1
|
Brinkley L, Brock MA, Stinson G, Bilgili A, Jacobs JP, Bleiweis M, Peek GJ. The biological role and future therapeutic uses of nitric oxide in extracorporeal membrane oxygenation, a narrative review. Perfusion 2025; 40:83-91. [PMID: 38226651 DOI: 10.1177/02676591241228169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
BACKGROUND Nitric oxide (NO) is a gas naturally produced by the human body that plays an important physiological role. Specifically, it binds guanylyl cyclase to induce smooth muscle relaxation. NO's other protective functions have been well documented, particularly its protective endothelial functions, effects on decreasing pulmonary vascular resistance, antiplatelet, and anticoagulation properties. The use of nitric oxide donors as vasodilators has been known since 1876. Inhaled nitric oxide has been used as a pulmonary vasodilator and to improve ventilation perfusion matching since the 1990s. It is currently approved by the United States Food and Drug Administration for neonates with hypoxic respiratory failure, however, it is used off-label for acute respiratory distress syndrome, acute bronchiolitis, and COVID-19. PURPOSE In this article we review the currently understood biological action and therapeutic uses of NO through nitric oxide donors such as inhaled nitric oxide. We will then explore recent studies describing use of NO in cardiopulmonary bypass and extracorporeal membrane oxygenation and speculate on NO's future uses.
Collapse
|
2
|
Fu X, Lu H, Gao M, Li P, He Y, He Y, Luo X, Rao X, Liu W. Nitric oxide in the cardio-cerebrovascular system: Source, regulation and application. Nitric Oxide 2024; 152:48-57. [PMID: 39299647 DOI: 10.1016/j.niox.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Nitric oxide (NO) plays a crucial role as a messenger or effector in the body, yet it presents a dual impact on cardio-cerebrovascular health. Under normal physiological conditions, NO exhibits vasodilatory effects, regulates blood pressure, inhibits platelet aggregation, and offers neuroprotective actions. However, in pathological situations, excessive NO production contributes to or worsens inflammation within the body. Moreover, NO may combine with reactive oxygen species (ROS), generating harmful substances that intensify physical harm. This paper succinctly reviews pertinent literature to clarify the in vivo and in vitro origins of NO, its regulatory function in the cardio-cerebrovascular system, and the advantages and disadvantages associated with NO donor drugs, NO delivery systems, and vascular stent materials for treating cardio-cerebrovascular disease. The findings provide a theoretical foundation for the application of NO in cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Xiaoming Fu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Haowei Lu
- Department of Pharmacy, The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Meng Gao
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Pinghe Li
- Lanzhou Foci Pharmaceutical Co., Ltd, Lanzhou, 730030, China
| | - Yan He
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Yu He
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China
| | - Xiaojian Luo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Xiaoyong Rao
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Wei Liu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
3
|
Brock MA, Ebraheem M, Jaudon A, Narasimhulu SS, Vazquez-Colon Z, Philip J, Lopez-Colon D, Jacobs JP, Bleiweis MS, Peek GJ. The safe addition of nitric oxide to the sweep gas of the extracorporeal membrane oxygenation circuit in a pediatric cardiac intensive care unit. Perfusion 2024:2676591241246079. [PMID: 38581646 DOI: 10.1177/02676591241246079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2024]
Abstract
Background: Nitric Oxide (NO) is a naturally occurring modulator of inflammation found in the human body. Several studies in the pediatric cardiothoracic surgery literature have demonstrated some beneficial clinical effects when NO is added to the sweep gas of the cardiopulmonary bypass circuit.Purpose: Our primary aim was to determine the safety of incorporating nitric oxide into the oxygenator sweep gas of the extracorporeal membrane oxygenation (ECMO) circuit. Secondarily, we looked at important clinical outcomes, such as survival, blood product utilization, and common complications related to ECMO.Methods: We performed a single center, retrospective review of all patients at our institution who received ECMO between January 1, 2017 and March 31, 2023. We began additing NO to the ECMO sweep gas in 2019. Results: There were no instances of clinically significant methemoglobinemia with the addition of NO to the sweep gas (0% vs 0%, p = 1). The median daily methemoglobin level was higher in those who received NO via the sweep gas when compared to those who did not (1.6 vs 1.1, p = <0.001). Conclusions: The addition of NO to the sweep gas of the ECMO circuit is safe.
Collapse
Affiliation(s)
- Michael A Brock
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Mohammed Ebraheem
- Department of Pediatrics, Division of Cardiology, Stanford University, Palo Alto, CA, USA
| | - Andrew Jaudon
- Department of Respiratory Care, ECMO coordinator, UF Health Shands Teaching Hospital, Gainvesville, FL, USA
| | | | - Zasha Vazquez-Colon
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Joseph Philip
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Dalia Lopez-Colon
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Jeffrey P Jacobs
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
- Department of Surgery, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Mark S Bleiweis
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
- Department of Surgery, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| | - Giles J Peek
- Department of Pediatrics, Congenital Heart Center, University of Florida, Gainesville, FL, USA
- Department of Surgery, Congenital Heart Center, University of Florida, Gainesville, FL, USA
| |
Collapse
|
4
|
Perez Lloveras E, Michelangelo JM, Videla CG, Gonzalez ML, Privitera V, Serra MM, Vazquez C. Severe thrombocytopenia associated to bevacizumab in a patient with scleroderma, gastrointestinal angiodysplasias and refractory gastrointestinal bleeding. Blood Coagul Fibrinolysis 2024; 35:141-146. [PMID: 38358904 DOI: 10.1097/mbc.0000000000001284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
This case report discusses the medical history of a 64-year-old woman diagnosed with scleroderma and diffuse gastrointestinal angiodysplasia. The patient received bevacizumab (BVZ) therapy to address gastrointestinal bleeding that was unresponsive to endoscopic treatment. Subsequently, she developed severe thrombocytopenia. Although there were suspicions of an immune-mediated mechanism resulting from BVZ treatment, the laboratory results did not provide conclusive evidence. The patient underwent transfusions, received gamma globulin, and was treated with Romiplostim. Over time, her platelet levels gradually improved, and the bleeding was successfully controlled. It's worth noting that BVZ-induced thrombocytopenia is a relatively rare yet severe adverse effect. Recognizing and understanding the mechanisms behind thrombocytopenia is essential for developing safer treatment approaches. Further research is required to identify potential risk factors associated with this condition.
Collapse
Affiliation(s)
| | | | | | - Maria Laura Gonzalez
- Gastroenterology Department
- Hereditary Hemorrhagic Unit, Hospital Italiano de Buenos Aires, Argentina
- Argentine Rendu Group (ARG)
| | - Veronica Privitera
- Hematology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Marcelo Martin Serra
- Internal Medicine Department
- Hereditary Hemorrhagic Unit, Hospital Italiano de Buenos Aires, Argentina
- Argentine Rendu Group (ARG)
| | | |
Collapse
|
5
|
Provenzale I, Solari FA, Schönichen C, Brouns SLN, Fernández DI, Kuijpers MJE, van der Meijden PEJ, Gibbins JM, Sickmann A, Jones C, Heemskerk JWM. Endothelium-mediated regulation of platelet activation: Involvement of multiple protein kinases. FASEB J 2024; 38:e23468. [PMID: 38334433 DOI: 10.1096/fj.202300360rr] [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: 02/25/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024]
Abstract
The endothelial regulation of platelet activity is incompletely understood. Here we describe novel approaches to find molecular pathways implicated on the platelet-endothelium interaction. Using high-shear whole-blood microfluidics, employing coagulant or non-coagulant conditions at physiological temperature, we observed that the presence of human umbilical vein endothelial cells (HUVEC) strongly suppressed platelet adhesion and activation, via the collagen receptor glycoprotein VI (GPVI) and the PAR receptors for thrombin. Real-time monitoring of the cytosolic Ca2+ rises in the platelets indicated no major improvement of inhibition by prostacyclin or nitric oxide. Similarly under stasis, exposure of isolated platelets to HUVEC reduced the Ca2+ responses by collagen-related peptide (CRP-XL, GPVI agonist) and thrombin (PAR agonist). We then analyzed the label-free phosphoproteome of platelets (three donors), exposed to HUVEC, CRP-XL, and/or thrombin. High-resolution mass spectrometry gave 5463 phosphopeptides, corresponding to 1472 proteins, with good correlation between biological and technical replicates (R > .86). Stringent filtering steps revealed 26 regulatory pathways (Reactome) and 143 regulated kinase substrates (PhosphoSitePlus), giving a set of protein phosphorylation sites that was differentially (44) or similarly (110) regulated by HUVEC or agonist exposure. The differential regulation was confirmed by stable-isotope analysis of platelets from two additional donors. Substrate analysis indicated major roles of poorly studied protein kinase classes (MAPK, CDK, DYRK, STK, PKC members). Collectively, these results reveal a resetting of the protein phosphorylation profile in platelets exposed to endothelium or to conventional agonists and to endothelium-promoted activity of a multi-kinase network, beyond classical prostacyclin and nitric oxide actors, that may contribute to platelet inhibition.
Collapse
Affiliation(s)
- Isabella Provenzale
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Fiorella A Solari
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Claudia Schönichen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Center for Thrombosis and Haemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sanne L N Brouns
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Delia I Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
- Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Chris Jones
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Synapse Research Institute Maastricht, Maastricht, The Netherlands
| |
Collapse
|
6
|
Stafuzza NB, Freitas ACD, Mioto MB, Silva RMDO, Fragomeni BDO, Pedrosa VB, Costa RLDD, Paz CCPD. Weighted single-step genome-wide association study and functional enrichment analyses for gastrointestinal nematode resistance traits in Santa Ines sheep. Vet Parasitol 2023; 323:110047. [PMID: 37857178 DOI: 10.1016/j.vetpar.2023.110047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
This study aimed to identify genomic regions, pathways, and putative candidate genes associated with resistance to gastrointestinal nematode in Santa Ines sheep. The phenotypic information comprised 5529 records from 1703 naturally infected animals. After genomic data quality control, 37,511 SNPs from 589 animals were available. The weighted single-step approach for genome-wide association study was performed to estimate the SNP effects and variances accounted by 10-SNP sliding windows. Confirming the polygenic nature of the studied traits, 20, 22, 21, and 19 genomic windows that explained more than 0.5% of the additive genetic variance were identified for fecal egg counts (FEC), Famacha© (FAM), packed cell volume (PCV), and total plasma protein (TPP), respectively. A total of 81, 122, 106, and 101 protein-coding genes were found in windows associated with FEC, FAM, PCV, and TPP, respectively. Several protein-coding genes related to the immune system and inflammatory response functions were identified within those genomic regions, such as ADCY9, ADRB2, BRAF, CADM1, CCL20, CD70, CREBBP, FNBP1, HTR4, IL16, IL22, IL26, MAPK8, NDFIP1, NLRC3, PAK5, PLCB1, PLCB4, ROCK1, TEK, TNFRSF12A, and VAV1. Functional enrichment analysis by DAVID tool also revealed many significant (P < 0.05) pathways and Gene Ontology terms that could be related to resistance to gastrointestinal nematode in Santa Ines sheep, such as chemokine signaling pathway (oas04062), cAMP signaling pathway (oas04024), cGMP-PKG signaling pathway (Oas04022), platelet activation (Oas04611), Rap1 signaling pathway (oas04015), and oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen (GO:0016705). These results contribute to improving the knowledge of the genetic architecture of resistance to gastrointestinal nematode in Santa Ines sheep.
