through P2 receptors as a neurotransmitter and neuromodulator and modulates heart contractility, while adenosine participates in neurotransmission, blood pressure, and many other mechanisms. Because of their capability to differentiate into mature cell types, they provide a unique therapeutic strategy for regenerating damaged tissue, such as in cardiovascular and neurodegenerative diseases. Purinergic signaling is pivotal for controlling stem cell differentiation and phenotype determination. Proliferation, differentiation, and apoptosis of stem cells of various origins are regulated by purinergic receptors. In this chapter, we selected neurodegenerative and cardiovascular diseases with clinical trials using cell therapy and purinergic receptor targeting. We discuss these approaches as therapeutic alternatives to neurodegenerative and cardiovascular diseases. For instance, promising results were demonstrated in the utilization of mesenchymal stem cells and bone marrow mononuclear cells in vascular regeneration. Regarding neurodegenerative diseases, in general, P2X7 and A2A receptors mostly worsen the degenerative state. Stem cell-based therapy, mainly through mesenchymal and hematopoietic stem cells, showed promising results in improving symptoms caused by neurodegeneration. We propose that purinergic receptor activity regulation combined with stem cells could enhance proliferative and differentiation rates as well as cell engraftment.
Collapse
Affiliation(s)
- Roberta Andrejew
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Talita Glaser
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Ágatha Oliveira-Giacomelli
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Deidiane Ribeiro
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Mariana Godoy
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.,Laboratory of Neurodegenerative Diseases, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro Granato
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Neuroscience Laboratory, Institute of Chemistry, Department of Biochemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
7
|
Abstract
Calcification is a regulated physiological process occurring in bones and teeth. However, calcification is commonly found in soft tissues in association with aging and in a variety of diseases. Over the last two decades, it has emerged that calcification occurring in diseased arteries is not simply an inevitable build-up of insoluble precipitates of calcium phosphate. In some cases, it is an active process in which transcription factors drive conversion of vascular cells to an osteoblast or chondrocyte-like phenotype, with the subsequent production of mineralizing "matrix vesicles." Early studies of bone and cartilage calcification suggested roles for cellular calcium signaling in several of the processes involved in the regulation of bone calcification. Similarly, calcium signaling has recently been highlighted as an important component in the mechanisms regulating pathological calcification. The emerging hypothesis is that ectopic/pathological calcification occurs in tissues in which there is an imbalance in the regulatory mechanisms that actively prevent calcification. This review highlights the various ways that calcium signaling regulates tissue calcification, with a particular focus on pathological vascular calcification.
Collapse
Affiliation(s)
- Diane Proudfoot
- Signalling Division, Babraham Institute, Babraham, Cambridge CB22 3AT, United Kingdom
| |
Collapse
|
8
|
Untargeted Metabolic Profiling Cell-Based Approach of Pulmonary Artery Smooth Muscle Cells in Response to High Glucose and the Effect of the Antioxidant Vitamins D and E. Metabolites 2018; 8:metabo8040087. [PMID: 30513640 PMCID: PMC6316736 DOI: 10.3390/metabo8040087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multi-factorial disease characterized by the hyperproliferation of pulmonary artery smooth muscle cells (PASMCs). Excessive reactive oxygen species (ROS) formation resulted in alterations of the structure and function of pulmonary arterial walls, leading to right ventricular failure and death. Diabetes mellitus has not yet been implicated in pulmonary hypertension. However, recently, variable studies have shown that diabetes is correlated with pulmonary hypertension pathobiology, which could participate in the modification of pulmonary artery muscles. The metabolomic changes in PASMCs were studied in response to 25 mM of D-glucose (high glucose, or HG) in order to establish a diabetic-like condition in an in vitro setting, and compared to five mM of D-glucose (normal glucose, or LG). The effect of co-culturing these cells with an ideal blood serum concentration of cholecalciferol-D3 and tocopherol was also examined. The current study aimed to examine the role of hyperglycemia in pulmonary arterial hypertension by the quantification and detection of the metabolomic alteration of smooth muscle cells in high-glucose conditions. Untargeted metabolomics was carried out using hydrophilic interaction liquid chromatography and high-resolution mass spectrometry. Cell proliferation was assessed by cell viability and the [³H] thymidine incorporation assay, and the redox state within the cells was examined by measuring reactive oxygen species (ROS) generation. The results demonstrated that PASMCs in high glucose (HG) grew, proliferated faster, and generated higher levels of superoxide anion (O₂·-) and hydrogen peroxide (H₂O₂). The metabolomics of cells cultured in HG showed that the carbohydrate pathway, especially that of the upper glycolytic pathway metabolites, was influenced by the activation of the oxidation pathway: the pentose phosphate pathway (PPP). The amount of amino acids such as aspartate and glutathione reduced via HG, while glutathione disulfide, N6-Acetyl-L-lysine, glutamate, and 5-aminopentanoate increased. Lipids either as fatty acids or glycerophospholipids were downregulated in most of the metabolites, with the exception of docosatetraenoic acid and PG (16:0/16:1(9Z)). Purine and pyrimidine were influenced by hyperglycaemia following PPP oxidation. The results in addition showed that cells exposed to 25 mM of glucose were oxidatively stressed comparing to those cultured in five mM of glucose. Cholecalciferol (D3, or vitamin D) and tocopherol (vitamin E) were shown to restore the redox status of many metabolic pathways.
Collapse
|
9
|
Nitschke Y, Yan Y, Buers I, Kintziger K, Askew K, Rutsch F. ENPP1-Fc prevents neointima formation in generalized arterial calcification of infancy through the generation of AMP. Exp Mol Med 2018; 50:1-12. [PMID: 30369595 PMCID: PMC6204430 DOI: 10.1038/s12276-018-0163-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/08/2018] [Accepted: 07/12/2018] [Indexed: 12/12/2022] Open
Abstract
Generalized arterial calcification of infancy (GACI) is associated with widespread arterial calcification and stenoses and is caused by mutations in ENPP1. ENPP1 encodes for ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which cleaves ATP to generate inorganic pyrophosphate (PPi) and adenosine monophosphate (AMP) extracellularly. The current study was designed to define the prevalence of arterial stenoses in GACI individuals and to identify the mechanism through which ENPP1 deficiency causes intimal proliferation. Furthermore, we aimed to effectively prevent and treat neointima formation in an animal model of GACI through the systemic administration of recombinant human (rh)ENPP1-Fc protein. Based on a literature review, we report that arterial stenoses are present in at least 72.4% of GACI cases. We evaluated the effect of rhENPP1-Fc on ENPP1-silenced human vascular smooth muscle cells (VSMCs) and on induced intimal proliferation in Enpp1-deficient ttw/ttw mice treated with carotid ligation. We demonstrate that silencing ENPP1 in VSMCs resulted in a tenfold increase in proliferation relative to that of cells transfected with negative control siRNA. The addition of rhENPP1-Fc, AMP or adenosine restored the silenced ENPP1-associated proliferation. In contrast, neither PPi nor etidronate, a current off-label treatment for GACI, had an effect on VSMC proliferation. Furthermore, subcutaneous rhENPP1-Fc protein replacement was effective in preventing and treating intimal hyperplasia induced by carotid ligation in an animal model of GACI. We conclude that ENPP1 inhibits neointima formation by generating AMP. RhENPP1-Fc may serve as an approach for the effective prevention and treatment of arterial stenoses in GACI. A protein replacement therapy may prove useful in tackling calcification and narrowing of the arteries in babies with a severe genetic disorder. Generalized Arterial Calcification of Infancy (GACI) is a rare condition in which infants’ arteries become calcified and their blood vessels internally scarred. It often leads to congestive heart failure. The ENPP1 gene encodes a protein that is crucial to preventing excess calcium build-up in the body. Mutations in the ENPP1 gene lead to GACI, but no therapies for the condition exist. Now, Frank Rutsch at Muenster University Children’s Hospital in Germany and co-workers have shown that administering a protein replacement can inhibit blood vessel scarring and arterial clogging in GACI mice models and in human stem cell cultures. The protein replacement boosts production of a key metabolic molecule called adenosine monophosphate.
Collapse
Affiliation(s)
- Yvonne Nitschke
- Department of General Pediatrics, Münster University Children's Hospital, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany.,Cells in Motion Cluster of Excellence, Münster University, Münster, Germany
| | - Yan Yan
- Alexion Pharmaceuticals, 100 College St, New Haven, CT, USA
| | - Insa Buers
- Department of General Pediatrics, Münster University Children's Hospital, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany.,Cells in Motion Cluster of Excellence, Münster University, Münster, Germany
| | - Kristina Kintziger
- Department of General Pediatrics, Münster University Children's Hospital, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany
| | - Kim Askew
- Alexion Pharmaceuticals, 100 College St, New Haven, CT, USA
| | - Frank Rutsch
- Department of General Pediatrics, Münster University Children's Hospital, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany. .,Cells in Motion Cluster of Excellence, Münster University, Münster, Germany.
| |
Collapse
|
10
|
Danger-Associated Molecular Patterns (DAMPs): the Derivatives and Triggers of Inflammation. Curr Allergy Asthma Rep 2018; 18:63. [PMID: 30267163 DOI: 10.1007/s11882-018-0817-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Allergen is an umbrella term for irritants of diverse origin. Along with other offenders such as pathogens, mutagens, xenobiotics, and pollutants, allergens can be grouped as inflammatory agents. Danger-associated molecular patterns (DAMPs) are altered metabolism products of necrotic or stressed cells, which are deemed as alarm signals by the innate immune system. Like inflammation, DAMPs play a role in correcting the altered physiological state, but in excess, they can be lethal due to their signal transduction roles. In a vicious loop, inflammatory agents are DAMP generators and DAMPs create a pro-inflammatory state. Only a handful of DAMPs such as uric acid, mtDNA, extracellular ATP, HSPs, amyloid β, S100, HMGB1, and ECM proteins have been studied till now. A large number of DAMPs are still obscure, in need to be unveiled. The identification and functional characterization of those DAMPs in inflammation pathways can be insightful. RECENT FINDINGS As inflammation and immune activation have been implicated in almost all pathologies, studies on them have been intensified in recent times. Consequently, the pathologic mechanisms of various DAMPs have emerged. Following PRR ligation, the activation of inflammasome, MAPK, and NF-kB is some of the common pathways. The limited number of recognized DAMPs are only a fraction of the vast array of other DAMPs. In fact, any misplaced or abnormal level of metabolite can be a DAMP. Sophisticated analysis studies can reveal the full profile of the DAMPs. Lowering the level of DAMPs is useful therapeutic intervention but certainly not as effective as avoiding the DAMP generators, i.e., the inflammatory agents. So, rather than mitigating DAMPs, efforts should be focused on the elimination of inflammatory agents.
