1
|
Matsushita K, Sato C, Bruckert C, Gong D, Amissi S, Hmadeh S, Fakih W, Remila L, Lessinger JM, Auger C, Jesel L, Ohlmann P, Kauffenstein G, Schini-Kerth VB, Morel O. Potential of dapagliflozin to prevent vascular remodeling in the rat carotid artery following balloon injury. Atherosclerosis 2024; 397:117595. [PMID: 38879387 DOI: 10.1016/j.atherosclerosis.2024.117595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND AIMS Sodium-glucose co-transporter 2 (SGLT2) inhibitors have been shown to reduce the risk of cardiovascular events independently of glycemic control. However, the possibility that SGLT2 inhibitors improve vascular restenosis is unknown. The aim of this study was to examine whether dapagliflozin could prevent neointima thickening following balloon injury and, if so, to determine the underlying mechanisms. METHODS Saline, dapagliflozin (1.5 mg/kg/day), or losartan (30 mg/kg/day) was administered orally for five weeks to male Wistar rats. Balloon injury of the left carotid artery was performed a week after starting the treatment and rats were sacrificed 4 weeks later. The extent of neointima was assessed by histomorphometric and immunofluorescence staining analyses. Vascular reactivity was assessed on injured and non-injured carotid artery rings, changes of target factors by immunofluorescence, RT-qPCR, and histochemistry. RESULTS Dapagliflozin and losartan treatments reduced neointima thickening by 32 % and 27 %, respectively. Blunted contractile responses to phenylephrine and relaxations to acetylcholine and down-regulation of eNOS were observed in the injured arteries. RT-qPCR investigations indicated an increased in gene expression of inflammatory (IL-1beta, VCAM-1), oxidative (p47phox, p22phox) and fibrotic (TGF-beta1) markers in the injured carotid. While these changes were not affected by dapagliflozin, increased levels of AT1R and NTPDase1 (CD39) and decreased levels of ENPP1 were observed in the restenotic carotid artery of the dapagliflozin group. CONCLUSIONS Dapagliflozin effectively reduced neointimal thickening. The present data suggest that dapagliflozin prevents restenosis through interfering with angiotensin and/or extracellular nucleotides signaling. SGLT2 represents potential new target for limiting vascular restenosis.
Collapse
Affiliation(s)
- Kensuke Matsushita
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Chisato Sato
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Christophe Bruckert
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - DalSeong Gong
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Said Amissi
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Sandy Hmadeh
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Walaa Fakih
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Lamia Remila
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Jean-Marc Lessinger
- CHU de Strasbourg, Laboratoire de Biochimie Clinique et Biologie Moléculaire, 67091, Strasbourg, France
| | - Cyril Auger
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Laurence Jesel
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Patrick Ohlmann
- Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France
| | - Gilles Kauffenstein
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Valérie B Schini-Kerth
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France
| | - Olivier Morel
- UR 3074, Translational CardioVascular Medicine, Biomedicine Research Centre of Strasbourg, FMTS, Université de Strasbourg, Strasbourg, France; Université de Strasbourg, Pôle D'Activité Médico-Chirurgicale Cardio-Vasculaire, Nouvel Hôpital Civil, Centre Hospitalier Universitaire, Strasbourg, France; Hanoï Medical University, Hanoi, Viet Nam.
| |
Collapse
|
2
|
Chen X, Obukhov AG, Weisman GA, Seye CI. Basal ATP release signals through the P2Y 2 receptor to maintain the differentiated phenotype of vascular smooth muscle cells. Atherosclerosis 2024; 395:117613. [PMID: 38889566 PMCID: PMC11254552 DOI: 10.1016/j.atherosclerosis.2024.117613] [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: 11/09/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Vascular smooth muscle cell (VSMC) dedifferentiation contributes substantively to vascular disease. VSMCs spontaneously release low levels of ATP that modulate vessel contractility, but it is unclear if autocrine ATP signaling in VSMCs is critical to the maintenance of the VSMC contractile phenotype. METHODS We used pharmacological inhibitors to block ATP release in human aortic smooth muscle cells (HASMCs) for studying changes in VSMC differentiation marker gene expression. We employed RNA interference and generated mice with SMC-specific inducible deletion of the P2Y2 receptor (P2Y2R) gene to evaluate resulting phenotypic alterations. RESULTS HASMCs constitutively release low levels of ATP that when blocked results in a significant decrease in VSMC differentiation marker gene expression, including smooth muscle actin (SMA), smooth muscle myosin heavy chain (SMMHC), SM-22α and calponin. Basal release of ATP represses transcriptional activation of the Krüppel-Like Factor 4 (KFL4) thereby preventing platelet-derived growth factor-BB (PDGF-BB) from inhibiting expression of SMC contractile phenotype markers. SMC-restricted conditional deletion of P2Y2R evoked dedifferentiation characterized by decreases in aortic contractility and contractile phenotype markers expression. This loss was accompanied by a transition to the synthetic phenotype with the acquisition of extracellular matrix (ECM) proteins characteristic of dedifferentiation, such as osteopontin and vimentin. CONCLUSIONS Our data establish the first direct evidence that an autocrine ATP release mechanism maintains SMC cytoskeletal protein expression by inhibiting VSMCs from transitioning to a synthetic phenotype, and further demonstrate that activation of the P2Y2R by basally released ATP is required for maintenance of the differentiated VSMC phenotype.
Collapse
Affiliation(s)
- Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 360A, Indianapolis, IN, 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 360A, Indianapolis, IN, 46202, USA
| | - Gary A Weisman
- Department of Biochemistry, Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Road, Columbia, MO, 65211, USA
| | - Cheikh I Seye
- Department of Biochemistry, Christopher S. Bond Life Sciences Center, University of Missouri, 1201 Rollins Road, Columbia, MO, 65211, USA.
| |
Collapse
|
3
|
Oliveira NF, Monteiro MMLV, Mainieri NS, Tamura AS, Pereira LM, Crepaldi LD, Coutinho-Silva R, Savio LEB, Silva CLM. P2Y 2-P2X7 receptors cross-talk in primed mesenteric endothelial cells upregulates NF-κB signaling favoring mononuclear cell adhesion in schistosomiasis. Front Immunol 2024; 14:1328897. [PMID: 38239348 PMCID: PMC10794548 DOI: 10.3389/fimmu.2023.1328897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
Schistosomiasis is an intravascular infectious disease that impacts over 200 million people globally. In its chronic stage, it leads to mesenteric inflammation with significant involvement of monocytes/macrophages. Endothelial cells lining the vessel lumens play a crucial role, and mount of evidence links this disease to a downregulation of endoprotective cell signaling favoring a primed and proinflammatory endothelial cell phenotype and therefore the loss of immunovascular homeostasis. One hallmark of infectious and inflammatory conditions is the release of nucleotides into the extracellular milieu, which, in turn, act as innate messengers, activating purinergic receptors and triggering cell-to-cell communication. ATP influences the progression of various diseases through P2X and P2Y purinergic receptor subtypes. Among these receptors, P2Y2 (P2Y2R) and P2X7 (P2X7R) receptors stand out, known for their roles in inflammation. However, their specific role in schistosomiasis has remained largely unexplored. Therefore, we hypothesized that endothelial P2Y2R and P2X7R could contribute to monocyte adhesion to mesenteric endothelial cells in schistosomiasis. Using a preclinical murine model of schistosomiasis associated with endothelial dysfunction and age-matched control mice, we showed that endothelial P2Y2R and P2X7R activation increased monocyte adhesion to cultured primary endothelial cells in both groups. However, a distinct upregulation of endothelial P2Y2R-driven canonical Ca2+ signaling was observed in the infected group, amplifying adhesion. In the control group, the coactivation of endothelial P2Y2R and P2X7R did not alter the maximal monocyte adhesion induced by each receptor individually. However, in the infected group, this coactivation induced a distinct upregulation of P2Y2R-P2X7R-driven canonical signaling, IL-1β release, and VCAM-1 expression, with underlying mechanisms involving inflammasome and NF-κB signaling. Therefore, current data suggest that schistosomiasis alters endothelial cell P2Y2R/P2X7R signaling during inflammation. These discoveries advance our understanding of schistosomiasis. This intricate interplay, driven by PAMP-triggered endothelial P2Y2R/P2X7R cross-talk, emerges as a potential key player in the mesenteric inflammation during schistosomiasis.
Collapse
Affiliation(s)
- Nathália Ferreira Oliveira
- Laboratório de Farmacologia Bioquímica e Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Nathália Santos Mainieri
- Laboratório de Farmacologia Bioquímica e Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Augusto Shuiti Tamura
- Laboratório de Imunofisiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Letícia Massimo Pereira
- Laboratório de Farmacologia Bioquímica e Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia Diniz Crepaldi
- Laboratório de Imunofisiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Laboratório de Imunofisiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Claudia Lucia Martins Silva
- Laboratório de Farmacologia Bioquímica e Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Li A, Xiong C, Gong C, Gou M, Wang Y, Yang Y, Xu H. Genetic polymorphisms of purinergic P2Y 2 receptor were associated with the susceptibility to essential hypertension in Chinese postmenopausal women. Purinergic Signal 2023; 19:663-671. [PMID: 36653592 PMCID: PMC10754786 DOI: 10.1007/s11302-023-09921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/07/2023] [Indexed: 01/20/2023] Open
Abstract
Hypertension has become a prominent public health concern. Essential hypertension (EH) is a polygenic disorder caused by multiple susceptibility genes. It has been previously shown that the purinergic P2Y2 receptor (P2Y2R) regulates blood pressure; however, whether P2Y2R genetic polymorphisms correlate with EH has not been investigated in Chinese. Our study included 500 EH cases and 504 controls who are Chinese postmenopausal women. We used allele-specific polymerase chain reaction (ASPCR) to genotype five single-nucleotide polymorphism (SNPs) in the P2Y2R gene, i.e., rs4944831, rs12366239, rs1783596, rs4382936, and rs10898909. We assessed the association of P2Y2R genetic polymorphisms with EH susceptibility. The results demonstrated that P2Y2R rs4382936A was correlated with a high risk of EH; particularly, the participants with the rs4382936A allele and CA/AA/(CA+AA) genotypes were at higher risks to EH compared to the subjects with the rs4382936C allele and CC genotype. Moreover, haplotype CAG combined by rs1783596-rs4382936-rs10898909 was a susceptible haplotype for EH, whereas haplotype CCG was a protective haplotype for EH. These results may provide new evidence for applying P2Y2R genetic polymorphisms as useful markers in clinic screening or monitoring potential EH cases in a population of Chinese postmenopausal women.
Collapse
Affiliation(s)
- Ao Li
- Department of Physiology, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
- Joint Program of Nanchang University and Queen Mary University of London, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Chaopeng Xiong
- Department of Intervention Center, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Chengxin Gong
- Department of Science and Education, Chest Hospital of Jiangxi Province, Nanchang, Jiangxi, China
| | - Mengyang Gou
- Department of Physiology, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Yaqun Wang
- Department of Physiology, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China
| | - Yuping Yang
- Medical Examination Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Hong Xu
- Department of Physiology, Jiangxi Medical College of Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
| |
Collapse
|
5
|
Zhang C, Zhao J, Mi W, Zhang Y, Zhong X, Tan G, Li F, Li X, Xu Y, Zhang Y. Comprehensive analysis of microglia gene and subpathway signatures for glioma prognosis and drug screening: linking microglia to glioma. Lab Invest 2022; 20:277. [PMID: 35729639 PMCID: PMC9210642 DOI: 10.1186/s12967-022-03475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/08/2022] [Indexed: 11/10/2022]
Abstract
Glioma is the most common malignant tumors in the brain. Previous studies have revealed that, as the innate immune cells in nervous system, microglia cells were involved in glioma pathology. And, the resident microglia displayed its specific biological roles which distinguished with peripheral macrophages. In this study, an integrated analysis was performed based on public resource database to explore specific biological of microglia within glioma. Through comprehensive analysis, the biological characterization underlying two conditions, glioma microglia compared to glioma macrophage (MicT/MacT) as well as glioma microglia compared to normal microglia (MicT/MicN), were revealed. Notably, nine core MicT/MicN genes displayed closely associations with glioma recurrence and prognosis, such as P2RY2, which was analyzed in more than 2800 glioma samples from 25 studies. Furthermore, we applied a random walk based strategy to identify microglia specific subpathways and developed SubP28 signature for glioma prognostic analysis. Multiple validation data sets confirmed the predictive performance of SubP28 and involvement in molecular subtypes. The associations between SuP28 score and microglia M1/M2 polarization were also explored for both GBM and LGG types. Finally, a comprehensive drug-subpathway network was established for screening candidate medicable molecules (drugs) and identifying therapeutic subpathway targets. In conclusions, the comprehensive analysis of microglia related gene and functional signatures in glioma pathobiologic events by large-scale data sets displayed a framework to dissect inner connection between microglia and glioma, and identify robust signature for glioma clinical implications.