Collapse
Affiliation(s)
- Nedenia Bonvino Stafuzza
- Sustainable Livestock Research Center, Animal Science Institute, 15130-000 São José do Rio Preto, SP, Brazil.
| | - Ana Claudia de Freitas
- São Paulo Agency of Agribusiness and Technology, Animal Science Institute, 13380-011 Nova Odessa, SP, Brazil; Agricultural Research Agency of the State of Minas Gerais, 38709-899 Patos de Minas, MG, Brazil
| | - Marina B Mioto
- Sustainable Livestock Research Center, Animal Science Institute, 15130-000 São José do Rio Preto, SP, Brazil
| | | | | | - Victor Breno Pedrosa
- Department of Animal Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Ricardo Lopes Dias da Costa
- São Paulo Agency of Agribusiness and Technology, Animal Science Institute, 13380-011 Nova Odessa, SP, Brazil
| | | |
Collapse
|
7
|
Bykov VV, Bykova AV, Motov VS, Larchenko VV, Chernysheva GA, Smol'yakova VI, Aliev OI, Khazanov VA, Vengerovskii AI, Udut VV. Pharmacological Effects of a New Soluble Guanylate Cyclase Stimulator in Experimental Ischemic Stroke. Bull Exp Biol Med 2023; 175:749-752. [PMID: 37978152 DOI: 10.1007/s10517-023-05938-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 11/19/2023]
Abstract
We studied the action of a new indolinone derivative GRS, acetylsalicylic acid (ASA), and their combination on platelet aggregation, vasodilatory endothelial function, neurological status, and cerebral infarction area in experimental focal cerebral ischemia/reperfusion in rats. GRS compound (10 mg/kg), ASA (10 mg/kg), and their combination in the same doses were administered orally once a day as a suspension in 1% starch solution over 5 days after pathology modeling. Sham-operated and control animals were administered 1% starch solution. On day 5 after pathology modeling, platelet aggregation and brain damage area were studied in a half of rats in each group, and the vasodilatory function of the endothelium was studied in the other half. Neurological deficit was assessed 4 h and 1, 3, and 5 days after pathology modeling. GRS compound and ASA equally effectively prevent platelet aggregation and the development of neurological deficit in rats. GRS compound restores the vasodilatory effects of the endothelium, but only ASA contributes to reduction of the cerebral infarction area. In case of combined administration, GRS and ASA do not exhibit synergy in their antiaggregant effect.
Collapse
Affiliation(s)
- V V Bykov
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia.
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - A V Bykova
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
| | - V S Motov
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
| | - V V Larchenko
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - G A Chernysheva
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - V I Smol'yakova
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - O I Aliev
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - V A Khazanov
- IPHAR (Innovative Pharmacology Research, LLC), Tomsk, Russia
| | - A I Vengerovskii
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - V V Udut
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
8
|
Gawaz M, Geisler T, Borst O. Current concepts and novel targets for antiplatelet therapy. Nat Rev Cardiol 2023; 20:583-599. [PMID: 37016032 DOI: 10.1038/s41569-023-00854-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
Platelets have a crucial role in haemostasis and atherothrombosis. Pharmacological control of platelet hyper-reactivity has become a cornerstone in the prevention of thrombo-ischaemic complications in atherosclerotic diseases. Current antiplatelet therapies substantially improve clinical outcomes in patients with coronary artery disease, but at the cost of increased risk of bleeding. Beyond their role in thrombosis, platelets are known to regulate inflammatory (thrombo-inflammatory) and microcirculatory pathways. Therefore, controlling platelet hyper-reactivity might have implications for both tissue inflammation (myocardial ischaemia) and vascular inflammation (vulnerable plaque formation) to prevent atherosclerosis. In this Review, we summarize the pathophysiological role of platelets in acute myocardial ischaemia, vascular inflammation and atherosclerotic progression. Furthermore, we highlight current clinical concepts of antiplatelet therapy that have contributed to improving patient care and have facilitated more individualized therapy. Finally, we discuss novel therapeutic targets and compounds for antiplatelet therapy that are currently in preclinical development, some of which have a more favourable safety profile than currently approved drugs with regard to bleeding risk. These novel antiplatelet targets might offer new strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Yalameha B, Reza Nejabati H. Urinary Exosomal Metabolites: Overlooked Clue for Predicting Cardiovascular Risk. Clin Chim Acta 2023:117445. [PMID: 37315726 DOI: 10.1016/j.cca.2023.117445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
Over the last decade, increasing research has focused on urinary exosomes (UEs) in biological fluids and their relationship with physiological and pathological processes. UEs are membranous vesicles with a size of 40-100 nm, containing a number of bioactive molecules such as proteins, lipids, mRNAs, and miRNAs. These vesicles are an inexpensive non-invasive source that can be used in clinical settings to differentiate healthy patients from diseased patients, thereby serving as potential biomarkers for the early identification of disease. Recent studies have reported the isolation of small molecules called exosomal metabolites from individuals' urine with different diseases. These metabolites could utilize for a variety of purposes, such as the discovery of biomarkers, investigation of mechanisms related to disease development, and importantly prediction of cardiovascular diseases (CVDs) risk factors, including thrombosis, inflammation, oxidative stress, hyperlipidemia as well as homocysteine. It has been indicated that alteration in urinary metabolites of N1-methylnicotinamide, 4-aminohippuric acid, and citric acid can be valuable in predicting cardiovascular risk factors, providing a novel approach to evaluating the pathological status of CVDs. Since the UEs metabolome has been clearly and precisely so far unexplored in CVDs, the present study has specifically addressed the role of the mentioned metabolites in the prediction of CVDs risk factors.
Collapse
Affiliation(s)
- Banafsheh Yalameha
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Prüschenk S, Majer M, Schlossmann J. Novel Functional Features of cGMP Substrate Proteins IRAG1 and IRAG2. Int J Mol Sci 2023; 24:9837. [PMID: 37372987 DOI: 10.3390/ijms24129837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The inositol triphosphate-associated proteins IRAG1 and IRAG2 are cGMP kinase substrate proteins that regulate intracellular Ca2+. Previously, IRAG1 was discovered as a 125 kDa membrane protein at the endoplasmic reticulum, which is associated with the intracellular Ca2+ channel IP3R-I and the PKGIβ and inhibits IP3R-I upon PKGIβ-mediated phosphorylation. IRAG2 is a 75 kDa membrane protein homolog of IRAG1 and was recently also determined as a PKGI substrate. Several (patho-)physiological functions of IRAG1 and IRAG2 were meanwhile elucidated in a variety of human and murine tissues, e.g., of IRAG1 in various smooth muscles, heart, platelets, and other blood cells, of IRAG2 in the pancreas, heart, platelets, and taste cells. Hence, lack of IRAG1 or IRAG2 leads to diverse phenotypes in these organs, e.g., smooth muscle and platelet disorders or secretory deficiency, respectively. This review aims to highlight the recent research regarding these two regulatory proteins to envision their molecular and (patho-)physiological tasks and to unravel their functional interplay as possible (patho-)physiological counterparts.
Collapse
Affiliation(s)
- Sally Prüschenk
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Michael Majer
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| | - Jens Schlossmann
- Department of Pharmacology and Toxicology, Institute of Pharmacy, University of Regensburg, 93040 Regensburg, Germany
| |
Collapse
|
11
|
Zhang P, Solari FA, Heemskerk JWM, Kuijpers MJE, Sickmann A, Walter U, Jurk K. Differential Regulation of GPVI-Induced Btk and Syk Activation by PKC, PKA and PP2A in Human Platelets. Int J Mol Sci 2023; 24:ijms24097776. [PMID: 37175486 PMCID: PMC10178361 DOI: 10.3390/ijms24097776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Bruton's tyrosine kinase (Btk) and spleen tyrosine kinase (Syk) are major signaling proteins in human platelets that are implicated in atherothrombosis and thrombo-inflammation, but the mechanisms controlling their activities are not well understood. Previously, we showed that Syk becomes phosphorylated at S297 in glycoprotein VI (GPVI)-stimulated human platelets, which limits Syk activation. Here, we tested the hypothesis that protein kinases C (PKC) and A (PKA) and protein phosphatase 2A (PP2A) jointly regulate GPVI-induced Btk activation in platelets. The GPVI agonist convulxin caused rapid, transient Btk phosphorylation at S180 (pS180↑), Y223 and Y551, while direct PKC activation strongly increased Btk pS180 and pY551. This increase in Btk pY551 was also Src family kinase (SFK)-dependent, but surprisingly Syk-independent, pointing to an alternative mechanism of Btk phosphorylation and activation. PKC inhibition abolished convulxin-stimulated Btk pS180 and Syk pS297, but markedly increased the tyrosine phosphorylation of Syk, Btk and effector phospholipase Cγ2 (PLCγ2). PKA activation increased convulxin-induced Btk activation at Y551 but strongly suppressed Btk pS180 and Syk pS297. PP2A inhibition by okadaic acid only increased Syk pS297. Both platelet aggregation and PLCγ2 phosphorylation with convulxin stimulation were Btk-dependent, as shown by the selective Btk inhibitor acalabrutinib. Together, these results revealed in GPVI-stimulated platelets a transient Syk, Btk and PLCγ2 phosphorylation at multiple sites, which are differentially regulated by PKC, PKA or PP2A. Our work thereby demonstrated the GPVI-Syk-Btk signalosome as a tightly controlled protein kinase network, in agreement with its role in atherothrombosis.
Collapse
Affiliation(s)
- Pengyu Zhang
- Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Fiorella A Solari
- Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
| | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands
- Synapse Research Institute Maastricht, 6217 KD Maastricht, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Albert Sickmann
- Leibniz Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
- Medizinische Fakultät, Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44780 Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| |
Collapse
|
12
|
Sharina I, Martin E. Cellular Factors That Shape the Activity or Function of Nitric Oxide-Stimulated Soluble Guanylyl Cyclase. Cells 2023; 12:471. [PMID: 36766813 PMCID: PMC9914232 DOI: 10.3390/cells12030471] [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: 01/05/2023] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
NO-stimulated guanylyl cyclase (SGC) is a hemoprotein that plays key roles in various physiological functions. SGC is a typical enzyme-linked receptor that combines the functions of a sensor for NO gas and cGMP generator. SGC possesses exclusive selectivity for NO and exhibits a very fast binding of NO, which allows it to function as a sensitive NO receptor. This review describes the effect of various cellular factors, such as additional NO, cell thiols, cell-derived small molecules and proteins on the function of SGC as cellular NO receptor. Due to its vital physiological function SGC is an important drug target. An increasing number of synthetic compounds that affect SGC activity via different mechanisms are discovered and brought to clinical trials and clinics. Cellular factors modifying the activity of SGC constitute an opportunity for improving the effectiveness of existing SGC-directed drugs and/or the creation of new therapeutic strategies.
Collapse
Affiliation(s)
| | - Emil Martin
- Department of Internal Medicine, Cardiology Division, The University of Texas—McGovern Medical School, 1941 East Road, Houston, TX 77054, USA
| |
Collapse
|
13
|
Affiliation(s)
- Zoltan Nagy
- Institute of Experimental Biomedicine, University of Würzburg, University Hospital, Würzburg, Germany.,Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University of Würzburg, University Hospital, Würzburg, Germany.,Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| |
Collapse
|
14
|
Anti-Atherosclerotic Action of a New Stimulator of Soluble Guanylate Cyclase in an Experiment. Bull Exp Biol Med 2023; 174:333-336. [PMID: 36723753 DOI: 10.1007/s10517-023-05703-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Indexed: 02/02/2023]
Abstract
We studied the effect of an indolinone derivative GRS on the development of experimental atherosclerosis in C57BL/6 mice. Atherosclerosis was modeled by intraperitoneal administration of endothelial lipoprotein lipase inhibitor Kolliphor P 407 micro Geismar over 5 months. GRS was administered orally in a dose of 10 mg/kg once a day throughout the experiment. In 5 months, the levels of total cholesterol, LDL, and triglycerides in blood serum, as well as histological composition of the ascending aorta were studied. In mice with experimental atherosclerosis, we observed pronounced dyslipidemia with an increase in serum cholesterol, LDL, and triglycerides and accumulation of xanthoma cells in the aorta wall. Repeat administration of GRS did not eliminate dyslipidemia, but prevented an increase in the number of xanthoma cells in the aorta wall (p<0.05). The stimulator of soluble guanylate cyclase GRS did not exhibit hypolipidemic activity, but restored impaired endothelial function in the atherosclerosis model and prevented atherosclerotic damage to blood vessels and vascular wall remodeling.
