1
|
Babou Kammoe RB, Sévigny J. Extracellular nucleotides in smooth muscle contraction. Biochem Pharmacol 2024; 220:116005. [PMID: 38142836 DOI: 10.1016/j.bcp.2023.116005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Extracellular nucleotides and nucleosides are crucial signalling molecules, eliciting diverse biological responses in almost all organs and tissues. These molecules exert their effects by activating specific nucleotide receptors, which are finely regulated by ectonucleotidases that break down their ligands. In this comprehensive review, we aim to elucidate the relevance of extracellular nucleotides as signalling molecules in the context of smooth muscle contraction, considering the modulatory influence of ectonucleotidases on this intricate process. Specifically, we provide a detailed examination of the involvement of extracellular nucleotides in the contraction of non-vascular smooth muscles, including those found in the urinary bladder, the airways, the reproductive system, and the gastrointestinal tract. Furthermore, we present a broader overview of the role of extracellular nucleotides in vascular smooth muscle contraction.
Collapse
Affiliation(s)
- Romuald Brice Babou Kammoe
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec City, QC G1V 0A6, Canada.
| |
Collapse
|
2
|
Biringer RG. Migraine signaling pathways: purine metabolites that regulate migraine and predispose migraineurs to headache. Mol Cell Biochem 2023; 478:2813-2848. [PMID: 36947357 DOI: 10.1007/s11010-023-04701-7] [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: 10/05/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
Migraine is a debilitating disorder that afflicts over 1 billion people worldwide, involving attacks that result in a throbbing and pulsating headache. Migraine is thought to be a neurovascular event associated with vasoconstriction, vasodilation, and neuronal activation. Understanding signaling in migraine pathology is central to the development of therapeutics for migraine prophylaxis and for mitigation of migraine in the prodrome phase before pain sets in. The fact that both vasoactivity and neural sensitization are involved in migraine indicates that agonists which promote these phenomena may very well be involved in migraine pathology. One such group of agonists is the purines, in particular, adenosine phosphates and their metabolites. This manuscript explores what is known about the relationship between these metabolites and migraine pathology and explores the potential for such relationships through their known signaling pathways. Reported receptor involvement in vasoaction and nociception.
Collapse
Affiliation(s)
- Roger Gregory Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
3
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [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: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
4
|
Ernst C, Bek T. Differential effects of purines and prostaglandins on hypoxia induced dilatation of porcine retinal vessels at different branching level ex vivo. Exp Eye Res 2023; 234:109584. [PMID: 37460082 DOI: 10.1016/j.exer.2023.109584] [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: 12/25/2022] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
The metabolic pathways leading from hypoxia to retinal vasodilatation can involve effects of both purines and prostaglandins, but the effects of these compounds at different vascular branching levels are unknown. The purpose of the present study was to investigate differential effects of purines and prostaglandins in hypoxia-induced dilatation of retinal arterioles, precapillary arterioles and capillaries ex vivo. Porcine hemiretinas were mounted in a tissue chamber while monitoring temperature, pH, and oxygen tension. The effect of hypoxia on the diameter of larger arterioles, precapillary arterioles and capillaries was studied in the presence of the ecto-nucleotidase inhibitor AOPCP, the nonselective P2 purinoreceptor antagonist PPADS, the A2B adenosine receptor antagonist MRS 1754, the A3 adenosine receptor antagonist MRS 1523, the EP1 receptor antagonist SC-19220, the EP2 receptor antagonist PF-04418948, the EP3 receptor antagonist L-798,106, the EP4 receptor antagonist L-161-982, the prostaglandin synthesis inhibitor ibuprofen, and ibuprofen combined with AOPCP or ATP. Hypoxia-induced dilatation in arterioles was reduced by the A2B adenosine receptor antagonist (p < 0.01) and increased by the EP2 and the EP3 receptor antagonists (p < 0.01 for both comparisons). In precapillary arterioles the dilatation was reduced by the EP2 receptor antagonist (p < 0.04) and increased by the EP1 receptor antagonist (p < 0.03), whereas in capillaries the dilatation was increased by both the A3 adenosine receptor antagonist (p < 0.01), by ibuprofen in combination with the unspecific ecto-nucleotidase inhibitor AOPCP (p = 0.04) and by the prostaglandin EP3 receptor antagonist. Hypoxia-induced dilatation of retinal vessels is influenced by adenosine A2B and A3 receptors, and by the prostaglandin EP1, EP2 and EP3 receptors. The effects mediated by these receptors differ at different branching levels of the resistance vessels.
Collapse
Affiliation(s)
- Charlotte Ernst
- Department of Ophthalmology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark.
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark
| |
Collapse
|
5
|
du Toit WL, Kruger R, Gafane-Matemane LF, Schutte AE, Louw R, Mels CMC. Markers of arterial stiffness and urinary metabolomics in young adults with early cardiovascular risk: the African-PREDICT study. Metabolomics 2023; 19:28. [PMID: 36988718 PMCID: PMC10060307 DOI: 10.1007/s11306-023-01987-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 03/04/2023] [Indexed: 03/30/2023]
Abstract
INTRODUCTION Increased exposure to risk factors in the young and healthy contributes to arterial changes, which may be accompanied by an altered metabolism. OBJECTIVES To increase our understanding of early metabolic alterations and how they associate with markers of arterial stiffness, we profiled urinary metabolites in young adults with cardiovascular disease (CVD) risk factor(s) and in a control group without CVD risk factors. METHODS We included healthy black and white women and men (N = 1202), aged 20-30 years with a detailed CVD risk factor profile, reflecting obesity, physical inactivity, smoking, excessive alcohol intake, masked hypertension, hyperglycemia, dyslipidemia and low socio-economic status, forming the CVD risk group (N = 1036) and the control group (N = 166). Markers of arterial stiffness, central systolic blood pressure (BP) and pulse wave velocity were measured. A targeted metabolomics approach was followed by measuring amino acids and acylcarnitines using a liquid chromatography-tandem mass spectrometry method. RESULTS In the CVD risk group, central systolic BP (adjusted for age, sex, ethnicity) was negatively associated with histidine, arginine, asparagine, serine, glutamine, dimethylglycine, threonine, GABA, proline, methionine, pyroglutamic acid, aspartic acid, glutamic acid, branched chain amino acids (BCAAs) and butyrylcarnitine (all P ≤ 0.048). In the same group, pulse wave velocity (adjusted for age, sex, ethnicity, mean arterial pressure) was negatively associated with histidine, lysine, threonine, 2-aminoadipic acid, BCAAs and aromatic amino acids (AAAs) (all P ≤ 0.044). In the control group, central systolic BP was negatively associated with pyroglutamic acid, glutamic acid and dodecanoylcarnitine (all P ≤ 0.033). CONCLUSION In a group with increased CVD risk, markers of arterial stiffness were negatively associated with metabolites related to AAA and BCAA as well as energy metabolism and oxidative stress. Our findings may suggest that metabolic adaptations may be at play in response to increased CVD risk to maintain cardiovascular integrity.
Collapse
Affiliation(s)
- Wessel L du Toit
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Lebo F Gafane-Matemane
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
- School of Population Health, University of New South Wales, Sydney, Australia
- The George Institute for Global Health, Sydney, Australia
| | - Roan Louw
- Human Metabolomics, North-West University, Potchefstroom Campus, Potchefstroom, South Africa
| | - Catharina M C Mels
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
6
|
Scharbarg E, Walter A, Lecoin L, Gallopin T, Lemaître F, Guille-Collignon M, Rouach N, Rancillac A. Prostaglandin D 2 Controls Local Blood Flow and Sleep-Promoting Neurons in the VLPO via Astrocyte-Derived Adenosine. ACS Chem Neurosci 2023; 14:1063-1070. [PMID: 36847485 DOI: 10.1021/acschemneuro.2c00660] [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: 03/01/2023] Open
Abstract
Prostaglandin D2 (PGD2) is one of the most potent endogenous sleep-promoting molecules. However, the cellular and molecular mechanisms of the PGD2-induced activation of sleep-promoting neurons in the ventrolateral preoptic nucleus (VLPO), the major nonrapid eye movement (NREM)-sleep center, still remains unclear. We here show that PGD2 receptors (DP1) are not only expressed in the leptomeninges but also in astrocytes from the VLPO. We further demonstrate, by performing real-time measurements of extracellular adenosine using purine enzymatic biosensors in the VLPO, that PGD2 application causes a 40% increase in adenosine level, via an astroglial release. Measurements of vasodilatory responses and electrophysiological recordings finally reveal that, in response to PGD2 application, adenosine release induces an A2AR-mediated dilatation of blood vessels and activation of VLPO sleep-promoting neurons. Altogether, our results unravel the PGD2 signaling pathway in the VLPO, controlling local blood flow and sleep-promoting neurons, via astrocyte-derived adenosine.
Collapse
Affiliation(s)
- Emeric Scharbarg
- Brain Plasticity Unit, CNRS, ESPCI-ParisTech, Labex Memolife, Université PSL, 75005 Paris, France
| | - Augustin Walter
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 75005 Paris, France
| | - Laure Lecoin
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 75005 Paris, France
| | - Thierry Gallopin
- Brain Plasticity Unit, CNRS, ESPCI-ParisTech, Labex Memolife, Université PSL, 75005 Paris, France
| | - Frédéric Lemaître
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University PSL, Sorbonne Université, CNRS, 75005 Paris, France
| | - Manon Guille-Collignon
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL University PSL, Sorbonne Université, CNRS, 75005 Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 75005 Paris, France
| | - Armelle Rancillac
- Brain Plasticity Unit, CNRS, ESPCI-ParisTech, Labex Memolife, Université PSL, 75005 Paris, France.,Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 75005 Paris, France
| |
Collapse
|
7
|
Zhuang Y, Yu ML, Lu SF. Purinergic signaling in myocardial ischemia-reperfusion injury. Purinergic Signal 2023; 19:229-243. [PMID: 35254594 PMCID: PMC9984618 DOI: 10.1007/s11302-022-09856-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/18/2022] [Indexed: 10/18/2022] Open
Abstract
Purines and their derivatives, extensively distributed in the body, act as a class of extracellular signaling molecules via a rich array of receptors, also known as purinoceptors (P1, P2X, and P2Y). They mediate multiple intracellular signal transduction pathways and participate in various physiological and pathological cell behaviors. Since the function in myocardial ischemia-reperfusion injury (MIRI), this review summarized the involvement of purinergic signal transduction in diversified pathological processes, including energy metabolism disorder, oxidative stress injury, calcium overload, inflammatory immune response, platelet aggregation, coronary vascular dysfunction, and cell necrosis and apoptosis. Moreover, increasing evidence suggests that purinergic signaling also mediates the prevention and treatment of MIRI, such as ischemic conditioning, pharmacological intervention, and some other therapies. In conclusion, this review exhibited that purinergic signaling mediates the complex processes of MIRI which shows its promising application and prospecting in the future.