Collapse
Affiliation(s)
- Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jiaxin Zhao
- Center of Cerebrovascular Disease, Zhuhai People's Hospital, Zhuhai Hospital Affiliated With Jinan University, Zhuhai, 519000, China
| | - Wanqi Mi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuxi Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xiaoling Zhong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Guiyuan Tan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
6
|
Li L, Jasmer KJ, Camden JM, Woods LT, Martin AL, Yang Y, Layton M, Petris MJ, Baker OJ, Weisman GA, Petris CK. Early Dry Eye Disease Onset in a NOD.H-2h4 Mouse Model of Sjögren's Syndrome. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 35727180 PMCID: PMC9233292 DOI: 10.1167/iovs.63.6.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To develop a mouse model of human dry eye disease (DED) for investigation of sex differences in autoimmune-associated dry eye pathology. Methods Ocular surface disease was assessed by quantifying corneal epithelial damage with lissamine green stain in the NOD.H-2h4,IFNγ−/−,CD28−/− (NOD.H-2h4 DKO) mouse model of Sjögren's syndrome (SS). Lacrimal gland function was assessed by tear volume quantification with phenol red thread and lacrimal gland inflammation (i.e., dacryoadenitis) was assessed by quantification of immune cell foci, flow cytometric analysis of immune cell composition, and expression of proinflammatory markers. Results The NOD.H-2h4 DKO mouse model of SS exhibits greater age-dependent increases in corneal damage than in NOD.H-2h4 parental mice and demonstrates an earlier disease onset in females compared to males. The severity of ocular surface disease correlates with loss of goblet cell density, increased conjunctivitis, and dacryoadenitis that is more pronounced in NOD.H-2h4 DKO than NOD.H-2h4 mice. B cells dominate lacrimal infiltrates in 16-week-old NOD.H-2h4 and NOD.H-2h4 DKO mice, but T helper cells and macrophages are also present. Lacrimal gland expression of proinflammatory genes, including the P2X7 and P2Y2 purinergic receptors, is greater in NOD.H-2h4 DKO than NOD.H-2h4 mice and correlates with dacryoadenitis. Conclusions Our results demonstrate for the first time that autoimmune dry eye disease occurs in both sexes of NOD.H-2h4 DKO and NOD.H-2h4 mice, with earlier onset in female NOD.H-2h4 DKO mice when compared to males of the same strain. This study demonstrates that both NOD.H-2h4 and NOD.H-2h4 DKO mice are novel models that closely resemble SS-related and sex-dependent DED.
Collapse
Affiliation(s)
- Lili Li
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Visual Science and Optometry Center, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Kimberly J Jasmer
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Jean M Camden
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Lucas T Woods
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Adam L Martin
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Yong Yang
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Maria Layton
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States
| | - Michael J Petris
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Ophthalmology, University of Missouri, Columbia, Missouri, United States
| | - Olga J Baker
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States.,Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri, United States
| | - Gary A Weisman
- Division of Biochemistry, University of Missouri, Columbia, Missouri, United States.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States
| | - Carisa K Petris
- Department of Ophthalmology, University of Missouri, Columbia, Missouri, United States.,Mason Eye Institute, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
7
|
The Interplay of Endothelial P2Y Receptors in Cardiovascular Health: From Vascular Physiology to Pathology. Int J Mol Sci 2022; 23:ijms23115883. [PMID: 35682562 PMCID: PMC9180512 DOI: 10.3390/ijms23115883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
The endothelium plays a key role in blood vessel health. At the interface of the blood, it releases several mediators that regulate local processes that protect against the development of cardiovascular disease. In this interplay, there is increasing evidence for a role of extracellular nucleotides and endothelial purinergic P2Y receptors (P2Y-R) in vascular protection. Recent advances have revealed that endothelial P2Y1-R and P2Y2-R mediate nitric oxide-dependent vasorelaxation as well as endothelial cell proliferation and migration, which are processes involved in the regeneration of damaged endothelium. However, endothelial P2Y2-R, and possibly P2Y1-R, have also been reported to promote vascular inflammation and atheroma development in mouse models, with endothelial P2Y2-R also being described as promoting vascular remodeling and neointimal hyperplasia. Interestingly, at the interface with lipid metabolism, P2Y12-R has been found to trigger HDL transcytosis through endothelial cells, a process known to be protective against lipid deposition in the vascular wall. Better characterization of the role of purinergic P2Y-R and downstream signaling pathways in determination of the endothelial cell phenotype in healthy and pathological environments has clinical potential for the prevention and treatment of cardiovascular diseases.
Collapse
|
8
|
Strassheim D, Verin A, Batori R, Nijmeh H, Burns N, Kovacs-Kasa A, Umapathy NS, Kotamarthi J, Gokhale YS, Karoor V, Stenmark KR, Gerasimovskaya E. P2Y Purinergic Receptors, Endothelial Dysfunction, and Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21186855. [PMID: 32962005 PMCID: PMC7555413 DOI: 10.3390/ijms21186855] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Purinergic G-protein-coupled receptors are ancient and the most abundant group of G-protein-coupled receptors (GPCRs). The wide distribution of purinergic receptors in the cardiovascular system, together with the expression of multiple receptor subtypes in endothelial cells (ECs) and other vascular cells demonstrates the physiological importance of the purinergic signaling system in the regulation of the cardiovascular system. This review discusses the contribution of purinergic P2Y receptors to endothelial dysfunction (ED) in numerous cardiovascular diseases (CVDs). Endothelial dysfunction can be defined as a shift from a “calm” or non-activated state, characterized by low permeability, anti-thrombotic, and anti-inflammatory properties, to a “activated” state, characterized by vasoconstriction and increased permeability, pro-thrombotic, and pro-inflammatory properties. This state of ED is observed in many diseases, including atherosclerosis, diabetes, hypertension, metabolic syndrome, sepsis, and pulmonary hypertension. Herein, we review the recent advances in P2Y receptor physiology and emphasize some of their unique signaling features in pulmonary endothelial cells.
Collapse
Affiliation(s)
- Derek Strassheim
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (R.B.); (A.K.-K.)
| | - Robert Batori
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (R.B.); (A.K.-K.)
| | - Hala Nijmeh
- The Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA;
| | - Nana Burns
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
| | - Anita Kovacs-Kasa
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; (A.V.); (R.B.); (A.K.-K.)
| | | | - Janavi Kotamarthi
- The Department of BioMedical Engineering, University of Wisconsin, Madison, WI 53706, USA; (J.K.); (Y.S.G.)
| | - Yash S. Gokhale
- The Department of BioMedical Engineering, University of Wisconsin, Madison, WI 53706, USA; (J.K.); (Y.S.G.)
| | - Vijaya Karoor
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
| | - Kurt R. Stenmark
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
- The Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA;
| | - Evgenia Gerasimovskaya
- The Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (N.B.); (V.K.); (K.R.S.)
- The Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA;
- Correspondence: ; Tel.: +1-303-724-5614
| |
Collapse
|
9
|
Khalafalla MG, Woods LT, Jasmer KJ, Forti KM, Camden JM, Jensen JL, Limesand KH, Galtung HK, Weisman GA. P2 Receptors as Therapeutic Targets in the Salivary Gland: From Physiology to Dysfunction. Front Pharmacol 2020; 11:222. [PMID: 32231563 PMCID: PMC7082426 DOI: 10.3389/fphar.2020.00222] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Although often overlooked in our daily lives, saliva performs a host of necessary physiological functions, including lubricating and protecting the oral cavity, facilitating taste sensation and digestion and maintaining tooth enamel. Therefore, salivary gland dysfunction and hyposalivation, often resulting from pathogenesis of the autoimmune disease Sjögren's syndrome or from radiotherapy of the head and neck region during cancer treatment, severely reduce the quality of life of afflicted patients and can lead to dental caries, periodontitis, digestive disorders, loss of taste and difficulty speaking. Since their initial discovery in the 1970s, P2 purinergic receptors for extracellular nucleotides, including ATP-gated ion channel P2X and G protein-coupled P2Y receptors, have been shown to mediate physiological processes in numerous tissues, including the salivary glands where P2 receptors represent a link between canonical and non-canonical saliva secretion. Additionally, extracellular nucleotides released during periods of cellular stress and inflammation act as a tissue alarmin to coordinate immunological and tissue repair responses through P2 receptor activation. Accordingly, P2 receptors have gained widespread clinical interest with agonists and antagonists either currently undergoing clinical trials or already approved for human use. Here, we review the contributions of P2 receptors to salivary gland function and describe their role in salivary gland dysfunction. We further consider their potential as therapeutic targets to promote physiological saliva flow, prevent salivary gland inflammation and enhance tissue regeneration.
Collapse
Affiliation(s)
- Mahmoud G. Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lucas T. Woods
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Kimberly J. Jasmer
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Kevin Muñoz Forti
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Jean M. Camden
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Janicke L. Jensen
- Institute of Clinical Dentistry, Section of Oral Surgery and Oral Medicine, University of Oslo, Oslo, Norway
| | - Kirsten H. Limesand
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Hilde K. Galtung
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Gary A. Weisman
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
10
|
Yuan LY, He ZY, Li L, Wang YZ. Association of G-protein coupled purinergic receptor P2Y2 with ischemic stroke in a Han Chinese population of North China. Neural Regen Res 2018; 14:506-512. [PMID: 30539820 PMCID: PMC6334602 DOI: 10.4103/1673-5374.245472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The G-protein-coupled purinergic receptor P2Y2 (P2RY2) plays an important role in the mechanism of atherosclerosis, which is relevant to ischemic stroke. This retrospective case-control study aimed to assess the relationship between P2RY2 gene polymorphisms and ischemic stroke risk in the northern Han Chinese population. In this study, clinical data and peripheral blood specimens were collected from 378 ischemic stroke patients and 344 controls. The ischemic stroke participants were recruited from the First Affiliated Hospital of China Medical University and the First Affiliated Hospital of Liaoning Medical University. The controls were recruited from the Health Check Center at the First Affiliated Hospital of China Medical University. Ischemic stroke patients were divided into two subgroups according to the Trial of ORG 10172 in Acute Stroke Treatment (TOAST) classification: large-artery atherosclerosis (n = 178) and small-artery occlusion (n = 200) strokes. All subjects were genotyped for three single nucleotide polymorphisms (rs4944831, rs1783596, and rs4944832) in the P2RY2 gene using peripheral venous blood samples. The distribution of the dominant rs4944832 phenotype (GG vs. GA+AA) differed significantly between small-artery occlusion patients and control subjects (odds ratio (OR) = 1.720, 95% confidence interval (CI): 1.203–2.458, P < 0.01). Multivariable logistic regression analysis revealed that the GG genotype of rs4944832 was significantly more prevalent in small-artery occlusion patients than in control subjects (OR = 1.807, 95% CI: 1.215–2.687, P < 0.01). The overall distribution of the haplotype established by rs4944831-rs1783596-rs4944832 was significantly different between ischemic stroke patients and controls (P < 0.01). In ischemic stroke patients, the frequency of the G-C-G haplotype was significantly higher than in control subjects (P = 0.028), whereas the frequency of the T-C-A haplotype was lower than in control subjects (P = 0.047). These results indicate that the G-C-G haplotype of P2RY2 is a susceptibility haplotype for ischemic stroke. In addition, the GG genotype of rs4944832 may be associated with the development of small-artery occlusion in the northern Han Chinese population. The study protocol was approved by the Ethics Committee of the First Affiliated Hospital of China Medical University on February 20, 2012 (No. 2012-38-1) and the First Affiliated Hospital of Liaoning Medical University, China, on March 1, 2013 (No. 2013-03-1). All participants gave their informed consent. This trial was registered with the ISRCTN Registry (ISRCTN11439124) on October 24, 2018. Protocol version (1.0).
Collapse
Affiliation(s)
- Li-Ying Yuan
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhi-Yi He
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lei Li
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yan-Zhe Wang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
11
|
Hitzel J, Lee E, Zhang Y, Bibli SI, Li X, Zukunft S, Pflüger B, Hu J, Schürmann C, Vasconez AE, Oo JA, Kratzer A, Kumar S, Rezende F, Josipovic I, Thomas D, Giral H, Schreiber Y, Geisslinger G, Fork C, Yang X, Sigala F, Romanoski CE, Kroll J, Jo H, Landmesser U, Lusis AJ, Namgaladze D, Fleming I, Leisegang MS, Zhu J, Brandes RP. Oxidized phospholipids regulate amino acid metabolism through MTHFD2 to facilitate nucleotide release in endothelial cells. Nat Commun 2018; 9:2292. [PMID: 29895827 PMCID: PMC5997752 DOI: 10.1038/s41467-018-04602-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 05/11/2018] [Indexed: 12/20/2022] Open
Abstract
Oxidized phospholipids (oxPAPC) induce endothelial dysfunction and atherosclerosis. Here we show that oxPAPC induce a gene network regulating serine-glycine metabolism with the mitochondrial methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) as a causal regulator using integrative network modeling and Bayesian network analysis in human aortic endothelial cells. The cluster is activated in human plaque material and by atherogenic lipoproteins isolated from plasma of patients with coronary artery disease (CAD). Single nucleotide polymorphisms (SNPs) within the MTHFD2-controlled cluster associate with CAD. The MTHFD2-controlled cluster redirects metabolism to glycine synthesis to replenish purine nucleotides. Since endothelial cells secrete purines in response to oxPAPC, the MTHFD2-controlled response maintains endothelial ATP. Accordingly, MTHFD2-dependent glycine synthesis is a prerequisite for angiogenesis. Thus, we propose that endothelial cells undergo MTHFD2-mediated reprogramming toward serine-glycine and mitochondrial one-carbon metabolism to compensate for the loss of ATP in response to oxPAPC during atherosclerosis.
Collapse
Affiliation(s)
- Juliane Hitzel
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Eunjee Lee
- Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Icahn School of Medicine, New York, 10029, NY, USA
- Sema4 Genomics (a Mount Sinai venture), Stamford, 06902, CT, USA
| | - Yi Zhang
- Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Icahn School of Medicine, New York, 10029, NY, USA
- Department of Mathematics, Hebei University of Science and Technology, Shijiazhuang, 050018, Hebei, China
| | - Sofia Iris Bibli
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, 60590, Germany
| | - Xiaogang Li
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Sven Zukunft
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, 60590, Germany
| | - Beatrice Pflüger
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Jiong Hu
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, 60590, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Andrea Estefania Vasconez
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - James A Oo
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Adelheid Kratzer
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, 12203, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Berlin), Berlin, 13316, Germany
| | - Sandeep Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, GA, USA
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Ivana Josipovic
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, Faculty of Medicine, Goethe University, Frankfurt am Main, 60590, Germany
| | - Hector Giral
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, 12203, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Berlin), Berlin, 13316, Germany
| | - Yannick Schreiber
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt am Main, 60596, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, Faculty of Medicine, Goethe University, Frankfurt am Main, 60590, Germany
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt am Main, 60596, Germany
| | - Christian Fork
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 90095, CA, USA
| | - Fragiska Sigala
- 1st Department of Propaedeutic Surgery, University of Athens Medical School, Hippocration Hospital, Athens, 11364, Greece
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, 85724, AZ, USA
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, GA, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, 12203, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Berlin), Berlin, 13316, Germany
- Berlin Institute of Health (BIH), Berlin, 10178, Germany
| | - Aldons J Lusis
- Departments of Medicine, Microbiology and Human Genetics, University of California, Los Angeles, 90095, CA, USA
| | - Dmitry Namgaladze
- Institute of Biochemistry I, Goethe University, Frankfurt am Main, 60590, Germany
| | - Ingrid Fleming
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, 60590, Germany
| | - Matthias S Leisegang
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany
| | - Jun Zhu
- Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Icahn School of Medicine, New York, 10029, NY, USA.