Collapse
|
11
|
Ng PY, McIntosh KA, Hargrave G, Ho KH, Paul A, Plevin R. Inhibition of cytokine-mediated JNK signalling by purinergic P2Y 11 receptors, a novel protective mechanism in endothelial cells. Cell Signal 2018; 51:59-71. [PMID: 30076967 DOI: 10.1016/j.cellsig.2018.07.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
Previous research from our laboratory has demonstrated a novel phenomenon whereby GPCRs play a role in inhibiting cytokine-mediated c-Jun N-terminal kinase (JNK) signalling. So far this novel phenomenon seems to have been vastly overlooked, with little research in the area. Therefore, in this study we explored this further; by assessing the potential of P2YRs to mediate inhibition of cytokine-mediated JNK signalling and related functional outcomes in human endothelial cells. We utilised primary endothelial cells, and employed the use of endogenous activators of P2YRs and well characterised pharmacological inhibitors, to assess signalling parameters mediated by P2YRs, Interleukin-1β (IL-1β), TNFα and JNK. Activation of P2YRs with adenosine tri-phosphate (ATP) resulted in a time- and concentration-dependent inhibition of IL-1β-mediated phosphorylation of JNK and associated kinase activity. The effect was specific for cytokine-mediated JNK signalling, as ATP was without effect on JNK induced by other non-specific activators (e.g. sorbitol, anisomycin), nor effective against other MAPK pathways such as p38 and the canonical NFκB cascade. Pharmacological studies demonstrated a role for the P2Y11 receptor in mediating this effect, but not the P2Y1 nor the adenosine receptors (A1, A2A, A2B & A3). The novel Gαq/11 inhibitor YM254890 and a protein kinase A (PKA) inhibitor H89 both partially reversed ATP-mediated inhibition of IL-1β-stimulated JNK indicating involvement of both Gαq/11 and Gαs mediated pathways. ATP also partially reversed IL-1β-mediated induction of cyclo‑oxygenase-2 (COX-2) and E-selectin. Collectively, these studies indicate the potential for activation of purinergic receptors to protect the endothelium from inflammatory driven JNK activation and may be a new target for inflammatory disease therapy.
Collapse
Affiliation(s)
- Pei Y Ng
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, Scotland, UK
| | - Kathryn A McIntosh
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, Scotland, UK.
| | - Gillian Hargrave
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, Scotland, UK
| | - Ka H Ho
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, Scotland, UK
| | - Andrew Paul
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, Scotland, UK
| | - Robin Plevin
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, Scotland, UK
| |
Collapse
|
12
|
Dautova Y, Kapustin AN, Pappert K, Epple M, Okkenhaug H, Cook SJ, Shanahan CM, Bootman MD, Proudfoot D. Calcium phosphate particles stimulate interleukin-1β release from human vascular smooth muscle cells: A role for spleen tyrosine kinase and exosome release. J Mol Cell Cardiol 2018; 115:82-93. [PMID: 29274344 PMCID: PMC5823844 DOI: 10.1016/j.yjmcc.2017.12.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022]
Abstract
AIMS Calcium phosphate (CaP) particle deposits are found in several inflammatory diseases including atherosclerosis and osteoarthritis. CaP, and other forms of crystals and particles, can promote inflammasome formation in macrophages leading to caspase-1 activation and secretion of mature interleukin-1β (IL-1β). Given the close association of small CaP particles with vascular smooth muscle cells (VSMCs) in atherosclerotic fibrous caps, we aimed to determine if CaP particles affected pro-inflammatory signalling in human VSMCs. METHODS AND RESULTS Using ELISA to measure IL-1β release from VSMCs, we demonstrated that CaP particles stimulated IL-1β release from proliferating and senescent human VSMCs, but with substantially greater IL-1β release from senescent cells; this required caspase-1 activity but not LPS-priming of cells. Potential inflammasome agonists including ATP, nigericin and monosodium urate crystals did not stimulate IL-1β release from VSMCs. Western blot analysis demonstrated that CaP particles induced rapid activation of spleen tyrosine kinase (SYK) (increased phospho-Y525/526). The SYK inhibitor R406 reduced IL-1β release and caspase-1 activation in CaP particle-treated VSMCs, indicating that SYK activation occurs upstream of and is required for caspase-1 activation. In addition, IL-1β and caspase-1 colocalised in intracellular endosome-like vesicles and we detected IL-1β in exosomes isolated from VSMC media. Furthermore, CaP particle treatment stimulated exosome secretion by VSMCs in a SYK-dependent manner, while the exosome-release inhibitor spiroepoxide reduced IL-1β release. CONCLUSIONS CaP particles stimulate SYK and caspase-1 activation in VSMCs, leading to the release of IL-1β, at least in part via exosomes. These novel findings in human VSMCs highlight the pro-inflammatory and pro-calcific potential of microcalcification.
Collapse
Affiliation(s)
- Yana Dautova
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | - Alexander N Kapustin
- Cardiovascular Division, James Black Centre, King's College London,125 Coldharbour Lane, London SE5 9NU, UK
| | - Kevin Pappert
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Essen-Duisburg, Essen 45117, Germany
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Essen-Duisburg, Essen 45117, Germany
| | - Hanneke Okkenhaug
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | - Simon J Cook
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | - Catherine M Shanahan
- Cardiovascular Division, James Black Centre, King's College London,125 Coldharbour Lane, London SE5 9NU, UK
| | - Martin D Bootman
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Diane Proudfoot
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, UK.
| |
Collapse
|
13
|
Shinozaki Y, Kashiwagi K, Namekata K, Takeda A, Ohno N, Robaye B, Harada T, Iwata T, Koizumi S. Purinergic dysregulation causes hypertensive glaucoma-like optic neuropathy. JCI Insight 2017; 2:93456. [PMID: 28978804 DOI: 10.1172/jci.insight.93456] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/24/2017] [Indexed: 12/24/2022] Open
Abstract
Glaucoma is an optic neuropathy characterized by progressive degeneration of retinal ganglion cells (RGCs) and visual loss. Although one of the highest risk factors for glaucoma is elevated intraocular pressure (IOP) and reduction in IOP is the only proven treatment, the mechanism of IOP regulation is poorly understood. We report that the P2Y6 receptor is critical for lowering IOP and that ablation of the P2Y6 gene in mice (P2Y6KO) results in hypertensive glaucoma-like optic neuropathy. Topically applied uridine diphosphate, an endogenous selective agonist for the P2Y6 receptor, decreases IOP. The P2Y6 receptor was expressed in nonpigmented epithelial cells of the ciliary body and controlled aqueous humor dynamics. P2Y6KO mice exhibited sustained elevation of IOP, age-dependent damage to the optic nerve, thinning of ganglion cell plus inner plexiform layers, and a reduction of RGC numbers. These changes in P2Y6KO mice were attenuated by an IOP lowering agent. Consistent with RGC damage, visual functions were impaired in middle-aged P2Y6KO mice. We also found that expression and function of P2Y6 receptors in WT mice were significantly reduced by aging, another important risk factor for glaucoma. In summary, our data show that dysfunctional purinergic signaling causes IOP dysregulation, resulting in glaucomatous optic neuropathy.
Collapse
Affiliation(s)
- Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, and
| | - Kenji Kashiwagi
- Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Akiko Takeda
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, and
| | - Nobuhiko Ohno
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Bernard Robaye
- Institute of Interdisciplinary Research and.,Institute of Biology and Molecular Medicine, Université Libre de Bruxelles, Belgium
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, and
| |
Collapse
|
14
|
Burnstock G. Purinergic Signaling in the Cardiovascular System. Circ Res 2017; 120:207-228. [PMID: 28057794 DOI: 10.1161/circresaha.116.309726] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023]
Abstract
There is nervous control of the heart by ATP as a cotransmitter in sympathetic, parasympathetic, and sensory-motor nerves, as well as in intracardiac neurons. Centers in the brain control heart activities and vagal cardiovascular reflexes involve purines. Adenine nucleotides and nucleosides act on purinoceptors on cardiomyocytes, AV and SA nodes, cardiac fibroblasts, and coronary blood vessels. Vascular tone is controlled by a dual mechanism. ATP, released from perivascular sympathetic nerves, causes vasoconstriction largely via P2X1 receptors. Endothelial cells release ATP in response to changes in blood flow (via shear stress) or hypoxia, to act on P2 receptors on endothelial cells to produce nitric oxide, endothelium-derived hyperpolarizing factor, or prostaglandins to cause vasodilation. ATP is also released from sensory-motor nerves during antidromic reflex activity, to produce relaxation of some blood vessels. Purinergic signaling is involved in the physiology of erythrocytes, platelets, and leukocytes. ATP is released from erythrocytes and platelets, and purinoceptors and ectonucleotidases are expressed by these cells. P1, P2Y1, P2Y12, and P2X1 receptors are expressed on platelets, which mediate platelet aggregation and shape change. Long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides promote migration and proliferation of vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis, vessel remodeling during restenosis after angioplasty and atherosclerosis. The involvement of purinergic signaling in cardiovascular pathophysiology and its therapeutic potential are discussed, including heart failure, infarction, arrhythmias, syncope, cardiomyopathy, angina, heart transplantation and coronary bypass grafts, coronary artery disease, diabetic cardiomyopathy, hypertension, ischemia, thrombosis, diabetes mellitus, and migraine.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- From the Autonomic Neuroscience Institute, Royal Free and University College Medical School, London, United Kingdom.
| |
Collapse
|
15
|
Gohar EY, Kasztan M, Pollock DM. Interplay between renal endothelin and purinergic signaling systems. Am J Physiol Renal Physiol 2017; 313:F666-F668. [PMID: 28179257 DOI: 10.1152/ajprenal.00639.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 12/19/2022] Open
Abstract
Alterations in extracellular fluid volume regulation and sodium balance may result in the development and maintenance of salt-dependent hypertension, a major risk factor for cardiovascular disease. Numerous pathways contribute to the regulation of sodium excretion and blood pressure, including endothelin and purinergic signaling. Increasing evidence suggests a link between purinergic receptor activation and endothelin production within the renal collecting duct as a means of promoting natriuresis. A better understanding of the relationship between these two systems, especially in regard to sodium homeostasis, will fill a significant knowledge gap and may provide novel antihypertensive treatment options. Therefore, this review focuses on the cross talk between endothelin and purinergic signaling as it relates to the renal regulation of sodium and blood pressure homeostasis.