Collapse
|
15
|
Donkin R, Fung YL, Singh I. Fibrinogen, Coagulation, and Ageing. Subcell Biochem 2023; 102:313-342. [PMID: 36600138 DOI: 10.1007/978-3-031-21410-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The World Health Organization estimates that the world's population over 60 years of age will nearly double in the next 30 years. This change imposes increasing demands on health and social services with increased disease burden in older people, hereafter defined as people aged 60 years or more. An older population will have a greater incidence of cardiovascular disease partly due to higher levels of blood fibrinogen, increased levels of some coagulation factors, and increased platelet activity. These factors lead to a hypercoagulable state which can alter haemostasis, causing an imbalance in appropriate coagulation, which plays a crucial role in the development of cardiovascular diseases. These changes in haemostasis are not only affected by age but also by gender and the effects of hormones, or lack thereof in menopause for older females, ethnicity, other comorbidities, medication interactions, and overall health as we age. Another confounding factor is how we measure fibrinogen and coagulation through laboratory and point-of-care testing and how our decision-making on disease and treatment (including anticoagulation) is managed. It is known throughout life that in normal healthy individuals the levels of fibrinogen and coagulation factors change, however, reference intervals to guide diagnosis and management are based on only two life stages, paediatric, and adult ranges. There are no specific diagnostic guidelines based on reference intervals for an older population. How ageing relates to alterations in haemostasis and the impact of the disease will be discussed in this chapter. Along with the effect of anticoagulation, laboratory testing of fibrinogen and coagulation, future directions, and implications will be presented.
Collapse
Affiliation(s)
- Rebecca Donkin
- The University of the Sunshine Coast, School of Health and Behavioural Sciences, Sippy Downs, QLD, Australia. .,Griffith University, School of Medicine and Dentistry, Gold Coast, QLD, Australia.
| | - Yoke Lin Fung
- The University of the Sunshine Coast, School of Health and Behavioural Sciences, Sippy Downs, QLD, Australia
| | - Indu Singh
- Griffith University, School of Pharmacy and Medical Science, Gold Coast, QLD, Australia
| |
Collapse
|
16
|
The Role of NO/sGC/cGMP/PKG Signaling Pathway in Regulation of Platelet Function. Cells 2022; 11:cells11223704. [PMID: 36429131 PMCID: PMC9688146 DOI: 10.3390/cells11223704] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Circulating blood platelets are controlled by stimulatory and inhibitory factors, and a tightly regulated equilibrium between these two opposing processes is essential for normal platelet and vascular function. NO/cGMP/ Protein Kinase G (PKG) pathways play a highly significant role in platelet inhibition, which is supported by a large body of studies and data. This review focused on inconsistent and controversial data of NO/sGC/cGMP/PKG signaling in platelets including sources of NO that activate sGC in platelets, the role of sGC/PKG in platelet inhibition/activation, and the complexity of the regulation of platelet inhibitory mechanisms by cGMP/PKG pathways. In conclusion, we suggest that the recently developed quantitative phosphoproteomic method will be a powerful tool for the analysis of PKG-mediated effects. Analysis of phosphoproteins in PKG-activated platelets will reveal many new PKG substrates. A future detailed analysis of these substrates and their involvement in different platelet inhibitory pathways could be a basis for the development of new antiplatelet drugs that may target only specific aspects of platelet functions.
Collapse
|
17
|
Wu G, Sharina I, Martin E. Soluble guanylyl cyclase: Molecular basis for ligand selectivity and action in vitro and in vivo. Front Mol Biosci 2022; 9:1007768. [PMID: 36304925 PMCID: PMC9592903 DOI: 10.3389/fmolb.2022.1007768] [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: 07/30/2022] [Accepted: 09/27/2022] [Indexed: 01/14/2023] Open
Abstract
Nitric oxide (NO), carbon monoxide (CO), oxygen (O2), hydrogen sulfide (H2S) are gaseous molecules that play important roles in the physiology and pathophysiology of eukaryotes. Tissue concentrations of these physiologically relevant gases vary remarkable from nM range for NO to high μM range of O2. Various hemoproteins play a significant role in sensing and transducing cellular signals encoded by gaseous molecules or in transporting them. Soluble guanylyl cyclase (sGC) is a hemoprotein that plays vital roles in a wide range of physiological functions and combines the functions of gaseous sensor and signal transducer. sGC uniquely evolved to sense low non-toxic levels of NO and respond to elevated NO levels by increasing its catalytic ability to generate the secondary signaling messenger cyclic guanosine monophosphate (cGMP). This review discusses sGC's gaseous ligand selectivity and the molecular basis for sGC function as high-affinity and selectivity NO receptor. The effects of other gaseous molecules and small molecules of cellular origin on sGC's function are also discussed.
Collapse
Affiliation(s)
- Gang Wu
- Hematology-Oncology Division, Department of Internal Medicine, The University of Texas—McGovern Medical School, Houston, TX, United States,*Correspondence: Gang Wu, ; Emil Martin,
| | - Iraida Sharina
- Cardiology Division, Department of Internal Medicine, The University of Texas—McGovern Medical School, Houston, TX, United States
| | - Emil Martin
- Cardiology Division, Department of Internal Medicine, The University of Texas—McGovern Medical School, Houston, TX, United States,*Correspondence: Gang Wu, ; Emil Martin,
| |
Collapse
|
18
|
Wang X, Li J, Liu L, Kan JM, Niu P, Yu ZQ, Ma C, Dong F, Han MX, Li J, Zhao DX. Pharmacological mechanism and therapeutic efficacy of Icariside II in the treatment of acute ischemic stroke: a systematic review and network pharmacological analysis. BMC Complement Med Ther 2022; 22:253. [PMID: 36180911 PMCID: PMC9526298 DOI: 10.1186/s12906-022-03732-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/19/2022] [Indexed: 01/23/2025] Open
Abstract
Background and objective Epimedii has long been used as a traditional medicine in Asia for the treatment of various common diseases, including Alzheimer's disease, cancer, erectile dysfunction, and stroke. Studies have reported the ameliorative effects of Icariside II (ICS II), a major metabolite of Epimedii, on acute ischemic stroke (AIS) in animal models. Based on network pharmacology, molecular docking, and molecular dynamics (MD) simulations, we conducted a systematic review to evaluate the effects and neuroprotective mechanisms of ICS II on AIS. Methods First, we have searched 6 databases using studies with ICS II treatment on AIS animal models to explore the efficacy of ICS II on AIS in preclinical studies. The literature retrieval time ended on March 8, 2022 (Systematic Review Registration ID: CRD42022306291). There were no restrictions on the language of the search strategy. Systematic review follows the Patient, Intervention, Comparison and Outcome (PICO) methodology and framework. SYCLE's RoB tool was used to evaluate the the risk of bias. In network pharmacology, AIS-related genes were identified and the target-pathway network was constructed. Then, these targets were used in the enrichments of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology (GO). Molecular docking and MD simulation were finally employed between ICS II and the potential target genes. Results Twelve publications were included describing outcomes of 1993 animals. The literature details, animal strains, induction models, doses administered, duration of administration, and outcome measures were extracted from the 12 included studies. ICS II has a good protective effect against AIS. Most of the studies in this systematic review had the appropriate methodological quality, but some did not clearly state the controlling for bias of potential study. Network pharmacology identified 246 targets with SRC, CTNNB1, HSP90AA1, MAPK1, and RELA as the core target proteins. Besides, 215 potential pathways of ICS II were identified, such as PI3K-Akt, MAPK, and cGMP-PKG signaling pathway. GO enrichment analysis showed that ICS II was significantly enriched in subsequent regulation such as MAPK cascade. Molecular docking and MD simulations showed that ICS II can closely bind with important targets. Conclusions ICS II is a promising drug in the treatment of AIS. However, this systematic review reveals key knowledge gaps (i.e., the protective role of ICS II in women) that ICS II must address before it can be used for the treatment of human AIS. Our study shows that ICS II plays a protective role in AIS through multi-target and multi-pathway characteristics, providing ideas for the development of drugs for the treatment of AIS. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03732-9.
Collapse
|
19
|
Granja TF, Köhler D, Leiss V, Eggstein C, Nürnberg B, Rosenberger P, Beer-Hammer S. Platelets and the Cybernetic Regulation of Ischemic Inflammatory Responses through PNC Formation Regulated by Extracellular Nucleotide Metabolism and Signaling. Cells 2022; 11:cells11193009. [PMID: 36230973 PMCID: PMC9561997 DOI: 10.3390/cells11193009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Ischemic events are associated with severe inflammation and are here referred to as ischemic inflammatory response (IIR). Recent studies identified the formation of platelet–neutrophil complexes (PNC) as key players in IIR. We investigated the role of extracellular platelet nucleotide signaling in the context of IIR and defined a cybernetic circle, including description of feedback loops. Cybernetic circles seek to integrate different levels of information to understand how biological systems function. Our study specifies the components of the cybernetic system of platelets in IIR and describes the theoretical progression of IIR passing the cybernetic cycle with positive and negative feedback loops based on nucleotide-dependent signaling and functional regulation. The cybernetic components and feedback loops were explored by cytometry, immunohistological staining, functional blocking antibodies, and ADP/ATP measurements. Using several ex vivo and in vivo approaches we confirmed cybernetic parameters, such as controller, sensor, and effector (VASP phosphorylation, P2Y12, ADORAs and GPIIb/IIIa activity), as well as set points (ADP, adenosine) and interfering control and disturbance variables (ischemia). We demonstrate the impact of the regulated platelet–neutrophil complex (PNC) formation in blood and the resulting damage to the affected inflamed tissue. Taken together, extracellular nucleotide signaling, PNC formation, and tissue damage in IIR can be integrated in a controlled cybernetic circle of platelet function, as introduced through this study.
Collapse
Affiliation(s)
- Tiago F. Granja
- Lusófona’s Research Center for Biosciences & Health Technologies, CBIOS–Universidade, Campo Grande 376, 1749-024 Lisboa, Portugal
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - David Köhler
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Veronika Leiss
- Department of Pharmacology and Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Claudia Eggstein
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology and Experimental Therapy and Toxicology and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen University Hospital, Wilhelmstrasse 56, D-72074 Tübingen, Germany
- Correspondence: ; Tel.: +49-7071-29-74594
| |
Collapse
|
20
|
Breitenbach T, Englert N, Osmanoglu Ö, Rukoyatkina N, Wangorsch G, Heinze K, Friebe A, Butt E, Feil R, Dittrich M, Gambaryan S, Dandekar T. A modular systems biological modelling framework studies cyclic nucleotide signaling in platelets. J Theor Biol 2022; 550:111222. [PMID: 35843440 DOI: 10.1016/j.jtbi.2022.111222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/29/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND The cyclic nucleotides cAMP and cGMP inhibit platelet activation. Different platelet signaling modules work together. We develop here a modelling framework to integrate different signaling modules and apply it to platelets. RESULTS We introduce a novel standardized bilinear coupling mechanism allowing sub model debugging and standardization of coupling with optimal data driven modelling by methods from optimization. Besides cAMP signaling our model considers specific cGMP effects including external stimuli by drugs. Moreover, the output of the cGMP module serves as input for a modular model of VASP phosphorylation and for the activity of cAMP and cGMP pathways in platelets. Experimental data driven modeling allows us to design models with quantitative output. We use the condensed information about involved regulation and system responses for modeling drug effects and obtaining optimal experimental settings. Stepwise further validation of our model is given by direct experimental data. CONCLUSIONS We present a general framework for model integration using modules and their stimulus responses. We demonstrate it by a multi-modular model for platelet signaling focusing on cGMP and VASP phosphorylation. Moreover, this allows to estimate drug action on any of the inhibitory cyclic nucleotide pathways (cGMP, cAMP) and is supported by experimental data.