- Sema4 Genomics (a Mount Sinai venture), Stamford, 06902, CT, USA.
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, 60590, Germany.
- German Center for Cardiovascular Research (DZHK) (Partner site Rhine-Main), Frankfurt am Main, 60590, Germany.
| |
Collapse
|
12
|
Bondu V, Bitting C, Poland VL, Hanson JA, Harkins MS, Lathrop S, Nolte KB, Lawrence DA, Buranda T. Upregulation of P2Y 2R, Active uPA, and PAI-1 Are Essential Components of Hantavirus Cardiopulmonary Syndrome. Front Cell Infect Microbiol 2018; 8:169. [PMID: 29930915 PMCID: PMC6001748 DOI: 10.3389/fcimb.2018.00169] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/03/2018] [Indexed: 12/13/2022] Open
Abstract
Sin Nombre virus (SNV) causes hantavirus cardiopulmonary pulmonary syndrome (HCPS) with the loss of pulmonary vascular endothelial integrity, and pulmonary edema without causing cytopathic effects on the vascular endothelium. HCPS is associated primarily with a dysregulated immune response. We previously found occult signs of hemostatic imbalance in the form of a sharp >30-100 fold increase in the expression of plasminogen activator inhibitor type 1 (PAI-1), in serial blood plasma draws of terminal stage-patients. However, the mechanism of the increase in PAI-1 remains unclear. PAI-1 is a primary inhibitor of fibrinolysis caused by tissue plasminogen activator (tPA) and urokinase plasminogen activator plasma (uPA). Here, we investigate factors that contribute to PAI-1 upregulation during HCPS. Using zymography, we found evidence of PAI-1-refractory uPA activity and no tPA activity in plasma samples drawn from HCPS patients. The sole prevalence of uPA activity suggested that severe inflammation drove PAI-1 activity. We have recently reported that the P2Y2 receptor (P2Y2R) mediates SNV infectivity by interacting in cis with β3 integrins, which activates the latter during infection. P2Y2R is a known effector for several biological processes relevant to HCPS pathogenesis, such as upregulation of tissue factor (TF), a primary initiator of the coagulation cascade, stimulating vascular permeability and leukocyte homing to sites of infection. As P2Y2R is prone to upregulation under conditions of inflammation, we compared the expression level of P2Y2R in formalin fixed tissues of HCPS decedents using a TaqMan assay and immunohistochemistry. Our TaqMan results show that the expression of P2Y2R is upregulated significantly in HCPS cases compared to non- HCPS controls (P < 0.001). Immunohistochemistry showed that lung macrophages were the primary reservoir of high and coincident localization of P2Y2R, uPA, PAI-1, and TF antigens. We also observed increased staining for SNV antigens in the same tissue segments where P2Y2R expression was upregulated. Conversely, sections of low P2Y2R expression showed weak manifestations of macrophages, SNV, PAI-1, and TF. Coincident localization of P2Y2R and PAI-1 on macrophage deposits suggests an inflammation-dependent mechanism of increasing pro-coagulant activity in HCPS in the absence of tissue injury.
Collapse
Affiliation(s)
- Virginie Bondu
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Casey Bitting
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Valerie L Poland
- Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Joshua A Hanson
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Michelle S Harkins
- Division of Infectious Disease, Pulmonary, Critical Care, and Sleep, Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Sarah Lathrop
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States.,Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Kurt B Nolte
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States.,Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Tione Buranda
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
13
|
Abstract
P2Y receptors (P2YRs) are a family of G protein-coupled receptors activated by extracellular nucleotides. Physiological P2YR agonists include purine and pyrimidine nucleoside di- and triphosphates, such as ATP, ADP, UTP, UDP, nucleotide sugars, and dinucleotides. Eight subtypes exist, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14, which represent current or potential future drug targets. Here we provide a comprehensive overview of ligands for the subgroup of the P2YR family that is activated by uracil nucleotides: P2Y2 (UTP, also ATP and dinucleotides), P2Y4 (UTP), P2Y6 (UDP), and P2Y14 (UDP, UDP-glucose, UDP-galactose). The physiological agonists are metabolically unstable due to their fast hydrolysis by ectonucleotidases. A number of agonists with increased potency, subtype-selectivity and/or enzymatic stability have been developed in recent years. Useful P2Y2R agonists include MRS2698 (6-01, highly selective) and PSB-1114 (6-05, increased metabolic stability). A potent and selective P2Y2R antagonist is AR-C118925 (10-01). For studies of the P2Y4R, MRS4062 (3-15) may be used as a selective agonist, while PSB-16133 (10-06) is a selective antagonist. Several potent P2Y6R agonists have been developed including 5-methoxyuridine 5'-O-((Rp)α-boranodiphosphate) (6-12), PSB-0474 (3-11), and MRS2693 (3-26). The isocyanate MRS2578 (10-08) is used as a selective P2Y6R antagonist, although its reactivity and low water-solubility are limiting. With MRS2905 (6-08), a potent and metabolically stable P2Y14R agonist is available, while PPTN (10-14) represents a potent and selective P2Y14R antagonist. The radioligand [3H]UDP can be used to label P2Y14Rs. In addition, several fluorescent probes have been developed. Uracil nucleotide-activated P2YRs show great potential as drug targets, especially in inflammation, cancer, cardiovascular and neurodegenerative diseases.
Collapse
|
14
|
Qian S, Regan JN, Shelton MT, Hoggatt A, Mohammad KS, Herring PB, Seye CI. The P2Y 2 nucleotide receptor is an inhibitor of vascular calcification. Atherosclerosis 2016; 257:38-46. [PMID: 28038380 DOI: 10.1016/j.atherosclerosis.2016.12.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/14/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Mutations in the 5'-nucleotidase ecto (NT5E) gene that encodes CD73, a nucleotidase that converts AMP to adenosine, are linked to arterial calcification. However, the role of purinergic receptor signaling in the pathology of intimal calcification is not well understood. In this study, we examined whether extracellular nucleotides acting via P2Y2 receptor (P2Y2R) modulate arterial intimal calcification, a condition highly correlated with cardiovascular morbidity. METHODS Apolipoprotein E, P2Y2R double knockout mice (ApoE-/-P2Y2R-/-) were used to determine the effect of P2Y2R deficiency on vascular calcification in vivo. Vascular smooth muscle cells (VSMC) isolated from P2Y2R-/- mice grown in high phosphate medium were used to assess the role of P2Y2R in the conversion of VSMC into osteoblasts. Luciferase-reporter assays were used to assess the effect of P2Y2R on the transcriptional activity of Runx2. RESULTS P2Y2R deficiency in ApoE-/- mice caused extensive intimal calcification despite a significant reduction in atherosclerosis and macrophage plaque content. The ectoenzyme apyrase that degrades nucleoside di- and triphosphates accelerated high phosphate-induced calcium deposition in cultured VSMC. Expression of P2Y2R inhibits calcification in vitro inhibited the osteoblastic trans-differentiation of VSMC. Mechanistically, expression of P2Y2R inhibited Runx2 transcriptional activation of an osteocalcin promoter driven luciferase reporter gene. CONCLUSIONS This study reveals a role for vascular P2Y2R as an inhibitor of arterial intimal calcification and provides a new mechanistic insight into the regulation of the osteoblastic trans-differentiation of SMC through P2Y2R-mediated Runx2 antagonism. Given that calcification of atherosclerotic lesions is a significant clinical problem, activating P2Y2R may be an effective therapeutic approach for treatment or prevention of vascular calcification.
Collapse
Affiliation(s)
- Shaomin Qian
- Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jenna N Regan
- Medicine/Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maxwell T Shelton
- Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - April Hoggatt
- Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Khalid S Mohammad
- Medicine/Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul B Herring
- Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cheikh I Seye
- Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
15
|
Chen X, Qian S, Hoggatt A, Tang H, Hacker TA, Obukhov AG, Herring PB, Seye CI. Endothelial Cell-Specific Deletion of P2Y2 Receptor Promotes Plaque Stability in Atherosclerosis-Susceptible ApoE-Null Mice. Arterioscler Thromb Vasc Biol 2016; 37:75-83. [PMID: 27856454 DOI: 10.1161/atvbaha.116.308561] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Nucleotide P2Y2 receptor (P2Y2R) contributes to vascular inflammation by increasing vascular cell adhesion molecule-1 expression in endothelial cells (EC), and global P2Y2R deficiency prevents fatty streak formation in apolipoprotein E null (ApoE-/-) mice. Because P2Y2R is ubiquitously expressed in vascular cells, we investigated the contribution of endothelial P2Y2R in the pathogenesis of atherosclerosis. APPROACH AND RESULTS EC-specific P2Y2R-deficient mice were generated by breeding VEcadherin5-Cre mice with the P2Y2R floxed mice. Endothelial P2Y2R deficiency reduced endothelial nitric oxide synthase activity and significantly altered ATP- and UTP (uridine 5'-triphosphate)-induced vasorelaxation without affecting vasodilatory responses to acetylcholine. Telemetric blood pressure and echocardiography measurements indicated that EC-specific P2Y2R-deficient mice did not develop hypertension. We investigated the role of endothelial P2Y2R in the development of atherosclerotic lesions by crossing the EC-specific P2Y2R knockout mice onto an ApoE-/- background and evaluated lesion development after feeding a standard chow diet for 25 weeks. Histopathologic examination demonstrated reduced atherosclerotic lesions in the aortic sinus and entire aorta, decreased macrophage infiltration, and increased smooth muscle cell and collagen content, leading to the formation of a subendothelial fibrous cap in EC-specific P2Y2R-deficient ApoE-/- mice. Expression and proteolytic activity of matrix metalloproteinase-2 was significantly reduced in atherosclerotic lesions from EC-specific P2Y2R-deficient ApoE-/- mice. Furthermore, EC-specific P2Y2R deficiency inhibited nitric oxide production, leading to significant increase in smooth muscle cell migration out of aortic explants. CONCLUSIONS EC-specific P2Y2R deficiency reduces atherosclerotic burden and promotes plaque stability in ApoE-/- mice through impaired macrophage infiltration acting together with reduced matrix metalloproteinase-2 activity and increased smooth muscle cell migration.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Cell Movement
- Cells, Cultured
- Collagen/metabolism
- Disease Models, Animal
- Disease Progression
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Fibrosis
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Matrix Metalloproteinase 2/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Plaque, Atherosclerotic
- Purinergic P2Y Receptor Agonists/pharmacology
- Receptors, Purinergic P2Y2/deficiency
- Receptors, Purinergic P2Y2/genetics
- Rupture, Spontaneous
- Signal Transduction
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Xingjuan Chen
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Shaomin Qian
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - April Hoggatt
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Hongying Tang
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Timothy A Hacker
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Alexander G Obukhov
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Paul B Herring
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Cheikh I Seye
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.).
| |
Collapse
|
16
|
Rafehi M, Burbiel JC, Attah IY, Abdelrahman A, Müller CE. Synthesis, characterization, and in vitro evaluation of the selective P2Y 2 receptor antagonist AR-C118925. Purinergic Signal 2016; 13:89-103. [PMID: 27766552 DOI: 10.1007/s11302-016-9542-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/29/2016] [Indexed: 01/18/2023] Open
Abstract
The Gq protein-coupled, ATP- and UTP-activated P2Y2 receptor is a potential drug target for a range of different disorders, including tumor metastasis, inflammation, atherosclerosis, kidney disorders, and osteoporosis, but pharmacological studies are impeded by the limited availability of suitable antagonists. One of the most potent and selective antagonists is the thiouracil derivative AR-C118925. However, this compound was until recently not commercially available and little is known about its properties. We therefore developed an improved procedure for the synthesis of AR-C118925 and two derivatives to allow up-scaling and assessed their potency in calcium mobilization assays on the human and rat P2Y2 receptors recombinantly expressed in 1321N1 astrocytoma cells. The compound was further evaluated for inhibition of P2Y2 receptor-induced β-arrestin translocation. AR-C118925 behaved as a competitive antagonist with pA 2 values of 37.2 nM (calcium assay) and 51.3 nM (β-arrestin assay). Selectivity was assessed vs. related receptors including P2X, P2Y, and adenosine receptor subtypes, as well as ectonucleotidases. AR-C118925 showed at least 50-fold selectivity against the other investigated targets, except for the P2X1 and P2X3 receptors which were blocked by AR-C118925 at concentrations of about 1 μM. AR-C118925 is soluble in buffer at pH 7.4 (124 μM) and was found to be metabolically highly stable in human and mouse liver microsomes. In Caco2 cell experiments, the compound displayed moderate permeability indicating that it may show limited peroral bioavailability. AR-C118925 appears to be a useful pharmacological tool for in vitro and in vivo studies.
