1
|
Nitric oxide bioavailability for red blood cell deformability in the microcirculation: A review of recent progress. Nitric Oxide 2022; 129:25-29. [PMID: 36184009 DOI: 10.1016/j.niox.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/20/2022]
Abstract
The rheological properties of red blood cells (RBCs) play an important role in their microcirculation. RBCs can elastically deform in response to mechanical forces to pass through narrow vessels for effective gas exchange in peripheral tissues. Decreased RBC deformability is observed in lifestyle-related diseases such as diabetes mellitus, hypercholesterolemia, and hypertension, which are pathological conditions linked to increased oxidative stress and decreased nitric oxide (NO) bioavailability. Redox-sensitive cysteine residues on RBC cytoskeletal proteins, such as α- and β-spectrins, responsible for membrane flexibility, are affected by prolonged oxidative stress, leading to reversible and irreversible oxidative modifications and decreased RBC deformability. However, endogenously, and exogenously generated NO protects RBC membrane flexibility from further oxidative modification by shielding redox-sensitive cysteine residues with a glutathione cap. Recent studies have shown that nitrate-rich diets and moderate exercise can enhance NO production to increase RBC deformability by increasing the interplay between RBCs and vascular endothelium-mediated NO bioavailability for microcirculation. This review focuses on the molecular mechanism of RBC- and non-RBC-mediated NO generation, and how diet- and exercise-derived NO exert prophylactic effects against decreased RBC deformability in lifestyle-related diseases with vascular endothelial dysfunction.
Collapse
|
2
|
Eligini S, Porro B, Werba JP, Capra N, Genovese S, Greco A, Cavalca V, Banfi C. Oxidative Stress and Arginine/Nitric Oxide Pathway in Red Blood Cells Derived from Patients with Prediabetes. Biomedicines 2022; 10:biomedicines10061407. [PMID: 35740426 PMCID: PMC9219800 DOI: 10.3390/biomedicines10061407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/25/2022] Open
Abstract
The effects of the oral glucose tolerance test (OGTT) on red blood cells (RBCs) have not been thoroughly investigated, although it is known that the ingestion of 75 g of glucose during OGTT results in a systemic state of inflammation and oxidative stress. Therefore, we evaluated the effect of OGTT on oxidative stress and L-arginine/Nitric Oxide (L-Arg/NO) metabolic pathway in RBCs obtained from patients with prediabetes. Blood samples were collected from all participants before (T0) and at 10 (T1), 20 (T2), 30 (T3), 60 (T4), 90 (T5), 120 (T6), 150 (T7), and 180 (T8) minutes after glucose loading. Results showed a significant increase in oxidative stress status characterized by a rise in the GSSG/GSH ratio at T4 and T6 that increased in parallel with a reduction of NO production in RBCs. In addition, in this time frame, increased exposure of phosphatidylserine on RBCs membrane was observed. These metabolic modifications were rescued at T8, together with an increase in activated RBC NO synthase expression. These findings provide a possible explanation of the phenomena occurring after glucose loading and suggest that, even in the early stages of diabetes, it may be important to avoid acute variations in glycemia in order to prevent diabetic complications.
Collapse
Affiliation(s)
- Sonia Eligini
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
| | - Benedetta Porro
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
| | - José Pablo Werba
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
- Correspondence:
| | - Nicolò Capra
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
| | - Stefano Genovese
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
| | - Arianna Greco
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
| | - Viviana Cavalca
- Dipartimento di Scienze Cliniche e di Comunità, Università Degli Studi di Milano, 20122 Milano, Italy;
| | - Cristina Banfi
- Centro Cardiologico Monzino, IRCCS, 20138 Milano, Italy; (S.E.); (B.P.); (N.C.); (S.G.); (A.G.); (C.B.)