Collapse
Affiliation(s)
- Yi Zhuang
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Mei-Ling Yu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China
| | - Sheng-Feng Lu
- College of Acupuncture and Tuina, Nanjing University of Chinese Medicine, 138 Xian-lin Avenue, Qixia District, Nanjing, 210023, Jiangsu Province, China. .,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
8
|
Moritz CEJ, Vieira AF, de Melo-Marins D, Figueiró F, Battastini AMO, Reischak-Oliveira A. Effects of physical exercise on the functionality of human nucleotidases: A systematic review. Physiol Rep 2022; 10:e15464. [PMID: 36117383 PMCID: PMC9483616 DOI: 10.14814/phy2.15464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023] Open
Abstract
Nucleotidases contribute to the regulation of inflammation, coagulation, and cardiovascular activity. Exercise promotes biological adaptations, but its effects on nucleotidase activities and expression are unclear. The objective of this study was to review systematically the effects of exercise on nucleotidase functionality in healthy and unhealthy subjects. The MEDLINE, EMBASE, Cochrane Library, and Web of Science databases were searched to identify, randomized clinical trials, non-randomized clinical trials, uncontrolled clinical trials, quasi-experimental, pre-, and post-interventional studies that evaluated the effects of exercise on nucleotidases in humans, and was not limited by language and date. Two independent reviewers performed the study selection, data extraction, and assessment of risk of bias. Of the 203 articles identified, 12 were included in this review. Eight studies reported that acute exercise, in healthy and unhealthy subjects, elevated the activities or expression of nucleotidases. Four studies evaluated the effects of chronic training on nucleotidase activities in the platelets and lymphocytes of patients with metabolic syndrome, chronic kidney disease, and hypertension and found a decrease in nucleotidase activities in these conditions. Acute and chronic exercise was able to modify the blood plasma and serum levels of nucleotides and nucleosides. Our results suggest that short- and long-term exercise modulate nucleotidase functionality. As such, purinergic signaling may represent a novel molecular adaptation in inflammatory, thrombotic, and vascular responses to exercise.
Collapse
Affiliation(s)
- Cesar Eduardo Jacintho Moritz
- Programa de Pós-Graduação em Ciências do Movimento Humano, Escola de Educação Física, Fisioterapia e Dança (ESEFID), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Alexandra Ferreira Vieira
- Programa de Pós-Graduação em Ciências do Movimento Humano, Escola de Educação Física, Fisioterapia e Dança (ESEFID), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Denise de Melo-Marins
- Programa de Pós-Graduação em Ciências do Movimento Humano, Escola de Educação Física, Fisioterapia e Dança (ESEFID), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Fabrício Figueiró
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Departamento do Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Departamento do Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Alvaro Reischak-Oliveira
- Programa de Pós-Graduação em Ciências do Movimento Humano, Escola de Educação Física, Fisioterapia e Dança (ESEFID), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
9
|
The purinergic signalling and inflammation in the pathogenesis and progression of diabetes: key factors and therapeutic targets. Inflamm Res 2022; 71:759-770. [PMID: 35648156 DOI: 10.1007/s00011-022-01587-x] [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: 02/23/2022] [Accepted: 05/10/2022] [Indexed: 11/27/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an important chronic disease around the world, and according to the World Health Organization, it is the 9th principal cause of global death. This pathology is characterized by high levels of circulating glucose as a result of insulin resistance, and it is well stated that inflammation related to obesity is directly associated with the development of the disease. The purinergic signalling is involved in both pancreatic destruction, which impairs insulin secretion, and the cytokine production that favors insulin resistance in T2DM. In this review, the purinergic signalling aspects will be discussed, showing the impact of the enzymes, nucleotides, nucleosides, and receptors of this system and the cytokines that result in inflammation, in the development and progression of T2DM, besides, pointing the purinergic receptors as a possible therapeutic approach.
Collapse
|
10
|
Erdling A, Johansson SE, Radziwon‐Balicka A, Ansar S, Edvinsson L. Changes in P2Y 6 receptor-mediated vasoreactivity following focal and global ischemia. Physiol Rep 2022; 10:e15283. [PMID: 35466569 PMCID: PMC9035753 DOI: 10.14814/phy2.15283] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023] Open
Abstract
Ischemia, both in the form of focal thromboembolic stroke and following subarachnoid hemorrhage (SAH), causes upregulation of vasoconstrictive receptor systems within the cerebral vasculature. Descriptions regarding changes in purinergic signaling following ischemia are lacking, especially when the importance of purinergic signaling in regulating vascular tone is taken into consideration. This prompted us to evaluate changes in P2Y6 -mediated vasomotor reactivity in two different stroke models in rat. We used wire myography to measure changes in cerebral vasoreactivity to the P2Y6 agonist UDP-β-S following either experimental SAH or transient middle cerebral artery occlusion. Changes in receptor localization or receptor expression were evaluated using immunohistochemistry and quantitative flow cytometry. Transient middle cerebral artery occlusion caused an increase in Emax when compared to sham (233.6 [206.1-258.5]% vs. 161.1 [147.1-242.6]%, p = 0.0365). No such change was seen following SAH. Both stroke models were associated with increased levels of P2Y6 receptor expression in the vascular smooth muscle cells (90.94 [86.99-99.15]% and 93.79 [89.96-96.39]% vs. 80.31 [70.80-80.86]%, p = 0.021) and p = 0.039 respectively. There was no change in receptor localization in either of the stroke models. Based on these findings, we conclude that focal ischemic stroke increases vascular sensitivity to UDP-β-S by upregulating P2Y6 receptors on vascular smooth muscle cells while experimental SAH did not induce changes in vasoreactivity in spite of increased P2Y6 receptor expression.
Collapse
Affiliation(s)
- André Erdling
- Department of Clinical SciencesDivision of Experimental Vascular ResearchLund UniversityLundSweden
- Department of Cardiothoracic Surgery, Anesthesiology and Intensive CareSkane University HospitalLundSweden
- Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Sara Ellinor Johansson
- Department of Clinical Experimental ResearchGlostrup Research InstituteRigshospitalet‐GlostrupGlostrupDenmark
| | - Aneta Radziwon‐Balicka
- Department of Clinical Experimental ResearchGlostrup Research InstituteRigshospitalet‐GlostrupGlostrupDenmark
| | - Saema Ansar
- Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Lars Edvinsson
- Department of Clinical SciencesDivision of Experimental Vascular ResearchLund UniversityLundSweden
- Department of Clinical Experimental ResearchGlostrup Research InstituteRigshospitalet‐GlostrupGlostrupDenmark
| |
Collapse
|
11
|
Hoel F, Hoel A, Pettersen IK, Rekeland IG, Risa K, Alme K, Sørland K, Fosså A, Lien K, Herder I, Thürmer HL, Gotaas ME, Schäfer C, Berge RK, Sommerfelt K, Marti HP, Dahl O, Mella O, Fluge Ø, Tronstad KJ. A map of metabolic phenotypes in patients with myalgic encephalomyelitis/chronic fatigue syndrome. JCI Insight 2021; 6:e149217. [PMID: 34423789 PMCID: PMC8409979 DOI: 10.1172/jci.insight.149217] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating disease usually presenting after infection. Emerging evidence supports that energy metabolism is affected in ME/CFS, but a unifying metabolic phenotype has not been firmly established. We performed global metabolomics, lipidomics, and hormone measurements, and we used exploratory data analyses to compare serum from 83 patients with ME/CFS and 35 healthy controls. Some changes were common in the patient group, and these were compatible with effects of elevated energy strain and altered utilization of fatty acids and amino acids as catabolic fuels. In addition, a set of heterogeneous effects reflected specific changes in 3 subsets of patients, and 2 of these expressed characteristic contexts of deregulated energy metabolism. The biological relevance of these metabolic phenotypes (metabotypes) was supported by clinical data and independent blood analyses. In summary, we report a map of common and context-dependent metabolic changes in ME/CFS, and some of them presented possible associations with clinical patient profiles. We suggest that elevated energy strain may result from exertion-triggered tissue hypoxia and lead to systemic metabolic adaptation and compensation. Through various mechanisms, such metabolic dysfunction represents a likely mediator of key symptoms in ME/CFS and possibly a target for supportive intervention.
Collapse
Affiliation(s)
| | - August Hoel
- Department of Biomedicine and.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Ingrid G Rekeland
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Kristin Risa
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Kine Alme
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Kari Sørland
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Alexander Fosså
- Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,KJ Jebsen Centre for B-cell malignancies, University of Oslo, Oslo, Norway
| | - Katarina Lien
- CFS/ME Center, Division of Medicine, Oslo University Hospital, Oslo, Norway
| | - Ingrid Herder
- CFS/ME Center, Division of Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Merete E Gotaas
- Department of Pain and Complex Disorders, St. Olav's Hospital, Trondheim, Norway
| | - Christoph Schäfer
- Department of Rehabilitation Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristian Sommerfelt
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Pediatrics and
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Olav Dahl
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Olav Mella
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Øystein Fluge
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | | |
Collapse
|
12
|
Effects of Dual Purinoceptor-dependent Approach on Release of Vascular Endothelial Growth Factor From Human Microvascular Endothelial Cell (HMEC-1) and Endothelial Cell Condition. J Cardiovasc Pharmacol 2021; 76:349-359. [PMID: 32569015 DOI: 10.1097/fjc.0000000000000866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the recent years, the awareness of the role purinergic signaling plays as a therapeutic target has increased considerably. The purinoceptor allows the action of extracellular nucleotides (P2 receptors) and intermediary products of their metabolism, such as adenosine (P1 receptors), regulating pivotal processes occurring in the cardiovascular system. This study focuses on a dual purinoreceptor-dependent approach, based on the activation of adenosine P1 receptors with the simultaneous inhibition of P2Y12 receptors that can be used as novel platelet inhibitors in antithrombotic therapy. Endothelial cells are directly exposed to the drugs circulating in the bloodstream. That is why effects of our concept on human microvascular endothelial cells (HMEC-1) were examined in in vitro studies, such as enzyme-linked immunosorbent assay and scratch assays. In response to adenosine receptor agonists, levels of secreted vascular endothelial growth factor varied. Two of them, 5'-N-ethylcarboxamidoadenosine and MRE0094 remarkably increased vascular endothelial growth factor release. The elevated levels were reduced when used together with the P2Y12 receptor antagonist. Also, rates of wound closure in a scratch assay were significantly reduced in these cases. The results suggest that the proposed treatment does not impair endothelial cell condition. In addition, it is suggested as a collateral benefit, namely solving the problem of excessive activation of endothelial cells during antiplatelet therapy.