Collapse
Affiliation(s)
- Muhammad Rafehi
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB), Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany.,Pharmazeutisches Institut, Pharmazeutische Chemie I, An der Immenburg 4, D-53121, Bonn, Germany
| | - Joachim C Burbiel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB), Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Isaac Y Attah
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB), Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany.,Pharmazeutisches Institut, Pharmazeutische Chemie I, An der Immenburg 4, D-53121, Bonn, Germany
| | - Aliaa Abdelrahman
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB), Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany.,Pharmazeutisches Institut, Pharmazeutische Chemie I, An der Immenburg 4, D-53121, Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB), Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany. .,Pharmazeutisches Institut, Pharmazeutische Chemie I, An der Immenburg 4, D-53121, Bonn, Germany.
| |
Collapse
|
17
|
Agca Y, Qian S, Agca C, Seye CI. Direct Evidence for P2Y2 Receptor Involvement in Vascular Response to Injury. J Vasc Res 2016; 53:163-171. [PMID: 27723650 DOI: 10.1159/000449059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 08/10/2016] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Extracellular nucleotide release at the site of arterial injury mediates the proliferation and migration of vascular smooth muscle cells. Our aim was to investigate the role of the P2Y2 nucleotide receptor (P2Y2R) in neointimal hyperplasia. Approach and Results: Vascular injury was induced by the implantation of a polyethylene cuff around the femoral artery in wild-type and P2Y2R-deficient mice (P2Y2R-/-). Electron microscopy was used to analyze monocyte and lymphocyte influx to the intima 36 h after injury. Compared to wild-type littermates, P2Y2R-/- mice exhibited a 3-fold decreased number of mononuclear leukocytes invading the intima (p < 0.05). Concomitantly, the migration of smooth muscle cells was decreased by more than 60% (p < 0.05), resulting in a sharp inhibition of intimal thickening formation in P2Y2R-/- mice (n = 15) 14 days after cuff placement. In vitro, loss of P2Y2R significantly impaired monocyte migration in response to nucleotide agonists. Furthermore, transgenic rats overexpressing the P2Y2R developed accelerated intimal lesions resulting in more than 95% luminal stenosis (p < 0.05, n = 10). CONCLUSIONS Loss- and gain-of-function approaches established direct evidence for P2Y2R involvement in neointimal hyperplasia. Specific anti-P2Y2R therapies may be used against restenosis and bypass graft failure.
Collapse
Affiliation(s)
- Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Mont., USA
| | | | | | | |
Collapse
|
18
|
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
|
19
|
Dissmore T, Seye CI, Medeiros DM, Weisman GA, Bradford B, Mamedova L. The P2Y2 receptor mediates uptake of matrix-retained and aggregated low density lipoprotein in primary vascular smooth muscle cells. Atherosclerosis 2016; 252:128-135. [PMID: 27522265 PMCID: PMC5060008 DOI: 10.1016/j.atherosclerosis.2016.07.927] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS The internalization of aggregated low-density lipoproteins (agLDL) mediated by low-density lipoprotein receptor related protein (LRP1) may involve the actin cytoskeleton in ways that differ from the endocytosis of soluble LDL by the LDL receptor (LDLR). This study aims to define novel mechanisms of agLDL uptake through modulation of the actin cytoskeleton, to identify molecular targets involved in foam cell formation in vascular smooth muscle cells (VSMCs). The critical observation that formed the basis for these studies is that under pathophysiological conditions, nucleotide release from blood-derived and vascular cells activates SMC P2Y2 receptors (P2Y2Rs) leading to rearrangement of the actin cytoskeleton and cell motility. Therefore, we tested the hypothesis that P2Y2R activation mediates agLDL uptake by VSMCs. METHODS Primary VSMCs were isolated from aortas of wild type (WT) C57BL/6 and.P2Y2R-/- mice to investigate whether P2Y2R activation modulates LRP1 expression. Cells were transiently transfected with cDNA encoding a hemagglutinin-tagged (HA-tagged) WT P2Y2R, or a mutant P2Y2R that unlike the WT P2Y2R does not bind the cytoskeletal actin-binding protein filamin-A (FLN-A). RESULTS P2Y2R activation significantly increased agLDL uptake, and LRP1 mRNA expression decreased in P2Y2R-/- VSMCs versus WT. SMCs, expressing P2Y2R defective in FLN-A binding, exhibit 3-fold lower LDLR expression levels than SMCs expressing WT P2Y2R, while cells transfected with WT P2Y2R show greater agLDL uptake in both WT and P2Y2R-/- VSMCs versus cells transfected with the mutant P2Y2R. CONCLUSIONS Together, these results show that both LRP1 and LDLR expression and agLDL uptake are regulated by P2Y2R in VSMCs, and that agLDL uptake due to P2Y2R activation is dependent upon cytoskeletal reorganization mediated by P2Y2R binding to FLN-A.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Aorta/metabolism
- Cell Movement
- Cells, Cultured
- Cytoskeleton/metabolism
- Dose-Response Relationship, Drug
- Endocytosis
- Filamins/metabolism
- Foam Cells/metabolism
- Humans
- Lipoproteins, LDL/blood
- Low Density Lipoprotein Receptor-Related Protein-1
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microfilament Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Receptors, LDL/metabolism
- Receptors, Purinergic P2Y2/metabolism
- Signal Transduction
- Tumor Suppressor Proteins/metabolism
- Uridine Triphosphate/chemistry
Collapse
Affiliation(s)
| | - Cheikh I Seye
- Indiana University School of Medicine, Indianapolis, IN, United States
| | - Denis M Medeiros
- School of Graduate Studies, University of Missouri, Kansas City, MO, United States
| | - Gary A Weisman
- Department of Biochemistry and Bond Life Sciences Center, University of Missouri, Columbia, United States
| | - Barry Bradford
- Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States
| | - Laman Mamedova
- Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States.
| |
Collapse
|
20
|
Qian S, Hoggatt A, Jones-Hall YL, Ware CF, Herring P, Seye CI. Deletion of P2Y2 receptor reveals a role for lymphotoxin-α in fatty streak formation. Vascul Pharmacol 2016; 85:11-20. [PMID: 27355755 PMCID: PMC5453728 DOI: 10.1016/j.vph.2016.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/02/2016] [Accepted: 06/02/2016] [Indexed: 11/15/2022]
Abstract
Background Lymphotoxin alpha (LTα) is expressed in human atherosclerotic lesions and genetic variations in the LTα pathway have been linked to myocardial infarction. Activation of the P2Y2 nucleotide receptor (P2Y2R) regulates the production of LTα. in vitro. We aimed to uncover a potential pathway linking purinergic receptor to LTα-mediated inflammatory processes pivotal to the early stages of atherosclerosis in apolipoprotein E (ApoE−/−) deficient mice. Methods and results En face immunostaining revealed that P2Y2R and VCAM-1 are preferentially expressed in the atherosclerosis prone site of the mouse aortic sinus. Deletion of the P2Y2R gene suppresses VCAM-1 expression. Compared with ApoE−/−mice, ApoE−/−mice lacking the P2Y2R gene (ApoE−/−/P2Y2R−/−) did not develop fatty streak lesions when fed a standard chow diet for 15 weeks. Systemic and CD4+ T cell production of the pro-inflammatory cytokine lymphotoxin-alpha (LTα) were specifically inhibited in ApoE−/−/P2Y2R−/− mice. Anti-LTα preventive treatment was initiated in ApoE−/− mice with intraperitoneal administration of recombinant human tumor necrosis factor receptor 1 fusion protein (TNFR1-Fc) on 5 consecutive days before the disease onset. Remarkably, none of the TNFR1:Fc-treated ApoE−/− mice exhibited atherosclerotic lesions at any developmental stage. Significance ApoE−/− mice deficient in P2Y2R exhibit low endothelial cell VCAM-1 levels, decreased production of LTα and delayed onset of atherosclerosis. These data suggest that targeting this nucleotide receptor could be an effective therapeutic approach in atherosclerosis.
Collapse
Affiliation(s)
- Shaomin Qian
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 332, Indianapolis, IN 46202, United States
| | - April Hoggatt
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 332, Indianapolis, IN 46202, United States
| | - Yava L Jones-Hall
- Department of Comparative Pathobiology, Purdue University, College of Veterinary Medicine, 725 Harrison Street VPTH 124, West Lafayette, IN 47907-2027, United States
| | - Carl F Ware
- Sanford-Burnham Medical Institute, La Jolla, CA 92037, United States
| | - Paul Herring
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 332, Indianapolis, IN 46202, United States
| | - Cheikh I Seye
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive MS 332, Indianapolis, IN 46202, United States.
| |
Collapse
|
21
|
Stachon P, Geis S, Peikert A, Heidenreich A, Michel NA, Ünal F, Hoppe N, Dufner B, Schulte L, Marchini T, Cicko S, Ayata K, Zech A, Wolf D, Hilgendorf I, Willecke F, Reinöhl J, von Zur Mühlen C, Bode C, Idzko M, Zirlik A. Extracellular ATP Induces Vascular Inflammation and Atherosclerosis via Purinergic Receptor Y2 in Mice. Arterioscler Thromb Vasc Biol 2016; 36:1577-86. [PMID: 27339459 DOI: 10.1161/atvbaha.115.307397] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 06/02/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE A solid body of evidence supports a role of extracellular ATP and its P2 receptors in innate and adaptive immunity. It promotes inflammation as a danger signal in various chronic inflammatory diseases. Thus, we hypothesize contribution of extracellular ATP and its receptor P2Y2 in vascular inflammation and atherosclerosis. APPROACH AND RESULTS Extracellular ATP induced leukocyte rolling, adhesion, and migration in vivo as assessed by intravital microscopy and in sterile peritonitis. To test the role of extracellular ATP in atherosclerosis, ATP or saline as control was injected intraperitoneally 3× a week in low-density lipoprotein receptor(-/-) mice consuming high cholesterol diet. Atherosclerosis significantly increased after 16 weeks in ATP-treated mice (n=13; control group, 0.26 mm2; ATP group, 0.33 mm2; P=0.01). To gain into the role of ATP-receptor P2Y2 in ATP-induced leukocyte recruitment, ATP was administered systemically in P2Y2-deficient or P2Y2-competent mice. In P2Y2-deficient mice, the ATP-induced leukocyte adhesion was significantly reduced as assessed by intravital microscopy. P2Y2 expression in atherosclerosis was measured by real-time polymerase chain reaction and immunohistochemistry and demonstrates an increased expression mainly caused by influx of P2Y2-expressing macrophages. To investigate the functional role of P2Y2 in atherogenesis, P2Y2-deficient low-density lipoprotein receptor(-/-) mice consumed high cholesterol diet. After 16 weeks, P2Y2-deficient mice showed significantly reduced atherosclerotic lesions with decreased macrophages compared with P2Y2-competent mice (n=11; aortic arch: control group, 0.25 mm(2); P2Y2-deficient, 0.14 mm2; P=0.04). Mechanistically, atherosclerotic lesions from P2Y2-deficient mice expressed less vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 RNA. CONCLUSIONS We show that extracellular ATP induces vascular inflammation and atherosclerosis via activation of P2Y2.
Collapse
Affiliation(s)
- Peter Stachon
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Serjosha Geis
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Alexander Peikert
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Adrian Heidenreich
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Nathaly Anto Michel
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Fatih Ünal
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Natalie Hoppe
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Bianca Dufner
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Lisa Schulte
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Timoteo Marchini
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Sanja Cicko
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Korcan Ayata
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Andreas Zech
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Florian Willecke
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Jochen Reinöhl
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Constantin von Zur Mühlen
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Marco Idzko
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany
| | - Andreas Zirlik
- From the Atherogenesis Research Group, University Heart Center Freiburg, Department of Cardiology and Angiology I (P.S., S.G., A.P., A.H., N.A.M., F.Ü., N.H., B.D., L.S., T.M., D.W., I.H., F.W., J.R., C.v.z.M., C.B., A.Z.) and Department of Pneumology (S.C., K.A., A.Z., M.I.), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
22
|
Schreier B, Schwerdt G, Heise C, Bethmann D, Rabe S, Mildenberger S, Gekle M. Substance-specific importance of EGFR for vascular smooth muscle cells motility in primary culture. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1519-33. [PMID: 27012600 DOI: 10.1016/j.bbamcr.2016.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/01/2016] [Accepted: 03/18/2016] [Indexed: 11/26/2022]
Abstract
Besides their importance for the vascular tone, vascular smooth muscle cells (VSMC) also contribute to pathophysiological vessel alterations. Various G-protein coupled receptor ligands involved in vascular dysfunction and remodeling can transactivate the epidermal growth factor receptor (EGFR) of VSMC, yet the importance of EGFR transactivation for the VSMC phenotype is incompletely understood. The aims of this study were (i) to characterize further the importance of the VSMC-EGFR for proliferation, migration and marker gene expression for inflammation, fibrosis and reactive oxygen species (ROS) homeostasis and (ii) to test the hypothesis that vasoactive substances (endothelin-1, phenylephrine, thrombin, vasopressin and ATP) rely differentially on the EGFR with respect to the abovementioned phenotypic alterations. In primary, aortic VSMC from mice without conditional deletion of the EGFR, proliferation, migration, marker gene expression (inflammation, fibrosis and ROS homeostasis) and cell signaling (ERK 1/2, intracellular calcium) were analyzed. VSMC-EGFR loss reduced collective cell migration and single cell migration probability, while no difference between the genotypes in single cell velocity, chemotaxis or marker gene expression could be observed under control conditions. EGF promoted proliferation, collective cell migration, chemokinesis and chemotaxis and leads to a proinflammatory gene expression profile in wildtype but not in knockout VSMC. Comparing the impact of five vasoactive substances (all reported to transactivate EGFR and all leading to an EGFR dependent increase in ERK1/2 phosphorylation), we demonstrate that the importance of EGFR for their action is substance-dependent and most apparent for crowd migration but plays a minor role for gene expression regulation.