| |
Collapse
|
3
|
Akamo AJ, Akinloye DI, Ugbaja RN, Adeleye OO, Dosumu OA, Eteng OE, Antiya MC, Amah G, Ajayi OA, Faseun SO. Naringin prevents cyclophosphamide-induced erythrocytotoxicity in rats by abrogating oxidative stress. Toxicol Rep 2021; 8:1803-1813. [PMID: 34760624 PMCID: PMC8567332 DOI: 10.1016/j.toxrep.2021.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/30/2021] [Accepted: 10/24/2021] [Indexed: 01/02/2023] Open
Abstract
Earlier reports have shown that Cyclophosphamide (CYCP), an anti-malignant drug, elicited cytotoxicity; and that naringin has several beneficial potentials against oxidative stress and dyslipidaemias. We investigated the influence of naringin on free radical scavenging, cellular integrity, cellular ATP, antioxidants, oxidative stress, and lipid profiles in the CYCP-induced erythrocytotoxicity rat model. Rats were pretreated orally by gavage for fourteen consecutive days with three doses (50, 100, and 200 mg/kg) naringin before single CYCP (200 mg/kg, i.p.) administration. Afterwards, the rats were sacrificed. Naringin concentrations required for 50 % scavenging hydrogen peroxide and nitric oxide radical were 0.27 mg/mL and 0.28 mg/mL, respectively. Naringin pretreatment significantly (p < 0.05) protected erythrocytes plasma membrane architecture and integrity by abolishing CYCP-induced decrease in the activity of erythrocyte LDH (a marker of ATP). Pretreatment with naringin remarkably (p < 0.05) reversed CYCP-induced decreases in the erythrocytes glutathione levels, activities of glutathione-S-transferase, catalase, glutathione peroxidase, and glutathione reductase; attenuated CYCP-mediated increases in erythrocytes levels of malondialdehyde, nitric oxide, and major lipids (cholesterol, triacylglycerol, phospholipids, and non-esterified fatty acids). Taken together, different acute pretreatment doses of naringin might avert CYCP-mediated erythrocytes dysfunctions via its antioxidant, free-radical scavenging, and anti-dyslipidaemia properties.
Collapse
Key Words
- AP-1, activator protein 1
- ATP, adenosine triphosphate
- Antioxidants
- BHT, butylated hydroxytoluene
- C31H28N2Na4O13S, xylenol tetrasodium
- C5FeN6Na2O, sodium nitroprusside
- CAT, catalase
- CDNB, 1-chloro-2,4-dinitrobenzene
- CYCP, cyclophosphamide
- Cu(NO3)2.3H2O, copper II nitrate
- Cyclophosphamide
- DNA, deoxyribonucleic acid
- DTNB, 5,5ˈ-dithiobis(2-nitrobenzoic acid)
- Erythrocytotoxicity
- FeSO4.7H2O, Iron (II) sulfate heptahydrate
- G6PDH, glucose-6-phosphate dehydrogenase
- GSH, reduced glutathione
- GSPx, glutathione peroxidase
- GSR, glutathione reductase
- GSSG, oxidized glutathione
- GST, glutathione-S-transferase
- H2O2, hydrogen peroxide
- H3PO3, phosphoric acid
- HO•, hydroxyl radical
- HSCs, hepatic stellate cells
- K2HPO4, dipotassium hydrogen phosphate
- KCl, potassium chloride
- LDH, lactate dehydrogenase
- Lipid profile
- MAPKs, mitogen-activated protein kinases
- MDA, malondialdehyde
- MMP, matrix metalloprotease
- NAD+, nicotinamide adenine dinucleotide
- NADH, nicotinamide adenine dinucleotide reduced
- NADPH, nicotinamide adenine dinucleotide phosphate reduced
- NF-κB, nuclear factor kappa B
- NH4OH, ammonium hydroxide
- NO, nitric oxide
- NO2−, nitrite
- NO3−, nitrate
- NOAEL, no-observed-adverse-effect level
- Na2HPO4, disodium hydrogen phosphate
- NaH2PO4, sodium dihydrogen phosphate
- Naringin
- Nrf2, nuclear factor-erythroid factor 2-related factor 2
- O2HbFe2+, oxyhemoglobin
- O2•–, superoxide radical
- OONO−, peroxynitrite radical
- Oxidative stress
- PBS, phosphate-buffered saline
- PUFA, Polyunsaturated fatty acids
- R-Smad, Smad activated receptor
- RNS, reactive nitrogen species
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- TBA, 2-thiobarbituric acid
- TBARS, thiobarbituric acid reactive substances
- TGF-β, transforming growth factor-β
- TLR, toll-like receptor
- TROOH, total hydroperoxide
- VLDL, very low density lipoprotein
- eNOS, endothelial nitric oxide synthase