Collapse
Affiliation(s)
- Tim Breitenbach
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Nils Englert
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; Department of Vegetative Physiology, University of Würzburg, Roentgenring 9, 97070 Würzburg, Germany
| | - Özge Osmanoglu
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Natalia Rukoyatkina
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Gaby Wangorsch
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, 63225 Langen, Germany
| | - Katrin Heinze
- Rudolf Virchow Zentrum, Universität Würzburg, Josef-Schneider-Str. 2, D15, 97080 Würzburg
| | - Andreas Friebe
- Department of Vegetative Physiology, University of Würzburg, Roentgenring 9, 97070 Würzburg, Germany
| | - Elke Butt
- Institute of Experimental Biomedicine II, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie (IFIB), University of Tübingen, Auf der Morgenstelle 34, 72076 Tübingen, Germany
| | - Marcus Dittrich
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; Department of Human Genetics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; European Molecular Biology Laboratory (EMBL), Postfach 102209, 69012 Heidelberg, Germany.
| |
Collapse
|
21
|
Prüschenk S, Schlossmann J. Function of IRAG2 Is Modulated by NO/cGMP in Murine Platelets. Int J Mol Sci 2022; 23:ijms23126695. [PMID: 35743138 PMCID: PMC9223716 DOI: 10.3390/ijms23126695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 01/27/2023] Open
Abstract
Inositol 1,4,5-triphosphate receptor-associated 2 (IRAG2) is a type II membrane protein located at the endoplasmic reticulum. It is a homologue of inositol 1,4,5-triphosphate receptor-associated cGMP kinase substrate 1 (IRAG1), a substrate protein of cGMP-dependent protein kinase I (PKGI), and is among others expressed in platelets. Here, we studied if IRAG2 is also located in platelets and might be a substrate protein of PKGI. IRAG2 was detected in platelets of IRAG2-WT animals but not in those of IRAG2-KO animals. Next, we validated by co-immunoprecipitation studies that IRAG2 is associated with IP3R1-3. No direct stable interaction with PKGIβ or with IRAG1 was observed. Phosphorylation of IRAG2 in murine platelets using a Ser/Thr-specific phospho-antibody was found in vitro and ex vivo upon cGMP stimulation. To gain insight into the function of IRAG2, platelet aggregation studies were performed using thrombin and collagen as agonists for treatment of isolated IRAG2-WT or IRAG2-KO platelets. Interestingly, platelet aggregation was reduced in the absence of IRAG2. Pretreatment of wild type or IRAG2-KO platelets with sodium nitroprusside (SNP) or 8-pCPT-cGMP revealed a further reduction in platelet aggregation in the absence of IRAG2. These results show that IRAG2 is a substrate of PKGI in murine platelets. Furthermore, our results indicate that IRAG2 is involved in the induction of thrombin- or collagen-induced platelet aggregation and that this effect is enhanced by cGMP-dependent phosphorylation of IRAG2. As IRAG1 was previously shown to inhibit platelet aggregation in a cGMP-dependent manner, it can be speculated that IRAG2 exerts an opposing function and might be an IRAG1 counterpart in murine platelets.
Collapse
|
22
|
Association of Genetic Variability in Selected Genes with Platelet Hyperaggregability and Arterial Thrombosis. ACTA MEDICA MARTINIANA 2022. [DOI: 10.2478/acm-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Introduction: Inherited platelet hyperaggregability, so called “Sticky platelet syndrome” (SPS), is a prothrombotic platelet disorder. The syndrome contributes more often to arterial than venous thrombosis. The most common localization of arterial occlusion involves cerebral or coronary arteries. However, SPS may also lead to thrombosis in the atypical sites of the circulation. This qualitative platelet alteration causes platelet hyperaggregability after a very low concentration of platelet inducers – adenosine diphosphate (ADP) and/or epinephrine (EPI). The precise genetic background of the syndrome has not been defined. In the present study we aimed to determine the association between selected single nucleotide polymorphisms (SNPs) within genes for platelet endothelial aggregation receptor 1 (PEAR1) and murine retrovirus integration site 1 (MRVI1) and the risk for arterial thrombosis in patients with SPS. The products of these selected genes play an important role in platelet aggregation.
Patients and methods: We examined 69 patients with SPS and a history of arterial thrombosis and 69 healthy blood donors who served as controls. SPS was confirmed by a light transmission aggregometry (LTA) according to the method and criteria described by Mammen and Bick. We assessed two SNPs within PEAR1 gene (rs12041331, rs1256888) and two SNPs within MRVI1 gene (rs1874445, rs7940646).
Results: Selected PEAR1 and MRVI1 polymorphisms seem not to be a risk factor for the development of SPS as the syndrome with an arterial thrombosis phenotype. However, in the subgroup of SPS1 patients there was found a decreased frequency of the minor A allele of SNP rs12041331 in PEAR1 gene (borderline p value, p=0.061) that can be hypothesized as protective against arterial thrombosis. In the same SPS1 subgroup the haplotype TA in PEAR1 gene also showed a decreased frequency with a borderline insignificance (p=0.056). We can theorize also about its protective role in SPS1 patients. We did not confirm the protective effect of polymorphism (T/T of rs 12566888) in PEAR1 against arterial thrombosis in SPS patients and SPS subgroups.
Conclusion: Our results support the idea that examined genetic variability of the selected SNPs in PEAR1 and MRVI1 genes is not associated with platelet hyperaggregability manifested as arterial thrombosis. The possible protective role of the minor A allele of SNP rs12041331 as well as a role of haplotype TA in PEAR1 gene related to the arterial thrombosis found in the subgroup of SPS1 patients needs to be verified in further research.
Collapse
|
23
|
Bykov VV, Smol'yakova VI, Chernysheva GA, Aliev OI, Anishchenko AM, Sidekhmenova AV, Dunaeva OI, Stankevich SA, Khazanov VA. Effects of a New Antithrombotic Drug GRS, a Soluble Guanylate Cyclase Stimulator, on Endothelial Dysfunction in Rats with Myocardial Infarction. Bull Exp Biol Med 2022; 172:709-712. [PMID: 35501639 DOI: 10.1007/s10517-022-05461-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Indexed: 10/18/2022]
Abstract
New antithrombotic drug GRS, a soluble guanylate cyclase stimulator, after repeated administration in a dose of 10 mg/kg alleviates the symptoms of endothelial dysfunction in rats with myocardial infarction; it restores antiplatelet activity of the blood vessel wall and vasodilatory function of the endothelium without producing significant effect on endothelium-independent vasodilation. GRS also has direct antiaggregant and antihypertensive effects in therapeutic doses. The obtained data suggest that GRS can be therapeutically useful in patients with cardiovascular diseases accompanied by endothelial dysfunction.
Collapse
Affiliation(s)
- V V Bykov
- LLC Innovative Pharmacology Research, Tomsk, Russia. .,Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - V I Smol'yakova
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - G A Chernysheva
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - O I Aliev
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - A M Anishchenko
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.,E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - A V Sidekhmenova
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - O I Dunaeva
- E. D. Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | | | - V A Khazanov
- LLC Innovative Pharmacology Research, Tomsk, Russia
| |
Collapse
|
24
|
Roy A, Tolone A, Hilhorst R, Groten J, Tomar T, Paquet-Durand F. Kinase activity profiling identifies putative downstream targets of cGMP/PKG signaling in inherited retinal neurodegeneration. Cell Death Dis 2022; 8:93. [PMID: 35241647 PMCID: PMC8894370 DOI: 10.1038/s41420-022-00897-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/15/2022]
Abstract
Inherited retinal diseases (IRDs) are a group of neurodegenerative disorders that lead to photoreceptor cell death and eventually blindness. IRDs are characterised by a high genetic heterogeneity, making it imperative to design mutation-independent therapies. Mutations in a number of IRD disease genes have been associated with a rise of cyclic 3’,5’-guanosine monophosphate (cGMP) levels in photoreceptors. Accordingly, the cGMP-dependent protein kinase (PKG) has emerged as a new potential target for the mutation-independent treatment of IRDs. However, the substrates of PKG and the downstream degenerative pathways triggered by its activity have yet to be determined. Here, we performed kinome activity profiling of different murine organotypic retinal explant cultures (diseased rd1 and wild-type controls) using multiplex peptide microarrays to identify proteins whose phosphorylation was significantly altered by PKG activity. In addition, we tested the downstream effect of a known PKG inhibitor CN03 in these organotypic retina cultures. Among the PKG substrates were potassium channels belonging to the Kv1 family (KCNA3, KCNA6), cyclic AMP-responsive element-binding protein 1 (CREB1), DNA topoisomerase 2-α (TOP2A), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (F263), and the glutamate ionotropic receptor kainate 2 (GRIK2). The retinal expression of these PKG targets was further confirmed by immunofluorescence and could be assigned to various neuronal cell types, including photoreceptors, horizontal cells, and ganglion cells. Taken together, this study confirmed the key role of PKG in photoreceptor cell death and identified new downstream targets of cGMP/PKG signalling that will improve the understanding of the degenerative mechanisms underlying IRDs.
Collapse
Affiliation(s)
- Akanksha Roy
- Division of Toxicology, Wageningen University and Research, 96708 WE, Wageningen, The Netherlands.,PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - Arianna Tolone
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, 72072, Germany
| | - Riet Hilhorst
- PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - John Groten
- Division of Toxicology, Wageningen University and Research, 96708 WE, Wageningen, The Netherlands.,PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - Tushar Tomar
- PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands.
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, 72072, Germany.
| |
Collapse
|
25
|
Comer SP. Turning Platelets Off and On: Role of RhoGAPs and RhoGEFs in Platelet Activity. Front Cardiovasc Med 2022; 8:820945. [PMID: 35071371 PMCID: PMC8770426 DOI: 10.3389/fcvm.2021.820945] [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: 11/23/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
Platelet cytoskeletal reorganisation is a critical component of platelet activation and thrombus formation in haemostasis. The Rho GTPases RhoA, Rac1 and Cdc42 are the primary drivers in the dynamic reorganisation process, leading to the development of filopodia and lamellipodia which dramatically increase platelet surface area upon activation. Rho GTPases cycle between their active (GTP-bound) and inactive (GDP-bound) states through tightly regulated processes, central to which are the guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). GEFs catalyse the dissociation of GDP by inducing changes in the nucleotide binding site, facilitating GTP binding and activating Rho GTPases. By contrast, while all GTPases possess intrinsic hydrolysing activity, this reaction is extremely slow. Therefore, GAPs catalyse the hydrolysis of GTP to GDP, reverting Rho GTPases to their inactive state. Our current knowledge of these proteins is constantly being updated but there is considerably less known about the functionality of Rho GTPase specific GAPs and GEFs in platelets. In the present review, we discuss GAP and GEF proteins for Rho GTPases identified in platelets, their regulation, biological function and present a case for their further study in platelets.
Collapse
Affiliation(s)
- Shane P Comer
- ConwaySPHERE Research Group, UCD Conway Institute, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
26
|
Rukoyatkina N, Shpakova V, Bogoutdinova A, Kharazova A, Mindukshev I, Gambaryan S. Curcumin by activation of adenosine A 2A receptor stimulates protein kinase a and potentiates inhibitory effect of cangrelor on platelets. Biochem Biophys Res Commun 2022; 586:20-26. [PMID: 34823218 DOI: 10.1016/j.bbrc.2021.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/28/2022]
Abstract
Curcumin is a natural polyphenol derived from the turmeric plant (Curcuma longa) which exhibits numerous beneficial effects on different cell types. Inhibition of platelet activation by curcumin is well known, however molecular mechanisms of its action on platelets are not fully defined. In this study, we used laser diffraction method for analysis of platelet aggregation and Western blot for analysis of intracellular signaling mechanisms of curcumin effects on platelets. We identified two new molecular mechanisms involved in the inhibitory effects of curcumin on platelet activation. Firstly, curcumin by activation of adenosine A2A receptor stimulated protein kinase A activation and phosphorylation of Vasodilator-stimulated phosphoprotein. Secondly, we demonstrated that curcumin even at low doses, which did not inhibit platelet aggregation, potentiated inhibitory effect of ADP receptor P2Y12 antagonist cangrelor which partly could be explained by activation of adenosine A2A receptor.