Collapse
Affiliation(s)
- Eman Y Gohar
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Malgorzata Kasztan
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
16
|
Abstract
Background: The nucleotide adenosine triphosphate (ATP) has long been known to drive and participate in countless intracellular processes. Extracellular ATP and its metabolite adenosine have also been shown to exert a variety of effects on nearly every cell type in human skin. Knowledge of the sources and effects of extracellular ATP in human skin may help shape new therapies for skin injury, inflammation, and numerous other cutaneous disorders. Objective: The objective of this review is to introduce the reader to current knowledge regarding the sources and effects of extracellular ATP in human skin and to outline areas in which further research is necessary to clarify the nature and mechanism of these effects. Conclusion: Extracellular ATP seems to play a direct role in triggering skin inflammatory, regenerative, and fibrotic responses to mechanical injury, an indirect role in melanocyte proliferation and apoptosis, and a complex role in Langerhans cell-directed adaptive immunity.
Collapse
Affiliation(s)
| | - Richard D. Granstein
- Department of Dermatology, Joan and Sanford I. Weill Medical College of Cornell University, New York, New York, USA
| |
Collapse
|
17
|
Aho J, Helenius M, Vattulainen-Collanus S, Alastalo TP, Koskenvuo J. Extracellular ATP protects endothelial cells against DNA damage. Purinergic Signal 2016; 12:575-81. [PMID: 27030122 DOI: 10.1007/s11302-016-9508-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/08/2016] [Indexed: 12/18/2022] Open
Abstract
Cell damage can lead to rapid release of ATP to extracellular space resulting in dramatic change in local ATP concentration. Evolutionary, this has been considered as a danger signal leading to adaptive responses in adjacent cells. Our aim was to demonstrate that elevated extracellular ATP or inhibition of ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1/CD39) activity could be used to increase tolerance against DNA-damaging conditions. Human endothelial cells, with increased extracellular ATP concentration in cell proximity, were more resistant to irradiation or chemically induced DNA damage evaluated with the DNA damage markers γH2AX and phosphorylated p53. In our rat models of DNA damage, inhibiting CD39-driven ATP hydrolysis with POM-1 protected the heart and lung tissues against chemically induced DNA damage. Interestingly, the phenomenon could not be replicated in cancer cells. Our results show that transient increase in extracellular ATP can promote resistance to DNA damage.
Collapse
Affiliation(s)
- Joonas Aho
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Mikko Helenius
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Sanna Vattulainen-Collanus
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Tero-Pekka Alastalo
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | - Juha Koskenvuo
- Research Center of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Gendaszewska-Darmach E, Szustak M. Thymidine 5'-O-monophosphorothioate induces HeLa cell migration by activation of the P2Y6 receptor. Purinergic Signal 2016; 12:199-209. [PMID: 26746211 DOI: 10.1007/s11302-015-9492-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 12/22/2015] [Indexed: 01/10/2023] Open
Abstract
ATP, ADP, UTP, and UDP acting as ligands of specific P2Y receptors activate intracellular signaling cascades to regulate a variety of cellular processes, including proliferation, migration, differentiation, and cell death. Contrary to a widely held opinion, we show here that nucleoside 5'-O-monophosphorothioate analogs, containing a sulfur atom in a place of one nonbridging oxygen atom in a phosphate group, act as ligands for selected P2Y subtypes. We pay particular attention to the unique activity of thymidine 5'-O-monophosphorothioate (TMPS) which acts as a specific partial agonist of the P2Y6 receptor (P2Y6R). We also collected evidence for the involvement of the P2Y6 receptor in human epithelial adenocarcinoma cell line (HeLa) cell migration induced by thymidine 5'-O-monophosphorothioate analog. The stimulatory effect of TMPS was abolished by siRNA-mediated P2Y6 knockdown and diisothiocyanate derivative MRS 2578, a selective antagonist of the P2Y6R. Our results indicate for the first time that increased stability of thymidine 5'-O-monophosphorothioate as well as its affinity toward the P2Y6R may be responsible for some long-term effects mediated by this receptor.
Collapse
Affiliation(s)
- Edyta Gendaszewska-Darmach
- Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924, Lodz, Poland.
| | - Marcin Szustak
- Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924, Lodz, Poland
| |
Collapse
|
19
|
Zimmermann H. Extracellular ATP and other nucleotides-ubiquitous triggers of intercellular messenger release. Purinergic Signal 2015; 12:25-57. [PMID: 26545760 DOI: 10.1007/s11302-015-9483-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
Extracellular nucleotides, and ATP in particular, are cellular signal substances involved in the control of numerous (patho)physiological mechanisms. They provoke nucleotide receptor-mediated mechanisms in select target cells. But nucleotides can considerably expand their range of action. They function as primary messengers in intercellular communication by stimulating the release of other extracellular messenger substances. These in turn activate additional cellular mechanisms through their own receptors. While this applies also to other extracellular messengers, its omnipresence in the vertebrate organism is an outstanding feature of nucleotide signaling. Intercellular messenger substances released by nucleotides include neurotransmitters, hormones, growth factors, a considerable variety of other proteins including enzymes, numerous cytokines, lipid mediators, nitric oxide, and reactive oxygen species. Moreover, nucleotides activate or co-activate growth factor receptors. In the case of hormone release, the initially paracrine or autocrine nucleotide-mediated signal spreads through to the entire organism. The examples highlighted in this commentary suggest that acting as ubiquitous triggers of intercellular messenger release is one of the major functional roles of extracellular nucleotides. While initiation of messenger release by nucleotides has been unraveled in many contexts, it may have been overlooked in others. It can be anticipated that additional nucleotide-driven messenger functions will be uncovered with relevance for both understanding physiology and development of therapy.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Institute of Cell Biology and Neuroscience, Molecular and Cellular Neurobiology, Goethe University, Max-von-Laue-Str. 13, Frankfurt am Main, Germany.
| |
Collapse
|
20
|
Kurokawa T, Zha X, Ito H, Aoki Y, Akino H, Kobayashi M, Yokoyama O. Underlying mechanisms of urine storage dysfunction in rats with salt-loading hypertension. Life Sci 2015; 141:8-12. [PMID: 26390819 DOI: 10.1016/j.lfs.2015.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 08/24/2015] [Accepted: 09/17/2015] [Indexed: 10/23/2022]
Abstract
AIMS Spontaneous hypertensive rats provide a genetic model for exploring the pathogenesis of urine storage dysfunction related to hypertension (HT). In humans, however, HT develops by both genetic and environmental factors including lifestyle factors such as a high-calorie diet, excessive salt intake and stress. We investigated the influence of salt-loading on bladder function and the underlying mechanisms of storage dysfunction related to HT. MAIN METHODS Six-week-old male Dahl salt-sensitive (DS) and Dahl salt-resistant (DR) rats were fed with a normal or high-salt diet for 12weeks. Micturition parameters were obtained from a metabolic cage. Whole bladders were excised from 18-week-old rats and distended in an organ bath. The releases of adenosine triphosphoric acid (ATP) and prostaglandin E2 (PGE2) from the distended bladder epithelia were measured. Changes in bladder blood flow (BBF) were determined with a laser-speckle-blood-flow imaging system. KEY FINDINGS An increase in mean blood pressure (BP) was noted only in DS rats after salt-loading. During the inactive (sleeping) period, voided volume per micturition gradually increased in DR rats fed a normal or high-salt diet and normal-diet DS rats, while it did not change in the DS rats fed a high-salt diet. Bladder distension significantly increased ATP and PGE2 release from the urothelium in DS rats fed a high-salt diet. BBF was significantly decreased in high-salt-diet DS rats. SIGNIFICANCE One mechanism behind the relationship between salt-sensitive HT and urine storage dysfunction may be an increase in ATP and PGE2 release from the urothelium via suppression of BBF.
Collapse
Affiliation(s)
- Tetsuyuki Kurokawa
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan
| | - Xinmin Zha
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan
| | - Hideaki Ito
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan
| | - Yoshitaka Aoki
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan
| | - Hironobu Akino
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan
| | - Motohiro Kobayashi
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan
| | - Osamu Yokoyama
- Department of Urology, Faculty of Medical Science, University of Fukui, 23-3 Matsuokashimoaizuki, Fukui 910-1193, Japan.
| |
Collapse
|
21
|
In vitro Study of a Novel Stent Coating Using Modified CD39 Messenger RNA to Potentially Reduce Stent Angioplasty-Associated Complications. PLoS One 2015; 10:e0138375. [PMID: 26381750 PMCID: PMC4575070 DOI: 10.1371/journal.pone.0138375] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/27/2015] [Indexed: 11/26/2022] Open
Abstract
Background Stent angioplasty provides a minimally invasive treatment for atherosclerotic vessels. However, no treatment option for atherosclerosis-associated endothelial dysfunction, which is accompanied by a loss of CD39, is available, and hence, adverse effects like thromboembolism and restenosis may occur. Messenger RNA (mRNA)-based therapy represents a novel strategy, whereby de novo synthesis of a desired protein is achieved after delivery of a modified mRNA to the target cells. Methods and Findings Our study aimed to develop an innovative bioactive stent coating that induces overexpression of CD39 in the atherosclerotic vessel. Therefore, a modified CD39-encoding mRNA was produced by in vitro transcription. Different endothelial cells (ECs) were transfected with the mRNA, and CD39 expression and functionality were analyzed using various assays. Furthermore, CD39 mRNA was immobilized using poly(lactic-co-glycolic-acid) (PLGA), and the transfection efficiency in ECs was analyzed. Our data show that ECs successfully translate in vitro-generated CD39 mRNA after transfection. The overexpressed CD39 protein is highly functional in hydrolyzing ADP and in preventing platelet activation. Furthermore, PLGA-immobilized CD39 mRNA can be delivered to ECs without losing its functionality. Summary In summary, we present a novel and promising concept for a stent coating for the treatment of atherosclerotic blood vessels, whereby patients could be protected against angioplasty-associated complications.