Collapse
|
13
|
Reichert KP, Castro MFV, Assmann CE, Bottari NB, Miron VV, Cardoso A, Stefanello N, Morsch VMM, Schetinger MRC. Diabetes and hypertension: Pivotal involvement of purinergic signaling. Biomed Pharmacother 2021; 137:111273. [PMID: 33524787 PMCID: PMC7846467 DOI: 10.1016/j.biopha.2021.111273] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/11/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus (DM) and hypertension are highly prevalent worldwide health problems and frequently associated with severe clinical complications, such as diabetic cardiomyopathy, nephropathy, retinopathy, neuropathy, stroke, and cardiac arrhythmia, among others. Despite all existing research results and reasonable speculations, knowledge about the role of purinergic system in individuals with DM and hypertension remains restricted. Purinergic signaling accounts for a complex network of receptors and extracellular enzymes responsible for the recognition and degradation of extracellular nucleotides and adenosine. The main components of this system that will be presented in this review are: P1 and P2 receptors and the enzymatic cascade composed by CD39 (NTPDase; with ATP and ADP as a substrate), CD73 (5'-nucleotidase; with AMP as a substrate), and adenosine deaminase (ADA; with adenosine as a substrate). The purinergic system has recently emerged as a central player in several physiopathological conditions, particularly those linked to inflammatory responses such as diabetes and hypertension. Therefore, the present review focuses on changes in both purinergic P1 and P2 receptor expression as well as the activities of CD39, CD73, and ADA in diabetes and hypertension conditions. It can be postulated that the manipulation of the purinergic axis at different levels can prevent or exacerbate the insurgency and evolution of diabetes and hypertension working as a compensatory mechanism.
Collapse
Affiliation(s)
- Karine Paula Reichert
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Milagros Fanny Vera Castro
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Charles Elias Assmann
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Nathieli Bianchin Bottari
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vanessa Valéria Miron
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Andréia Cardoso
- Academic Coordination, Medicine, Campus Chapecó, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Naiara Stefanello
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vera Maria Melchiors Morsch
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Rosa Chitolina Schetinger
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
14
|
Molecular Dambusters: What Is Behind Hyperpermeability in Bradykinin-Mediated Angioedema? Clin Rev Allergy Immunol 2021; 60:318-347. [PMID: 33725263 PMCID: PMC7962090 DOI: 10.1007/s12016-021-08851-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2021] [Indexed: 02/08/2023]
Abstract
In the last few decades, a substantial body of evidence underlined the pivotal role of bradykinin in certain types of angioedema. The formation and breakdown of bradykinin has been studied thoroughly; however, numerous questions remained open regarding the triggering, course, and termination of angioedema attacks. Recently, it became clear that vascular endothelial cells have an integrative role in the regulation of vessel permeability. Apart from bradykinin, a great number of factors of different origin, structure, and mechanism of action are capable of modifying the integrity of vascular endothelium, and thus, may participate in the regulation of angioedema formation. Our aim in this review is to describe the most important permeability factors and the molecular mechanisms how they act on endothelial cells. Based on endothelial cell function, we also attempt to explain some of the challenging findings regarding bradykinin-mediated angioedema, where the function of bradykinin itself cannot account for the pathophysiology. By deciphering the complex scenario of vascular permeability regulation and edema formation, we may gain better scientific tools to be able to predict and treat not only bradykinin-mediated but other types of angioedema as well.
Collapse
|
15
|
Milne K, Ivens A, Reid AJ, Lotkowska ME, O'Toole A, Sankaranarayanan G, Munoz Sandoval D, Nahrendorf W, Regnault C, Edwards NJ, Silk SE, Payne RO, Minassian AM, Venkatraman N, Sanders MJ, Hill AVS, Barrett M, Berriman M, Draper SJ, Rowe JA, Spence PJ. Mapping immune variation and var gene switching in naive hosts infected with Plasmodium falciparum. eLife 2021; 10:e62800. [PMID: 33648633 PMCID: PMC7924948 DOI: 10.7554/elife.62800] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Falciparum malaria is clinically heterogeneous and the relative contribution of parasite and host in shaping disease severity remains unclear. We explored the interaction between inflammation and parasite variant surface antigen (VSA) expression, asking whether this relationship underpins the variation observed in controlled human malaria infection (CHMI). We uncovered marked heterogeneity in the host response to blood challenge; some volunteers remained quiescent, others triggered interferon-stimulated inflammation and some showed transcriptional evidence of myeloid cell suppression. Significantly, only inflammatory volunteers experienced hallmark symptoms of malaria. When we tracked temporal changes in parasite VSA expression to ask whether variants associated with severe disease rapidly expand in naive hosts, we found no transcriptional evidence to support this hypothesis. These data indicate that parasite variants that dominate severe malaria do not have an intrinsic growth or survival advantage; instead, they presumably rely upon infection-induced changes in their within-host environment for selection.
Collapse
Affiliation(s)
- Kathryn Milne
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
| | - Adam J Reid
- Wellcome Sanger InstituteCambridgeUnited Kingdom
| | | | - Aine O'Toole
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
- Institute of Evolutionary Biology, University of EdinburghEdinburghUnited Kingdom
| | | | - Diana Munoz Sandoval
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Instituto de Microbiologia, Universidad San Francisco de QuitoQuitoEcuador
| | - Wiebke Nahrendorf
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
| | - Clement Regnault
- Wellcome Centre for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Glasgow Polyomics, University of GlasgowGlasgowUnited Kingdom
| | - Nick J Edwards
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - Sarah E Silk
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - Ruth O Payne
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | | | | | | | - Adrian VS Hill
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - Michael Barrett
- Wellcome Centre for Integrative Parasitology, University of GlasgowGlasgowUnited Kingdom
- Glasgow Polyomics, University of GlasgowGlasgowUnited Kingdom
| | | | - Simon J Draper
- The Jenner Institute, University of OxfordOxfordUnited Kingdom
| | - J Alexandra Rowe
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
| | - Philip J Spence
- Institute of Immunology and Infection Research, University of EdinburghEdinburghUnited Kingdom
- Centre for Immunity, Infection and Evolution, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
16
|
Thakore P, Alvarado MG, Ali S, Mughal A, Pires PW, Yamasaki E, Pritchard HA, Isakson BE, Tran CHT, Earley S. Brain endothelial cell TRPA1 channels initiate neurovascular coupling. eLife 2021; 10:63040. [PMID: 33635784 PMCID: PMC7935492 DOI: 10.7554/elife.63040] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cerebral blood flow is dynamically regulated by neurovascular coupling to meet the dynamic metabolic demands of the brain. We hypothesized that TRPA1 channels in capillary endothelial cells are stimulated by neuronal activity and instigate a propagating retrograde signal that dilates upstream parenchymal arterioles to initiate functional hyperemia. We find that activation of TRPA1 in capillary beds and post-arteriole transitional segments with mural cell coverage initiates retrograde signals that dilate upstream arterioles. These signals exhibit a unique mode of biphasic propagation. Slow, short-range intercellular Ca2+ signals in the capillary network are converted to rapid electrical signals in transitional segments that propagate to and dilate upstream arterioles. We further demonstrate that TRPA1 is necessary for functional hyperemia and neurovascular coupling within the somatosensory cortex of mice in vivo. These data establish endothelial cell TRPA1 channels as neuronal activity sensors that initiate microvascular vasodilatory responses to redirect blood to regions of metabolic demand.
Collapse
Affiliation(s)
- Pratish Thakore
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - Michael G Alvarado
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - Sher Ali
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - Amreen Mughal
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, United States
| | - Paulo W Pires
- Department of Physiology, College of Medicine, University of Arizona, Tucson, United States
| | - Evan Yamasaki
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - Harry At Pritchard
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States.,Institute of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, United States
| | - Cam Ha T Tran
- Department of Physiology & Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| | - Scott Earley
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, United States
| |
Collapse
|
17
|
Purinergic Regulation of Endothelial Barrier Function. Int J Mol Sci 2021; 22:ijms22031207. [PMID: 33530557 PMCID: PMC7865261 DOI: 10.3390/ijms22031207] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Increased vascular permeability is a hallmark of several cardiovascular anomalies, including ischaemia/reperfusion injury and inflammation. During both ischaemia/reperfusion and inflammation, massive amounts of various nucleotides, particularly adenosine 5'-triphosphate (ATP) and adenosine, are released that can induce a plethora of signalling pathways via activation of several purinergic receptors and may affect endothelial barrier properties. The nature of the effects on endothelial barrier function may depend on the prevalence and type of purinergic receptors activated in a particular tissue. In this review, we discuss the influence of the activation of various purinergic receptors and downstream signalling pathways on vascular permeability during pathological conditions.
Collapse
|
18
|
Segura-Collar B, Mata-Martínez P, Hernández-Laín A, Sánchez-Gómez P, Gargini R. Blood-Brain Barrier Disruption: A Common Driver of Central Nervous System Diseases. Neuroscientist 2021; 28:222-237. [PMID: 33446074 DOI: 10.1177/1073858420985838] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The brain is endowed with a unique cellular composition and organization, embedded within a vascular network and isolated from the circulating blood by a specialized frontier, the so-called blood-brain barrier (BBB), which is necessary for its proper function. Recent reports have shown that increments in the permeability of the blood vessels facilitates the entry of toxic components and immune cells to the brain parenchyma and alters the phenotype of the supporting astrocytes. All of these might contribute to the progression of different pathologies such as brain cancers or neurodegenerative diseases. Although it is well known that BBB breakdown occurs due to pericyte malfunctioning or to the lack of stability of the blood vessels, its participation in the diverse neural diseases needs further elucidation. This review summarizes what it is known about BBB structure and function and how its instability might trigger or promote neuronal degeneration and glioma progression, with a special focus on the role of pericytes as key modulators of the vasculature. Moreover, we will discuss some recent reports that highlights the participation of the BBB alterations in glioma growth. This pan-disease analysis might shed some light into these otherwise untreatable diseases and help to design better therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Ricardo Gargini
- Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
| |
Collapse
|
19
|
Yegutkin GG. Adenosine metabolism in the vascular system. Biochem Pharmacol 2020; 187:114373. [PMID: 33340515 DOI: 10.1016/j.bcp.2020.114373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
The concept of extracellular purinergic signaling was first proposed by Geoffrey Burnstock in the early 1970s. Since then, extracellular ATP and its metabolites ADP and adenosine have attracted an enormous amount of attention in terms of their involvement in a wide range of immunomodulatory, thromboregulatory, angiogenic, vasoactive and other pathophysiological activities in different organs and tissues, including the vascular system. In addition to significant progress in understanding the properties of nucleotide- and adenosine-selective receptors, recent studies have begun to uncover the complexity of regulatory mechanisms governing the duration and magnitude of the purinergic signaling cascade. This knowledge has led to the development of new paradigms in understanding the entire purinome by taking into account the multitude of signaling and metabolic pathways involved in biological effects of ATP and adenosine and compartmentalization of the adenosine system. Along with the "canonical route" of ATP breakdown to adenosine via sequential ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39) and ecto-5'-nucleotidase/CD73 activities, it has now become clear that purine metabolism is the result of concerted effort between ATP release, its metabolism through redundant nucleotide-inactivating and counteracting ATP-regenerating ectoenzymatic pathways, as well as cellular nucleoside uptake and phosphorylation of adenosine to ATP through complex phosphotransfer reactions. In this review I provide an overview of key enzymes involved in adenosine metabolic network, with special emphasis on the emerging roles of purine-converting ectoenzymes as novel targets for cancer and vascular therapies.