Collapse
Affiliation(s)
- Natalia Rukoyatkina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Thorez Prospect 44, Saint Petersburg, 194223, Russia.
| | - Valentina Shpakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Thorez Prospect 44, Saint Petersburg, 194223, Russia.
| | - Alina Bogoutdinova
- Saint Petersburg State Chemical Pharmaceutical University, Professora Popova Street 14, Saint Petersburg, 197376, Russia.
| | - Alexandra Kharazova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., Saint Petersburg, 199034, Russia.
| | - Igor Mindukshev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Thorez Prospect 44, Saint Petersburg, 194223, Russia.
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Thorez Prospect 44, Saint Petersburg, 194223, Russia.
| |
Collapse
|
27
|
Godwin MD, Aggarwal A, Hilt Z, Shah S, Gorski J, Cameron SJ. Sex-Dependent Effect of Platelet Nitric Oxide: Production and Platelet Reactivity in Healthy Individuals. JACC Basic Transl Sci 2022; 7:14-25. [PMID: 35128205 PMCID: PMC8807728 DOI: 10.1016/j.jacbts.2021.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/17/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022]
Abstract
Platelet reactivity is greater in healthy women compared with men. Following an oral nitrate load, platelet nitric oxide production increased disproportionately more in healthy women than healthy men with attenuated platelet reactivity in women and enhanced platelet reactivity in men.
A nitrate-rich diet has many cardiovascular benefits, but the mechanism behind this is unclear. We hypothesized that the ingestion of nitrate augments nitrate to nitrite reduction, leading to nitric oxide (NO) production, which may suppress platelet reactivity. In a randomized, double-blinded, placebo-controlled study involving healthy individuals, ingestion of nitrate augmented saliva and plasma nitrite/nitrate concentration and enhanced platelet NO production disproportionately in women compared with men. The response of elevated platelet NO in men was increased platelet reactivity and the response of markedly elevated platelet NO in women slightly inhibited platelet reactivity.
Collapse
Affiliation(s)
- Matthew D. Godwin
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Anu Aggarwal
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Zachary Hilt
- Department of Medicine, Aab Cardiovascular Research Center, University of Rochester School of Medicine, Rochester, New York, USA
| | - Shalini Shah
- Department of Medicine, Division of Cardiology, University of Rochester School of Medicine, Rochester, New York, USA
| | - Joshua Gorski
- Department of Medicine, Division of Cardiology, University of Rochester School of Medicine, Rochester, New York, USA
| | - Scott J. Cameron
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Department of Medicine, Aab Cardiovascular Research Center, University of Rochester School of Medicine, Rochester, New York, USA
- Department of Medicine, Division of Cardiology, University of Rochester School of Medicine, Rochester, New York, USA
- Heart, Vascular, and Thoracic Institute, Department of Cardiovascular Medicine, Section of Vascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Taussig Institute, Department Hematology, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Address for correspondence: Dr Scott J. Cameron, Cleveland Clinic Foundation, Heart Vascular and Thoracic Institute, Department of Cardiovascular Medicine, Section of Vascular Medicine, J3-5, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA.
| |
Collapse
|
28
|
Belleville-Rolland T, Leuci A, Mansour A, Decouture B, Martin F, Poirault-Chassac S, Rouaud M, Guerineau H, Dizier B, Pidard D, Gaussem P, Bachelot-Loza C. Role of Membrane Lipid Rafts in MRP4 (ABCC4) Dependent Regulation of the cAMP Pathway in Blood Platelets. Thromb Haemost 2021; 121:1628-1636. [PMID: 33851387 DOI: 10.1055/a-1481-2663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Platelet cytosolic cyclic adenosine monophosphate (cAMP) levels are balanced by synthesis, degradation, and efflux. Efflux can occur via multidrug resistant protein-4 (MRP4; ABCC4) present on dense granule and/or plasma membranes. As lipid rafts have been shown to interfere on cAMP homeostasis, we evaluated the relationships between the distribution and activity of MRP4 in lipid rafts and cAMP efflux. METHODS Platelet activation and cAMP homeostasis were analyzed in human and wild-type or MRP4-deleted mouse platelets in the presence of methyl-β-cyclodextrin (MßCD) to disrupt lipid rafts, and of activators of the cAMP signalling pathways. Human platelet MRP4 and effector proteins of the cAMP pathway were analyzed by immunoblots in lipid rafts isolated by differential centrifugation. RESULTS MßCD dose dependently inhibited human and mouse platelet aggregation without affecting per se cAMP levels. An additive inhibitory effect existed between the adenylate cyclase (AC) activator forskolin and MßCD that was accompanied by an overincrease of cAMP, and which was significantly enhanced upon MRP4 deletion. Finally, an efflux of cAMP out of resting platelets incubated with prostaglandin E1 (PGE1) was observed that was partly dependent on MRP4. Lipid rafts contained a small fraction (≈15%) of MRP4 and most of the inhibitory G-protein Gi, whereas Gs protein, AC3, and phosphodiesterases PDE2 and PDE3A were all present as only trace amounts. CONCLUSION Our results are in favour of part of MRP4 present at the platelet surface, including in lipid rafts. Lipid raft integrity is necessary for cAMP signalling regulation, although MRP4 and most players of cAMP homeostasis are essentially located outside rafts.
Collapse
Affiliation(s)
- Tiphaine Belleville-Rolland
- Service d'hématologie biologique, AH-HP, Hopital Européen Georges Pompidou, Paris, France
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Alexandre Leuci
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Alexandre Mansour
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Benoit Decouture
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Fanny Martin
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | | | - Margot Rouaud
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Hippolyte Guerineau
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Blandine Dizier
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Dominique Pidard
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | - Pascale Gaussem
- Service d'hématologie biologique, AH-HP, Hopital Européen Georges Pompidou, Paris, France
- Université de Paris, Innovative Therapies in Haemostasis, INSERM U1140, Paris, France
| | | |
Collapse
|
29
|
Shpakova V, Rukoyatkina N, Walter U, Gambaryan S. Potential and limitations of PKA/ PKG inhibitors for platelet studies. Platelets 2021; 33:859-868. [PMID: 34845961 DOI: 10.1080/09537104.2021.2003316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cyclic nucleotides (cAMP and cGMP) and corresponding protein kinases, protein kinase A (PKA) and protein kinase G (PKG), are the main intracellular mediators of endothelium-derived platelet inhibitors. Pharmacological PKA/PKG inhibitors are often used to discriminate between these two kinase activities and to analyze their underlying mechanisms. Previously we showed that all widely used PKG inhibitors (KT5823, DT3, RP isomers) either did not inhibit PKG or inhibited and even activated platelets independently from PKG. In this study, we examined several PKA inhibitors as well as inhibitors of adenylate and guanylate cyclases to reveal their effects on platelets and establish whether they are mediated by PKA/PKG. The commonly used PKA inhibitor H89 inhibited both PKA and PKG but PKA-independently inhibited thrombin-induced platelet activation. In our experiments, KT5720 did not inhibit PKA and had no effect on platelet activation. PKI inhibited PKA activity in platelets but also strongly PKA-independently activated platelets. Inhibition of adenylate and guanylate cyclases may be an alternative approach to analyze PKA/PKG function. Based on our previous and presented data, we conclude that all results where the mentioned PKA inhibitors were used for the analysis of PKA activity in intact platelets should be considered with caution.
Collapse
Affiliation(s)
- Valentina Shpakova
- Laboratory of cellular mechanisms of blood homeostasis, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Natalia Rukoyatkina
- Laboratory of cellular mechanisms of blood homeostasis, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stepan Gambaryan
- Laboratory of cellular mechanisms of blood homeostasis, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
30
|
Molecular Proteomics and Signalling of Human Platelets in Health and Disease. Int J Mol Sci 2021; 22:ijms22189860. [PMID: 34576024 PMCID: PMC8468031 DOI: 10.3390/ijms22189860] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/21/2022] Open
Abstract
Platelets are small anucleate blood cells that play vital roles in haemostasis and thrombosis, besides other physiological and pathophysiological processes. These roles are tightly regulated by a complex network of signalling pathways. Mass spectrometry-based proteomic techniques are contributing not only to the identification and quantification of new platelet proteins, but also reveal post-translational modifications of these molecules, such as acetylation, glycosylation and phosphorylation. Moreover, target proteomic analysis of platelets can provide molecular biomarkers for genetic aberrations with established or non-established links to platelet dysfunctions. In this report, we review 67 reports regarding platelet proteomic analysis and signalling on a molecular base. Collectively, these provide detailed insight into the: (i) technical developments and limitations of the assessment of platelet (sub)proteomes; (ii) molecular protein changes upon ageing of platelets; (iii) complexity of platelet signalling pathways and functions in response to collagen, rhodocytin, thrombin, thromboxane A2 and ADP; (iv) proteomic effects of endothelial-derived mediators such as prostacyclin and the anti-platelet drug aspirin; and (v) molecular protein changes in platelets from patients with congenital disorders or cardiovascular disease. However, sample sizes are still low and the roles of differentially expressed proteins are often unknown. Based on the practical and technical possibilities and limitations, we provide a perspective for further improvements of the platelet proteomic field.
Collapse
|
31
|
Ebermeyer T, Cognasse F, Berthelot P, Mismetti P, Garraud O, Hamzeh-Cognasse H. Platelet Innate Immune Receptors and TLRs: A Double-Edged Sword. Int J Mol Sci 2021; 22:ijms22157894. [PMID: 34360659 PMCID: PMC8347377 DOI: 10.3390/ijms22157894] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Platelets are hematopoietic cells whose main function has for a long time been considered to be the maintenance of vascular integrity. They have an essential role in the hemostatic response, but they also have functional capabilities that go far beyond it. This review will provide an overview of platelet functions. Indeed, stress signals may induce platelet apoptosis through proapoptotis or hemostasis receptors, necrosis, and even autophagy. Platelets also interact with immune cells and modulate immune responses in terms of activation, maturation, recruitment and cytokine secretion. This review will also show that platelets, thanks to their wide range of innate immune receptors, and in particular toll-like receptors, and can be considered sentinels actively participating in the immuno-surveillance of the body. We will discuss the diversity of platelet responses following the engagement of these receptors as well as the signaling pathways involved. Finally, we will show that while platelets contribute significantly, via their TLRs, to immune response and inflammation, these receptors also participate in the pathophysiological processes associated with various pathogens and diseases, including cancer and atherosclerosis.
Collapse
Affiliation(s)
- Théo Ebermeyer
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
| | - Fabrice Cognasse
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Etablissement Français du Sang Auvergne-Rhône-Alpes, 25 bd Pasteur, F-42100 Saint-Étienne, France
| | - Philippe Berthelot
- Team GIMAP, CIRI—Centre International de Recherche en Infectiologie, Université de Lyon, U1111, UMR5308, F-69007 Lyon, France;
- Infectious Diseases Department, CHU de St-Etienne, F-42055 Saint-Etienne, France
| | - Patrick Mismetti
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Department of Vascular Medicine and Therapeutics, INNOVTE, CHU de St-Etienne, F-42055 Saint-Etienne, France
| | - Olivier Garraud
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
| | - Hind Hamzeh-Cognasse
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Correspondence:
| |
Collapse
|
32
|
Shiravand Y, Walter U, Jurk K. Fine-Tuning of Platelet Responses by Serine/Threonine Protein Kinases and Phosphatases-Just the Beginning. Hamostaseologie 2021; 41:206-216. [PMID: 34192779 DOI: 10.1055/a-1476-7873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Comprehensive proteomic analyses of human and murine platelets established an extraordinary intracellular repertoire of signaling components, which control crucial functions. The spectrum of platelet serine/threonine protein kinases (more than 100) includes the AGC family (protein kinase A, G, C [PKA, PKG, PKC]), the mitogen-activated protein kinases (MAPKs), and others. PKA and PKG have multiple significantly overlapping substrates in human platelets, which possibly affect functions with clear "signaling nodes" of regulation by multiple protein kinases/phosphatases. Signaling nodes are intracellular Ca2+ stores, the contractile system (myosin light chains), and other signaling components such as G-proteins, protein kinases, and protein phosphatases. An example for this fine-tuning is the tyrosine kinase Syk, a crucial component of platelet activation, which is controlled by several serine/threonine and tyrosine protein kinases as well as phosphatases. Other protein kinases including PKA/PKG modulate protein phosphatase 2A, which may be a master regulator of MAPK signaling in human platelets. Protein kinases and in particular MAPKs are targeted by an increasing number of clinically used inhibitors. However, the precise regulation and fine-tuning of these protein kinases and their effects on other signaling components in platelets are only superficially understood-just the beginning. However, promising future approaches are in sight.