Collapse
|
22
|
Vénéreau E, Ceriotti C, Bianchi ME. DAMPs from Cell Death to New Life. Front Immunol 2015; 6:422. [PMID: 26347745 PMCID: PMC4539554 DOI: 10.3389/fimmu.2015.00422] [Citation(s) in RCA: 445] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/02/2015] [Indexed: 12/13/2022] Open
Abstract
Our body handles tissue damage by activating the immune system in response to intracellular molecules released by injured tissues [damage-associated molecular patterns (DAMPs)], in a similar way as it detects molecular motifs conserved in pathogens (pathogen-associated molecular patterns). DAMPs are molecules that have a physiological role inside the cell, but acquire additional functions when they are exposed to the extracellular environment: they alert the body about danger, stimulate an inflammatory response, and finally promote the regeneration process. Beside their passive release by dead cells, some DAMPs can be secreted or exposed by living cells undergoing a life-threatening stress. DAMPs have been linked to inflammation and related disorders: hence, inhibition of DAMP-mediated inflammatory responses is a promising strategy to improve the clinical management of infection- and injury-elicited inflammatory diseases. However, it is important to consider that DAMPs are not only danger signals but also central players in tissue repair. Indeed, some DAMPs have been studied for their role in tissue healing after sterile or infection-associated inflammation. This review is focused on two exemplary DAMPs, HMGB1 and adenosine triphosphate, and their contribution to both inflammation and tissue repair.
Collapse
Affiliation(s)
- Emilie Vénéreau
- Chromatin Dynamics Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute , Milan , Italy ; HMGBiotech Srl , Milan , Italy
| | - Chiara Ceriotti
- Chromatin Dynamics Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute , Milan , Italy
| | - Marco Emilio Bianchi
- Chromatin Dynamics Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute , Milan , Italy ; Università Vita-Salute San Raffaele , Milan , Italy
| |
Collapse
|
23
|
Signalling functions of coenzyme A and its derivatives in mammalian cells. Biochem Soc Trans 2015; 42:1056-62. [PMID: 25110002 DOI: 10.1042/bst20140146] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In all living organisms, CoA (coenzyme A) is synthesized in a highly conserved process that requires pantothenic acid (vitamin B5), cysteine and ATP. CoA is uniquely designed to function as an acyl group carrier and a carbonyl-activating group in diverse biochemical reactions. The role of CoA and its thioester derivatives, including acetyl-CoA, malonyl-CoA and HMG-CoA (3-hydroxy-3-methylglutaryl-CoA), in the regulation of cellular metabolism has been extensively studied and documented. The main purpose of the present review is to summarize current knowledge on extracellular and intracellular signalling functions of CoA/CoA thioesters and to speculate on future developments in this area of research.
Collapse
|
24
|
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
|
25
|
Helenius MH, Vattulainen S, Orcholski M, Aho J, Komulainen A, Taimen P, Wang L, de Jesus Perez VA, Koskenvuo JW, Alastalo TP. Suppression of endothelial CD39/ENTPD1 is associated with pulmonary vascular remodeling in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1046-57. [PMID: 25820525 DOI: 10.1152/ajplung.00340.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 03/24/2015] [Indexed: 11/22/2022] Open
Abstract
Endothelial cell (EC) dysfunction plays a role in the pathobiology of occlusive vasculopathy in pulmonary arterial hypertension (PAH). Purinergic signaling pathways, which consist of extracellular nucleotide and nucleoside-mediated cell signaling through specific receptors, are known to be important regulators of vascular tone and remodeling. Therefore, we hypothesized that abnormalities in the vascular purinergic microenvironment are associated with PAH. Enzymatic clearance is crucial to terminate unnecessary cell activation; one of the most abundantly expressed enzymes on the EC surface is E-NTPDase1/CD39, which hydrolyzes ATP and ADP to AMP. we used histological samples from patients and healthy donors, radioisotope-labeled substrates to measure ectoenzyme activity, and a variety of in vitro approaches to study the role of CD39 in PAH. Immunohistochemistry on human idiopathic PAH (IPAH) patients' lungs demonstrated that CD39 was significantly downregulated in the endothelium of diseased small arteries. Similarly, CD39 expression and activity were decreased in cultured pulmonary ECs from IPAH patients. Suppression of CD39 in vitro resulted in EC phenotypic switch that gave rise to apoptosis-resistant pulmonary arterial endothelial cells and promoted a microenvironment that induced vascular smooth muscle cell migration. we also identified that the ATP receptor P2Y11 is essential for ATP-mediated EC survival. Furthermore, we report that apelin, a known regulator of pulmonary vascular homeostasis, can potentiate the activity of CD39 both in vitro and in vivo. we conclude that sustained attenuation of CD39 activity through ATP accumulation is tightly linked to vascular dysfunction and remodeling in PAH and could represent a novel target for therapy.
Collapse
Affiliation(s)
- Mikko H Helenius
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Sanna Vattulainen
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Mark Orcholski
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California
| | - Joonas Aho
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Anne Komulainen
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Pekka Taimen
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland; and
| | - Lingli Wang
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California
| | | | - Juha W Koskenvuo
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland; Department of Clinical Physiology and Nuclear Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Tero-Pekka Alastalo
- Children's Hospital Helsinki, Pediatric Cardiology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland;
| |
Collapse
|
26
|
Uemura Y, Shibata R, Kanemura N, Ohashi K, Kambara T, Hiramatsu-Ito M, Enomoto T, Yuasa D, Joki Y, Matsuo K, Ito M, Hayakawa S, Ogawa H, Murohara T, Ouchi N. Adipose-derived protein omentin prevents neointimal formation after arterial injury. FASEB J 2014; 29:141-51. [PMID: 25300621 DOI: 10.1096/fj.14-258129] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Obesity is highly linked with the development of vascular diseases. Omentin is a circulating adipokine that is downregulated in patients with cardiovascular diseases. In this study, we investigated the role of omentin in regulation of vascular remodeling in response to injury. Wild-type (WT) mice were treated intravenously with adenoviral vectors encoding human omentin (Ad-OMT) or control β-gal and subjected to arterial wire injury. Ad-OMT treatment reduced the neointimal thickening and the frequencies of bromodeoxyuridine-positive proliferating cells in injured arteries. Treatment of vascular smooth muscle cells (VSMCs) with human omentin protein at a physiologic concentration led to suppression of growth and ERK phosphorylation after stimulation with various growth factors. Omentin stimulated AMPK signaling in VSMCs, and blockade of AMPK reversed omentin-mediated inhibition of VSMC growth and ERK phosphorylation. Furthermore, fat-specific human omentin transgenic (OMT-TG) mice exhibited reduced neointimal thickening and vascular cell growth following vascular injury. AMPK activation was enhanced in injured arteries in OMT-TG mice, and administration of AMPK inhibitor reversed the reduction of neointimal hyperplasia in OMT-TG mice. These data indicate that omentin attenuates neointimal formation after arterial injury and suppresses VSMC growth through AMPK-dependent mechanisms. Thus, omentin can represent a novel target molecule for the prevention of vascular disorders.
Collapse
Affiliation(s)
| | | | | | - Koji Ohashi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | - Noriyuki Ouchi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
27
|
Jia S, Chen Z, Li J, Chi Y, Wang J, Li S, Luo Y, Geng B, Wang C, Cui Q, Guan Y, Yang J. FAM3A promotes vascular smooth muscle cell proliferation and migration and exacerbates neointima formation in rat artery after balloon injury. J Mol Cell Cardiol 2014; 74:173-82. [PMID: 24857820 DOI: 10.1016/j.yjmcc.2014.05.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/07/2014] [Accepted: 05/14/2014] [Indexed: 11/17/2022]
Abstract
The biological function of FAM3A, the first member of family with sequence similarity 3 (FAM3) gene family, remains largely unknown. This study aimed to determine its role in the proliferation and migration of vascular smooth muscle cells (VSMCs). Immunohistochemical staining revealed that FAM3A protein is expressed in the tunica media of rodent arteries, and its expression is reduced with an increase in prostaglandin E receptor 2 (EP2) expression after injury. In vitro, FAM3A overexpression promotes proliferation and migration of VSMCs, whereas FAM3A silencing inhibits these processes. In vivo, FAM3A overexpression results in exaggerated neointima formation of rat carotid artery after balloon injury. FAM3A activates Akt in a PI3K-dependent manner. In contrast, FAM3A induces ERK1/2 activation independent of PI3K. FAM3A protein is subcellularly located in mitochondria, where it affects ATP production and release. Activation of EP2 represses FAM3A expression, leading to impaired ATP production and release in VSMCs. FAM3A-induced activation of Akt and ERK1/2 pathways, proliferation and migration of VSMCs are inhibited by P2 receptor antagonist suramin. Furthermore, inhibition or knockdown of P2Y1 receptor inihibits FAM3A-induced proliferation and migration of VSMCs. In conclusion, FAM3A promotes proliferation and migration of VSMCs via P2Y1 receptor-mediated activation of Akt and ERK1/2 pathways. In injured vessels, FAM3A was repressed by upregulated EP2 expression, leading to the attenuation of ATP-P2Y1 receptor signaling, which is beneficial for preventing excessive proliferation and migration of VSMCs.