Collapse
|
20
|
Kreft E, Sałaga-Zaleska K, Sakowicz-Burkiewicz M, Dąbkowski K, Szczepánska-Konkel M, Jankowski M. Diabetes Affects the A1 Adenosine Receptor-Dependent Action of Diadenosine Tetraphosphate (Ap4A) on Cortical and Medullary Renal Blood Flow. J Vasc Res 2020; 58:38-48. [PMID: 33207336 DOI: 10.1159/000511461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 09/07/2020] [Indexed: 11/19/2022] Open
Abstract
Diabetes through adenosine A1 receptor (A1R) and P2 receptors (P2Rs) may lead to disturbances in renal microvasculature. We investigated the renal microvascular response to Ap4A, an agonist of P2Rs, in streptozotocin-induced diabetic rats. Using laser Doppler flowmetry, renal blood perfusion (RBP) was measured during infusion of Ap4A alone or in the presence of A1R antagonist, either DPCPX (8-cyclopentyl-1,3-dipropylxanthine) or 8-cyclopentyltheophylline (CPT). Ap4A induced a biphasic response in RBP: a phase of rapid decrease was followed by a rapid increase, which was transient in diabetic rats but extended for 30 min in nondiabetic rats. Phase of decreased RBP was not affected by DPCPX or CPT in either group. Early and extended increases in RBP were prevented by DPCPX and CPT in nondiabetic rats, while in diabetic rats, the early increase in RBP was not affected by these antagonists. A1R mRNA and protein levels were increased in isolated glomeruli of diabetic rats, but no changes were detected in P2Y1R and P2Y2R mRNA. Presence of unblocked A1R is a prerequisite for the P2R-mediated relaxing effect of Ap4A in nondiabetic conditions, but influence of A1R on P2R-mediated renal vasorelaxation is abolished under diabetic conditions.
Collapse
Affiliation(s)
- Ewelina Kreft
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Kamil Dąbkowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Maciej Jankowski
- Department of Clinical Chemistry, Medical University of Gdańsk, Gdańsk, Poland,
| |
Collapse
|
21
|
Adenosine and ATPγS protect against bacterial pneumonia-induced acute lung injury. Sci Rep 2020; 10:18078. [PMID: 33093565 PMCID: PMC7581771 DOI: 10.1038/s41598-020-75224-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Lipopolysaccharide (LPS), a component of the outer membrane of gram-negative bacteria, disrupts the alveolar-capillary barrier, triggering pulmonary vascular leak thus inducing acute lung injury (ALI). Extracellular purines, adenosine and ATP, protected against ALI induced by purified LPS. In this study, we investigated whether these purines can impact vascular injury in more clinically-relevant E.coli (non-sterile LPS) murine ALI model. Mice were inoculated with live E. coli intratracheally (i.t.) with or without adenosine or a non-hydrolyzable ATP analog, adenosine 5'-(γ-thio)-triphosphate (ATPγS) added intravenously (i.v.). After 24 h of E. coli treatment, we found that injections of either adenosine or ATPγS 15 min prior or adenosine 3 h after E.coli insult significantly attenuated the E.coli-mediated increase in inflammatory responses. Furthermore, adenosine prevented weight loss, tachycardia, and compromised lung function in E. coli-exposed mice. Accordingly, treatment with adenosine or ATPγS increased oxygen saturation and reduced histopathological signs of lung injury in mice exposed to E. coli. Lastly, lung-targeting gene delivery of adenosine or ATPγS downstream effector, myosin phosphatase, significantly attenuated the E. coli-induced compromise of lung function. Collectively, our study has demonstrated that adenosine or ATPγS mitigates E. coli-induced ALI in mice and may be useful as an adjuvant therapy in future pre-clinical studies.
Collapse
|
22
|
Kumar G, Dey SK, Kundu S. Functional implications of vascular endothelium in regulation of endothelial nitric oxide synthesis to control blood pressure and cardiac functions. Life Sci 2020; 259:118377. [PMID: 32898526 DOI: 10.1016/j.lfs.2020.118377] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 11/29/2022]
Abstract
The endothelium is the innermost vascular lining performing significant roles all over the human body while maintaining the blood pressure at physiological levels. Malfunction of endothelium is thus recognized as a biomarker linked with many vascular diseases including but not limited to atherosclerosis, hypertension and thrombosis. Alternatively, prevention of endothelial malfunctioning or regulating the functions of its associated physiological partners like endothelial nitric oxide synthase can prevent the associated vascular disorders which account for the highest death toll worldwide. While many anti-hypertensive drugs are available commercially, a comprehensive description of the key physiological roles of the endothelium and its regulation by endothelial nitric oxide synthase or vice versa is the need of the hour to understand its contribution in vascular homeostasis. This, in turn, will help in designing new therapeutics targeting endothelial nitric oxide synthase or its interacting partners present in the cellular pool. This review describes the central role of vascular endothelium in the regulation of endothelial nitric oxide synthase while outlining the emerging drug targets present in the vasculature with potential to treat vascular disorders including hypertension.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India
| | - Sanjay Kumar Dey
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India; Center for Advanced Biotechnology and Medicine, Rutgers University, NJ 08854, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi, South Campus, New Delhi 110021, India.
| |
Collapse
|
23
|
Abstract
Pulmonary arterial hypertension (PAH) is a life‐threatening disease characterized by increased pulmonary arterial pressure and pulmonary vascular resistance, which result in an increase in afterload imposed onto the right ventricle, leading to right heart failure. Current therapies are incapable of reversing the disease progression. Thus, the identification of novel and potential therapeutic targets is urgently needed. An alteration of nucleotide‐ and nucleoside‐activated purinergic signaling has been proposed as a potential contributor in the pathogenesis of PAH. Adenosine‐mediated purinergic 1 receptor activation, particularly A2AR activation, reduces pulmonary vascular resistance and attenuates pulmonary vascular remodeling and right ventricle hypertrophy, thereby exerting a protective effect. Conversely, A2BR activation induces pulmonary vascular remodeling, and is therefore deleterious. ATP‐mediated P2X7R activation and ADP‐mediated activation of P2Y1R and P2Y12R play a role in pulmonary vascular tone, vascular remodeling, and inflammation in PAH. Recent studies have revealed a role of ectonucleotidase nucleoside triphosphate diphosphohydrolase, that degrades ATP/ADP, in regulation of pulmonary vascular remodeling. Interestingly, existing evidence that adenosine activates erythrocyte A2BR signaling, counteracting hypoxia‐induced pulmonary injury, and that ATP release is impaired in erythrocyte in PAH implies erythrocyte dysfunction as an important trigger to affect purinergic signaling for pathogenesis of PAH. The present review focuses on current knowledge on alteration of nucleot(s)ide‐mediated purinergic signaling as a potential disease mechanism underlying the development of PAH.
Collapse
Affiliation(s)
- Zongye Cai
- Division of Experimental Cardiology Department of Cardiology Erasmus MCUniversity Medical Center Rotterdam Rotterdam the Netherlands
| | - Ly Tu
- INSERM UMR_S 999Hôpital Marie Lannelongue Le Plessis-Robinson France.,School of Medicine Université Paris-Saclay Kremlin-Bicêtre France
| | - Christophe Guignabert
- INSERM UMR_S 999Hôpital Marie Lannelongue Le Plessis-Robinson France.,School of Medicine Université Paris-Saclay Kremlin-Bicêtre France
| | - Daphne Merkus
- Division of Experimental Cardiology Department of Cardiology Erasmus MCUniversity Medical Center Rotterdam Rotterdam the Netherlands.,Walter Brendel Center of Experimental Medicine LMU Munich Munich Germany.,German Center for Cardiovascular Research, Partner Site MunichMunich Heart Alliance Munich Germany
| | - Zhichao Zhou
- Division of Cardiology Department of Medicine Karolinska University HospitalKarolinska Institutet Stockholm Sweden
| |
Collapse
|
24
|
Xavier FE. Nitrergic perivascular innervation in health and diseases: Focus on vascular tone regulation. Acta Physiol (Oxf) 2020; 230:e13484. [PMID: 32336027 DOI: 10.1111/apha.13484] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
For a long time, the vascular tone was considered to be regulated exclusively by tonic innervation of vasoconstrictor adrenergic nerves. However, accumulating experimental evidence has revealed the existence of nerves mediating vasodilatation, including perivascular nitrergic nerves (PNN), in a wide variety of mammalian species. Functioning of nitrergic vasodilator nerves is evidenced in several territories, including cerebral, mesenteric, pulmonary, renal, penile, uterine and cutaneous arteries. Nitric oxide (NO) is the main neurogenic vasodilator in cerebral arteries and acts as a counter-regulatory mechanism for adrenergic vasoconstriction in other vascular territories. In the penis, NO relaxes the vascular and cavernous smooth muscles leading to penile erection. Furthermore, when interacting with other perivascular nerves, NO can act as a neuromodulator. PNN dysfunction is involved in the genesis and maintenance of vascular disorders associated with arterial and portal hypertension, diabetes, ageing, obesity, cirrhosis and hormonal changes. For example defective nitrergic function contributes to enhanced sympathetic neurotransmission, vasoconstriction and blood pressure in some animal models of hypertension. In diabetic animals and humans, dysfunctional nitrergic neurotransmission in the corpus cavernosum is associated with erectile dysfunction. However, in some vascular beds of hypertensive and diabetic animals, an increased PNN function has been described as a compensatory mechanism to the increased vascular resistance. The present review summarizes current understanding on the role of PNN in control of vascular tone, its alterations under different conditions and the associated mechanisms. The knowledge of these changes can serve to better understand the mechanisms involved in these disorders and help in planning new treatments.
Collapse
Affiliation(s)
- Fabiano E. Xavier
- Departamento de Fisiologia e Farmacologia Centro de Biociências Universidade Federal de Pernambuco Recife Brazil
| |
Collapse
|
25
|
Feliu C, Peyret H, Vautier D, Djerada Z. Simultaneous quantification of 8 nucleotides and adenosine in cells and their medium using UHPLC-HRMS. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1148:122156. [PMID: 32446186 DOI: 10.1016/j.jchromb.2020.122156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Purinergic signalling is involved in physiological processes, particularly during ischemia-reperfusion injuries for which it has a protective effect. The purpose of this work was to develop a method for simultaneous quantification of eight nucleotides and adenosine in biological matrices by liquid chromatography coupled with high-resolution mass spectrometry. A method was developed that was sufficiently robust to quantify the targeted analytes in 20 min with good sensitivity. Analysis of extracellular media from cultured endothelial cells detected the release of nucleotides and adenosine during 2 h of hypoxia. The quantification of cylic adenosine monophosphate (cAMP) allowed to establish a dose-response curve after receptor stimulation. Therefore, our method allows us to study the involvement of nucleotides in various processes in both the intracellular and extracellular compartment.