Collapse
Affiliation(s)
- Barbara Schreier
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany.
| | - Gerald Schwerdt
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany
| | - Christian Heise
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany
| | - Daniel Bethmann
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany
| | - Sindy Rabe
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany
| | - Sigrid Mildenberger
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, University of Halle-Wittenberg, Magdeburger Strasse 6, 06112 Halle/Saale, Germany
| |
Collapse
|
23
|
Ruchel JB, Rezer JFP, Thorstenberg ML, Dos Santos CB, Cabral FL, Lopes STA, da Silva CB, Machado AK, da Cruz IBM, Schetinger MRC, Gonçalves JF, Leal DBR. Hypercholesterolemia and Ecto-enzymes of Purinergic System: Effects of Paullinia cupana. Phytother Res 2015; 30:49-57. [PMID: 26514663 DOI: 10.1002/ptr.5499] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 08/30/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022]
Abstract
Hypercholesterolemia is a metabolic disorder characterized by high levels of low-density lipoprotein and blood cholesterol, causing inflammatory lesion. Purinergic signaling modulates the inflammatory and immune responses through adenine nucleotides and nucleoside. Guaraná has hypocholesterolemic and antiinflammatory properties. Considering that there are few studies demonstrating the effects of guaraná powder on the metabolism of adenine nucleotides, we investigated its effects on the activity of ecto-nucleoside triphosphate diphosphohydrolase (E-NTPDase) and ecto-adenosine deaminase activity in lymphocytes of rats with diet-induced hypercholesterolemia. The rats were divided into hypercholesterolemic and normal diet groups. Each group was subdivided by treatment: saline, guaraná powder 12.5, 25, or 50 mg/kg/day and caffeine concentration equivalent to highest dose of guaraná, fed orally for 30 days. An increase in adenosine triphosphate hydrolysis was observed in the lymphocytes of rats with hypercholesterolemia and treated with 25 or 50 mg/kg/day when compared with the other groups. The hypercholesterolemic group treated with the highest concentration of guaraná powder showed decreased ecto-adenosine deaminase activity compared with the normal diet groups. Guaraná was able to reduce the total cholesterol and low-density lipoprotein cholesterol to basal levels in hypercholesterolemic rats. High concentrations of guaraná associated with a hypercholesterolemic diet are likely to have contributed to the reduction of the inflammatory process.
Collapse
Affiliation(s)
- J B Ruchel
- Centro de Ciências Naturais e Exatas, Departamento de Química, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil.,Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - J F P Rezer
- Centro de Ciências Naturais e Exatas, Departamento de Química, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil.,Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - M L Thorstenberg
- Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - C B Dos Santos
- Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - F L Cabral
- Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - S T A Lopes
- Centro de Ciências Rurais, Departamento de Patologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - C B da Silva
- Centro de Ciências Rurais, Departamento de Patologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - A K Machado
- Centro de Ciências da Saúde, Departamento de Morfologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - I B M da Cruz
- Centro de Ciências da Saúde, Departamento de Morfologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - M R C Schetinger
- Centro de Ciências Naturais e Exatas, Departamento de Química, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - J F Gonçalves
- Centro de Ciências Naturais e Exatas, Departamento de Química, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil.,Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - D B R Leal
- Centro de Ciências da Saúde, Departamento de Microbiologia e Parasitologia, Universidade Federal de Santa Maria, Campus Universitário, Camobi, Santa Maria, RS, 97105-900, Brazil
| |
Collapse
|
24
|
Eun SY, Ko YS, Park SW, Chang KC, Kim HJ. IL-1β enhances vascular smooth muscle cell proliferation and migration via P2Y2 receptor-mediated RAGE expression and HMGB1 release. Vascul Pharmacol 2015; 72:108-17. [PMID: 25956731 DOI: 10.1016/j.vph.2015.04.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 01/11/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessel walls, and their proliferation and migration play important roles in the development of atherosclerosis. Recently, it has been reported that IL-1β mediates the inflammatory response through the upregulation of the P2Y2 receptor (P2Y2R). Thus, we examined the role of P2Y2R in IL-1β-mediated proliferation and migration of VSMCs and the underlying molecular mechanisms. VSMCs were pretreated with IL-1β for 24h to upregulate P2Y2R expression. The cells were then stimulated with UTP or ATP for the indicated times, and cell proliferation and migration and the related signaling pathways were examined. The equipotent P2Y2R agonists ATP and UTP enhanced proliferation, RAGE expression and HMGB1 secretion in IL-1β-pretreated VSMCs. Additionally, pretreatment with IL-1β enhanced UTP-mediated VSMC migration and MMP-2 release, but these effects were not observed in the P2Y2R-siRNA- or RAGE-siRNA-transfected VSMCs. Next, the signaling molecules involved in P2Y2R-mediated cell proliferation and migration were determined. The ERK, AKT, PKC, Rac-1 and ROCK2 pathways were involved in UTP-induced cell proliferation and migration, MMP-2 and HMGB1 secretion and RAGE expression in the IL-1β-pretreated VSMCs. UTP induced the phosphorylation of ERK, AKT and PKC and the translocation of Rac-1 and ROCK2 from cytosol to membrane as well as stress fiber formation, which were markedly increased in the IL-1β-pretreated VSMCs but not in the P2Y2R-siRNA-transfected VSMCs. These results demonstrate that pro-inflammatory cytokines associated with atherosclerosis, such as IL-1β, can accelerate the process of atherosclerosis through the upregulation of P2Y2R.
Collapse
Affiliation(s)
- So Young Eun
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Young Shin Ko
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Ki Churl Chang
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea.
| |
Collapse
|
25
|
Infection by Toxoplasma gondii, a severe parasite in neonates and AIDS patients, causes impaired anion secretion in airway epithelia. Proc Natl Acad Sci U S A 2015; 112:4435-40. [PMID: 25831498 DOI: 10.1073/pnas.1503474112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The airway epithelia initiate and modulate the inflammatory responses to various pathogens. The cystic fibrosis transmembrane conductance regulator-mediated Cl(-) secretion system plays a key role in mucociliary clearance of inhaled pathogens. We have explored the effects of Toxoplasma gondii, an opportunistic intracellular protozoan parasite, on Cl(-) secretion of the mouse tracheal epithelia. In this study, ATP-induced Cl(-) secretion indicated the presence of a biphasic short-circuit current (Isc) response, which was mediated by a Ca(2+)-activated Cl(-) channel (CaCC) and the cystic fibrosis transmembrane conductance regulator. However, the ATP-evoked Cl(-) secretion in T. gondii-infected mouse tracheal epithelia and the elevation of [Ca(2+)]i in T. gondii-infected human airway epithelial cells were suppressed. Quantitative reverse transcription-PCR revealed that the mRNA expression level of the P2Y2 receptor (P2Y2-R) increased significantly in T. gondii-infected mouse tracheal cells. This revealed the influence that pathological changes in P2Y2-R had on the downstream signal, suggesting that P2Y2-R was involved in the mechanism underlying T. gondii infection in airways. These results link T. gondii infection as well as other pathogen infections to Cl(-) secretion, via P2Y2-R, which may provide new insights for the treatment of pneumonia caused by pathogens including T. gondii.
Collapse
|
26
|
Ferrari D, Vitiello L, Idzko M, la Sala A. Purinergic signaling in atherosclerosis. Trends Mol Med 2015; 21:184-92. [PMID: 25637413 DOI: 10.1016/j.molmed.2014.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/19/2014] [Accepted: 12/19/2014] [Indexed: 12/28/2022]
Abstract
Cell surface expression of specific receptors and ecto-nucleotidases makes extracellular nucleotides such as ATP, ADP, UTP, and adenosine suitable as signaling molecules for physiological and pathological events, including tissue stress and damage. Recent data have revealed the participation of purinergic signaling in atherosclerosis, depicting a scenario in which, in addition to some exceptions reflecting dual effects of individual receptor subtypes, adenosine and most P1 receptors, as well as ecto-nucleotidases, show a protective, anti-atherosclerotic function. By contrast, P2 receptors promote atherosclerosis. In consideration of these findings, modulation of purinergic signaling would represent an innovative and valuable tool to counteract atherosclerosis. We summarize recent developments on the participation of the purinergic network in atheroma formation and evolution.
Collapse
Affiliation(s)
- Davide Ferrari
- Department of Life Sciences and Biotechnology, Biotechnology Centre, University of Ferrara, 44121 Ferrara, Italy.
| | - Laura Vitiello
- Laboratory of Molecular and Cellular Immunology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Pisana, 00166 Rome, Italy
| | - Marco Idzko
- Department of Pneumology, Freiburg University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - Andrea la Sala
- Laboratory of Molecular and Cellular Immunology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Pisana, 00166 Rome, Italy
| |
Collapse
|
27
|
Erb L, Cao C, Ajit D, Weisman GA. P2Y receptors in Alzheimer's disease. Biol Cell 2014; 107:1-21. [PMID: 25179475 DOI: 10.1111/boc.201400043] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/27/2014] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting more than 10% of people over the age of 65. Age is the greatest risk factor for AD, although a combination of genetic, lifestyle and environmental factors also contribute to disease development. Common features of AD are the formation of plaques composed of beta-amyloid peptides (Aβ) and neuronal death in brain regions involved in learning and memory. Although Aβ is neurotoxic, the primary mechanisms by which Aβ affects AD development remain uncertain and controversial. Mouse models overexpressing amyloid precursor protein and Aβ have revealed that Aβ has potent effects on neuroinflammation and cerebral blood flow that contribute to AD progression. Therefore, it is important to consider how endogenous signalling in the brain responds to Aβ and contributes to AD pathology. In recent years, Aβ has been shown to affect ATP release from brain and blood cells and alter the expression of G protein-coupled P2Y receptors that respond to ATP and other nucleotides. Accumulating evidence reveals a prominent role for P2Y receptors in AD pathology, including Aβ production and elimination, neuroinflammation, neuronal function and cerebral blood flow.
Collapse
Affiliation(s)
- Laurie Erb
- Department of Biochemistry, Life Sciences Center, University of Missouri, Columbia, MO, 65211, U.S.A
| | | | | | | |
Collapse
|
28
|
McEnaney RM, Shukla A, Madigan MC, Sachdev U, Tzeng E. P2Y2 nucleotide receptor mediates arteriogenesis in a murine model of hind limb ischemia. J Vasc Surg 2014; 63:216-25. [PMID: 25088742 DOI: 10.1016/j.jvs.2014.06.112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/13/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Arteriogenesis represents the maturation of preformed vascular connections in response to flow changes and shear stress. These collateral vessels can restore up to 60% of the native blood flow. Shear stress and vascular injury can induce the release of nucleotides from vascular smooth muscle cells and platelets that can serve as signaling ligands, suggesting they may be involved in mediating arteriogenesis. The P2Y2 nucleotide receptor (P2Y2R) has also been shown to mediate smooth muscle migration and arterial remodeling. Thus, we hypothesize that P2Y2R mediates arteriogenesis in response to ischemia. METHODS Hind limb ischemia was induced by femoral artery ligation (FAL) in C57Bl/6NJ or P2Y2R negative mice (P2Y2(-/-)). Hind limb perfusion was measured with laser Doppler perfusion imaging and compared with the sham-operated contralateral limb immediately and at 3, 7, 14, 21, and 28 days after ligation. Collateral vessel size was measured by Microfil casting. Muscle specimens were harvested and analyzed with immunohistochemistry for Ki67, vascular cell adhesion molecule, macrophages, and muscle viability by hematoxylin and essoin stain. RESULTS Hind limb ischemia induced by FAL in C57Bl/6NJ mice resulted in significant ischemia as measured by laser Doppler perfusion imaging. There was rapid recovery to nearly normal levels of perfusion by 2 weeks. FAL in P2Y2(-/-) mice resulted in severe ischemia with greater tissue loss. Recovery of perfusion was impaired, achieving only 40% compared with wild-type mice by 28 days. Collateral vessels in the P2Y2(-/-) mice were underdeveloped, with reduced vascular cell proliferation and smaller vessel size. The collaterals were ∼65% the size of wild-type collateral vessels (P = .011). Angiogenesis at 28 days in the ischemic muscle, however, was greater in the P2Y2(-/-) mice (P < .001), possibly related to persistent ischemia leading and angiogenic drive. Early macrophage recruitment was reduced by nearly 70% in P2Y2(-/-) despite significantly more myocyte necrosis. However, inflammation was greater at 28 days in the P2Y2(-/-) mice. CONCLUSIONS P2Y2R deficiency does not alter baseline collateral vessel formation but does significantly impair collateral maturation, with resultant persistent limb ischemia despite enhanced angiogenesis. These findings reinforce the importance of arteriogenesis in the recovery of perfusion in ischemic tissues compared with angiogenesis. They also support the role of P2Y2R in mediating this process. The mechanism by which P2Y2R mediates arteriogenesis may involve the recruitment of inflammatory cells to the ischemic tissues, which is essential to arteriogenesis. Approaches to target P2Y2R may yield new therapeutic strategies for the treatment of arterial occlusive disease.
Collapse
Affiliation(s)
- Ryan M McEnaney
- Surgery Services, Department of Veterans Affairs Medical Center, Pittsburgh, Pa; Division of Vascular Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Ankur Shukla
- Surgery Services, Department of Veterans Affairs Medical Center, Pittsburgh, Pa; Division of Vascular Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Michael C Madigan
- Surgery Services, Department of Veterans Affairs Medical Center, Pittsburgh, Pa; Division of Vascular Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Ulka Sachdev
- Division of Vascular Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Edith Tzeng
- Surgery Services, Department of Veterans Affairs Medical Center, Pittsburgh, Pa; Division of Vascular Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pa.
| |
Collapse
|
29
|
Eun SY, Park SW, Lee JH, Chang KC, Kim HJ. P2Y(2)R activation by nucleotides released from oxLDL-treated endothelial cells (ECs) mediates the interaction between ECs and immune cells through RAGE expression and reactive oxygen species production. Free Radic Biol Med 2014; 69:157-66. [PMID: 24486339 DOI: 10.1016/j.freeradbiomed.2014.01.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 01/14/2014] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
Lipoprotein oxidation, inflammation, and immune responses involving the vascular endothelium and immune cells contribute to the pathogenesis of atherosclerosis. In an atherosclerotic animal model, P2Y2 receptor (P2Y2R) upregulation and stimulation were previously shown to induce intimal hyperplasia and increased intimal monocyte infiltration. Thus, we investigated the role of P2Y2R in oxidized low-density lipoprotein (oxLDL)-mediated oxidative stress and the subsequent interaction between endothelial cells (ECs) and immune cells. The treatment of human ECs with oxLDL caused the rapid release of ATP (maximum after 5 min). ECs treated with oxLDL or the P2Y2R agonists ATP/UTP for 1h exhibited significant reactive oxygen species (ROS) production, but this effect was not observed in P2Y2R siRNA-transfected ECs. In addition, oxLDL and ATP/UTP both induced RAGE expression, which was P2Y2R dependent. Oxidized LDL- and ATP/UTP-mediated ROS production was diminished in RAGE siRNA-transfected ECs, suggesting that RAGE is an important mediator in P2Y2R-mediated ROS production. Treatment with oxLDL for 24h induced P2Y2R expression in the human monocyte cell line THP-1 and increased THP-1 cell migration toward ECs. The addition of apyrase, an enzyme that hydrolyzes nucleotides, or diphenyleneiodonium (DPI), a well-known inhibitor of NADPH oxidase, significantly inhibited the increase in cell migration caused by oxLDL. P2Y2R siRNA-transfected THP-1 cells did not migrate in response to oxLDL or ATP/UTP treatment, indicating a critical role for P2Y2R and nucleotide release in oxLDL-induced monocyte migration. Last, oxLDL and ATP/UTP effectively increased ICAM-1 and VCAM-1 expression and the subsequent binding of THP-1 cells to ECs, which was inhibited by pretreatment with DPI or by siRNA against P2Y2R or RAGE, suggesting that P2Y2R is an important mediator in oxLDL-mediated monocyte adhesion to ECs through the regulation of ROS-dependent adhesion molecule expression in ECs. Taken together, our findings suggest that P2Y2R could be a therapeutic target for the prevention of vascular disorders, including atherosclerosis.