Collapse
Affiliation(s)
- Yavar Shiravand
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
33
|
Hofmann F. The cGMP system: components and function. Biol Chem 2021; 401:447-469. [PMID: 31747372 DOI: 10.1515/hsz-2019-0386] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of a variety of physiological and pathophysiological processes in many mammalian and non-mammalian tissues. Targeting this pathway by increasing cGMP levels has been a very successful approach in pharmacology as shown for nitrates, phosphodiesterase (PDE) inhibitors and stimulators of nitric oxide-guanylyl cyclase (NO-GC) and particulate GC (pGC). This is an introductory review to the cGMP signaling system intended to introduce those readers to this system, who do not work in this area. This article does not intend an in-depth review of this system. Signal transduction by cGMP is controlled by the generating enzymes GCs, the degrading enzymes PDEs and the cGMP-regulated enzymes cyclic nucleotide-gated ion channels, cGMP-dependent protein kinases and cGMP-regulated PDEs. Part A gives a very concise introduction to the components. Part B gives a very concise introduction to the functions modulated by cGMP. The article cites many recent reviews for those who want a deeper insight.
Collapse
Affiliation(s)
- Franz Hofmann
- Pharmakologisches Institut, Technische Universität München, Biedersteiner Str. 29, D-80802 München, Germany
| |
Collapse
|
34
|
Tsikas D, Gambaryan S. Nitrous anhydrase activity of carbonic anhydrase II: cysteine is required for nitric oxide (NO) dependent phosphorylation of VASP in human platelets. J Enzyme Inhib Med Chem 2021; 36:525-534. [PMID: 33508993 PMCID: PMC7875556 DOI: 10.1080/14756366.2021.1874946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The carbonic anhydrase (CA) family does not only catalyse the reversible hydration of CO2 to bicarbonate, but it also possesses esterase and phosphatase activity. Recently, bovine CA II and human CA II have been reported to convert inorganic nitrite (O=N-O−) to nitric oxide (NO) and nitrous anhydride (N2O3). Given the ability of NO to mediate vasodilation and inhibit platelet aggregation, this CA II activity would represent a bioactivation of nitrite. There are contradictory reports in the literature and the physiological role of CA II nitrite bioactivation is still disputed. Here, we provide new experimental data in support of the nitrous anhydrase activity of CA II and the key role L-cysteine in the bioactivation of nitrite by CA II. Using washed human platelets and by measuring VASP phosphorylation we provide evidence that exogenous nitrite (10 µM) is bioactivated to NO in a manner strongly depending on L-cysteine (100 and 200 µM). The process is not inhibitable by acetazolamide, a potent CA inhibitor. The contradictory results of recently published studies in this area are thoroughly discussed.
Collapse
Affiliation(s)
- Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Petersburg, Russia
| |
Collapse
|
35
|
Li X, Chen X, Hu X, Shen Y, Xu R, Wu L, Shen X. Overexpression of GUCY1A2 Correlates With Poor Prognosis in Gastric Cancer Patients. Front Oncol 2021; 11:632172. [PMID: 34113559 PMCID: PMC8185334 DOI: 10.3389/fonc.2021.632172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 01/06/2023] Open
Abstract
Background Nitric oxide (NO) and cyclic guanosine phosphate (cGMP) play important roles in blood pressure regulation, neurotransmitter delivery, renal function, and tumorigenesis and development. The intermediate link of this signaling pathway, soluble guanylyl cyclase (sGC), is particularly important. However, the role of the GUCY1A2 gene encoding the sGC α2 subunit is unknown. Methods Gene expression and clinical data were obtained from The Cancer Genome Atlas (TCGA) database. After screening for GUCY1A2 expression, the expression differences between gastric cancer (GC) tissues and adjacent noncancerous tissues were determined using R software. Quantitative real-time polymerase chain reaction (qRT-PCR) and meta-analysis were used to verify the result. The correlation between the expression of GUCY1A2 and clinicopathological parameters was explored by logistic regression. Then, Kaplan-Meier survival analysis and the Cox proportional hazards regression were used to evaluate the relationship between the expression of GUCY1A2 and the survival of GC patients. Finally, gene set enrichment analysis (GSEA) was used to explore and analyze the GC-related signaling pathways affected by high GUCY1A2 expression. Results We found that GUCY1A2 was highly expressed in GC tissues compared to adjacent noncancerous tissues (P < 0.001). qRT-PCR (P < 0.001) and meta-analysis (SMD = 0.65, 95% CI: 0.20-1.10) confirmed the difference in GUCY1A2 expression. Logistic regression analysis showed that high expression of GUCY1A2 was associated with histological grade (OR=1.858 for poor vs. well or moderate, P = 0.004) and T stage (OR = 3.389 for T3 vs. T1, P = 0.025; OR = 3.422 for T4 vs. T1, P = 0.028). Kaplan-Meier curves indicated that GC patients with high expression of GUCY1A2 had a poor prognosis than that of patients with low expression. Univariate analysis indicated that GUCY1A2 and some clinicopathological parameters, such as age, pathological stage, and TNM stage, may predict poor prognosis. Multivariate analysis further confirmed that GUCY1A2 was an independent prognostic marker (HR = 1.699; 95%CI, 1.175-2.456; P = 0.005). GSEA showed that the high GUCY1A2 phenotype is significantly enriched for tumor-associated signaling pathways. Conclusions GUCY1A2 is highly expressed in GC and may be used as a potential prognostic marker.
Collapse
Affiliation(s)
- Xin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xiaowei Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Xueju Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yan Shen
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Rui Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Leilei Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xiaobing Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
36
|
Mahdi A, Cortese-Krott MM, Kelm M, Li N, Pernow J. Novel perspectives on redox signaling in red blood cells and platelets in cardiovascular disease. Free Radic Biol Med 2021; 168:95-109. [PMID: 33789125 DOI: 10.1016/j.freeradbiomed.2021.03.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 12/13/2022]
Abstract
The fundamental physiology of circulating red blood cells (RBCs) and platelets involving regulation of oxygen transport and hemostasis, respectively, are well-described in the literature. Their abundance in the circulation and their interaction with the vascular wall and each other have attracted the attention of other putative physiological and pathophysiological effects of these cells. RBCs and platelets are both important regulators of redox balance harboring powerful pro-oxidant and anti-oxidant (enzymatic and non-enzymatic) capacities. They are also involved in the regulation of vascular tone mainly via export of nitric oxide bioactivity and adenosine triphosphate. Of further importance are emerging observations that these cells undergo functional alterations when exposed to risk factors for cardiovascular disease and during developed cardiometabolic diseases. Under these conditions, the RBCs and platelets contribute to increased oxidative stress by their formation of reactive species including superoxide anion radical, hydrogen peroxide and peroxynitrite. These alterations trigger key changes in the vascular wall characterized by enhanced oxidative stress, reduced nitric oxide bioavailability and endothelial dysfunction. Additional pathophysiological effects are triggered in the heart resulting in increased susceptibility to ischemia-reperfusion injury with impairment in cardiac function. Pharmacological interventions aiming at restoring circulating cell function has been shown to exert marked beneficial effects on cardiovascular function. In this review, we summarize the current knowledge of RBC and platelet biology with special focus on redox biology, their roles in the development of cardiovascular disease and potential therapeutic strategies targeting RBC and platelet dysfunction. Finally, the complex and scarcely understood interaction between RBCs and platelets is discussed.
Collapse
Affiliation(s)
- Ali Mahdi
- Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Nailin Li
- Department of Medicine, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Department of Cardiology, Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
37
|
Beavo JA, Golkowski M, Shimizu-Albergine M, Beltejar MC, Bornfeldt KE, Ong SE. Phosphoproteomic Analysis as an Approach for Understanding Molecular Mechanisms of cAMP-Dependent Actions. Mol Pharmacol 2021; 99:342-357. [PMID: 33574048 PMCID: PMC8058506 DOI: 10.1124/molpharm.120.000197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/23/2020] [Indexed: 12/26/2022] Open
Abstract
In recent years, highly sensitive mass spectrometry-based phosphoproteomic analysis is beginning to be applied to identification of protein kinase substrates altered downstream of increased cAMP. Such studies identify a very large number of phosphorylation sites regulated in response to increased cAMP. Therefore, we now are tasked with the challenge of determining how many of these altered phosphorylation sites are relevant to regulation of function in the cell. This minireview describes the use of phosphoproteomic analysis to monitor the effects of cyclic nucleotide phosphodiesterase (PDE) inhibitors on cAMP-dependent phosphorylation events. More specifically, it describes two examples of this approach carried out in the authors' laboratories using the selective PDE inhibitor approach. After a short discussion of several likely conclusions suggested by these analyses of cAMP function in steroid hormone-producing cells and also in T-cells, it expands into a discussion about some newer and more speculative interpretations of the data. These include the idea that multiple phosphorylation sites and not a single rate-limiting step likely regulate these and, by analogy, many other cAMP-dependent pathways. In addition, the idea that meaningful regulation requires a high stoichiometry of phosphorylation to be important is discussed and suggested to be untrue in many instances. These new interpretations have important implications for drug design, especially for targeting pathway agonists. SIGNIFICANCE STATEMENT: Phosphoproteomic analyses identify thousands of altered phosphorylation sites upon drug treatment, providing many possible regulatory targets but also highlighting questions about which phosphosites are functionally important. These data imply that multistep processes are regulated by phosphorylation at not one but rather many sites. Most previous studies assumed a single step or very few rate-limiting steps were changed by phosphorylation. This concept should be changed. Previous interpretations also assumed substoichiometric phosphorylation was not of regulatory importance. This assumption also should be changed.
Collapse
Affiliation(s)
- Joseph A Beavo
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Martin Golkowski
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Masami Shimizu-Albergine
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Michael-Claude Beltejar
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Karin E Bornfeldt
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| | - Shao-En Ong
- Departments of Pharmacology and Medicine (J.A.B., M.G., M.S.-A., M.-C.B., S.-E.O.), and Division of Metabolism, Endocrinology and Nutrition (K.E.B.), University of Washington, Seattle, Washington
| |
Collapse
|
38
|
Shevchuk O, Begonja AJ, Gambaryan S, Totzeck M, Rassaf T, Huber TB, Greinacher A, Renne T, Sickmann A. Proteomics: A Tool to Study Platelet Function. Int J Mol Sci 2021; 22:ijms22094776. [PMID: 33946341 PMCID: PMC8125008 DOI: 10.3390/ijms22094776] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022] Open
Abstract
Platelets are components of the blood that are highly reactive, and they quickly respond to multiple physiological and pathophysiological processes. In the last decade, it became clear that platelets are the key components of circulation, linking hemostasis, innate, and acquired immunity. Protein composition, localization, and activity are crucial for platelet function and regulation. The current state of mass spectrometry-based proteomics has tremendous potential to identify and quantify thousands of proteins from a minimal amount of material, unravel multiple post-translational modifications, and monitor platelet activity during drug treatments. This review focuses on the role of proteomics in understanding the molecular basics of the classical and newly emerging functions of platelets. including the recently described role of platelets in immunology and the development of COVID-19.The state-of-the-art proteomic technologies and their application in studying platelet biogenesis, signaling, and storage are described, and the potential of newly appeared trapped ion mobility spectrometry (TIMS) is highlighted. Additionally, implementing proteomic methods in platelet transfusion medicine, and as a diagnostic and prognostic tool, is discussed.
Collapse
Affiliation(s)
- Olga Shevchuk
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
- Department of Immunodynamics, Institute of Experimental Immunology and Imaging, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
- Correspondence: (O.S.); (A.S.)
| | - Antonija Jurak Begonja
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia;
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Torez pr. 44, 194223 St. Petersburg, Russia;
| | - Matthias Totzeck
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; (M.T.); (T.R.)
| | - Tienush Rassaf
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; (M.T.); (T.R.)