Collapse
MESH Headings
- Animals
- Balloon Occlusion
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cell Movement
- Cell Proliferation
- Cytokines/genetics
- Cytokines/metabolism
- Gene Expression Regulation
- Male
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/genetics
- Neointima/metabolism
- Neointima/pathology
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E, EP2 Subtype/genetics
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Purinergic P2Y1/genetics
- Receptors, Purinergic P2Y1/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Shi Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Zhenzhen Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Jing Li
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Yujing Chi
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, Beijing 100044, China
| | - Jinyu Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Sha Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Yanjin Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Bin Geng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Cheng Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
28
|
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
|
29
|
Hinze AV, Mayer P, Harst A, von Kügelgen I. P2X1 receptor-mediated inhibition of the proliferation of human coronary smooth muscle cells involving the transcription factor NR4A1. Purinergic Signal 2013; 9:677-86. [PMID: 23873636 PMCID: PMC3889386 DOI: 10.1007/s11302-013-9380-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/03/2013] [Indexed: 01/28/2023] Open
Abstract
Adenine nucleotides acting at P2X1 receptors are potent vasoconstrictors. Recently, we demonstrated that activation of adenosine A2B receptors on human coronary smooth muscle cells inhibits cell proliferation by the induction of the nuclear receptor subfamily 4, group A, member 1 (NR4A1; alternative notation Nur77). In the present study, we searched for long-term effects mediated by P2X1 receptors by analyzing receptor-mediated changes in cell proliferation and in the expression of NR4A1. Cultured human coronary smooth muscle cells were treated with selective receptor ligands. Effects on proliferation were determined by counting cells and measuring changes in impedance. The induction of transcription factors was assessed by qPCR. The P2X receptor agonist α,β-methylene-ATP and its analog β,γ-methylene-ATP inhibited cell proliferation by about 50 % after 5 days in culture with half-maximal concentrations of 0.3 and 0.08 μM, respectively. The effects were abolished or markedly attenuated by the P2X1 receptor antagonist NF449 (carbonylbis-imino-benzene-triylbis-(carbonylimino)tetrakis-benzene-1,3-disulfonic acid; 100 nM and 1 μM). α,β-methylene-ATP and β,γ-methylene-ATP applied for 30 min to 4 h increased the expression of NR4A1; NF449 blocked or attenuated this effect. Small interfering RNA directed against NR4A1 diminished the antiproliferative effects of α,β-methylene-ATP and β,γ-methylene-ATP. α,β-methylene-ATP (0.1 to 30 μM) decreased migration of cultured human coronary smooth muscle cells in a chamber measuring changes in impedance; NF449 blocked the effect. In conclusion, our results demonstrate for the first time that adenine nucleotides acting at P2X1 receptors inhibit the proliferation of human coronary smooth muscle cells via the induction of the early gene NR4A1.
Collapse
Affiliation(s)
- Annette Viktoria Hinze
- />Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
- />Department of Pharmacology and Toxicology, University of Bonn, 53105 Bonn, Germany
- />Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, 53105 Bonn, Germany
| | - Peter Mayer
- />Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Anja Harst
- />Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Ivar von Kügelgen
- />Department of Pharmacology and Toxicology, University of Bonn, 53105 Bonn, Germany
| |
Collapse
|
30
|
Development of longitudinal smooth muscle in the posterior mesenteric artery and purinergic regulation of its contractile responses in chickens. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2013; 199:857-65. [PMID: 23963637 DOI: 10.1007/s00359-013-0848-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 05/30/2013] [Accepted: 08/08/2013] [Indexed: 10/26/2022]
Abstract
This study was designed to clarify development and the neural regulation of longitudinal smooth muscle in the chicken posterior mesenteric artery to generate new hypotheses for the roles of arterial longitudinal muscles. The existence of longitudinal muscles was examined with hematoxylin-eosin staining. A well-developed longitudinal muscle layer exists in the posterior mesenteric artery of adult female chickens but not adult male chickens. The muscle layer is poorly developed in chickens aged < 15 weeks, even in female chickens. Mechanical responses of muscles were recorded and perivascular nerves were stimulated by electrical field stimulation (EFS). EFS induced monophasic contractions in longitudinal muscle of the posterior mesenteric artery segment, and those responses were inhibited by pretreatment with tetrodotoxin. Blockers for cholinoceptors and adrenoceptors did not affect EFS-evoked contractions but an antagonist for P2X purinoceptors blocked them. The present study demonstrated that the longitudinal muscle in the posterior mesenteric artery of the domestic fowl develops between the 5th and 15th week of life, suggesting that its development is involved in oviposition. The longitudinal muscle might have a role in resisting extensional stress from the oviduct containing eggs. Moreover, the arterial longitudinal muscle is regulated by purinergic neurons via P2X purinoceptors.
Collapse
|
31
|
Sawosz F, Pineda L, Hotowy A, Jaworski S, Prasek M, Sawosz E, Chwalibog A. Nano-nutrition of chicken embryos. The effect of silver nanoparticles and ATP on expression of chosen genes involved in myogenesis. Arch Anim Nutr 2013; 67:347-55. [PMID: 23952606 DOI: 10.1080/1745039x.2013.830520] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
It has been suggested that the quantity and quality of nutrients stored in the egg might not be optimal for the fast rate of chicken embryo development in modern broilers, and embryos could be supplemented with nutrients by in ovo injection. Recent experiments showed that in ovo feeding reduces post-hatch mortality and skeletal disorders and increases muscle growth and breast meat yield. Adenosine triphosphate (ATP) is a "ready for use" energetic molecule, while nanoparticles of silver (Nano-Ag) may penetrate tissues as well as cells and localise inside cells. In this investigation, we hypothesised that silver nanoparticles could be used as a protective carrier for ATP as well as an active agent. ATP and/or an ATP complex with Nano-Ag would be delivered to the muscle cells as a gene expression regulator and promoter of growth and development of embryo breast muscle. A collection of 160 broiler eggs was randomly divided into a Control group without injection and injected groups with hydrocolloids of Nano-Ag, ATP or a complex of Nano-Ag and ATP (Nano-Ag/ATP). The embryos were evaluated on day 20 of incubation. The results indicate that the application of ATP to chicken embryos increases expression of fibroblast growth factor 2 (FGF2), vascular endothelial growth factor (VEGF) and Na(+)/K(+) transporting ATPase (ATP1A1), which may indicate that an extra energy source can enhance molecular mechanisms of muscle cell proliferation. Nano-Ag also up-regulated expression of FGF2, VEGF, ATP1A1 and, also up-regulated expression of myogenic differentiation 1(MyoD1), affecting cell differentiation. The results indicate that ATP and Nano-Ag may accelerate growth and maturation of muscle cells.
Collapse
Affiliation(s)
- Filip Sawosz
- a Department of Veterinary Clinical and Animal Sciences , University of Copenhagen , Frederiksberg C , Denmark
| | | | | | | | | | | | | |
Collapse
|
32
|
P2Y2 receptor agonist with enhanced stability protects the heart from ischemic damage in vitro and in vivo. Purinergic Signal 2013; 9:633-42. [PMID: 23828651 DOI: 10.1007/s11302-013-9374-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/19/2013] [Indexed: 12/15/2022] Open
Abstract
Extracellular nucleotides acting via P2 receptors play important roles in cardiovascular physiology/pathophysiology. Pyrimidine nucleotides activate four G protein-coupled P2Y receptors (P2YRs): P2Y2 and P2Y4 (UTP-activated), P2Y6, and P2Y14. Previously, we showed that uridine 5'-triphosphate (UTP) activating P2Y2R reduced infarct size and improved mouse heart function after myocardial infarct (MI). Here, we examined the cardioprotective role of P2Y2R in vitro and in vivo following MI using uridine-5'-tetraphosphate δ-phenyl ester tetrasodium salt (MRS2768), a selective and more stable P2Y2R agonist. Cultured rat cardiomyocytes pretreated with MRS2768 displayed protection from hypoxia [as revealed by lactate dehydrogenase (LDH) release and propidium iodide (PI) binding], which was reduced by P2Y2R antagonist, AR-C118925 (5-((5-(2,8-dimethyl-5H-dibenzo[a,d][7]annulen-5-yl)-2-oxo-4-thioxo-3,4-dihydropyrimidin-1(2H)-yl)methyl)-N-(1H-tetrazol-5-yl)furan-2-carboxamide). In vivo, echocardiography and infarct size staining of triphenyltetrazolium chloride (TTC) in 3 groups of mice 24 h post-MI: sham, MI, and MI+MRS2768 indicated protection. Fractional shortening (FS) was higher in MRS2768-treated mice than in MI alone (40.0 ± 3.1 % vs. 33.4 ± 2.7 %, p < 0.001). Troponin T and tumor necrosis factor-α (TNF-α) measurements demonstrated that MRS2768 pretreatment reduced myocardial damage (p < 0.05) and c-Jun phosphorylation increased. Thus, P2Y2R activation protects cardiomyocytes from hypoxia in vitro and reduces post-ischemic myocardial damage in vivo.
Collapse
|
33
|
Scodelaro Bilbao P, Boland R. Extracellular ATP regulates FoxO family of transcription factors and cell cycle progression through PI3K/Akt in MCF-7 cells. Biochim Biophys Acta Gen Subj 2013; 1830:4456-69. [PMID: 23742826 DOI: 10.1016/j.bbagen.2013.05.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Forkhead Box-O (FoxO) transcription factors regulate the expression of many genes involved in suppression. Released nucleotides can regulate intracellular signaling pathways through membrane-bound purinergic receptors, to promote or prevent malignant cell transformation. We studied the role of extracellular ATP in the modulation of Forkhead Box O (FoxO) transcription factors and of cell cycle progression in MCF-7 breast cancer cells. METHODS Western blot analysis, cell transfections with siRNA against Akt, immunocytochemistry, subcellular fractionation studies and flow cytometry analysis were performed. RESULTS ATP induced the phosphorylation of FoxO1/3a at threonine 24/32, whereas reduced the expression of FoxO1. In addition, ATP increased the expression of the cyclins D1 and D3 and down-regulated the cell cycle inhibitory proteins p21Cip1 and p27Kip1. The use of the phosphatidylinositol 3 kinase (PI3K) inhibitor, Ly294002, and/or of siRNA to reduce the expression of the serine/threonine kinase Akt showed that these effects are mediated by the PI3K/Akt signaling pathway. ATP induced the translocation of FoxO3a from the nucleus to the cytoplasm. Also, ATP increased the number of cells in the S phase of cell cycle; this effect was reverted by the use of Ly294002 and the proteasome inhibitor bortezomib. CONCLUSION Extracellular ATP induces the inactivation of FoxO transcription factors and cell cycle progression through the PI3K/Akt pathway in MCF-7 cells. GENERAL SIGNIFICANCE These findings provide new molecular basis for further understanding the mechanisms involved in ATP signal transduction in breast cancer cells, and should be considered for the development of effective breast cancer therapeutic strategies.