- i.p., intraperitoneally
- mRNA, messenger ribonucleic acid
- metHb, methemoglobin
- α-SMA, alpha smooth muscle actin
- •NO, nitric oxide radical
Collapse
Affiliation(s)
- Adio J. Akamo
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Dorcas I. Akinloye
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Regina N. Ugbaja
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Oluwagbemiga O. Adeleye
- Department of Animal Production and Health, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Oluwatosin A. Dosumu
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Ofem E. Eteng
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Moses C. Antiya
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Gogonte Amah
- Department of Biochemistry, Benjamin Carson (SRN) School of Medicine, Babcock University, Ilisan, Ogun State, Nigeria
| | - Oluwafunke A. Ajayi
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Samuel O. Faseun
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| |
Collapse
|
4
|
van de Wouw J, Sorop O, van Drie RWA, Joles JA, Danser AHJ, Verhaar MC, Merkus D, Duncker DJ. Reduced nitric oxide bioavailability impairs myocardial oxygen balance during exercise in swine with multiple risk factors. Basic Res Cardiol 2021; 116:50. [PMID: 34435256 PMCID: PMC8387273 DOI: 10.1007/s00395-021-00890-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/20/2021] [Indexed: 10/28/2022]
Abstract
In the present study, we tested the hypothesis that multiple risk factors, including diabetes mellitus (DM), dyslipidaemia and chronic kidney disease (CKD) result in a loss of nitric oxide (NO) signalling, thereby contributing to coronary microvascular dysfunction. Risk factors were induced in 12 female swine by intravenous streptozotocin injections (DM), a high fat diet (HFD) and renal artery embolization (CKD). Female healthy swine (n = 13) on normal diet served as controls (Normal). After 5 months, swine were chronically instrumented and studied at rest and during exercise. DM + HFD + CKD swine demonstrated significant hyperglycaemia, dyslipidaemia and impaired kidney function compared to Normal swine. These risk factors were accompanied by coronary microvascular endothelial dysfunction both in vivo and in isolated small arteries, due to a reduced NO bioavailability, associated with perturbations in myocardial oxygen balance at rest and during exercise. NO synthase inhibition caused coronary microvascular constriction in exercising Normal swine, but had no effect in DM + HFD + CKD animals, while inhibition of phosphodiesterase 5 produced similar vasodilator responses in both groups, indicating that loss of NO bioavailability was principally responsible for the observed coronary microvascular dysfunction. This was associated with an increase in myocardial 8-isoprostane levels and a decrease in antioxidant capacity, while antioxidants restored the vasodilation to bradykinin in isolated coronary small arteries, suggesting that oxidative stress was principally responsible for the reduced NO bioavailability. In conclusion, five months of combined exposure to DM + HFD + CKD produces coronary endothelial dysfunction due to impaired NO bioavailability, resulting in impaired myocardial perfusion at rest and during exercise.
Collapse
Affiliation(s)
- Jens van de Wouw
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands
| | - Oana Sorop
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands
| | - Ruben W A van Drie
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne Merkus
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands.,Walter Brendel Center of Experimental Medicine (WBex), University Clinic Munich, 81377, LMU Munich, Germany.,German Center for Cardiovascular Research (DZHK), Munich Heart Alliance (MHA), Partner Site Munich, 81377, Munich, Germany
| | - Dirk J Duncker
- Department of Cardiology, Division of Experimental Cardiology, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, Netherlands.