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Andreas Greinacher
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany;
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany;
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
- Medizinisches Proteom-Center (MPC), Medizinische Fakultät, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
- Correspondence: (O.S.); (A.S.)
| |
Collapse
|
39
|
Xu Y, Liu J, Liu Z, Chen G, Li X, Ren H. Damaging Tumor Vessels with an Ultrasound-Triggered NO Release Nanosystem to Enhance Drug Accumulation and T Cells Infiltration. Int J Nanomedicine 2021; 16:2597-2613. [PMID: 33833514 PMCID: PMC8021257 DOI: 10.2147/ijn.s295445] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Limited by tumor vascular barriers, restricted intratumoural T cell infiltration and nanoparticles accumulation remain major bottlenecks for anticancer therapy. Platelets are now known to maintain tumor vascular integrity. Therefore, inhibition of tumor-associated platelets may be an effective method to increase T cell infiltration and drug accumulation at tumor sites. Herein, we designed an ultrasound-responsive nitric oxide (NO) release nanosystem, SNO-HSA-PTX, which can release NO in response to ultrasound (US) irradiation, thereby inhibiting platelet function and opening the tumor vascular barrier, promoting drug accumulation and T cell infiltration. METHODS We evaluated the ability of SNO-HSA-PTX to release NO in response to US irradiation. We also tested the effect of SNO-HSA-PTX on platelet function. Plenty of studies including cytotoxicity, pharmacokinetics study, biodistribution, blood perfusion, T cell infiltration, in vivo antitumor efficacy and safety assessment were conducted to investigate the antitumor effect of SNO-HSA-PTX. RESULTS SNO-HSA-PTX with US irradiation inhibited tumor-associated platelets activation and induced openings in the tumor vascular barriers, which promoted the accumulation of SNO-HSA-PTX nanoparticles to the tumor sites. Meanwhile, the damaged vascular barriers allowed oxygen-carrying hemoglobin to infiltrate tumor regions, alleviating hypoxia of the tumor microenvironment. In addition, the intratumoral T cell infiltration was augmented, together with chemotherapy and NO therapy, which greatly inhibited tumor growth. DISCUSSION Our research designed a simple strategy to open the vascular barrier by inhibiting the tumor-associated platelets, which provide new ideas for anti-tumor treatment.
Collapse
Affiliation(s)
- Yan Xu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Jiwei Liu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Zhangya Liu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Guoguang Chen
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Xueming Li
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| | - Hao Ren
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
40
|
Vara D, Mailer RK, Tarafdar A, Wolska N, Heestermans M, Konrath S, Spaeth M, Renné T, Schröder K, Pula G. NADPH Oxidases Are Required for Full Platelet Activation In Vitro and Thrombosis In Vivo but Dispensable for Plasma Coagulation and Hemostasis. Arterioscler Thromb Vasc Biol 2021; 41:683-697. [PMID: 33267663 PMCID: PMC7837688 DOI: 10.1161/atvbaha.120.315565] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Using 3KO (triple NOX [NADPH oxidase] knockout) mice (ie, NOX1-/-/NOX2-/-/NOX4-/-), we aimed to clarify the role of this family of enzymes in the regulation of platelets in vitro and hemostasis in vivo. Approach and Results: 3KO mice displayed significantly reduced platelet superoxide radical generation, which was associated with impaired platelet aggregation, adhesion, and thrombus formation in response to the key agonists collagen and thrombin. A comparison with single-gene knockouts suggested that the phenotype of 3KO platelets is the combination of the effects of the genetic deletion of NOX1 and NOX2, while NOX4 does not show any significant function in platelet regulation. 3KO platelets displayed significantly higher levels of cGMP-a negative platelet regulator that activates PKG (protein kinase G). The inhibition of PKG substantially but only partially rescued the defective phenotype of 3KO platelets, which are responsive to both collagen and thrombin in the presence of the PKG inhibitors KT5823 or Rp-8-pCPT-cGMPs, but not in the presence of the NOS (NO synthase) inhibitor L-NG-monomethyl arginine. In vivo, triple NOX deficiency protected against ferric chloride-driven carotid artery thrombosis and experimental pulmonary embolism, while hemostasis tested in a tail-tip transection assay was not affected. Procoagulatory activity of platelets (ie, phosphatidylserine surface exposure) and the coagulation cascade in platelet-free plasma were normal. CONCLUSIONS This study indicates that inhibiting NOXs has strong antithrombotic effects partially caused by increased intracellular cGMP but spares hemostasis. NOXs are, therefore, pharmacotherapeutic targets to develop new antithrombotic drugs without bleeding side effects.
Collapse
Affiliation(s)
- Dina Vara
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, United Kingdom (D.V.)
| | - Reiner K. Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.M., N.W., M.H., S.K., T.R., G.P.)
| | - Anuradha Tarafdar
- Cancer Research UK Manchester Institute, University of Manchester (A.T.)
| | - Nina Wolska
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.M., N.W., M.H., S.K., T.R., G.P.)
| | - Marco Heestermans
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.M., N.W., M.H., S.K., T.R., G.P.)
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.M., N.W., M.H., S.K., T.R., G.P.)
| | - Manuela Spaeth
- Institute of Cardiovascular Physiology, Goethe-University, Frankfurt, Germany (M.S., K.S.)
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.M., N.W., M.H., S.K., T.R., G.P.)
| | - Katrin Schröder
- Institute of Cardiovascular Physiology, Goethe-University, Frankfurt, Germany (M.S., K.S.)
| | - Giordano Pula
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.M., N.W., M.H., S.K., T.R., G.P.)
| |
Collapse
|
41
|
Infante T, Costa D, Napoli C. Novel Insights Regarding Nitric Oxide and Cardiovascular Diseases. Angiology 2021; 72:411-425. [PMID: 33478246 DOI: 10.1177/0003319720979243] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a powerful mediator with biological activities such as vasodilation and prevention of vascular smooth muscle cell proliferation as well as functional regulation of cardiac cells. Thus, impaired production or reduced bioavailability of NO predisposes to the onset of different cardiovascular (CV) diseases. Alterations in the redox balance associated with excitation-contraction coupling have been identified in heart failure (HF), thus contributing to contractile abnormalities and arrhythmias. For its ability to influence cell proliferation and angiogenesis, NO may be considered a therapeutic option for the management of several CV diseases. Several clinical studies and trials investigated therapeutic NO strategies for systemic hypertension, atherosclerosis, and/or prevention of in stent restenosis, coronary heart disease (CHD), pulmonary arterial hypertension (PAH), and HF, although with mixed results in long-term treatment and effective dose administered in selected groups of patients. Tadalafil, sildenafil, and cinaguat were evaluated for the treatment of PAH, whereas vericiguat was investigated in the treatment of HF patients with reduced ejection fraction. Furthermore, supplementation with hydrogen sulfide, tetrahydrobiopterin, and nitrite/nitrate has shown beneficial effects at the vascular level.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, 18994University of Campania "L. Vanvitelli," Naples, Italy
| | - Claudio Napoli
- Department of Advanced Clinical and Surgical Sciences, 18994University of Campania "Luigi Vanvitelli," Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
42
|
Gorman S, Weller RB. Investigating the Potential for Ultraviolet Light to Modulate Morbidity and Mortality From COVID-19: A Narrative Review and Update. Front Cardiovasc Med 2020; 7:616527. [PMID: 33426009 PMCID: PMC7786057 DOI: 10.3389/fcvm.2020.616527] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
During the COVID-19 (coronavirus disease of 2019) pandemic, researchers have been seeking low-cost and accessible means of providing protection from its harms, particularly for at-risk individuals such as those with cardiovascular disease, diabetes and obesity. One possible way is via safe sun exposure, and/or dietary supplementation with induced beneficial mediators (e.g., vitamin D). In this narrative review, we provide rationale and updated evidence on the potential benefits and harms of sun exposure and ultraviolet (UV) light that may impact COVID-19. We review recent studies that provide new evidence for any benefits (or otherwise) of UV light, sun exposure, and the induced mediators, vitamin D and nitric oxide, and their potential to modulate morbidity and mortality induced by infection with SARS-CoV-2 (severe acute respiratory disease coronavirus-2). We identified substantial interest in this research area, with many commentaries and reviews already published; however, most of these have focused on vitamin D, with less consideration of UV light (or sun exposure) or other mediators such as nitric oxide. Data collected to-date suggest that ambient levels of both UVA and UVB may be beneficial for reducing severity or mortality due to COVID-19, with some inconsistent findings. Currently unresolved are the nature of the associations between blood 25-hydroxyvitamin D and COVID-19 measures, with more prospective data needed that better consider lifestyle factors, such as physical activity and personal sun exposure levels. Another short-coming has been a lack of measurement of sun exposure, and its potential to influence COVID-19 outcomes. We also discuss possible mechanisms by which sun exposure, UV light and induced mediators could affect COVID-19 morbidity and mortality, by focusing on likely effects on viral pathogenesis, immunity and inflammation, and potential cardiometabolic protective mechanisms. Finally, we explore potential issues including the impacts of exposure to high dose UV radiation on COVID-19 and vaccination, and effective and safe doses for vitamin D supplementation.
Collapse
Affiliation(s)
- Shelley Gorman
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Richard B. Weller
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Makhoul S, Kumm E, Zhang P, Walter U, Jurk K. The Serine/Threonine Protein Phosphatase 2A (PP2A) Regulates Syk Activity in Human Platelets. Int J Mol Sci 2020; 21:ijms21238939. [PMID: 33255747 PMCID: PMC7728356 DOI: 10.3390/ijms21238939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/14/2020] [Accepted: 11/19/2020] [Indexed: 12/21/2022] Open
Abstract
Distinct membrane receptors activate platelets by Src-family-kinase (SFK)-, immunoreceptor-tyrosine-based-activation-motif (ITAM)-dependent stimulation of spleen tyrosine kinase (Syk). Recently, we reported that platelet activation via glycoprotein (GP) VI or GPIbα stimulated the well-established Syk tyrosine (Y)-phosphorylation, but also stoichiometric, transient protein kinase C (PKC)-mediated Syk serine(S)297 phosphorylation in the regulatory interdomain-B, suggesting possible feedback inhibition. The transient nature of Syk S297 phosphorylation indicated the presence of an unknown Syk pS297 protein phosphatase. In this study, we hypothesize that the S-protein phosphatase 2A (PP2A) is responsible for Syk pS297 dephosphorylation, thereby affecting Syk Y-phosphorylation and activity in human washed platelets. Using immunoblotting, we show that specific inhibition of PP2A by okadaic acid (OA) alone leads to stoichiometric Syk S297 phosphorylation, as analyzed by Zn2+-Phos-tag gels, without affecting Syk Y-phosphorylation. Pharmacological inhibition of Syk by PRT060318 or PKC by GF109203X only minimally reduced OA-induced Syk S297 phosphorylation. PP2A inhibition by OA preceding GPVI-mediated platelet activation induced by convulxin extended Syk S297 phosphorylation but inhibited Syk Y-phosphorylation. Our data demonstrate a novel biochemical and functional link between the S-protein phosphatase PP2A and the Y-protein kinase Syk in human platelets, and suggest that PP2A represents a potential enhancer of GPVI-mediated Syk activity caused by Syk pS297 dephosphorylation.
Collapse
Affiliation(s)
- Stephanie Makhoul
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, D-55131 Mainz, Germany; (S.M.); (E.K.); (P.Z.)
| | - Elena Kumm
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, D-55131 Mainz, Germany; (S.M.); (E.K.); (P.Z.)
| | - Pengyu Zhang
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, D-55131 Mainz, Germany; (S.M.); (E.K.); (P.Z.)
- Leibniz-Institut für Analytische Wissenschaften, D-44227 Dortmund, Germany
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, D-55131 Mainz, Germany; (S.M.); (E.K.); (P.Z.)
- Correspondence: (U.W.); (K.J.)