Collapse
Affiliation(s)
- Catherine Feliu
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Hélène Peyret
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Damien Vautier
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France
| | - Zoubir Djerada
- Department of Pharmacology, E.A.3801, SFR CAP-santé, Reims University Hospital, 51, rue Cognacq-Jay, 51095 Reims Cedex, France.
| |
Collapse
|
26
|
Zhou R, Dang X, Sprague RS, Mustafa SJ, Zhou Z. Alteration of purinergic signaling in diabetes: Focus on vascular function. J Mol Cell Cardiol 2020; 140:1-9. [PMID: 32057736 DOI: 10.1016/j.yjmcc.2020.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/02/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022]
Abstract
Diabetes is an important risk factor for the development of cardiovascular disease including atherosclerosis and ischemic heart disease. Vascular complications including macro- and micro-vascular dysfunction are the leading causes of morbidity and mortality in diabetes. Disease mechanisms at present are unclear and no ideal therapies are available, which urgently calls for the identification of novel therapeutic targets/agents. An altered nucleotide- and nucleoside-mediated purinergic signaling has been implicated to cause diabetes-associated vascular dysfunction in major organs. Alteration of both purinergic P1 and P2 receptor sensitivity rather than the changes in receptor expression accounts for vascular dysfunction in diabetes. Activation of P2X7 receptors plays a crucial role in diabetes-induced retinal microvascular dysfunction. Recent findings have revealed that both ecto-nucleotidase CD39, a key enzyme hydrolyzing ATP, and CD73, an enzyme regulating adenosine turnover, are involved in the renal vascular injury in diabetes. Interestingly, erythrocyte dysfunction in diabetes by decreasing ATP release in response to physiological stimuli may serve as an important trigger to induce vascular dysfunction. Nucleot(s)ide-mediated purinergic activation also exerts long-term actions including inflammatory and atherogenic effects in hyperglycemic and diabetic conditions. This review highlights the current knowledge regarding the altered nucleot(s)ide-mediated purinergic signaling as an important disease mechanism for the diabetes-associated vascular complications. Better understanding the role of key receptor-mediated signaling in diabetes will provide more insights into their potential as targets for the treatment.
Collapse
Affiliation(s)
- Rui Zhou
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology of Ministry of Education, Southwest Medical University, Luzhou, PR China
| | - Xitong Dang
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology of Ministry of Education, Southwest Medical University, Luzhou, PR China
| | - Randy S Sprague
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
27
|
Martin-Aragon Baudel M, Espinosa-Tanguma R, Nieves-Cintron M, Navedo MF. Purinergic Signaling During Hyperglycemia in Vascular Smooth Muscle Cells. Front Endocrinol (Lausanne) 2020; 11:329. [PMID: 32528416 PMCID: PMC7256624 DOI: 10.3389/fendo.2020.00329] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
The activation of purinergic receptors by nucleotides and/or nucleosides plays an important role in the control of vascular function, including modulation of vascular smooth muscle excitability, and vascular reactivity. Accordingly, purinergic receptor actions, acting as either ion channels (P2X) or G protein-coupled receptors (GCPRs) (P1, P2Y), target diverse downstream effectors, and substrates to regulate vascular smooth muscle function and vascular reactivity. Both vasorelaxant and vasoconstrictive effects have been shown to be mediated by different purinergic receptors in a vascular bed- and species-specific manner. Purinergic signaling has been shown to play a key role in altering vascular smooth muscle excitability and vascular reactivity following acute and short-term elevations in extracellular glucose (e.g., hyperglycemia). Moreover, there is evidence that vascular smooth muscle excitability and vascular reactivity is severely impaired during diabetes and that this is mediated, at least in part, by activation of purinergic receptors. Thus, purinergic receptors present themselves as important candidates mediating vascular reactivity in hyperglycemia, with potentially important clinical and therapeutic potential. In this review, we provide a narrative summarizing our current understanding of the expression, function, and signaling of purinergic receptors specifically in vascular smooth muscle cells and discuss their role in vascular complications following hyperglycemia and diabetes.
Collapse
Affiliation(s)
- Miguel Martin-Aragon Baudel
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
- *Correspondence: Miguel Martin-Aragon Baudel
| | - Ricardo Espinosa-Tanguma
- Departamento de Fisiologia y Biofisca, Universidad Autónoma San Luis Potosí, San Luis Potosí, Mexico
| | | | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
- Manuel F. Navedo
| |
Collapse
|
28
|
Werner S, Mesch S, Hillig RC, Ter Laak A, Klint J, Neagoe I, Laux-Biehlmann A, Dahllöf H, Bräuer N, Puetter V, Nubbemeyer R, Schulz S, Bairlein M, Zollner TM, Steinmeyer A. Discovery and Characterization of the Potent and Selective P2X4 Inhibitor N-[4-(3-Chlorophenoxy)-3-sulfamoylphenyl]-2-phenylacetamide (BAY-1797) and Structure-Guided Amelioration of Its CYP3A4 Induction Profile. J Med Chem 2019; 62:11194-11217. [PMID: 31746599 DOI: 10.1021/acs.jmedchem.9b01304] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The P2X4 receptor is a ligand-gated ion channel that is expressed on a variety of cell types, especially those involved in inflammatory and immune processes. High-throughput screening led to a new class of P2X4 inhibitors with substantial CYP 3A4 induction in human hepatocytes. A structure-guided optimization with respect to decreased pregnane X receptor (PXR) binding was started. It was found that the introduction of larger and more polar substituents on the ether linker led to less PXR binding while maintaining the P2X4 inhibitory potency. This translated into significantly reduced CYP 3A4 induction for compounds 71 and 73. Unfortunately, the in vivo pharmacokinetic (PK) profiles of these compounds were insufficient for the desired profile in humans. However, BAY-1797 (10) was identified and characterized as a potent and selective P2X4 antagonist. This compound is suitable for in vivo studies in rodents, and the anti-inflammatory and anti-nociceptive effects of BAY-1797 were demonstrated in a mouse complete Freund's adjuvant (CFA) inflammatory pain model.
Collapse
Affiliation(s)
- Stefan Werner
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Stefanie Mesch
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Roman C Hillig
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Antonius Ter Laak
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | | | | | | | - Henrik Dahllöf
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Nico Bräuer
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Vera Puetter
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | | | - Simone Schulz
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Michaela Bairlein
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Thomas M Zollner
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| | - Andreas Steinmeyer
- Bayer AG, Research & Development, Pharmaceuticals , 13353 Berlin , Germany
| |
Collapse
|
29
|
Fu LW, Tjen-A-Looi SC, Barvarz S, Guo ZL, Malik S. Role of opioid receptors in modulation of P2X receptor-mediated cardiac sympathoexcitatory reflex response. Sci Rep 2019; 9:17224. [PMID: 31748569 PMCID: PMC6868205 DOI: 10.1038/s41598-019-53754-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023] Open
Abstract
Myocardial ischemia evokes powerful reflex responses through activation of vagal and sympathetic afferents in the heart through the release of ischemic metabolites. We have demonstrated that extracellular ATP stimulates cardiac sympathetic afferents through P2 receptor-mediated mechanism, and that opioid peptides suppress these afferents' activity. However, the roles of both P2 receptor and endogenous opioids in cardiac sympathoexcitatory reflex (CSR) responses remain unclear. We therefore hypothesized that activation of cardiac P2 receptor evokes CSR responses by stimulating cardiac sympathetic afferents and these CSR responses are modulated by endogenous opioids. We observed that intrapericardial injection of α,β-methylene ATP (α,β-meATP, P2X receptor agonist), but not ADP (P2Y receptor agonist), caused a graded increase in mean arterial pressure in rats with sinoaortic denervation and vagotomy. This effect of α,β-meATP was abolished by blockade of cardiac neural transmission with intrapericardial procaine treatment and eliminated by intrapericardial A-317491, a selective P2X2/3 and P2X3 receptor antagonist. Intrapericardial α,β-meATP also evoked CSR response in vagus-intact rats. Furthermore, the P2X receptor-mediated CSR responses were enhanced by intrapericardial naloxone, a specific opioid receptor antagonist. These data suggest that stimulation of cardiac P2X2/3 and P2X3, but not P2Y receptors, powerfully evokes CSR responses through activation of cardiac spinal afferents, and that endogenous opioids suppress the P2X receptor-mediated CSR responses.
Collapse
Affiliation(s)
- Liang-Wu Fu
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA.
| | - Stephanie C Tjen-A-Looi
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| | - Sherwin Barvarz
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| | - Zhi-Ling Guo
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| | - Shaista Malik
- Susan Samueli Integrative Health Institute and Department of Medicine, School of Medicine, University of California at Irvine, Irvine, CA, 92697, USA
| |
Collapse
|
30
|
Abstract
Perivascular adipose tissue (PVAT) refers to the local aggregate of adipose tissue surrounding the vascular tree, exhibiting phenotypes from white to brown and beige adipocytes. Although PVAT has long been regarded as simply a structural unit providing mechanical support to vasculature, it is now gaining reputation as an integral endocrine/paracrine component, in addition to the well-established modulator endothelium, in regulating vascular tone. Since the discovery of anti-contractile effect of PVAT in 1991, the use of multiple rodent models of reduced amounts of PVAT has revealed its regulatory role in vascular remodeling and cardiovascular implications, including atherosclerosis. PVAT does not only release PVAT-derived relaxing factors (PVRFs) to activate multiple subsets of endothelial and vascular smooth muscle potassium channels and anti-inflammatory signals in the vasculature, but it does also provide an interface for neuron-adipocyte interactions in the vascular wall to regulate arterial vascular tone. In this review, we outline our current understanding towards PVAT and attempt to provide hints about future studies that can sharpen the therapeutic potential of PVAT against cardiovascular diseases and their complications.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China
- Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hamidah Abu Bakar
- Health Sciences Department, Universiti Selangor, 40000, Shah Alam, Selangor, Malaysia
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC)-a joint cooperation between the Charité-University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.
- Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Yu Huang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China.