| |
Collapse
|
5
|
Suriany S, Xu I, Liu H, Ulker P, Fernandez GE, Sposto R, Borzage M, Wenby R, Meiselman HJ, Forman HJ, Coates TD, Detterich JA. Individual red blood cell nitric oxide production in sickle cell anemia: Nitric oxide production is increased and sickle shaped cells have unique morphologic change compared to discoid cells. Free Radic Biol Med 2021; 171:143-155. [PMID: 33974976 DOI: 10.1016/j.freeradbiomed.2021.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
Sickle cell anemia (SCA) is characterized by decreased red blood cell (RBC) deformability due to polymerization of deoxygenated hemoglobin, leading to abnormal mechanical properties of RBC, increased cellular adhesion, and microcirculatory obstruction. Prior work has demonstrated that NO• influences RBC hydration and deformability and is produced at a basal rate that increases under shear stress in normal RBC. Nevertheless, the origin and physiological relevance of nitric oxide (NO•) production and scavenging in RBC remains unclear. We aimed to assess the basal and shear-mediated production of NO• in RBC from SCA patients and control (CTRL) subjects. RBCs loaded with a fluorescent NO• detector, DAF-FM (4-Amino-5-methylamino- 2',7'-difluorofluorescein diacetate), were imaged in microflow channels over 30-min without shear stress, followed by a 30-min period under 0.5Pa shear stress. We utilized non-specific nitric oxide synthase (NOS) blockade and carbon monoxide (CO) saturation of hemoglobin to assess the contribution of NOS and hemoglobin, respectively, to NO• production. Quantification of DAF-FM fluorescence intensity in individual RBC showed an increase in NO• in SCA RBC at the start of the basal period; however, both SCA and CTRL RBC increased NO• by a similar quantity under shear. A subpopulation of sickle-shaped RBC exhibited lower basal NO• production compared to discoid RBC from SCA group, and under shear became more circular in the direction of shear when compared to discoid RBC from SCA and CTRL, which elongated. Both CO and NOS inhibition caused a decrease in basal NO• production. Shear-mediated NO• production was decreased by CO in all RBC, but was decreased by NOS blockade only in SCA. In conclusion, total NO• production is increased and shear-mediated NO• production is preserved in SCA RBC in a NOS-dependent manner. Sickle shaped RBC with inclusions have higher NO• production and they become more circular rather than elongated with shear.
Collapse
Affiliation(s)
- Silvie Suriany
- Division of Cardiology, Children's Hospital of Los Angeles, USA
| | - Iris Xu
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Honglei Liu
- Division of Cardiology, Children's Hospital of Los Angeles, USA
| | - Pinar Ulker
- Department of Physiology, Akdeniz University, Turkey
| | | | - Richard Sposto
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Matthew Borzage
- Fetal and Neonatal Institute, Division of Neonatology Children's Hospital Los Angeles, USA
| | - Rosalinda Wenby
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, USA
| | - Herbert J Meiselman
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, USA
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, USA
| | - Thomas D Coates
- Division of Hematology, Children's Hospital of Los Angeles, USA
| | - Jon A Detterich
- Division of Cardiology, Children's Hospital of Los Angeles, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
6
|
Barbarino F, Wäschenbach L, Cavalho-Lemos V, Dillenberger M, Becker K, Gohlke H, Cortese-Krott MM. Targeting spectrin redox switches to regulate the mechanoproperties of red blood cells. Biol Chem 2020; 402:317-331. [PMID: 33544503 DOI: 10.1515/hsz-2020-0293] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022]
Abstract
The mechanical properties of red blood cells (RBCs) are fundamental for their physiological role as gas transporters. RBC flexibility and elasticity allow them to survive the hemodynamic changes in the different regions of the vascular tree, to dynamically contribute to the flow thereby decreasing vascular resistance, and to deform during the passage through narrower vessels. RBC mechanoproperties are conferred mainly by the structural characteristics of their cytoskeleton, which consists predominantly of a spectrin scaffold connected to the membrane via nodes of actin, ankyrin and adducin. Changes in redox state and treatment with thiol-targeting molecules decrease the deformability of RBCs and affect the structure and stability of the spectrin cytoskeleton, indicating that the spectrin cytoskeleton may contain redox switches. In this perspective review, we revise current knowledge about the structural and functional characterization of spectrin cysteine redox switches and discuss the current lines of research aiming to understand the role of redox regulation on RBC mechanical properties. These studies may provide novel functional targets to modulate RBC function, blood viscosity and flow, and tissue perfusion in disease conditions.