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, D-55131 Mainz, Germany; (S.M.); (E.K.); (P.Z.)
- Correspondence: (U.W.); (K.J.)
| |
Collapse
|
44
|
Olas B, Urbańska K, Bryś M. Saponins as Modulators of the Blood Coagulation System and Perspectives Regarding Their Use in the Prevention of Venous Thromboembolic Incidents. Molecules 2020; 25:molecules25215171. [PMID: 33172028 PMCID: PMC7664220 DOI: 10.3390/molecules25215171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Saponins comprise a heterogenous group of chemical compounds containing a triterpene or steroid aglycone group and at least one sugar chain. They exist as secondary metabolites, occurring frequently in dicotyledonous plants and lower marine animals. Plant saponin extracts or single saponins have indicated antiplatelet and anticoagulant activity. Venous thromboembolism (VTE), including deep venous thrombosis and pulmonary embolism, is a multifactorial disease influenced by various patient characteristics such as age, immobility, previous thromboembolism and inherited thrombophilia. This mini-review (1) evaluates the current literature on saponins as modulators of the coagulation system, (2) discusses the impact of chemical structure on the modulation of the coagulation system, which may further provide a basis for drug or supplement design, (3) examines perspectives of their use in the prevention of VTE. It also describes the molecular mechanisms of action of the saponins involved in the prevention of VTE.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/3, 90-236 Lodz, Poland
- Correspondence: ; Tel./Fax: +48-42-6354485
| | - Karina Urbańska
- Faculty of Medicine, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Magdalena Bryś
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/3, 90-236 Lodz, Poland;
| |
Collapse
|
45
|
Comer S, Nagy Z, Bolado A, von Kriegsheim A, Gambaryan S, Walter U, Pagel O, Zahedi RP, Jurk K, Smolenski A. The RhoA regulators Myo9b and GEF-H1 are targets of cyclic nucleotide-dependent kinases in platelets. J Thromb Haemost 2020; 18:3002-3012. [PMID: 32692911 DOI: 10.1111/jth.15028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/15/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Circulating platelets are maintained in an inactive state by the endothelial lining of the vasculature. Endothelium-derived prostacyclin and nitric oxide stimulate cAMP- and cGMP-dependent kinases, PKA and PKG, to inhibit platelets. PKA and PKG effects include the inhibition of the GTPase RhoA, which has been suggested to involve the direct phosphorylation of RhoA on serine 188. OBJECTIVES We wanted to confirm RhoA S188 phosphorylation by cyclic nucleotide-dependent kinases and to identify possible alternative mechanisms of RhoA regulation in platelets. METHODS Phosphoproteomics data of human platelets were used to identify candidate PKA and PKG substrates. Phosphorylation of individual proteins was studied by Western blotting and Phos-tag gel electrophoresis in human platelets and transfected HEK293T cells. Pull-down assays were performed to analyze protein interaction and function. RESULTS Our data indicate that RhoA is not phosphorylated by PKA in platelets. Instead, we provide evidence that cyclic nucleotide effects are mediated through the phosphorylation of the RhoA-specific GTPase-activating protein Myo9b and the guanine nucleotide exchange factor GEF-H1. We identify Myo9b S1354 and guanine nucleotide exchange factor-H1 (GEF-H1) S886 as PKA and PKG phosphorylation sites. Myo9b S1354 phosphorylation enhances its GTPase activating protein function leading to reduced RhoA-GTP levels. GEF-H1 S886 phosphorylation stimulates binding of 14-3-3β and has been shown to inhibit GEF function by facilitating binding of GEF-H1 to microtubules. Microtubule disruption increases RhoA-GTP levels confirming the importance of GEF-H1 in platelets. CONCLUSION Phosphorylation of RhoA regulatory proteins Myo9b and GEF-H1, but not RhoA itself, is involved in cyclic nucleotide-mediated control of RhoA in human platelets.
Collapse
Affiliation(s)
- Shane Comer
- UCD School of Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Zoltan Nagy
- UCD School of Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Alfonso Bolado
- Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| | | | - Stepan Gambaryan
- Sechenov Institute for Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Oliver Pagel
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Albert Smolenski
- UCD School of Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
46
|
A new paradigm for gaseous ligand selectivity of hemoproteins highlighted by soluble guanylate cyclase. J Inorg Biochem 2020; 214:111267. [PMID: 33099233 DOI: 10.1016/j.jinorgbio.2020.111267] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023]
Abstract
Nitric oxide (NO), carbon monoxide (CO), and oxygen (O2) are important physiological messengers whose concentrations vary in a remarkable range, [NO] typically from nM to several μM while [O2] reaching to hundreds of μM. One of the machineries evolved in living organisms for gas sensing is sensor hemoproteins whose conformational change upon gas binding triggers downstream response cascades. The recently proposed "sliding scale rule" hypothesis provides a general interpretation for gaseous ligand selectivity of hemoproteins, identifying five factors that govern gaseous ligand selectivity. Hemoproteins have intrinsic selectivity for the three gases due to a neutral proximal histidine ligand while proximal strain of heme and distal steric hindrance indiscriminately adjust the affinity of these three gases for heme. On the other hand, multiple-step NO binding and distal hydrogen bond donor(s) specifically enhance affinity for NO and O2, respectively. The "sliding scale rule" hypothesis provides clear interpretation for dramatic selectivity for NO over O2 in soluble guanylate cyclase (sGC) which is an important example of sensor hemoproteins and plays vital roles in a wide range of physiological functions. The "sliding scale rule" hypothesis has so far been validated by all experimental data and it may guide future designs for heme-based gas sensors.
Collapse
|
47
|
Nechipurenko DY, Shibeko AM, Sveshnikova AN, Panteleev MA. In Silico Hemostasis Modeling and Prediction. Hamostaseologie 2020; 40:524-535. [PMID: 32916753 DOI: 10.1055/a-1213-2117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Computational physiology, i.e., reproduction of physiological (and, by extension, pathophysiological) processes in silico, could be considered one of the major goals in computational biology. One might use computers to simulate molecular interactions, enzyme kinetics, gene expression, or whole networks of biochemical reactions, but it is (patho)physiological meaning that is usually the meaningful goal of the research even when a single enzyme is its subject. Although exponential rise in the use of computational and mathematical models in the field of hemostasis and thrombosis began in the 1980s (first for blood coagulation, then for platelet adhesion, and finally for platelet signal transduction), the majority of their successful applications are still focused on simulating the elements of the hemostatic system rather than the total (patho)physiological response in situ. Here we discuss the state of the art, the state of the progress toward the efficient "virtual thrombus formation," and what one can already get from the existing models.
Collapse
Affiliation(s)
- Dmitry Y Nechipurenko
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia.,Center for Theoretical Problems of Physicochemical Pharmacology of the Russian Academy of Sciences, Moscow, Russia.,Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Aleksey M Shibeko
- Center for Theoretical Problems of Physicochemical Pharmacology of the Russian Academy of Sciences, Moscow, Russia.,Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Anastasia N Sveshnikova
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia.,Center for Theoretical Problems of Physicochemical Pharmacology of the Russian Academy of Sciences, Moscow, Russia.,Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mikhail A Panteleev
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia.,Center for Theoretical Problems of Physicochemical Pharmacology of the Russian Academy of Sciences, Moscow, Russia.,Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| |
Collapse
|
48
|
Gulati G, Kiernan MS. Phosphodiesterase-5 Inhibitor Therapy for Left Ventricular Assist Device Patients: More Data, More Questions. J Am Heart Assoc 2020; 9:e017585. [PMID: 32648504 PMCID: PMC7660721 DOI: 10.1161/jaha.120.017585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Gaurav Gulati
- CardioVascular Center Tufts Medical Center Boston MA
| | | |
Collapse
|
49
|
Kapil V, Khambata RS, Jones DA, Rathod K, Primus C, Massimo G, Fukuto JM, Ahluwalia A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol Rev 2020; 72:692-766. [PMID: 32576603 DOI: 10.1124/pr.120.019240] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
In contrast to nitric oxide, which has well established and important roles in the regulation of blood flow and thrombosis, neurotransmission, the normal functioning of the genitourinary system, and the inflammation response and host defense, its oxidized metabolites nitrite and nitrate have, until recently, been considered to be relatively inactive. However, this view has been radically revised over the past decade and more. Much evidence has now accumulated demonstrating that nitrite serves as a storage form of nitric oxide, releasing nitric oxide preferentially under acidic and/or hypoxic conditions but also occurring under physiologic conditions: a phenomenon that is catalyzed by a number of distinct mammalian nitrite reductases. Importantly, preclinical studies demonstrate that reduction of nitrite to nitric oxide results in a number of beneficial effects, including vasodilatation of blood vessels and lowering of blood pressure, as well as cytoprotective effects that limit the extent of damage caused by an ischemia/reperfusion insult, with this latter issue having been translated more recently to the clinical setting. In addition, research has demonstrated that the other main metabolite of the oxidation of nitric oxide (i.e., nitrate) can also be sequentially reduced through processing in vivo to nitrite and then nitrite to nitric oxide to exert a range of beneficial effects-most notably lowering of blood pressure, a phenomenon that has also been confirmed recently to be an effective method for blood pressure lowering in patients with hypertension. This review will provide a detailed description of the pathways involved in the bioactivation of both nitrate and nitrite in vivo, their functional effects in preclinical models, and their mechanisms of action, as well as a discussion of translational exploration of this pathway in diverse disease states characterized by deficiencies in bioavailable nitric oxide. SIGNIFICANCE STATEMENT: The past 15 years has seen a major revision in our understanding of the pathways for nitric oxide synthesis in the body with the discovery of the noncanonical pathway for nitric oxide generation known as the nitrate-nitrite-nitric oxide pathway. This review describes the molecular components of this pathway, its role in physiology, potential therapeutics of targeting this pathway, and their impact in experimental models, as well as the clinical translation (past and future) and potential side effects.
Collapse
Affiliation(s)
- V Kapil
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - R S Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - D A Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - K Rathod
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - C Primus
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - G Massimo
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - J M Fukuto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - A Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| |
Collapse
|
50
|
Pan C, Hu Y, Gong Z, Yang Y, Liu S, Quan L, Yang Z, Wei Y, Ye W. Improved Blood Compatibility and Endothelialization of Titanium Oxide Nanotube Arrays on Titanium Surface by Zinc Doping. ACS Biomater Sci Eng 2020; 6:2072-2083. [PMID: 33455341 DOI: 10.1021/acsbiomaterials.0c00187] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Titanium dioxide nanotube arrays are widely used in biomaterials due to their unique tubular structure and tunable biocompatibility. In the present study, titanium oxide nanotube arrays with different diameters were prepared on the titanium surface by anodization, followed by zinc doping using hydrothermal treatment to enhance the biocompatibility. Both the nanotube dimensions and zinc doping had obvious influences on the hydrophilicity, protein adsorption, blood compatibility, and endothelial cell behaviors of the titanium surface. The increase of the diameter and zinc doping can improve the hydrophilicity of the titanium surface. The increase of nanotube diameter could reduce the albumin adsorption while increasing the fibrinogen adsorption. However, zinc doping can simultaneously promote the adsorption of albumin and fibrinogen, and the effect was more obvious for albumin. Zinc doping can significantly improve the blood compatibility of the titanium oxide nanotubes because it cannot only increase the activity of cyclophosphate guanylate (cGMP) but also significantly reduce the platelets adhesion and hemolysis rate. Moreover, it was also found that both the smaller diameter and zinc doping nanotubes can enhance the endothelial cell adhesion and proliferation as well as up-regulate the expression of NO and VEGF. Therefore, the zinc doped titanium dioxide nanotube array can be used to simultaneously improve the blood compatibility and promote endothelialization of the titanium-based biomaterials and implants, such as intravascular stents.
Collapse
Affiliation(s)
- Changjiang Pan
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Zhihao Gong
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Ya Yang
- Department of Geriatrics, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Sen Liu
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Li Quan
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Zhongmei Yang
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Yanchun Wei
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Wei Ye
- Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huai'an 223003, China
| |
Collapse
|