- Institute of Vascular Medicine, Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
31
|
Guilherme A, Henriques F, Bedard AH, Czech MP. Molecular pathways linking adipose innervation to insulin action in obesity and diabetes mellitus. Nat Rev Endocrinol 2019; 15:207-225. [PMID: 30733616 PMCID: PMC7073451 DOI: 10.1038/s41574-019-0165-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissue comprises adipocytes and many other cell types that engage in dynamic crosstalk in a highly innervated and vascularized tissue matrix. Although adipose tissue has been studied for decades, it has been appreciated only in the past 5 years that extensive arborization of nerve fibres has a dominant role in regulating the function of adipose tissue. This Review summarizes the latest literature, which suggests that adipocytes signal to local sensory nerve fibres in response to perturbations in lipolysis and lipogenesis. Such adipocyte signalling to the central nervous system causes sympathetic output to distant adipose depots and potentially other metabolic tissues to regulate systemic glucose homeostasis. Paracrine factors identified in the past few years that mediate such adipocyte-neuron crosstalk are also reviewed. Similarly, immune cells and endothelial cells within adipose tissue communicate with local nerve fibres to modulate neurotransmitter tone, blood flow, adipocyte differentiation and energy expenditure, including adipose browning to produce heat. This understudied field of neurometabolism related to adipose tissue biology has great potential to reveal new mechanistic insights and potential therapeutic strategies for obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alexander H Bedard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
32
|
Zhou Z, Matsumoto T, Jankowski V, Pernow J, Mustafa SJ, Duncker DJ, Merkus D. Uridine adenosine tetraphosphate and purinergic signaling in cardiovascular system: An update. Pharmacol Res 2019; 141:32-45. [PMID: 30553823 PMCID: PMC6685433 DOI: 10.1016/j.phrs.2018.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/26/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Uridine adenosine tetraphosphate (Up4A), biosynthesized by activation of vascular endothelial growth factor receptor (VEGFR) 2, was initially identified as a potent endothelium-derived vasoconstrictor in perfused rat kidney. Subsequently, the effect of Up4A on vascular tone regulation was intensively investigated in arteries isolated from different vascular beds in rodents including rat pulmonary arteries, aortas, mesenteric and renal arteries as well as mouse aortas, in which Up4A produces vascular contraction. In contrast, Up4A produces vascular relaxation in porcine coronary small arteries and rat aortas. Intravenous infusion of Up4A into conscious rats or mice decreases blood pressure, and intravenous bolus injection of Up4A into anesthetized mice increases coronary blood flow, indicating an overall vasodilator influence in vivo. Although Up4A is the first dinucleotide described that contains both purine and pyrimidine moieties, its cardiovascular effects are exerted mainly through activation of purinergic receptors. These effects not only encompass regulation of vascular tone, but also endothelial angiogenesis, smooth muscle cell proliferation and migration, and vascular calcification. Furthermore, this review discusses a potential role for Up4A in cardiovascular pathophysiology, as plasma levels of Up4A are elevated in juvenile hypertensive patients and Up4A-mediated vascular purinergic signaling changes in cardiovascular disease such as hypertension, diabetes, atherosclerosis and myocardial infarction. Better understanding the vascular effect of the novel dinucleotide Up4A and the purinergic signaling mechanisms mediating its effects will enhance its potential as target for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo, Japan
| | - Vera Jankowski
- RWTH-Aachen, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - S Jamal Mustafa
- Department of Physiology, Pharmacology & Neuroscience, Center for Cardiovascular and Respiratory Sciences, Clinical and Translational Science Institute, West Virginia University, Morgantown, WV, United States
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
33
|
Best KA, Bone DB, Vilas G, Gros R, Hammond JR. Changes in aortic reactivity associated with the loss of equilibrative nucleoside transporter 1 (ENT1) in mice. PLoS One 2018; 13:e0207198. [PMID: 30408123 PMCID: PMC6224178 DOI: 10.1371/journal.pone.0207198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/26/2018] [Indexed: 01/23/2023] Open
Abstract
Slc29a1 encodes for equilibrative nucleoside transporter subtype 1 (ENT1), the primary mechanism of adenosine transfer across cell membranes. Previous studies showed that tissues isolated from Slc29a1-null mice are relatively resistant to injury caused by vascular ischemia-reperfusion. To determine if there are similar changes in the microvasculature, and investigate underlying mechanism, we examined aortas isolated from wildtype and Slc29a1-null mice. Aorta macrostructure and gene expression were examined histologically and by qPCR, respectively. Wire myography was used to assess the contractile properties of isolated thoracic aortic rings and their response to adenosine under both normoxic and hypoxic conditions. In vivo haemodynamic parameters were assessed using the tail-cuff method. Slc29a1-null mice had significantly (P<0.05) increased plasma adenosine (2.75-fold) and lower blood pressure (~15% ↓) than wild-type mice. Aortas from Slc29a1-null mice were stiffer with a smaller circumference (11% ↓), and had an enhanced contractile response to KCl and receptor-mediated stimuli. Blockade of ENT1 with nitrobenzylthioinosine significantly enhanced (by ~3.5-fold) the response of aorta from wild-type mice to phenylephrine, but had minimal effect on aortas from Slc29a1-null mice. Adenosine enhanced phenylephrine-mediated constriction in the wild-type tissue under both normoxic (11.7-fold) and hypoxic (3.6-fold) conditions, but had no effect on the Slc29a1-null aortic aorta. In conclusion, aortas from Slc29a1-null mice respond to hypoxic insult in a manner comparable to wild-type tissues that have been pharmacologically preconditioned with adenosine. These data also support a role for ENT1 in the regulation of the protective effects of adenosine on contractile function in elastic conduit arteries such as thoracic aorta.
Collapse
Affiliation(s)
- K. Arielle Best
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Derek B. Bone
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Gonzalo Vilas
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Robert Gros
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Molecular Medicine Research Group, Robarts Research Institute, London, Ontario, Canada
| | - James R. Hammond
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
34
|
Willson C. The clinical toxicology of caffeine: A review and case study. Toxicol Rep 2018; 5:1140-1152. [PMID: 30505695 PMCID: PMC6247400 DOI: 10.1016/j.toxrep.2018.11.002] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 10/09/2018] [Accepted: 11/01/2018] [Indexed: 02/08/2023] Open
Abstract
Caffeine is a widely recognized psychostimulant compound with a long history of consumption by humans. While it has received a significant amount of attention there is still much to be learned with respect to its toxicology in humans, especially in cases of overdose. A review of the history of consumption and the clinical toxicology of caffeine including clinical features, pharmacokinetics, toxicokinetics, a thorough examination of mechanism of action and management/treatment strategies are undertaken. While higher (i.e., several grams) quantities of caffeine are known to cause toxicity and potentially lethality, cases of mainly younger individuals who have experienced severe side effects and death despite consuming doses not otherwise known to cause such harm is troubling and deserves further study. An attempted case reconstruction is performed in an effort to shed light on this issue with a focus on the pharmacokinetics and pharmacodynamics of caffeine.
Collapse
|
35
|
Sarhan M, Land WG, Tonnus W, Hugo CP, Linkermann A. Origin and Consequences of Necroinflammation. Physiol Rev 2018; 98:727-780. [PMID: 29465288 DOI: 10.1152/physrev.00041.2016] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When cells undergo necrotic cell death in either physiological or pathophysiological settings in vivo, they release highly immunogenic intracellular molecules and organelles into the interstitium and thereby represent the strongest known trigger of the immune system. With our increasing understanding of necrosis as a regulated and genetically determined process (RN, regulated necrosis), necrosis and necroinflammation can be pharmacologically prevented. This review discusses our current knowledge about signaling pathways of necrotic cell death as the origin of necroinflammation. Multiple pathways of RN such as necroptosis, ferroptosis, and pyroptosis have been evolutionary conserved most likely because of their differences in immunogenicity. As the consequence of necrosis, however, all necrotic cells release damage associated molecular patterns (DAMPs) that have been extensively investigated over the last two decades. Analysis of necroinflammation allows characterizing specific signatures for each particular pathway of cell death. While all RN-pathways share the release of DAMPs in general, most of them actively regulate the immune system by the additional expression and/or maturation of either pro- or anti-inflammatory cytokines/chemokines. In addition, DAMPs have been demonstrated to modulate the process of regeneration. For the purpose of better understanding of necroinflammation, we introduce a novel classification of DAMPs in this review to help detect the relative contribution of each RN-pathway to certain physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Walter G Land
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Wulf Tonnus
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Christian P Hugo
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Andreas Linkermann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
36
|
Schmid R, Evans RJ. ATP-Gated P2X Receptor Channels: Molecular Insights into Functional Roles. Annu Rev Physiol 2018; 81:43-62. [PMID: 30354932 DOI: 10.1146/annurev-physiol-020518-114259] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the nervous system, ATP is co-stored in vesicles with classical transmitters and released in a regulated manner. ATP from the intracellular compartment can also exit the cell through hemichannels and following shear stress or membrane damage. In the past 30 years, the action of ATP as an extracellular transmitter at cell-surface receptors has evolved from somewhat of a novelty that was treated with skepticism to purinergic transmission being accepted as having widespread important functional roles mediated by ATP-gated ionotropic P2X receptors (P2XRs). This review focuses on work published in the last five years and provides an overview of ( a) structural studies, ( b) the molecular basis of channel properties and regulation of P2XRs, and ( c) the physiological and pathophysiological roles of ATP acting at defined P2XR subtypes.
Collapse
Affiliation(s)
- Ralf Schmid
- Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 7RH, United Kingdom; .,Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester LE1 7RH, United Kingdom
| | - Richard J Evans
- Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 7RH, United Kingdom;
| |
Collapse
|
37
|
Nemkov T, Reisz JA, Xia Y, Zimring JC, D’Alessandro A. Red blood cells as an organ? How deep omics characterization of the most abundant cell in the human body highlights other systemic metabolic functions beyond oxygen transport. Expert Rev Proteomics 2018; 15:855-864. [DOI: 10.1080/14789450.2018.1531710] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Aurora, CO, USA
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Aurora, CO, USA
| | - Yang Xia
- Department of Biochemistry, University of Texas Houston – McGovern Medical School , Houston, TX, USA
| | | | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Aurora, CO, USA
| |
Collapse
|
38
|
Akintunde JK, Irondi AE, Ajani EO, Olayemi TV. Neuroprotective effect of dietary black seed flour on key enzymes linked with neuronal signaling molecules in rats exposed to mixture of environmental metals. J Food Biochem 2018. [DOI: 10.1111/jfbc.12573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- J. K. Akintunde
- Toxicology and Safety Unit, Faculty of Public Health, College of Medicine, Department of Environmental Health Sciences; University of Ibadan; Inadan Nigeria
- Department of Biochemistry, School of Basic Medical Sciences, College of Pure and Applied Sciences; Kwara State University; Malete P.M.B 1530 Nigeria
| | - A. E. Irondi
- Department of Biochemistry, School of Basic Medical Sciences, College of Pure and Applied Sciences; Kwara State University; Malete P.M.B 1530 Nigeria
| | - E. O. Ajani
- Department of Biochemistry, School of Basic Medical Sciences, College of Pure and Applied Sciences; Kwara State University; Malete P.M.B 1530 Nigeria
| | - T. V. Olayemi
- Chemistry Unit, Department of Chemical, Physical and Geological, College of Pure and Applied Sciences; Kwara State University; Malete P.M.B 1530 Nigeria
| |
Collapse
|
39
|
Klein A, Joseph PD, Christensen VG, Jensen LJ, Jacobsen JCB. Lack of tone in mouse small mesenteric arteries leads to outward remodeling, which can be prevented by prolonged agonist-induced vasoconstriction. Am J Physiol Heart Circ Physiol 2018; 315:H644-H657. [PMID: 29775408 DOI: 10.1152/ajpheart.00111.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inward remodeling of resistance vessels is an independent risk factor for cardiovascular events. Thus far, the remodeling process remains incompletely elucidated, but the activation level of the vascular smooth muscle cell appears to play a central role. Accordingly, previous data have suggested that an antagonistic and supposedly beneficial response, outward remodeling, may follow prolonged vasodilatation. The present study aimed to determine whether 1) outward remodeling follows 3 days of vessel culture without tone, 2) a similar response can be elicited in a much shorter 4-h timeframe, and, finally, 3) whether a 4-h response can be prevented or reversed by the presence of vasoconstrictors in the medium. Cannulated mouse small mesenteric arteries were organocultured for 3 days in the absence of tone, leading to outward remodeling that continued throughout the culture period. In more acute experiments in which cannulated small mesenteric arteries were maintained in physiological saline without tone for 4 h, we detected a similar outward remodeling that proceeded at a rate several times faster. In the 4-h experimental setting, continuous vasoconstriction to ~50% tone by abluminal application of UTP or norepinephrine + neuropeptide Y prevented outward remodeling but did not cause inward remodeling. Computational modeling was used to simulate and interpret these findings and to derive time constants of the remodeling processes. It is suggested that depriving resistance arteries of activation will lead to eutrophic outward remodeling, which can be prevented by vascular smooth muscle cell activation induced by prolonged vasoconstrictor exposure. NEW & NOTEWORTHY We have established an effective 4-h method for studying outward remodeling in pressurized mouse resistance vessels ex vivo and have determined conditions that block the remodeling response. This allows for investigating the subtle but clinically highly relevant phenomenon of outward remodeling while avoiding both laborious 3-day organoid culture of cannulated vessels and in vivo experiments lasting several weeks.