Collapse
Affiliation(s)
- Frederik Barbarino
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Postfach 128, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Lucas Wäschenbach
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Virginia Cavalho-Lemos
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Postfach 128, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Melissa Dillenberger
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, D-35392, Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, D-35392, Giessen, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
- John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, D-52425, Jülich, Germany
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Postfach 128, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| |
Collapse
|
7
|
Wischmann P, Kuhn V, Suvorava T, Muessig JM, Fischer JW, Isakson BE, Haberkorn SM, Flögel U, Schrader J, Jung C, Cortese-Krott MM, Heusch G, Kelm M. Anaemia is associated with severe RBC dysfunction and a reduced circulating NO pool: vascular and cardiac eNOS are crucial for the adaptation to anaemia. Basic Res Cardiol 2020; 115:43. [PMID: 32533377 PMCID: PMC7293199 DOI: 10.1007/s00395-020-0799-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023]
Abstract
Anaemia is frequently present in patients with acute myocardial infarction (AMI) and contributes to an adverse prognosis. We hypothesised that, besides reduced oxygen carrying capacity, anaemia is associated with (1) red blood cell (RBC) dysfunction and a reduced circulating nitric oxide (NO) pool, (2) compensatory enhancement of vascular and cardiac endothelial nitric oxide synthase (eNOS) activity, and (3) contribution of both, RBC dysfunction and reduced circulatory NO pool to left ventricular (LV) dysfunction and fatal outcome in AMI. In mouse models of subacute and chronic anaemia from repeated mild blood loss the circulating NO pool, RBC, cardiac and vascular function were analysed at baseline and in reperfused AMI. In anaemia, RBC function resulted in profound changes in membrane properties, enhanced turnover, haemolysis, dysregulation of intra-erythrocytotic redox state, and RBC-eNOS. RBC from anaemic mice and from anaemic patients with acute coronary syndrome impaired the recovery of contractile function of isolated mouse hearts following ischaemia/reperfusion. In anaemia, the circulating NO pool was reduced. The cardiac and vascular adaptation to anaemia was characterised by increased arterial eNOS expression and activity and an eNOS-dependent increase of end-diastolic left ventricular volume. Endothelial dysfunction induced through genetic or pharmacologic reduction of eNOS-activity abrogated the anaemia-induced cardio-circulatory compensation. Superimposed AMI was associated with decreased survival. In summary, moderate blood loss anaemia is associated with severe RBC dysfunction and reduced circulating NO pool. Vascular and cardiac eNOS are crucial for the cardio-circulatory adaptation to anaemia. RBC dysfunction together with eNOS dysfunction may contribute to adverse outcomes in AMI.
Collapse
Affiliation(s)
- Patricia Wischmann
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Viktoria Kuhn
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Johanna M Muessig
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jens W Fischer
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Department of Pharmacology and Clinical Pharmacology, Heinrich-Heine University, Düsseldorf, Germany
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sebastian M Haberkorn
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Ulrich Flögel
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Jürgen Schrader
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Department of Molecular Cardiology, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany.,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonary Diseases, and Vascular Medicine, Medical Faculty, CARID Cardiovascular Research Institute of Duesseldorf, Heinrich Heine University of Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Division of Cardiology, Pulmonary Diseases and Vascular Medicine, University Hospital of Duesseldorf, Düsseldorf, Germany. .,Cardiovascular Research Laboratory, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
8
|
Tai YH, Chu YH, Wu HL, Lin SM, Tsou MY, Huang CH, Chang HH, Lu CC. High-dose nitroglycerin administered during rewarming preserves erythrocyte deformability in cardiac surgery with cardiopulmonary bypass. Microcirculation 2020; 27:e12608. [PMID: 31991513 DOI: 10.1111/micc.12608] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We aimed to determine whether high-dose nitroglycerin, a nitric oxide donor, preserves erythrocyte deformability during cardiopulmonary bypass and examines the signaling pathway of nitric oxide in erythrocytes. METHODS In a randomized and controlled fashion, forty-two patients undergoing cardiac surgery with hypothermic cardiopulmonary bypass were allocated to high-dose (N = 21) and low-dose groups (N = 21). During rewarming period, patients were given intravenous nitroglycerin with an infusion rate 5 and 1 µg·kg-1 ·min-1 in high-dose and low-dose groups, respectively. Tyrosine phosphorylation level of non-muscle myosin IIA in erythrocyte membrane was used as an index of erythrocyte deformability and analyzed using immunoblotting. RESULTS Tyrosine phosphorylation of non-muscle myosin IIA was significantly enhanced after bypass in high-dose group (3.729 ± 1.700 folds, P = .011) but not low-dose group (1.545 ± 0.595 folds, P = .076). Phosphorylation of aquaporin 1, vasodilator-stimulated phosphoprotein, and focal adhesion kinase in erythrocyte membrane was also upregulated in high-dose group after bypass. Besides, plasma nitric oxide level was highly correlated with fold change of non-muscle myosin IIA phosphorylation (Pearson's correlation coefficient .871). CONCLUSIONS High-dose nitroglycerin administered during cardiopulmonary bypass improves erythrocyte deformability through activating phosphorylation of aquaporin 1, vasodilator-stimulated phosphoprotein, and focal adhesion kinase in erythrocytes.