Collapse
Affiliation(s)
- So Young Eun
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-701, Korea
| | - Sang Won Park
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-701, Korea
| | - Jae Heun Lee
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-701, Korea
| | - Ki Churl Chang
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-701, Korea
| | - Hye Jung Kim
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-701, Korea.
| |
Collapse
|
30
|
Li N, Lu ZY, Yu LH, Burnstock G, Deng XM, Ma B. Inhibition of G protein-coupled P2Y2 receptor induced analgesia in a rat model of trigeminal neuropathic pain. Mol Pain 2014; 10:21. [PMID: 24642246 PMCID: PMC3995183 DOI: 10.1186/1744-8069-10-21] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 03/04/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUNDS ATP and P2X receptors play important roles in the modulation of trigeminal neuropathic pain, while the role of G protein-coupled P2Y₂ receptors and the underlying mechanisms are less clear. The threshold and frequency of action potentials, fast inactivating transient K+ channels (IA) are important regulators of membrane excitability in sensory neurons because of its vital role in the control of the spike onset. In this study, pain behavior tests, QT-RT-PCR, immunohistochemical staining, and patch-clamp recording, were used to investigate the role of P2Y₂ receptors in pain behaviour. RESULTS In control rats: 1) UTP, an agonist of P2Y₂/P2Y₄ receptors, caused a significant decrease in the mean threshold intensities for evoking action potentials and a striking increase in the mean number of spikes evoked by TG neurons. 2) UTP significantly inhibited IA and the expression of Kv1.4, Kv3.4 and Kv4.2 subunits in TG neurons, which could be reversed by the P2 receptor antagonist suramin and the ERK antagonist U0126. In ION-CCI (chronic constriction injury of infraorbital nerve) rats: 1) mRNA levels of Kv1.4, Kv3.4 and Kv4.2 subunits were significantly decreased, while the protein level of phosphorylated ERK was significantly increased. 2) When blocking P2Y₂ receptors by suramin or injection of P2Y2R antisense oligodeoxynucleotides both led to a time- and dose-dependent reverse of allodynia in ION-CCI rats. 3) Injection of P2Y₂ receptor antisense oligodeoxynucleotides induced a pronounced decrease in phosphorylated ERK expression and a significant increase in Kv1.4, Kv3.4 and Kv4.2 subunit expression in trigeminal ganglia. CONCLUSIONS Our data suggest that inhibition of P2Y₂ receptors leads to down-regulation of ERK-mediated phosphorylation and increase of the expression of I(A)-related Kv channels in trigeminal ganglion neurons, which might contribute to the clinical treatment of trigeminal neuropathic pain.
Collapse
Affiliation(s)
| | | | | | | | - Xiao-ming Deng
- Department of Physiology, The Key Laboratory of Molecular Neurobiology, Ministry of Education, Second Military Medical University, Shanghai 200433, P,R, China.
| | | |
Collapse
|
31
|
Liao Z, Cao C, Wang J, Huxley VH, Baker O, Weisman GA, Erb L. The P2Y 2 Receptor Interacts with VE-Cadherin and VEGF Receptor-2 to Regulate Rac1 Activity in Endothelial Cells. ACTA ACUST UNITED AC 2014; 7:1105-1121. [PMID: 25657827 PMCID: PMC4314728 DOI: 10.4236/jbise.2014.714109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Vascular endothelial cadherin (VE-cadherin) mediates homophylic adhesion between endothelial cells and is an important regulator of angiogenesis, blood vessel permeability and leukocyte trafficking. Rac1, a member of the Rho family of GTPases, controls VE-cadherin adhesion by acting downstream of several growth factors, including angiopoietin-1 and vascular endothelial growth factor (VEGF). Here we show that UTP-induced activation of the Gq protein-coupled P2Y2 nucleotide receptor (P2Y2R) in human coronary artery endothelial cells (HCAECs) activated Rac1 and caused a transient complex to form between P2Y2R, VE-cadherin and VEGF receptor-2 (VEGFR-2). Knockdown of VE-cadherin expression with siRNA did not affect UTP-induced activation of extracellular signal-regulated kinases 1/2 (ERK1/2) but led to a loss of UTP-induced Rac1 activation and tyrosine phosphorylation of p120 catenin, a cytoplasmic protein known to interact with VE-cadherin. Activation of the P2Y2R by UTP also caused a prolonged interaction between p120 catenin and vav2 (a guanine nucleotide exchange factor for Rac) that correlated with the kinetics of UTP-induced tyrosine phosphorylation of p120 catenin and VE-cadherin. Inhibitors of VEGFR-2 (SU1498) or Src (PP2) significantly diminished UTP-induced Rac1 activation, tyrosine phosphorylation of p120 catenin and VE-cadherin, and association of the P2Y2R with VE-cadherin and p120 catenin with vav2. These findings suggest that the P2Y2R uses Src and VEGFR-2 to mediate association of the P2Y2R with VE-cadherin complexes in endothelial adherens junctions to activate Rac1.
Collapse
Affiliation(s)
- Zhongji Liao
- Department of Medicine, University of California, San Diego, USA
| | - Chen Cao
- Department of Biochemistry, Life Sciences Center, University of Missouri, Columbia, USA
| | - Jianjie Wang
- Department of Biomedical Sciences, Missouri State University, Springfield, USA
| | - Virginia H Huxley
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, USA
| | - Olga Baker
- School of Dentistry, University of Utah, Salt Lake City, USA
| | - Gary A Weisman
- Department of Biochemistry, Life Sciences Center, University of Missouri, Columbia, USA
| | - Laurie Erb
- Department of Biochemistry, Life Sciences Center, University of Missouri, Columbia, USA
| |
Collapse
|
32
|
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
|
33
|
Ajit D, Woods LT, Camden JM, Thebeau CN, El-Sayed FG, Greeson GW, Erb L, Petris MJ, Miller DC, Sun GY, Weisman GA. Loss of P2Y₂ nucleotide receptors enhances early pathology in the TgCRND8 mouse model of Alzheimer's disease. Mol Neurobiol 2013; 49:1031-42. [PMID: 24193664 DOI: 10.1007/s12035-013-8577-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/21/2013] [Indexed: 11/26/2022]
Abstract
Neuroinflammation is a prominent feature in Alzheimer's disease (AD) and activation of the brain's innate immune system, particularly microglia, has been postulated to both retard and accelerate AD progression. Recent studies indicate that the G protein-coupled P2Y2 nucleotide receptor (P2Y2R) is an important regulator of innate immunity by assisting in the recruitment of monocytes to injured tissue, neutrophils to bacterial infections and eosinophils to allergen-infected lungs. In this study, we investigated the role of the P2Y2R in progression of an AD-like phenotype in the TgCRND8 mouse model that expresses Swedish and Indiana mutations in amyloid precursor protein (APP). Our results indicate that P2Y 2 R expression is upregulated in TgCRND8 mouse brain within 10 weeks of age and then decreases after 25 weeks of age, as compared to littermate controls expressing low levels of the P2Y 2 R. TgCRND8 mice with homozygous P2Y 2 R deletion survive less than 5 weeks, whereas mice with heterozygous P2Y 2 R deletion survive for 12 weeks, a time point when TgCRND8 mice are fully viable. Heterozygous P2Y 2 R deletion in TgCRND8 mice increased β-amyloid (Aβ) plaque load and soluble Aβ1-42 levels in the cerebral cortex and hippocampus, decreased the expression of the microglial marker CD11b in these brain regions and caused neurological deficits within 10 weeks of age, as compared to age-matched TgCRND8 mice. These findings suggest that the P2Y2R is important for the recruitment and activation of microglial cells in the TgCRND8 mouse brain and that the P2Y2R may regulate neuroprotective mechanisms through microglia-mediated clearance of Aβ that when lost can accelerate the onset of an AD-like phenotype in the TgCRND8 mouse.
Collapse
Affiliation(s)
- Deepa Ajit
- Department of Biochemistry, University of Missouri, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO, 65211-7310, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Matsumoto T, Watanabe S, Kawamura R, Taguchi K, Kobayashi T. Enhanced uridine adenosine tetraphosphate-induced contraction in renal artery from type 2 diabetic Goto-Kakizaki rats due to activated cyclooxygenase/thromboxane receptor axis. Pflugers Arch 2013; 466:331-42. [PMID: 23900807 DOI: 10.1007/s00424-013-1330-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/15/2013] [Accepted: 07/16/2013] [Indexed: 12/28/2022]
Abstract
The dinucleotide uridine adenosine tetraphosphate (Up4A), which has both purine and pyrimidine moieties, was reported as a novel endothelium-derived contracting factor. Recently, growing evidence has suggested that Up4A plays an important role in regulation of the cardiovascular function. We previously demonstrated that Up4A-induced vasoconstrictions are altered in arteries from DOCA-salt hypertensive rats. We have assessed responses to Up4A shown by renal arteries from type 2 diabetic Goto-Kakizaki (GK) rats (42-46 weeks old) and identified the molecular mechanisms involved. Concentration-dependent contractions to Up4A were greater in renal arterial rings from the GK than age-matched control Wistar group. In both groups, the inhibition of nitric oxide synthase (with N (G)-nitro-L-arginine) increased the response to Up4A, whereas the inhibition of cyclooxygenase (COX) (with indomethacin) decreased the response. Specific inhibitors of COX-1 (valeroyl salicylate) and COX-2 (NS398), a thromboxane (TX) receptor (TP) antagonist (SQ29548), and P2 receptor antagonist (suramin) also decreased the response to Up4A. Protein expressions of COXs in renal arteries were greater in the GK than Wistar group. The production of TXB2 (a metabolite of TXA2) by Up4A did not differ between these groups. Concentration-dependent contractions to U46619, an agonist of the TP receptor, were greater in renal arteries from the GK than Wistar group. The expression of P2X1 and P2Y2 receptors did not differ between these groups. These results suggest that enhancement of the Up4A-induced contraction in renal arteries from GK rats may be attributable to the increased activation of COXs/TP receptor signaling.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | | | | | | | | |
Collapse
|
35
|
Weisman GA, Woods LT, Erb L, Seye CI. P2Y receptors in the mammalian nervous system: pharmacology, ligands and therapeutic potential. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2013; 11:722-38. [PMID: 22963441 DOI: 10.2174/187152712803581047] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 06/14/2012] [Accepted: 06/14/2012] [Indexed: 11/22/2022]
Abstract
P2Y receptors for extracellular nucleotides are coupled to activation of a variety of G proteins and stimulate diverse intracellular signaling pathways that regulate functions of cell types that comprise the central nervous system (CNS). There are 8 different subtypes of P2Y receptor expressed in cells of the CNS that are activated by a select group of nucleotide agonists. Here, the agonist selectivity of these 8 P2Y receptor subtypes is reviewed with an emphasis on synthetic agonists with high potency and resistance to degradation by extracellular nucleotidases that have potential applications as therapeutic agents. In addition, the recent identification of a wide variety of subtype-selective antagonists is discussed, since these compounds are critical for discerning cellular responses mediated by activation of individual P2Y receptor subtypes. The functional expression of P2Y receptor subtypes in cells that comprise the CNS is also reviewed and the role of each subtype in the regulation of physiological and pathophysiological responses is considered. Other topics include the role of P2Y receptors in the regulation of blood-brain barrier integrity and potential interactions between different P2Y receptor subtypes that likely impact tissue responses to extracellular nucleotides in the CNS. Overall, current research suggests that P2Y receptors in the CNS regulate repair mechanisms that are triggered by tissue damage, inflammation and disease and thus P2Y receptors represent promising targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, 540E Life Sciences Center, 1201 Rollins Road, University of Missouri, Columbia, MO 65211-7310, USA.