Collapse
Affiliation(s)
- Anika Klein
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Philomeena Daphne Joseph
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Vibeke Grøsfjeld Christensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Lars Jørn Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens Christian Brings Jacobsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
40
|
In Vivo PET Imaging of Adenosine 2A Receptors in Neuroinflammatory and Neurodegenerative Disease. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:6975841. [PMID: 29348737 PMCID: PMC5733838 DOI: 10.1155/2017/6975841] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023]
Abstract
Adenosine receptors are G-protein coupled P1 purinergic receptors that are broadly expressed in the peripheral immune system, vasculature, and the central nervous system (CNS). Within the immune system, adenosine 2A (A2A) receptor-mediated signaling exerts a suppressive effect on ongoing inflammation. In healthy CNS, A2A receptors are expressed mainly within the neurons of the basal ganglia. Alterations in A2A receptor function and expression have been noted in movement disorders, and in Parkinson's disease pharmacological A2A receptor antagonism leads to diminished motor symptoms. Although A2A receptors are expressed only at a low level in the healthy CNS outside striatum, pathological challenge or inflammation has been shown to lead to upregulation of A2A receptors in extrastriatal CNS tissue, and this has been successfully quantitated using in vivo positron emission tomography (PET) imaging and A2A receptor-binding radioligands. Several radioligands for PET imaging of A2A receptors have been developed in recent years, and A2A receptor-targeting PET imaging may thus provide a potential additional tool to evaluate various aspects of neuroinflammation in vivo. This review article provides a brief overview of A2A receptors in healthy brain and in a selection of most important neurological diseases and describes the recent advances in A2A receptor-targeting PET imaging studies.
Collapse
|
41
|
Jiang Y, Tang J, Wang X, Shen C. Developing a Wireless, High Precision and Processing Speed Pulse Monitoring Headset Using Photoplethysmography. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE-JTEHM 2017; 5:2700311. [PMID: 29285419 PMCID: PMC5739535 DOI: 10.1109/jtehm.2017.2761873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/23/2017] [Accepted: 09/14/2017] [Indexed: 11/06/2022]
Abstract
In this paper, thorough improvement of pulse monitoring and analysis equipment with a headset structure is presented. In order to study the most suitable infrared wavelength for the acquisition of the pulse wave at the earlobe, Monte Carlo simulation was adapted. Both high frequency noise and baseline drift, generated in the signal acquisition process, are considered. To further optimize the system design and improve accuracy, for the sensor's dimensional drift, the corresponding compensation was carried on in the software. This paper introduced nonlinear quantization, especially in terms of very weak pulse signal, in the time domain analysis process. A quick extraction method named table look-up combing with interpolation was utilized to obtain frequency domain information whose processing speed can be increased by about 30 times compared with fast Fourier transformation setting the sampling point as 300. The results demonstrated the sensor's excellent performance in pulse signal acquisition whose maximum residual is less than 0.004 mV. The test on a random sample of 300 people indicates that the system had high correlation with reference, validating the system accuracy is extremely high. Overall, this paper provides a practical pulse monitoring and analysis system with high precision and processing speed that can be widely applied in the field of health management or medical measurement.
Collapse
|
42
|
Shestopalov VI, Panchin Y, Tarasova OS, Gaynullina D, Kovalzon VM. Pannexins Are Potential New Players in the Regulation of Cerebral Homeostasis during Sleep-Wake Cycle. Front Cell Neurosci 2017; 11:210. [PMID: 28769767 PMCID: PMC5511838 DOI: 10.3389/fncel.2017.00210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
During brain homeostasis, both neurons and astroglia release ATP that is rapidly converted to adenosine in the extracellular space. Pannexin-1 (Panx1) hemichannels represent a major conduit of non-vesicular ATP release from brain cells. Previous studies have shown that Panx1−/− mice possess severe disruption of the sleep-wake cycle. Here, we review experimental data supporting the involvement of pannexins (Panx) in the coordination of fundamental sleep-associated brain processes, such as neuronal activity and regulation of cerebrovascular tone. Panx1 hemichannels are likely implicated in the regulation of the sleep-wake cycle via an indirect effect of released ATP on adenosine receptors and through interaction with other somnogens, such as IL-1β, TNFα and prostaglandin D2. In addition to the recently established role of Panx1 in the regulation of endothelium-dependent arterial dilation, similar signaling pathways are the major cellular component of neurovascular coupling. The new discovered role of Panx in sleep regulation may have broad implications in coordinating neuronal activity and homeostatic housekeeping processes during the sleep-wake cycle.
Collapse
Affiliation(s)
- Valery I Shestopalov
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of MedicineMiami, FL, United States.,Microbiology and Bioengineering Laboratory, Department of Genomics and Biotechnology, Vavilov Institute of General Genetics, Russian Academy of SciencesMoscow, Russia
| | - Yuri Panchin
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Mathematical Methods in Biology, Belozersky Institute, M.V. Lomonosov Moscow State UniversityMoscow, Russia
| | - Olga S Tarasova
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State UniversityMoscow, Russia.,State Research Center of the Russian Federation, Institute for Biomedical Problems, Russian Academy of SciencesMoscow, Russia
| | - Dina Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State UniversityMoscow, Russia.,Department of Physiology, Russian National Research Medical UniversityMoscow, Russia
| | - Vladimir M Kovalzon
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Severtsov Institute Ecology and Evolution, Russian Academy of SciencesMoscow, Russia
| |
Collapse
|
43
|
Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci 2017; 18:419-434. [PMID: 28515434 PMCID: PMC5759779 DOI: 10.1038/nrn.2017.48] [Citation(s) in RCA: 774] [Impact Index Per Article: 110.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cerebral blood flow (CBF) regulation is essential for normal brain function. The mammalian brain has evolved a unique mechanism for CBF control known as neurovascular coupling. This mechanism ensures a rapid increase in the rate of CBF and oxygen delivery to activated brain structures. The neurovascular unit is composed of astrocytes, mural vascular smooth muscle cells and pericytes, and endothelia, and regulates neurovascular coupling. This Review article examines the cellular and molecular mechanisms within the neurovascular unit that contribute to CBF control, and neurovascular dysfunction in neurodegenerative disorders such as Alzheimer disease.
Collapse
Affiliation(s)
- Kassandra Kisler
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Axel Montagne
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| |
Collapse
|
44
|
Loesch A, Dashwood MR. Nerve-perivascular fat communication as a potential influence on the performance of blood vessels used as coronary artery bypass grafts. J Cell Commun Signal 2017; 12:181-191. [PMID: 28601937 PMCID: PMC5842173 DOI: 10.1007/s12079-017-0393-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/09/2017] [Indexed: 12/14/2022] Open
Abstract
Perivascular fat, the cushion of adipose tissue surrounding blood vessels, possesses dilator, anti-contractile and constrictor actions. The majority of these effects have been demonstrated in vitro and may depend on the vessel and/or the experimental method or species used. In general, the relaxant effect of perivascular adipose tissue is local and may be either endothelium-dependent or endothelium-independent. However, nerve stimulation studies show that, in general, perivascular adipose tissue (PVAT) has an anti-contractile vascular effect likely to involve an action of the autonomic vascular nerves. Apart from a direct effect of perivascular fat-derived factors on bypass conduits, an interaction with a number of neurotransmitters and other agents may play an important role in graft performance. Although the vascular effects of PVAT are now well-established there is a lack of information regarding the role and/or involvement of peripheral nerves including autonomic nerves. For example, are perivascular adipocytes innervated and does PVAT affect neuronal control of vessels used as grafts? To date there is a paucity of electrophysiological studies into nerve-perivascular fat control. This review provides an overview of the vascular actions of PVAT, focussing on its potential relevance on blood vessels used as bypass grafts. In particular, the anatomical relationship between the perivascular nerves and fat are considered and the role of the perivascular-nerve/fat axis in the performance of bypass grafts is also discussed.