Collapse
Affiliation(s)
- Ying-Hsuan Tai
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - You-Hsiang Chu
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsiang-Ling Wu
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Su-Man Lin
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Mei-Yung Tsou
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Hsiung Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsiao-Huang Chang
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Cherng Lu
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Aerospace Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
9
|
Premont RT, Reynolds JD, Zhang R, Stamler JS. Role of Nitric Oxide Carried by Hemoglobin in Cardiovascular Physiology: Developments on a Three-Gas Respiratory Cycle. Circ Res 2019; 126:129-158. [PMID: 31590598 DOI: 10.1161/circresaha.119.315626] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A continuous supply of oxygen is essential for the survival of multicellular organisms. The understanding of how this supply is regulated in the microvasculature has evolved from viewing erythrocytes (red blood cells [RBCs]) as passive carriers of oxygen to recognizing the complex interplay between Hb (hemoglobin) and oxygen, carbon dioxide, and nitric oxide-the three-gas respiratory cycle-that insures adequate oxygen and nutrient delivery to meet local metabolic demand. In this context, it is blood flow and not blood oxygen content that is the main driver of tissue oxygenation by RBCs. Herein, we review the lines of experimentation that led to this understanding of RBC function; from the foundational understanding of allosteric regulation of oxygen binding in Hb in the stereochemical model of Perutz, to blood flow autoregulation (hypoxic vasodilation governing oxygen delivery) observed by Guyton, to current understanding that centers on S-nitrosylation of Hb (ie, S-nitrosohemoglobin; SNO-Hb) as a purveyor of oxygen-dependent vasodilatory activity. Notably, hypoxic vasodilation is recapitulated by native S-nitrosothiol (SNO)-replete RBCs and by SNO-Hb itself, whereby SNO is released from Hb and RBCs during deoxygenation, in proportion to the degree of Hb deoxygenation, to regulate vessels directly. In addition, we discuss how dysregulation of this system through genetic mutation in Hb or through disease is a common factor in oxygenation pathologies resulting from microcirculatory impairment, including sickle cell disease, ischemic heart disease, and heart failure. We then conclude by identifying potential therapeutic interventions to correct deficits in RBC-mediated vasodilation to improve oxygen delivery-steps toward effective microvasculature-targeted therapies. To the extent that diseases of the heart, lungs, and blood are associated with impaired tissue oxygenation, the development of new therapies based on the three-gas respiratory system have the potential to improve the well-being of millions of patients.