Collapse
Affiliation(s)
- Paola Scodelaro Bilbao
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan, Argentina
| | | |
Collapse
|
34
|
Stenmark KR, Yeager ME, El Kasmi KC, Nozik-Grayck E, Gerasimovskaya EV, Li M, Riddle SR, Frid MG. The adventitia: essential regulator of vascular wall structure and function. Annu Rev Physiol 2012; 75:23-47. [PMID: 23216413 PMCID: PMC3762248 DOI: 10.1146/annurev-physiol-030212-183802] [Citation(s) in RCA: 273] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and is composed of a variety of cells, including fibroblasts, immunomodulatory cells (dendritic cells and macrophages), progenitor cells, vasa vasorum endothelial cells and pericytes, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to influence the tone and structure of the vessel wall; to initiate and perpetuate chronic vascular inflammation; and to stimulate expansion of the vasa vasorum, which can act as a conduit for continued inflammatory and progenitor cell delivery to the vessel wall. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of vascular wall function and structure from the outside in.
Collapse
Affiliation(s)
- Kurt R. Stenmark
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Michael E. Yeager
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Karim C. El Kasmi
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Eva Nozik-Grayck
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | | | - Min Li
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Suzette R. Riddle
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| | - Maria G. Frid
- University of Colorado Denver, Division of Pediatric Critical Care, Aurora, CO 80045
| |
Collapse
|
35
|
Uemura Y, Shibata R, Ohashi K, Enomoto T, Kambara T, Yamamoto T, Ogura Y, Yuasa D, Joki Y, Matsuo K, Miyabe M, Kataoka Y, Murohara T, Ouchi N. Adipose-derived factor CTRP9 attenuates vascular smooth muscle cell proliferation and neointimal formation. FASEB J 2012; 27:25-33. [PMID: 22972916 DOI: 10.1096/fj.12-213744] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity is closely associated with the progression of vascular disorders, including atherosclerosis and postangioplasty restenosis. C1q/TNF-related protein (CTRP) 9 is an adipocytokine that is down-regulated in obese mice. Here we investigated whether CTRP9 modulates neointimal hyperplasia and vascular smooth muscle cell (VSMC) proliferation in vivo and in vitro. Left femoral arteries of wild-type (WT) mice were injured by a steel wire. An adenoviral vector expressing CTRP9 (Ad-CTRP9) or β-galactosidase as a control was intravenously injected into WT mice 3 d before vascular injury. Delivery of Ad-CTRP9 significantly attenuated the neointimal thickening and the number of bromodeoxyuridine-positive proliferating cells in the injured arteries compared with that of control. Treatment of VSMCs with CTRP9 protein attenuated the proliferative and chemotactic activities induced by growth factors including platelet-derived growth factor (PDGF)-BB, and suppressed PDGF-BB-stimulated phosphorylation of ERK. CTRP9 treatment dose-dependently increased cAMP levels in VSMCs. Blockade of cAMP-PKA pathway reversed the inhibitory effect of CTRP9 on DNA synthesis and ERK phosphorylation in response to PDGF-BB. The present data indicate that CTRP9 functions to attenuate neointimal formation following vascular injury through its ability to inhibit VSMC growth via cAMP-dependent mechanism, suggesting that the therapeutic approaches to enhance CTRP9 production could be valuable for prevention of vascular restenosis after angioplasty.
Collapse
Affiliation(s)
- Yusuke Uemura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Lohman AW, Billaud M, Straub AC, Johnstone SR, Best AK, Lee M, Barr K, Penuela S, Laird DW, Isakson BE. Expression of pannexin isoforms in the systemic murine arterial network. J Vasc Res 2012; 49:405-16. [PMID: 22739252 DOI: 10.1159/000338758] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Accepted: 04/04/2012] [Indexed: 12/20/2022] Open
Abstract
AIMS Pannexins (Panx) form ATP release channels and it has been proposed that they play an important role in the regulation of vascular tone. However, distribution of Panx across the arterial vasculature is not documented. METHODS We tested antibodies against Panx1, Panx2 and Panx3 on human embryonic kidney cells (which do not endogenously express Panx proteins) transfected with plasmids encoding each Panx isoform and Panx1(-/-) mice. Each of the Panx antibodies was found to be specific and was tested on isolated arteries using immunocytochemistry. RESULTS We demonstrated that Panx1 is the primary isoform detected in the arterial network. In large arteries, Panx1 is primarily in endothelial cells, whereas in small arteries and arterioles it localizes primarily to the smooth muscle cells. Panx1 was the predominant isoform expressed in coronary arteries, except in arteries less than 100 µm where Panx3 became detectable. Only Panx3 was expressed in the juxtaglomerular apparatus and cortical arterioles. The pulmonary artery and alveoli had expression of all 3 Panx isoforms. No Panx isoforms were detected at the myoendothelial junctions. CONCLUSION We conclude that the specific localized expression of Panx channels throughout the vasculature points towards an important role for these channels in regulating the release of ATP throughout the arterial network.
Collapse
Affiliation(s)
- Alexander W Lohman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lohman AW, Billaud M, Isakson BE. Mechanisms of ATP release and signalling in the blood vessel wall. Cardiovasc Res 2012; 95:269-80. [PMID: 22678409 DOI: 10.1093/cvr/cvs187] [Citation(s) in RCA: 239] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The nucleotide adenosine 5'-triphosphate (ATP) has classically been considered the cell's primary energy currency. Importantly, a novel role for ATP as an extracellular autocrine and/or paracrine signalling molecule has evolved over the past century and extensive work has been conducted to characterize the ATP-sensitive purinergic receptors expressed on almost all cell types in the body. Extracellular ATP elicits potent effects on vascular cells to regulate blood vessel tone but can also be involved in vascular pathologies such as atherosclerosis. While the effects of purinergic signalling in the vasculature have been well documented, the mechanism(s) mediating the regulated release of ATP from cells in the blood vessel wall and circulation are now a key target of investigation. The aim of this review is to examine the current proposed mechanisms of ATP release from vascular cells, with a special emphasis on the transporters and channels involved in ATP release from vascular smooth muscle cells, endothelial cells, circulating red blood cells, and perivascular sympathetic nerves, including vesicular exocytosis, plasma membrane F(1)/F(0)-ATP synthase, ATP-binding cassette (ABC) transporters, connexin hemichannels, and pannexin channels.
Collapse
Affiliation(s)
- Alexander W Lohman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
38
|
Guzman-Aranguez A, Díez LM, Martín-Gil A, Gualix J, Miras-Portugal MT, Pintor J. Effect of diinosine polyphosphates on intraocular pressure in normotensive rabbits. Exp Eye Res 2012; 101:49-55. [PMID: 22677090 DOI: 10.1016/j.exer.2012.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/27/2012] [Accepted: 05/29/2012] [Indexed: 12/01/2022]
Abstract
The ability of diinosine polyphosphates, diinosine triphosphate (Ip(3)I), diinosine tetraphosphate (Ip(4)I) and diinosine pentaphosphate (Ip(5)I) to modify intraocular pressure in normotensive New Zealand white rabbits was tested. Ip(5)I produced increase in intraocular pressure, while Ip(3)I and Ip(4)I produced a decrease. Ip(4)I was the most effective reducing intraocular pressure inducing a maximal decrease of intraocular pressure to 74.2 ± 2.5% compared with the control value. Dose-response analysis demonstrated a concentration dependent pattern which presented a pD(2) value of 6.19 ± 0.18, equivalent to an EC(50) of 0.63 μM. Regarding the underlying mechanism used by Ip(4)I to reduce intraocular pressure, studies with agonists and antagonists revealed that Ip(4)I reduces intraocular pressure via P2Y receptors in the eye. We suggest that topical application of Ip(4)I to the cornea has therapeutic potential for lowering intraocular pressure, a major risk factor for glaucoma.
Collapse
Affiliation(s)
- Ana Guzman-Aranguez
- Dep. Bioquímica y Biología Molecular, Escuela Universitaria de Óptica, Universidad Complutense de Madrid, Spain
| | | | | | | | | | | |
Collapse
|
39
|
Signaling mechanisms mediating uridine adenosine tetraphosphate-induced proliferation of human vascular smooth muscle cells. J Cardiovasc Pharmacol 2012; 58:654-62. [PMID: 21885991 DOI: 10.1097/fjc.0b013e318231e929] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Proliferation of vascular smooth muscle cells (SMCs) plays an important role in the development of atherosclerosis and restenosis. Extracellular mononucleotides, such as adenosine triphosphate and uridine-5'-triphosphate stimulate SMC proliferation. However, the effects of dinucleotides on SMC proliferation and their underlying signaling mechanisms are less well defined. Recently, increasing evidence suggests that the dinucleotide, uridine adenosine tetraphosphate (Up4A) plays a role in the regulation of cardiovascular function. We have previously demonstrated that Up4A stimulates DNA synthesis and proliferation of human SMCs. This study investigated the signaling mechanisms underlying the proliferative effect of Up4A. Up4A-induced increase in bromodeoxyuridine incorporation was blocked by the mammalian target of rapamycin inhibitor, rapamycin, and the MEK inhibitor, PD98059. Up4A-stimulated phosphorylation and kinase activity of S6 kinase (S6K) and Erk1/2 were inhibited by PD98059, whereas phosphorylation and kinase activity of S6K, but not Erk1/2, were inhibited by rapamycin. Up4A also increased the phosphorylation of Akt, which was blocked by the PI3-kinase inhibitor, LY294002. Up4A-stimulated activation of S6K, but not Erk1/2, was also prevented by LY294002. Furthermore, Up4A-stimulated phosphorylation and kinase activity of S6K and Erk1/2 were inhibited by the P2 receptor antagonist, suramin, but not by the P2X receptor antagonist, Ip5I. Up4A also stimulated an increase in the protein expression of cycle-dependent kinase 2, which was prevented by rapamycin, PD98059, and suramin. These results suggest that the signaling mechanisms underlying the Up4A-stimulated proliferation of SMCs are mediated by P2Y receptors and involve the PI3-K/Akt and mitogen-activated protein kinase pathways, leading to the independent activation of S6K and an increase in cycle-dependent kinase 2 expression. This work stresses the concept that dinucleotides, like mononucleotides, play potentially important roles in the regulation of vascular function.