Collapse
Affiliation(s)
- Andrzej Loesch
- Centre for Rheumatology and Connective Tissue Diseases, Division of Medicine, University College London Medical School, Royal Free Campus, Rowland Hill Street, NW3 2PF, London, UK.
| | - Michael R Dashwood
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London Medical School, Royal Free Campus, Rowland Hill Street, NW3 2PF, London, UK
| |
Collapse
|
45
|
Leiva A, Guzmán-Gutiérrez E, Contreras-Duarte S, Fuenzalida B, Cantin C, Carvajal L, Salsoso R, Gutiérrez J, Pardo F, Sobrevia L. Adenosine receptors: Modulators of lipid availability that are controlled by lipid levels. Mol Aspects Med 2017; 55:26-44. [DOI: 10.1016/j.mam.2017.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
|
46
|
Burnstock G. Purinergic Signaling in the Cardiovascular System. Circ Res 2017; 120:207-228. [PMID: 28057794 DOI: 10.1161/circresaha.116.309726] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023]
Abstract
There is nervous control of the heart by ATP as a cotransmitter in sympathetic, parasympathetic, and sensory-motor nerves, as well as in intracardiac neurons. Centers in the brain control heart activities and vagal cardiovascular reflexes involve purines. Adenine nucleotides and nucleosides act on purinoceptors on cardiomyocytes, AV and SA nodes, cardiac fibroblasts, and coronary blood vessels. Vascular tone is controlled by a dual mechanism. ATP, released from perivascular sympathetic nerves, causes vasoconstriction largely via P2X1 receptors. Endothelial cells release ATP in response to changes in blood flow (via shear stress) or hypoxia, to act on P2 receptors on endothelial cells to produce nitric oxide, endothelium-derived hyperpolarizing factor, or prostaglandins to cause vasodilation. ATP is also released from sensory-motor nerves during antidromic reflex activity, to produce relaxation of some blood vessels. Purinergic signaling is involved in the physiology of erythrocytes, platelets, and leukocytes. ATP is released from erythrocytes and platelets, and purinoceptors and ectonucleotidases are expressed by these cells. P1, P2Y1, P2Y12, and P2X1 receptors are expressed on platelets, which mediate platelet aggregation and shape change. Long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides promote migration and proliferation of vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis, vessel remodeling during restenosis after angioplasty and atherosclerosis. The involvement of purinergic signaling in cardiovascular pathophysiology and its therapeutic potential are discussed, including heart failure, infarction, arrhythmias, syncope, cardiomyopathy, angina, heart transplantation and coronary bypass grafts, coronary artery disease, diabetic cardiomyopathy, hypertension, ischemia, thrombosis, diabetes mellitus, and migraine.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- From the Autonomic Neuroscience Institute, Royal Free and University College Medical School, London, United Kingdom.
| |
Collapse
|
47
|
Sunggip C, Nishimura A, Shimoda K, Numaga-Tomita T, Tsuda M, Nishida M. Purinergic P2Y 6 receptors: A new therapeutic target of age-dependent hypertension. Pharmacol Res 2017; 120:51-59. [PMID: 28336370 DOI: 10.1016/j.phrs.2017.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/13/2017] [Accepted: 03/13/2017] [Indexed: 01/04/2023]
Abstract
Aging has a remarkable effect on cardiovascular homeostasis and it is known as the major non-modifiable risk factor in the development of hypertension. Medications targeting sympathetic nerve system and/or renin-angiotensin-aldosterone system are widely accepted as a powerful therapeutic strategy to improve hypertension, although the control rates remain unsatisfactory especially in the elder patients with hypertension. Purinergic receptors, activated by adenine, uridine nucleotides and nucleotide sugars, play pivotal roles in many biological processes, including platelet aggregation, neurotransmission and hormone release, and regulation of cardiovascular contractility. Since clopidogrel, a selective inhibitor of G protein-coupled purinergic P2Y12 receptor (P2Y12R), achieved clinical success as an anti-platelet drug, P2YRs has been attracted more attention as new therapeutic targets of cardiovascular diseases. We have revealed that UDP-responsive P2Y6R promoted angiotensin type 1 receptor (AT1R)-stimulated vascular remodeling in mice, in an age-dependent manner. Moreover, the age-related formation of heterodimer between AT1R and P2Y6R was disrupted by MRS2578, a P2Y6R-selective inhibitor. These findings suggest that P2Y6R is a therapeutic target to prevent age-related hypertension.
Collapse
Affiliation(s)
- Caroline Sunggip
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Biomedical Science & Therapeutic, Faculty of Medicine and Health Sciences, University Malaysia Sabah, 88400 Kota Kinabalu Sabah, Malaysia
| | - Akiyuki Nishimura
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| | - Kakeru Shimoda
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan; Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takuro Numaga-Tomita
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan; Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
48
|
Dănilă MD, Privistirescu A, Duicu OM, Rațiu CD, Angoulvant D, Muntean DM, Sturza A. The effect of purinergic signaling via the P 2Y 11 receptor on vascular function in a rat model of acute inflammation. Mol Cell Biochem 2017; 431:37-44. [PMID: 28213772 DOI: 10.1007/s11010-017-2973-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 02/02/2017] [Indexed: 01/22/2023]
Abstract
There is a growing body of evidence pointing to the role of purinergic signaling in the development and progression of various conditions that have inflammation as a common pathogenetic denominator. The aim of the present study was to assess the involvement of P2Y11 purinergic receptors in the regulation of vascular function in aortic segments obtained using an experimental model of acute inflammation, the lipopolysaccharide (LPS, 8 mg/kg, i.p)-treated rats. Twelve hours after LPS administration, thoracic aortas were isolated and used for studies of vascular reactivity in the organ bath and for the measurement of reactive oxygen species (ROS) generation, respectively. LPS treatment significantly increased contractility to phenylephrine and attenuated the endothelium-dependent relaxation of the vascular segments in response to acetylcholine; an increased production of hydrogen peroxide (H2O2) was also recorded. The P2Y11 activator, NF546, decreased the LPS-induced aortic H2O2 release and partially normalized the vasomotor function, namely reduced contractility and improved relaxation. The effect was abolished by co-treatment with the P2Y11 inhibitor, NF340, and also after endothelium denudation. Importantly, NF546 did not elicit an antioxidant effect by acting as a H2O2 scavenger, suggesting that the beneficial outcome of this treatment on the vasculature is the consequence of P2Y11 stimulation. In conclusion, purinergic P2Y11 receptors stimulation improves vascular function and mitigates oxidative stress in the setting of acute systemic inflammation, revealing salutary effects and therapeutic potential in pathologies associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Maria D Dănilă
- Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania.,Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania
| | - Andreea Privistirescu
- Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania
| | - Oana M Duicu
- Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania.,Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania
| | - Corina D Rațiu
- Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania
| | - Denis Angoulvant
- EA 4245 Cellules Dendritiques, Immunomodulation et Greffes, Université François-Rabelais de Tours, 37032, Tours, France.,Service de Cardiologie, Hôpital Trousseau, Centre Hospitalier Régional Universitaire de Tours, 37044, Tours, France
| | - Danina M Muntean
- Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania. .,Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania.
| | - Adrian Sturza
- Department of Functional Sciences - Pathophysiology, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania.,Center for Translational Research and Systems Medicine, "Victor Babeş" University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Sq., 300041, Timişoara, Romania
| |
Collapse
|
49
|
Watanabe S, Matsumoto T, Ando M, Kobayashi S, Iguchi M, Taguchi K, Kobayashi T. A Comparative Study of Vasorelaxant Effects of ATP, ADP, and Adenosine on the Superior Mesenteric Artery of SHR. Biol Pharm Bull 2017; 39:1374-80. [PMID: 27476946 DOI: 10.1248/bpb.b16-00260] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated superior mesenteric arteries from spontaneously hypertensive rats (SHR) to determine the relaxation responses induced by ATP, ADP, and adenosine and the relationship between the relaxant effects of these compounds and nitric oxide (NO) or cyclooxygenase (COX)-derived prostanoids. In rat superior mesenteric artery, relaxation induced by ATP and ADP but not by adenosine was completely eliminated by endothelial denudation. In the superior mesenteric arteries isolated from SHR [vs. age-matched control Wistar Kyoto rats (WKY)], a) ATP- and ADP-induced relaxations were weaker, whereas adenosine-induced relaxation was similar in both groups, b) ATP- and ADP-induced relaxations were substantially and partly reduced by N(G)-nitro-L-arginine [a NO synthase (NOS) inhibitor], respectively, c) indomethacin, an inhibitor of COX, increased ATP- and ADP-induced relaxations, d) ADP-induced relaxation was weaker under combined inhibition by NOS and COX, and e) adenosine-induced relaxation was not altered by treatment with these inhibitors. These data indicate that levels of responsiveness to these nucleotides/adenosine vary in the superior mesenteric arteries from SHR and WKY and are differentially modulated by NO and COX-derived prostanoids.
Collapse
Affiliation(s)
- Shun Watanabe
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | | | | | | | | | | | | |
Collapse
|
50
|
Greaney JL, Kenney WL, Alexander LM. Neurovascular mechanisms underlying augmented cold-induced reflex cutaneous vasoconstriction in human hypertension. J Physiol 2017; 595:1687-1698. [PMID: 27891612 DOI: 10.1113/jp273487] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS In hypertensive adults (HTN), cardiovascular risk increases disproportionately during environmental cold exposure. Despite ample evidence of dysregulated sympathetic control of the peripheral vasculature in hypertension, no studies have examined integrated neurovascular function during cold stress in HTN. The findings of the present study show that whole-body cold stress elicits greater increases in sympathetic outflow directed to the cutaneous vasculature and, correspondingly, greater reductions in skin blood flow in HTN. We further demonstrate an important role for non-adrenergic sympathetic co-transmitters in mediating the vasoconstrictor response to cold stress in hypertension. In the context of thermoregulation and the maintenance of core temperature, sympathetically-mediated control of the cutaneous vasculature is not only preserved, but also exaggerated in hypertension. Given the increasing prevalence of hypertension, clarifying the mechanistic underpinnings of hypertension-induced alterations in neurovascular function during cold exposure is clinically relevant. ABSTRACT Despite ample evidence of dysregulated sympathetic control of the peripheral vasculature in hypertension, no studies have examined integrated neurovascular function during cold stress in hypertensive adults (HTN). We hypothesized that (i) whole-body cooling would elicit greater cutaneous vasoconstriction and greater increases in skin sympathetic nervous system activity (SSNA) in HTN (n = 14; 56 ± 2 years) compared to age-matched normotensive adults (NTN; n = 14; 55 ± 2 years) and (ii) augmented reflex vasoconstriction in HTN would be mediated by an increase in cutaneous vascular adrenergic sensitivity and a greater contribution of non-adrenergic sympathetic co-transmitters. SSNA (peroneal microneurography) and red cell flux (laser Doppler flowmetry; dorsum of foot) were measured during whole-body cooling (water-perfused suit). Sympathetic adrenergic- and non-adrenergic-dependent contributions to reflex cutaneous vasoconstriction and vascular adrenergic sensitivity were assessed pharmacologically using intradermal microdialysis. Cooling elicited greater increases in SSNA (NTN: +64 ± 13%baseline vs. HTN: +194 ± 26%baseline ; P < 0.01) and greater reductions in skin blood flow (NTN: -16 ± 2%baseline vs. HTN: -28 ± 3%baseline ; P < 0.01) in HTN compared to NTN, reflecting an increased response range for sympathetic reflex control of cutaneous vasoconstriction in HTN. Norepinephrine dose-response curves showed no HTN-related difference in cutaneous adrenergic sensitivity (logEC50 ; NTN: -7.4 ± 0.3 log M vs. HTN: -7.5 ± 0.3 log M; P = 0.84); however, non-adrenergic sympathetic co-transmitters mediated a significant portion of the vasoconstrictor response to cold stress in HTN. Collectively, these findings indicate that hypertension increases the peripheral cutaneous vasoconstrictor response to cold via greater increases in skin sympathetic outflow coupled with an increased reliance on non-adrenergic neurotransmitters.
Collapse
Affiliation(s)
- Jody L Greaney
- Department of Kinesiology, Noll Laboratory, The Pennsylvania State University, University Park, PA, USA
| | - W Larry Kenney
- Department of Kinesiology, Noll Laboratory, The Pennsylvania State University, University Park, PA, USA
| | - Lacy M Alexander
- Department of Kinesiology, Noll Laboratory, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|