| | | | | | | |
Collapse
|
36
|
Peterson TS, Thebeau CN, Ajit D, Camden JM, Woods LT, Wood WG, Petris MJ, Sun GY, Erb L, Weisman GA. Up-regulation and activation of the P2Y(2) nucleotide receptor mediate neurite extension in IL-1β-treated mouse primary cortical neurons. J Neurochem 2013; 125:885-96. [PMID: 23550835 DOI: 10.1111/jnc.12252] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 03/26/2013] [Accepted: 03/28/2013] [Indexed: 12/12/2022]
Abstract
The pro-inflammatory cytokine interleukin-1β (IL-1β), whose levels are elevated in the brain in Alzheimer's and other neurodegenerative diseases, has been shown to have both detrimental and beneficial effects on disease progression. In this article, we demonstrate that incubation of mouse primary cortical neurons (mPCNs) with IL-1β increases the expression of the P2Y2 nucleotide receptor (P2Y2R) and that activation of the up-regulated receptor with UTP, a relatively selective agonist of the P2Y2R, increases neurite outgrowth. Consistent with the accepted role of cofilin in the regulation of neurite extension, results indicate that incubation of IL-1β-treated mPCNs with UTP increases the phosphorylation of cofilin, a response absent in PCNs isolated from P2Y2R(-/-) mice. Other findings indicate that function-blocking anti-αv β3/5 integrin antibodies prevent UTP-induced cofilin activation in IL-1β-treated mPCNs, suggesting that established P2Y2R/αv β3/5 interactions that promote G12 -dependent Rho activation lead to cofilin phosphorylation involved in neurite extension. Cofilin phosphorylation induced by UTP in IL-1β-treated mPCNs is also decreased by inhibitors of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), suggesting a role for P2Y2R-mediated and Gq-dependent calcium mobilization in neurite outgrowth. Taken together, these studies indicate that up-regulation of P2Y2Rs in mPCNs under pro-inflammatory conditions can promote cofilin-dependent neurite outgrowth, a neuroprotective response that may be a novel pharmacological target in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Troy S Peterson
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, Missouri 65211-7310, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Erb L, Weisman GA. Coupling of P2Y receptors to G proteins and other signaling pathways. ACTA ACUST UNITED AC 2012; 1:789-803. [PMID: 25774333 DOI: 10.1002/wmts.62] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
P2Y receptors are G protein-coupled receptors (GPCRs) that are activated by adenine and uridine nucleotides and nucleotide sugars. There are eight subtypes of P2Y receptors (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14), which activate intracellular signaling cascades to regulate a variety of cellular processes, including proliferation, differentiation, phagocytosis, secretion, nociception, cell adhesion, and cell migration. These signaling cascades operate mainly by the sequential activation or deactivation of heterotrimeric and monomeric G proteins, phospholipases, adenylyl and guanylyl cyclases, protein kinases, and phosphodiesterases. In addition, there are numerous ion channels, cell adhesion molecules, and receptor tyrosine kinases that are modulated by P2Y receptors and operate to transmit an extracellular signal to an intracellular response.
Collapse
Affiliation(s)
- Laurie Erb
- Department of Biochemistry, Life Sciences Center, University of Missouri-Columbia, Columbia, MO 65211, USA
| | - Gary A Weisman
- Department of Biochemistry, Life Sciences Center, University of Missouri-Columbia, Columbia, MO 65211, USA
| |
Collapse
|
38
|
Weisman GA, Ajit D, Garrad R, Peterson TS, Woods LT, Thebeau C, Camden JM, Erb L. Neuroprotective roles of the P2Y(2) receptor. Purinergic Signal 2012; 8:559-78. [PMID: 22528682 DOI: 10.1007/s11302-012-9307-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/04/2011] [Indexed: 02/07/2023] Open
Abstract
Purinergic signaling plays a unique role in the brain by integrating neuronal and glial cellular circuits. The metabotropic P1 adenosine receptors and P2Y nucleotide receptors and ionotropic P2X receptors control numerous physiological functions of neuronal and glial cells and have been implicated in a wide variety of neuropathologies. Emerging research suggests that purinergic receptor interactions between cells of the central nervous system (CNS) have relevance in the prevention and attenuation of neurodegenerative diseases resulting from chronic inflammation. CNS responses to chronic inflammation are largely dependent on interactions between different cell types (i.e., neurons and glia) and activation of signaling molecules including P2X and P2Y receptors. Whereas numerous P2 receptors contribute to functions of the CNS, the P2Y(2) receptor is believed to play an important role in neuroprotection under inflammatory conditions. While acute inflammation is necessary for tissue repair due to injury, chronic inflammation contributes to neurodegeneration in Alzheimer's disease and occurs when glial cells undergo prolonged activation resulting in extended release of proinflammatory cytokines and nucleotides. This review describes cell-specific and tissue-integrated functions of P2 receptors in the CNS with an emphasis on P2Y(2) receptor signaling pathways in neurons, glia, and endothelium and their role in neuroprotection.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, University of Missouri, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO 65211-7310, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Weisman GA, Camden JM, Peterson TS, Ajit D, Woods LT, Erb L. P2 receptors for extracellular nucleotides in the central nervous system: role of P2X7 and P2Y₂ receptor interactions in neuroinflammation. Mol Neurobiol 2012; 46:96-113. [PMID: 22467178 DOI: 10.1007/s12035-012-8263-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/21/2012] [Indexed: 12/16/2022]
Abstract
Extracellular nucleotides induce cellular responses in the central nervous system (CNS) through the activation of ionotropic P2X and metabotropic P2Y nucleotide receptors. Activation of these receptors regulates a wide range of physiological and pathological processes. In this review, we present an overview of the current literature regarding P2X and P2Y receptors in the CNS with a focus on the contribution of P2X7 and P2Y(2) receptor-mediated responses to neuroinflammatory and neuroprotective mechanisms.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, University of Missouri, 540E Life Sciences Center, 1201 Rollins Road, Columbia, MO 65211-7310, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Kim HJ, Ajit D, Peterson TS, Wang Y, Camden JM, Gibson Wood W, Sun GY, Erb L, Petris M, Weisman GA. Nucleotides released from Aβ₁₋₄₂ -treated microglial cells increase cell migration and Aβ₁₋₄₂ uptake through P2Y₂ receptor activation. J Neurochem 2012; 121:228-38. [PMID: 22353164 DOI: 10.1111/j.1471-4159.2012.07700.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Amyloid β-protein (Aβ) deposits in brains of Alzheimer's disease patients generate proinflammatory cytokines and chemokines that recruit microglial cells to phagocytose Aβ. Nucleotides released from apoptotic cells activate P2Y(2) receptors (P2Y(2) Rs) in macrophages to promote clearance of dead cells. In this study, we investigated the role of P2Y(2) Rs in the phagocytosis and clearance of Aβ. Treatment of mouse primary microglial cells with fibrillar (fAβ(1-42) ) and oligomeric (oAβ(1-42) ) Aβ(1-42) aggregation solutions caused a rapid release of ATP (maximum after 10 min). Furthermore, fAβ(1-42) and oAβ(1-42) treatment for 24 h caused an increase in P2Y(2) R gene expression. Treatment with fAβ(1-42) and oAβ(1-42) aggregation solutions increased the motility of neighboring microglial cells, a response inhibited by pre-treatment with apyrase, an enzyme that hydrolyzes nucleotides. The P2Y(2) R agonists ATP and UTP caused significant uptake of Aβ(1-42) by microglial cells within 30 min, which reached a maximum within 1 h, but did not increase Aβ(1-42) uptake by primary microglial cells isolated from P2Y(2) R(-/-) mice. Inhibitors of α(v) integrins, Src and Rac decreased UTP-induced Aβ(1-42) uptake, suggesting that these previously identified components of the P2Y(2) R signaling pathway play a role in Aβ phagocytosis by microglial cells. Finally, we found that UTP treatment enhances Aβ(1-42) degradation by microglial cells, but not in cells isolated from P2Y(2) R(-/-) mice. Taken together, our findings suggest that P2Y(2) Rs can activate microglial cells to enhance Aβ clearance and highlight the P2Y(2) R as a therapeutic target in Alzheimer's disease.
Collapse
Affiliation(s)
- Hye Jung Kim
- Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Seye CI, Agca Y, Agca C, Derbigny W. P2Y2 receptor-mediated lymphotoxin-α secretion regulates intercellular cell adhesion molecule-1 expression in vascular smooth muscle cells. J Biol Chem 2012; 287:10535-10543. [PMID: 22298782 DOI: 10.1074/jbc.m111.313189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The proinflammatory cytokine lymphotoxin-α (LTA) is thought to contribute to the pathogenesis of atherosclerosis. However, the mechanisms that regulate its expression in vascular smooth muscle cells (VSMC) are poorly understood. The ability of exogenous nucleotides to stimulate LTA production was evaluated in VSMC by ELISA. The P2Y(2) nucleotide receptor (P2Y(2)R) agonist UTP stimulates a strong and sustained release of LTA from WT but not P2Y(2)R(-/-) SMC. Assessment of LTA gene transcription by LTA promoter-luciferase construct indicated that LTA levels are controlled at the level of transcription. We show using RNAi techniques that knockdown of the actin-binding protein filamin-A (FLNa) severely impaired nucleotide-induced Rho activation and consequent Rho-mediated LTA secretion. Reintroduction of FLNa in FLNa RNAi SMC rescued UTP-induced LTA expression. In addition, we found that UTP-stimulated LTA secretion is not sensitive to brefeldin A, which blocks the formation of vesicles involved in protein transport from the endoplasmic reticulum to the Golgi apparatus, suggesting that P2Y(2)R/filamin-mediated secretion of LTA is independent of the endoplasmic reticulum/Golgi secretory vesicle route. Furthermore, UTP selectively induces ICAM-1 expression in WT but not SMC expressing a truncated P2Y(2)R deficient in LTA secretion. These data suggest that P2Y(2)R recruits FLNa to provide a cytoskeletal scaffold necessary for Rho signaling pathway upstream of LTA release and subsequent stimulation of ICAM-1 expression on vascular smooth muscle cells.
Collapse
Affiliation(s)
- Cheikh I Seye
- Departments of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202.
| | - Yuksel Agca
- Department of Veterinary Pathobiology, University of Missouri-Columbia, Columbia, Missouri 65201
| | - Cansu Agca
- Department of Veterinary Pathobiology, University of Missouri-Columbia, Columbia, Missouri 65201
| | - Wilbert Derbigny
- Departments of Immunology and Microbiology, Indiana University School of Medicine, Indianapolis, Indiana 46202 and
| |
Collapse
|
42
|
Ding L, Ma W, Littmann T, Camp R, Shen J. The P2Y(2) nucleotide receptor mediates tissue factor expression in human coronary artery endothelial cells. J Biol Chem 2011; 286:27027-38. [PMID: 21652710 DOI: 10.1074/jbc.m111.235176] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The discovery of the role of P2Y(12) receptor in platelet aggregation leads to a new anti-thrombotic drug Plavix; however, little is known about non-platelet P2Y receptors in thrombosis. This study tested the hypothesis that endothelial P2Y receptor(s) mediates up-regulation of tissue factor (TF), the initiator of coagulation cascade. Stimulation of human coronary artery endothelial cells (HCAEC) by UTP/ATP increased the mRNA level of TF but not of its counterpart-tissue factor pathway inhibitor, which was accompanied by up-regulation of TF protein and cell surface activity. RT-PCR revealed a selective expression of P2Y(2) and P2Y(11) receptors in HCAEC. Consistent with this, TF up-regulation was inhibited by suramin or by siRNA silencing of P2Y(2) receptor, but not by NF-157, a P2Y(11)-selective antagonist, suggesting a role for the P2Y(2) receptor. In addition, P2Y(2) receptor activated ERK1/2, JNK, and p38 MAPK pathways without affecting the positive NF-κB and negative AKT regulatory pathways of TF expression. Furthermore, TF up-regulation was abolished or partially suppressed by inhibition of p38 or JNK but not ERK1/2. Interestingly, blockade of the PLC/Ca(2+) pathway did not affect P2Y(2) receptor activation of p38, JNK, and TF induction. However, blockade of Src kinase reduced phosphorylation of p38 but not JNK, eliminating TF induction. In contrast, inhibition of Rho kinase reduced phosphorylation of JNK but not p38, decreasing TF expression. These findings demonstrate that P2Y(2) receptor mediates TF expression in HCAEC through new mechanisms involving Src/p38 and Rho/JNK pathways, possibly contributing to a pro-thrombotic status after vascular injury.
Collapse
Affiliation(s)
- Ling Ding
- Division of Pharmacology, Department of Pharmacal Sciences, Harrison School of Pharmacy, Auburn University, Auburn, Alabama 36849, USA
| | | | | | | | | |
Collapse
|
43
|
Zerr M, Hechler B, Freund M, Magnenat S, Lanois I, Cazenave JP, Léon C, Gachet C. Major contribution of the P2Y₁receptor in purinergic regulation of TNFα-induced vascular inflammation. Circulation 2011; 123:2404-13. [PMID: 21576651 DOI: 10.1161/circulationaha.110.002139] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Atherosclerosis is an inflammatory disease, and extracellular nucleotides are one of the factors possibly involved in vascular inflammation. The P2Y(1) receptor for adenosine 5'-diphosphate has been shown to be involved in the development of atherosclerosis in apolipoprotein E--deficient mice. Our aim is to determine whether the endothelial P2Y(1) receptor plays a role in leukocyte recruitment during vascular inflammation and characterize underlying mechanisms. METHODS AND RESULTS We show here that the P2Y(1) receptor plays a role in leukocyte recruitment in inflamed mouse femoral arteries. Moreover, in wild-type bone marrow--transplanted chimeric P2Y(1)-deficient mice with an apolipoprotein E--deficient background, a strong reduction of adhesion molecule--dependent leukocyte recruitment was observed after local injection of tumor necrosis factor and interleukin 1β, excluding a role for the platelet or other hematopoietic cell type P2Y(1) in these events. Similarly, the in vitro adhesion of isolated mouse monocytes to tumor necrosis factor α--stimulated murine endothelial cell monolayers and their migration across the cell layers were strongly reduced in P2Y(1)-deficient compared with wild-type endothelial cells, as was the expression of the adhesion molecules P-selectin, Vascular cell adhesion molecule 1, and intercellular adhesion molecule 1. Pharmacological inhibition using the selective antagonist MRS2500 also resulted in decreased expression of adhesion molecules. These events are related to the p38 mitogen-activated protein kinase and activating transcription factor 2 pathway. Finally, in vivo administration of MRS2500 resulted in strong reduction of leukocyte recruitment in inflamed femoral arteries of apolipoprotein E--deficient mice. CONCLUSIONS The data highlight a key role of the endothelial P2Y(1) receptor in acute vascular inflammation. Pharmacological targeting the P2Y(1) receptor could represent a promising approach for the treatment of vascular inflammation.