Collapse
|
40
|
Lin CC, Lin WN, Cheng SE, Tung WH, Wang HH, Yang CM. Transactivation of EGFR/PI3K/Akt involved in ATP-induced inflammatory protein expression and cell motility. J Cell Physiol 2012; 227:1628-38. [PMID: 21678415 DOI: 10.1002/jcp.22880] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Phenotype transition of vascular smooth muscle cells (VSMCs) is important in vascular diseases, such as atherosclerosis and restenosis. Once released, ATP may promote activation of VSMCs by stimulating cyclooxygenase-2 (COX-2), cytosolic phospholipase A(2) (cPLA(2)) expression and prostaglandin (PG)E(2) synthesis via activation of MAPKs and NF-κB. However, whether alternative signaling pathways participated in regulating COX-2 and cPLA(2) expression associated with cell migration were investigated in rat VSMCs. Western blot analysis, RT-PCR, promoter assay and PGE(2) ELISA were used to determine expression of COX-2, cPLA(2) and PGE(2). Specific inhibitors and siRNAs against various protein kinases or transcription factors were used to investigate the related signaling components in inflammatory protein induction by ATPγS. We found that ATPγS-induced COX-2 and cPLA(2) expression and PGE(2) release was attenuated by the pharmacological inhibitors or transfection with siRNA against PKCδ, c-Src, EGFR, PI3-K, Akt, p44/p42 MAPK or Elk-1. Moreover, ATPγS-stimulated phosphorylation of PKCδ, c-Src, EGFR, Akt, p42/p44 MAPK and Elk-1, suggesting the participation of PKCδ/c-Src/EGFR/PI3-K/Akt/p42/p44 MAPK cascade in mediating Elk-1 activities in VSMCs. In addition, migration assay revealed that ATPγS promoted cell mobility through up-regulation of COX-2 and cPLA(2) expression and PGE(2) release, which was attenuated by pretreatment with PGE(2) receptor antagonists. Taken together, these data showed that ATPγS up-regulated the expression of COX-2 and cPLA(2) through transactivation of PKCδ/c-Src/EGFR/PI3K/Akt/Elk-1 pathway. Newly synthesized PGE(2) acted on its receptors to promote cell motility of ATPγS-stimulated VSMCs.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Cohen R, Shainberg A, Hochhauser E, Cheporko Y, Tobar A, Birk E, Pinhas L, Leipziger J, Don J, Porat E. UTP reduces infarct size and improves mice heart function after myocardial infarct via P2Y2 receptor. Biochem Pharmacol 2011; 82:1126-33. [PMID: 21839729 DOI: 10.1016/j.bcp.2011.07.094] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/06/2011] [Accepted: 07/21/2011] [Indexed: 10/18/2022]
Abstract
Pyrimidine nucleotides are signaling molecules, which activate G protein-coupled membrane receptors of the P2Y family. P2Y(2) and P2Y(4) receptors are part of the P2Y family, which is composed of 8 subtypes that have been cloned and functionally defined. We have previously found that uridine-5'-triphosphate (UTP) reduces infarct size and improves cardiac function following myocardial infarct (MI). The aim of the present study was to determine the role of P2Y(2) receptor in cardiac protection following MI using knockout (KO) mice, in vivo and wild type (WT) for controls. In both experimental groups used (WT and P2Y(2)(-/-) receptor KO mice) there were 3 subgroups: sham, MI, and MI+UTP. 24h post MI we performed echocardiography and measured infarct size using triphenyl tetrazolium chloride (TTC) staining on all mice. Fractional shortening (FS) was higher in WT UTP-treated mice than the MI group (44.7±4.08% vs. 33.5±2.7% respectively, p<0.001). However, the FS of P2Y(2)(-/-) receptor KO mice were not affected by UTP treatment (34.7±5.3% vs. 35.9±2.9%). Similar results were obtained with TTC and hematoxylin and eosin stainings. Moreover, troponin T measurements demonstrated reduced myocardial damage in WT mice pretreated with UTP vs. untreated mice (8.8±4.6 vs. 12±3.1 p<0.05). In contrast, P2Y(2)(-/-) receptor KO mice pretreated with UTP did not demonstrate reduced myocardial damage. These results indicate that the P2Y(2) receptor mediates UTP cardioprotection, in vivo.
Collapse
Affiliation(s)
- R Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wagner MCE. The therapeutic potential of adenosine triphosphate as an immune modulator in the treatment of HIV/AIDS: a combination approach with HAART. Curr HIV Res 2011; 9:209-22. [PMID: 21675943 PMCID: PMC3343418 DOI: 10.2174/157016211796320289] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 04/27/2011] [Accepted: 06/08/2011] [Indexed: 12/11/2022]
Abstract
Extracellular adenosine triphosphate (eATP) is a potent molecule that has the capacity to modulate various aspects of cell functions including gene expression. This element of modulation is essential to the role of ATP as a therapeutic agent. The hypothesis presented is that ATP can have an important impact on the treatment of HIV infection. This is supported in part by published research, although a much greater role for ATP is suggested than prior authors ever thought possible. ATP has the ability to enhance the immune system and could thus improve the host's own defense mechanisms to eradicate the virus-infected cells and restore normal immune function. This could provide effective therapy when used in conjunction with highly active antiretroviral therapies (HAART) to eliminate the latently infected cells. The key lies in applying ATP through the methodology described. This article presents a strategy for using ATP therapeutically along with background evidence to substantiate the importance of using ATP in the treatment of HIV infection.
Collapse
|
43
|
Aberrant, differential and bidirectional regulation of the unfolded protein response towards cell survival by 3'-deoxyadenosine. Cell Death Differ 2011; 18:1876-88. [PMID: 21597460 DOI: 10.1038/cdd.2011.63] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The unfolded protein response (UPR) is involved in a diverse range of pathologies triggered by endoplasmic reticulum (ER) stress. Endeavor to seek selective regulators of the UPR is a promising challenge towards therapeutic intervention in ER stress-related disorders. In the present report, we describe aberrant, differential and bidirectional regulation of the UPR by 3'-deoxyadenosine (cordycepin) towards cell survival. 3'-Deoxyadenosine blocked ER stress-induced apoptosis via inhibiting the IRE1-JNK pro-apoptotic pathway. 3'-Deoxyadenosine also inhibited apoptosis through reinforcement of the pro-survival eIF2α signaling without affecting PERK activity. It was associated with depression of GADD34 that dephosphorylates eIF2α, and dephosphorylation of eIF2α by salubrinal mimicked the anti-apoptotic effect of 3'-deoxyadenosine. Unexpectedly, although 3'-deoxyadenosine caused activation of eIF2α, it inhibited downstream pro-apoptotic events including induction of ATF4 and expression of CHOP. Cooperation of adenosine transporter and A3 adenosine receptor, but not A1/A2 receptors, mediated the pluripotent effects of 3'-deoxyadenosine. In mice, ER stress caused activation of JNK, expression of CHOP and induction of apoptosis in renal tubules. The apoptosis was significantly attenuated by administration with 3'-deoxyadenosine, and it was correlated with blunted induction of JNK and CHOP in the kidney. These results disclosed atypical pro-survival regulation of the UPR by 3'-deoxyadenosine, which may be advantageous for the treatment of intractable, ER stress-related disorders.
Collapse
|
44
|
Gendaszewska-Darmach E, Kucharska M. Nucleotide receptors as targets in the pharmacological enhancement of dermal wound healing. Purinergic Signal 2011; 7:193-206. [PMID: 21519856 PMCID: PMC3146642 DOI: 10.1007/s11302-011-9233-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 04/10/2011] [Indexed: 12/15/2022] Open
Abstract
With a growing interest of the involvement of extracellular nucleotides in both normal physiology and pathology, it has become evident that P2 receptor agonists and antagonists may have therapeutic potential. The P2Y2 receptor agonists (diquafosol tetrasodium and denufosol tetrasodium) are in the phase 3 of clinical trials for dry eye and cystic fibrosis, respectively. The thienopyridine derivatives clopidogrel and ticlopidine (antagonists of the platelet P2Y12 receptor) have been used in cardiovascular medicine for nearly a decade. Purines and pyrimidines may be of therapeutic potential also in wound healing since ATP and UTP have been shown to have many hallmarks of wound healing factors. Recent studies have demonstrated that extracellular nucleotides take part in all phases of wound repair: hemostasis, inflammation, tissue formation, and tissue remodeling. This review is focused on the potent purines and pyrimidines which regulate many physiological processes important for wound healing.