Collapse
Affiliation(s)
- Murielle Zerr
- UMR_S949 INSERM, Université de Strasbourg, Etablissement Français du Sang-Alsace, Strasbourg, France
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Matsumoto T, Tostes RC, Webb RC. Uridine adenosine tetraphosphate-induced contraction is increased in renal but not pulmonary arteries from DOCA-salt hypertensive rats. Am J Physiol Heart Circ Physiol 2011; 301:H409-17. [PMID: 21551273 DOI: 10.1152/ajpheart.00084.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Uridine adenosine tetraphosphate (Up(4)A) was reported as a novel endothelium-derived contracting factor. Up(4)A contains both purine and pyrimidine moieties, which activate purinergic (P2)X and P2Y receptors. However, alterations in the vasoconstrictor responses to Up(4)A in hypertensive states remain unclear. The present study examined the effects of Up(4)A on contraction of isolated renal arteries (RA) and pulmonary arteries (PA) from DOCA-salt rats using isometric tension recording. RA from DOCA-salt rats exhibited increased contraction to Up(4)A versus arteries from control uninephrectomized rats in the absence and presence of N(G)-nitro-l-arginine (nitric oxide synthase inhibitor). On the other hand, the Up(4)A-induced contraction in PA was similar between the two groups. Up(4)A-induced contraction was inhibited by suramin (nonselective P2 antagonist) but not by diinosine pentaphosphate pentasodium salt hydrate (Ip(5)I; P2X(1) antagonist) in RA from both groups. Furthermore, 2-thiouridine 5'-triphosphate tetrasodium salt (2-ThioUTP; P2Y(2) agonist)-, uridine-5'-(γ-thio)-triphosphate trisodium salt (UTPγS; P2Y(2)/P2Y(4) agonist)-, and 5-iodouridine-5'-O-diphosphate trisodium salt (MRS 2693; P2Y(6) agonist)-induced contractions were all increased in RA from DOCA-salt rats. Protein expression of P2Y(2)-, P2Y(4)-, and P2Y(6) receptors in RA was similar between the two groups. In DOCA-salt RA, the enhanced Up(4)A-induced contraction was reduced by PD98059, an ERK pathway inhibitor, and Up(4)A-stimulated ERK activation was increased. These data are the first to indicate that Up(4)A-induced contraction is enhanced in RA from DOCA-salt rats. Enhanced P2Y receptor signaling and activation of the ERK pathway together represent a likely mechanism mediating the enhanced Up(4)A-induced contraction. Up(4)A might be of relevance in the pathophysiology of vascular tone regulation and renal dysfunction in arterial hypertension.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Dept. of Physiology, Georgia Health Sciences Univ., 1120 15th St., Rm. CA-3147, Augusta, GA 30912, USA.
| | | | | |
Collapse
|
45
|
Heterologous down-regulation of angiotensin type 1 receptors by purinergic P2Y2 receptor stimulation through S-nitrosylation of NF-kappaB. Proc Natl Acad Sci U S A 2011; 108:6662-7. [PMID: 21464294 DOI: 10.1073/pnas.1017640108] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cross-talk between G protein-coupled receptor (GPCR) signaling pathways serves to fine tune cellular responsiveness by neurohumoral factors. Accumulating evidence has implicated nitric oxide (NO)-based signaling downstream of GPCRs, but the molecular details are unknown. Here, we show that adenosine triphosphate (ATP) decreases angiotensin type 1 receptor (AT(1)R) density through NO-mediated S-nitrosylation of nuclear factor κB (NF-κB) in rat cardiac fibroblasts. Stimulation of purinergic P2Y(2) receptor by ATP increased expression of inducible NO synthase (iNOS) through activation of nuclear factor of activated T cells, NFATc1 and NFATc3. The ATP-induced iNOS interacted with p65 subunit of NF-κB in the cytosol through flavin-binding domain, which was indispensable for the locally generated NO-mediated S-nitrosylation of p65 at Cys38. β-Arrestins anchored the formation of p65/IκBα/β-arrestins/iNOS quaternary complex. The S-nitrosylated p65 resulted in decreases in NF-κB transcriptional activity and AT(1)R density. In pressure-overloaded mouse hearts, ATP released from cardiomyocytes led to decrease in AT(1)R density through iNOS-mediated S-nitrosylation of p65. These results show a unique regulatory mechanism of heterologous regulation of GPCRs in which cysteine modification of transcriptional factor rather than protein phosphorylation plays essential roles.
Collapse
|
46
|
Abstract
Airway epithelia are continuously damaged by airborne pollutants, pathogens and allergens, and they rely on intrinsic mechanisms to restore barrier integrity. Epithelial repair is a multi-step process including cell migration into the wounded area, proliferation, differentiation and matrix deposition. Each step requires the secretion of various molecules, including growth factors, integrins and matrix metalloproteinases. Evidence is emerging that purinergic signaling promotes repair in human airway epithelia. An injury induces ATP release, which binds P2Y(2) receptors (P2Y(2)Rs) to initiate protein kinase C (PKC)-dependent oxidative activation of TNFα-converting enzyme (TACE), which then releases the membrane-bound ligands of the epidermal growth factor receptor (EGFR). The P2Y(2)R- and EGFR-dependent signaling cascades converge to induce mediator release, whereas the latter also induces cytoskeletal rearrangement for cell migration and proliferation. Similar roles for purinergic signaling are reported in pulmonary endothelial cells, smooth muscle cells and fibroblasts. In chronic airway diseases, the aberrant regulation of extracellular purines is implicated in the development of airway remodeling by mucus cell metaplasia and hypersecretion, excess collagen deposition, fibrosis and neovascularization. This chapter describes the crosstalk between these signaling cascades and discusses the impact of deregulated purinergic signaling in chronic lung diseases.
Collapse
|
47
|
Abstract
The purine- and pyrimidine-sensitive P2Y receptors belong to the large group of G-protein-coupled receptors that are the target of approximately one-third of the pharmaceutical drugs used in the clinic today. It is therefore not unexpected that the P2Y receptors could be useful targets for drug development. This chapter will discuss P2Y receptor-based therapies currently used, in development and possible future developments. The platelet inhibitors blocking the ADP-receptor P2Y(12) reduce myocardial infarction, stroke, and mortality in patients with cardiovascular disease. Clopidogrel (Plavix) was for many years the second most selling drug in the world. The improved P2Y(12) inhibitors prasugrel, ticagrelor, and elinogrel are now entering the clinic with even more pronounced protective effects. The UTP-activated P2Y(2) receptor stimulates ciliary movement and secretion from epithelial cells. Cystic fibrosis is a monogenetic disease where reduced chloride ion secretion results in a severe lung disease and early death. No specific treatment has been available, but the P2Y(2) agonist Denufosol has been shown to improve lung function and is expected to be introduced as treatment for cystic fibrosis soon. In preclinical studies, there are indications that P2Y receptors can be important for diabetes, osteoporosis, cardiovascular, and atherosclerotic disease. In conclusion, P2Y receptors are important for the health of humans for many diseases, and we can expect even more beneficial drugs targeting P2Y receptors in the future.
Collapse
Affiliation(s)
- David Erlinge
- Department of Cardiology, Lund University, Skane University Hospital, Sweden
| |
Collapse
|
48
|
Peterson TS, Camden JM, Wang Y, Seye CI, Wood WG, Sun GY, Erb L, Petris MJ, Weisman GA. P2Y2 nucleotide receptor-mediated responses in brain cells. Mol Neurobiol 2010; 41:356-66. [PMID: 20387013 PMCID: PMC3086510 DOI: 10.1007/s12035-010-8115-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 03/01/2010] [Indexed: 01/14/2023]
Abstract
Acute inflammation is important for tissue repair; however, chronic inflammation contributes to neurodegeneration in Alzheimer's disease (AD) and occurs when glial cells undergo prolonged activation. In the brain, stress or damage causes the release of nucleotides and activation of the G(q) protein-coupled P2Y(2) nucleotide receptor subtype (P2Y(2)R) leading to pro-inflammatory responses that can protect neurons from injury, including the stimulation and recruitment of glial cells. P2Y(2)R activation induces the phosphorylation of the epidermal growth factor receptor (EGFR), a response dependent upon the presence of a SH3 binding domain in the intracellular C terminus of the P2Y(2)R that promotes Src binding and transactivation of EGFR, a pathway that regulates the proliferation of cortical astrocytes. Other studies indicate that P2Y(2)R activation increases astrocyte migration. P2Y(2)R activation by UTP increases the expression in astrocytes of alpha(V)beta(3/5) integrins that bind directly to the P2Y(2)R via an Arg-Gly-Asp (RGD) motif in the first extracellular loop of the P2Y(2)R, an interaction required for G(o) and G(12) protein-dependent astrocyte migration. In rat primary cortical neurons (rPCNs) P2Y(2)R expression is increased by stimulation with interleukin-1beta (IL-1beta), a pro-inflammatory cytokine whose levels are elevated in AD, in part due to nucleotide-stimulated release from glial cells. Other results indicate that oligomeric beta-amyloid peptide (Abeta(1-42)), a contributor to AD, increases nucleotide release from astrocytes, which would serve to activate upregulated P2Y(2)Rs in neurons. Data with rPCNs suggest that P2Y(2)R upregulation by IL-1beta and subsequent activation by UTP are neuroprotective, since this increases the non-amyloidogenic cleavage of amyloid precursor protein. Furthermore, activation of IL-1beta-upregulated P2Y(2)Rs in rPCNs increases the phosphorylation of cofilin, a cytoskeletal protein that stabilizes neurite outgrowths. Thus, activation of pro-inflammatory P2Y(2)Rs in glial cells can promote neuroprotective responses, suggesting that P2Y(2)Rs represent a novel pharmacological target in neurodegenerative and other pro-inflammatory diseases.
Collapse
Affiliation(s)
- Troy S Peterson
- Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Guns PJDF, Hendrickx J, Van Assche T, Fransen P, Bult H. P2Y receptors and atherosclerosis in apolipoprotein E-deficient mice. Br J Pharmacol 2009; 159:326-36. [PMID: 20050854 DOI: 10.1111/j.1476-5381.2009.00497.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE P2Y nucleotide receptors are involved in the regulation of vascular tone, smooth muscle cell (SMC) proliferation and inflammatory responses. The present study investigated whether they are involved in atherosclerosis. EXPERIMENTAL APPROACH mRNA of P2Y receptors was quantified (RT-PCR) in atherosclerotic and plaque-free aorta segments of apolipoprotein E-deficient (apoE(-/-)) mice. Macrophage activation was assessed in J774 macrophages, and effects of non-selective purinoceptor antagonists on atherosclerosis were evaluated in cholesterol-fed apoE(-/-) mice. KEY RESULTS P2Y(6) receptor mRNA was consistently elevated in segments with atherosclerosis, whereas P2Y(2) receptor expression remained unchanged. Expression of P2Y(1) or P2Y(4) receptor mRNA was low or undetectable, and not influenced by atherosclerosis. P2Y(6) mRNA expression was higher in cultured J774 macrophages than in cultured aortic SMCs. Furthermore, immunohistochemical staining of plaques demonstrated P2Y(6)-positive macrophages, but few SMCs, suggesting that macrophage recruitment accounted for the increase in P2Y(6) receptor mRNA during atherosclerosis. In contrast to ATP, the P2Y(6)-selective agonist UDP increased mRNA expression and activity of inducible nitric oxide synthase and interleukin-6 in J774 macrophages; this effect was blocked by suramin (100-300 microM) or pyridoxal-phosphate-6-azophenyl-2'-4'-disulphonic acid (PPADS, 10-30 microM). Finally, 4-week treatment of cholesterol-fed apoE(-/-) mice with suramin or PPADS (50 and 25 mg.kg(-1).day(-1) respectively) reduced plaque size, without changing plaque composition (relative SMC and macrophage content) or cell replication. CONCLUSIONS AND IMPLICATIONS These results suggest involvement of nucleotide receptors, particularly P2Y(6) receptors, during atherosclerosis, and warrant further research with selective purinoceptor antagonists or P2Y(6) receptor-deficient mice.
Collapse
|
50
|
Langlois C, Gendron FP. Promoting MPhi transepithelial migration by stimulating the epithelial cell P2Y(2) receptor. Eur J Immunol 2009; 39:2895-905. [PMID: 19634190 DOI: 10.1002/eji.200939369] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In intestine, neutrophils are recruited in response to bacterial infiltration and their anti-cellular activities contribute to inflammatory bowel diseases. In contrast, little is known regarding the recruitment of MPhi to the intestinal epithelium. Extracellular adenosine and uridine 5'-triphosphate (ATP and UTP) can function as leukocyte chemoattractants. We investigated the effects of these nucleotides on the ability of intestinal epithelial cells (IEC) to promote MPhi transepithelial migration and adhesion. ATP and UTP promoted the migration of neutrophil-like PLB-985 cells and MPhi across a Caco-2 monolayer. The MPhi-like U-937 cells adhered to nucleotide-stimulated IEC monolayers. In mice with intestinal inflammation, there were infiltrating CD68(+) MPhi in the colonic epithelium and CD68(+) MPhi present at the apical surface of colonocytes. We determined that ATP and UTP activated the P2Y(2) receptor P (P2Y(2)R) to increase ICAM-1 expression, which mediated the adhesion of MPhi to the apical surface of IEC. Intriguingly, stimulation of IEC with nucleotides did not increase the adhesion of neutrophils. However, in the presence of adherent MPhi, there was adhesion of neutrophils, suggesting that MPhi may serve as anchors for neutrophil adhesion. These studies provide insight into the inflammatory mechanisms that contribute to inflammatory bowel diseases and identify potential therapeutic targets for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Christine Langlois
- Canadian Institutes of Health Research Team on the Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, J1H5N4, Canada
| | | |
Collapse
|