Collapse
Affiliation(s)
- Edyta Gendaszewska-Darmach
- Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Technical University of Lodz, Stefanowskiego 4/10, 90-924, Lodz, Poland,
| | | |
Collapse
|
45
|
Mayer P, Hinze AV, Harst A, von Kügelgen I. A₂B receptors mediate the induction of early genes and inhibition of arterial smooth muscle cell proliferation via Epac. Cardiovasc Res 2010; 90:148-56. [PMID: 21109603 DOI: 10.1093/cvr/cvq371] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIMS Extracellular adenosine and adenine nucleotides play important roles in the regulation of the blood vessel tonus and platelet aggregation. Less is known about the effects of these extracellular signalling molecules on gene expression in vascular smooth muscle cells involved in long-term vascular effects. In the present study, we therefore searched for adenosine-induced changes in the expression of early genes in cultured human coronary artery smooth muscle cells (HCASMCs). METHODS AND RESULTS Whole-genome DNA array hybridization revealed that adenosine induced a set of early genes including the nuclear receptor subfamily 4, group A, member 1 (NR4A1/Nur77/TR3). The pattern of the effects of adenosine on gene expression resembles the change in expression induced by the direct activator of adenylate cyclase forskolin. Real-time reverse-transcriptase PCR confirmed that adenosine and its analogue N-ethyl-carboxamidoadenosine elicited a strong induction of NR4A1. These effects were markedly attenuated by A(2B) receptor antagonists including 8-[4-(4-benzylpiperazide-1-sulfonyl)phenyl]-1-propylxanthine (PSB-601) and were mimicked by a cyclic AMP (cAMP) analogue [8-(4-chlorophenylthio)-2'-O-methyl-cAMP, 8CPT] acting on the exchange protein activated by cAMP (Epac). Long-term experiments over 5 days showed that 2-chloroadenosine decreased cell proliferation in the presence of platelet-derived growth factor. This effect of 2-chloroadenosine was also attenuated by PSB-601 and mimicked by 8CPT. Treatment with small interfering RNA directed against NR4A1 attenuated the inhibitory effect of 8CPT on proliferation. CONCLUSIONS In summary, our results demonstrate the operation of adenosine A₂(B) receptors mediating an early induction of NR4A1 and a decrease in cell proliferation via the cAMP/Epac pathway in HCASMCs.
Collapse
Affiliation(s)
- Peter Mayer
- Federal Institute for Drugs and Medical Devices, Bonn, Germany.
| | | | | | | |
Collapse
|
46
|
Govindan S, Taylor EJA, Taylor CW. Ca(2+) signalling by P2Y receptors in cultured rat aortic smooth muscle cells. Br J Pharmacol 2010; 160:1953-62. [PMID: 20649593 PMCID: PMC2913105 DOI: 10.1111/j.1476-5381.2010.00763.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background and purpose: P2Y receptors evoke Ca2+ signals in vascular smooth muscle cells and regulate contraction and proliferation, but the roles of the different P2Y receptor subtypes are incompletely resolved. Experimental approach: Quantitative PCR was used to define expression of mRNA encoding P2Y receptor subtypes in freshly isolated and cultured rat aortic smooth muscle cells (ASMC). Fluorescent indicators in combination with selective ligands were used to measure the changes in cytosolic free [Ca2+] in cultured ASMC evoked by each P2Y receptor subtype. Key results: The mRNA for all rat P2Y receptor subtypes are expressed at various levels in cultured ASMC. Four P2Y receptor subtypes (P2Y1, P2Y2, P2Y4 and P2Y6) evoke Ca2+ signals that require activation of phospholipase C and comprise both release of Ca2+ from stores and Ca2+ entry across the plasma membrane. Conclusions and implications: Combining analysis of P2Y receptor expression with functional analyses using selective agonists and antagonists, we isolated the Ca2+ signals evoked in ASMC by activation of P2Y1, P2Y2, P2Y4 and P2Y6 receptors.
Collapse
|
47
|
Abstract
The purpose of this study was to investigate the sources of ATP in the 1A arteriole, and to investigate age-related changes in ATP overflow. Arterioles (1A) from the red portion of the gastrocnemius muscle were isolated, cannulated and pressurized in a microvessel chamber with field stimulation electrodes. ATP overflow was determined using probes specific for ATP and null probes that were constructed similar to the ATP probes, but did not contain the enzyme coating. ATP concentrations were determined using a normal curve (0.78 to 25 micromol l(-1) ATP). ATP overflow occurred in two phases. Phase one began in the first 20 s following stimulation and phase two started 35 s after field stimulation. Tetrodotoxin, a potent neurotoxin that blocks action potential generation in nerves, abolished both phases of ATP overflow. alpha1-Receptor blockade resulted in a small decrease in ATP overflow in phase two, but endothelial removal resulted in an increase in ATP overflow. ATP overflow was lowest in 6-month-old rats and highest in 12- and 2-month-old rats (P<0.05). ATP overflow measured via biosensors was of neural origin with a small contribution from the vascular smooth muscle. The endothelium seems to play an important role in attenuating ATP overflow in 1A arterioles.
Collapse
Affiliation(s)
- Heidi A Kluess
- Department of Health Science, Kinesiology, Recreation and Dance, 308V HPER Building, 1 University of Arkansas, Fayetteville, AR 72701, USA.
| | | | | |
Collapse
|
48
|
Tsukimoto M, Homma T, Ohshima Y, Kojima S. Involvement of purinergic signaling in cellular response to gamma radiation. Radiat Res 2010; 173:298-309. [PMID: 20199215 DOI: 10.1667/rr1732.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Recent studies have suggested a bystander effect in nonirradiated cells adjacent to irradiated cells; however, the mechanism is poorly understood. In this study, we investigated the involvement of both extracellular nucleotides and activation of P2 receptors in cellular responses to gamma radiation using human HaCaT keratinocytes. The concentration of ATP in culture medium was increased after gamma irradiation (0.1-1.0 Gy), suggesting that radiation induces ATP release from cells. Intracellular Ca(2+) concentration was elevated when conditioned medium from irradiated cells was transferred to nonirradiated cells, and this elevation was suppressed by apyrase (ecto-nucleotidase), indicating the involvement of extracellular nucleotides in this event. Further, we examined the activation of ERK1/2 by gamma radiation and nucleotides (ATP and UTP). Both gamma radiation and nucleotides induced activation of ERK1/2. Next, the effect of inhibitors of P2 receptors on radiation-induced activation of ERK1/2 was examined. The activation of ERK1/2 was blocked by suramin (P2Y inhibitor), MRS2578 (P2Y(6) antagonist) and apyrase. These results suggest that both released nucleotides and activation of P2Y receptors are involved in gamma-radiation-induced activation of ERK1/2. We conclude that ionizing radiation induces release of nucleotides from cells, leading to activation of P2Y receptors, which in turn would result in a variety of biological effects.
Collapse
|
49
|
Lim Y, Kwon JS, Kim DW, Lee SH, Park RK, Lee JJ, Hong JT, Yoo HS, Kwon BM, Yun YP. Obovatol from Magnolia obovata inhibits vascular smooth muscle cell proliferation and intimal hyperplasia by inducing p21Cip1. Atherosclerosis 2009; 210:372-80. [PMID: 20022323 DOI: 10.1016/j.atherosclerosis.2009.11.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 11/24/2009] [Accepted: 11/24/2009] [Indexed: 11/16/2022]
Abstract
AIMS Obovatol is isolated from Magnolia obovata leaves and this active component has various pharmacological properties such as anti-oxidant, anti-platelet, anti-fungal and anti-inflammatory activities. In the present study, we investigated the inhibitory effects of obovatol on in vitro vascular smooth muscle cell (VSMC) proliferation and in vivo neointimal formation in a rat carotid artery injury model. METHODS AND RESULTS Obovatol (1-5 microM) exerted concentration-dependent inhibition on platelet-derived growth factor (PDGF)-BB-induced rat VSMC proliferation, without exhibiting any cellular toxicity or apoptosis, as determined by cell count, [3H]thymidine incorporation and Annexin-V-binding analyses. Treatment with obovatol blocked the cell cycle in G1 phase by down-regulating the expression of cyclins and CDKs, and selectively up-regulating the expression of p21Cip1, a well-known CDK inhibitor. Effects of perivascular delivery of obovatol were assessed 14 days after injury. The angiographic mean luminal diameters of the obovatol-treated groups (100 microg and 1 mg: 0.78+/-0.06 and 0.77+/-0.07AU, respectively) were significantly larger than that of the control group (0.58+/-0.07AU). The obovatol-treated groups (100 microg and 1mg: 0.14+/-0.04 and 0.09+/-0.03 mm2, respectively) showed significant reduction in neointimal formation versus the control group (0.17+/-0.02 mm2). Immunohistochemical staining demonstrated strong expression of p21Cip1 in the neointima of the obovatol-treated groups. CONCLUSIONS These data suggest that obovatol inhibits VSMC proliferation by perturbing cell cycle progression, possibly due to activation of p21Cip1 pathway. These results also show that obovatol may have potential as an anti-proliferative agent for the treatment of restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Yong Lim
- Research Institute of Veterinary Medicine, Department of Internal Medicine, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Buzzi N, Bilbao PS, Boland R, de Boland AR. Extracellular ATP activates MAP kinase cascades through a P2Y purinergic receptor in the human intestinal Caco-2 cell line. Biochim Biophys Acta Gen Subj 2009; 1790:1651-9. [PMID: 19836435 DOI: 10.1016/j.bbagen.2009.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 09/14/2009] [Accepted: 10/08/2009] [Indexed: 11/26/2022]
Abstract
BACKGROUND ATP exerts diverse effects on various cell types via specific purinergic P2Y receptors. Intracellular signaling cascades are the main routes of communication between P2Y receptors and regulatory targets in the cell. METHODS AND RESULTS We examined the role of ATP in the modulation of ERK1/2, JNK1/2, and p38 MAP kinases (MAPKs) in human colon cancer Caco-2 cells. Immunoblot analysis showed that ATP induces the phosphorylation of MAPKs in a time- and dose-dependent manner, peaking at 5 min at 10 microM ATP. Moreover, ATPgammaS, UTP, and UDP but not ADP or ADPbetaS increased phosphorylation of MAPKs, indicating the involvement of, at least, P2Y2/P2Y4 and P2Y6 receptor subtypes. RT-PCR studies and PCR product sequencing supported the expression of P2Y2 and P2Y4 receptors in this cell line. Spectrofluorimetric measurements showed that cell stimulation with ATP induced transient elevations in intracellular calcium concentration. In addition, ATP-induced phosphorylation of MAPKs in Caco-2 cells was dependent on Src family tyrosine kinases, calcium influx, and intracellular Ca2+ release and was partially dependent on the cAMP/PKA and PKC pathways and the EGFR. GENERAL SIGNIFICANCE These findings provide new molecular basis for further understanding the mechanisms involved in ATP functions, as a signal transducer and activator of MAP kinase cascades, in colon adenocarcinoma Caco-2 cells.
Collapse
Affiliation(s)
- Natalia Buzzi
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, 8000 Bahía Blanca, Argentina
| | | | | | | |
Collapse
|