Collapse
Affiliation(s)
- Richard T Premont
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| | - James D Reynolds
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Anesthesiology and Perioperative Medicine (J.D.R.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| | - Rongli Zhang
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Medicine, Cardiovascular Research Institute (R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH
| | - Jonathan S Stamler
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Medicine, Cardiovascular Research Institute (R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| |
Collapse
|
10
|
Diederich L, Suvorava T, Sansone R, Keller TCS, Barbarino F, Sutton TR, Kramer CM, Lückstädt W, Isakson BE, Gohlke H, Feelisch M, Kelm M, Cortese-Krott MM. On the Effects of Reactive Oxygen Species and Nitric Oxide on Red Blood Cell Deformability. Front Physiol 2018; 9:332. [PMID: 29867516 PMCID: PMC5958211 DOI: 10.3389/fphys.2018.00332] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/16/2018] [Indexed: 01/08/2023] Open
Abstract
The main function of red blood cells (RBCs) is the transport of respiratory gases along the vascular tree. To fulfill their task, RBCs are able to elastically deform in response to mechanical forces and, pass through the narrow vessels of the microcirculation. Decreased RBC deformability was observed in pathological conditions linked to increased oxidative stress or decreased nitric oxide (NO) bioavailability, like hypertension. Treatments with oxidants and with NO were shown to affect RBC deformability ex vivo, but the mechanisms underpinning these effects are unknown. In this study we investigate whether changes in intracellular redox status/oxidative stress or nitrosation reactions induced by reactive oxygen species (ROS) or NO may affect RBC deformability. In a case-control study comparing RBCs from healthy and hypertensive participants, we found that RBC deformability was decreased, and levels of ROS were increased in RBCs from hypertensive patients as compared to RBCs from aged-matched healthy controls, while NO levels in RBCs were not significantly different. To study the effects of oxidants on RBC redox state and deformability, RBCs from healthy volunteers were treated with increasing concentrations of tert-butylhydroperoxide (t-BuOOH). We found that high concentrations of t-BuOOH (≥ 1 mM) significantly decreased the GSH/GSSG ratio in RBCs, decreased RBC deformability and increased blood bulk viscosity. Moreover, RBCs from Nrf2 knockout (KO) mice, a strain genetically deficient in a number of antioxidant/reducing enzymes, were more susceptible to t-BuOOH-induced impairment in RBC deformability as compared to wild type (WT) mice. To study the role of NO in RBC deformability we treated RBC suspensions from human volunteers with NO donors and nitrosothiols and analyzed deformability of RBCs from mice lacking the endothelial NO synthase (eNOS). We found that NO donors induced S-nitrosation of the cytoskeletal protein spectrin, but did not affect human RBC deformability or blood bulk viscosity; moreover, under unstressed conditions RBCs from eNOS KO mice showed fully preserved RBC deformability as compared to WT mice. Pre-treatment of human RBCs with nitrosothiols rescued t-BuOOH-mediated loss of RBC deformability. Taken together, these findings suggest that NO does not affect RBC deformability per se, but preserves RBC deformability in conditions of oxidative stress.
Collapse
Affiliation(s)
- Lukas Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Roberto Sansone
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - T C Stevenson Keller
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Frederik Barbarino
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Thomas R Sutton
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Wiebke Lückstädt
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Holger Gohlke
- Faculty of Mathematics and Natural Sciences, Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
11
|
Abstract
Erythrocytes regulate vascular function through the modulation of oxygen delivery and the scavenging and generation of nitric oxide (NO). First, hemoglobin inside the red blood cell binds oxygen in the lungs and delivers it to tissues throughout the body in an allosterically regulated process, modulated by oxygen, carbon dioxide and proton concentrations. The vasculature responds to low oxygen tensions through vasodilation, further recruiting blood flow and oxygen carrying erythrocytes. Research has shown multiple mechanisms are at play in this classical hypoxic vasodilatory response, with a potential role of red cell derived vasodilatory molecules, such as nitrite derived nitric oxide and red blood cell ATP, considered in the last 20 years. According to these hypotheses, red blood cells release vasodilatory molecules under low oxygen pressures. Candidate molecules released by erythrocytes and responsible for hypoxic vasodilation are nitric oxide, adenosine triphosphate and S-nitrosothiols. Our research group has characterized the biochemistry and physiological effects of the electron and proton transfer reactions from hemoglobin and other ferrous heme globins with nitrite to form NO. In addition to NO generation from nitrite during deoxygenation, hemoglobin has a high affinity for NO. Scavenging of NO by hemoglobin can cause vasoconstriction, which is greatly enhanced by cell free hemoglobin outside of the red cell. Therefore, compartmentalization of hemoglobin inside red blood cells and localization of red blood cells in the blood stream are important for healthy vascular function. Conditions where erythrocyte lysis leads to cell free hemoglobin or where erythrocytes adhere to the endothelium can result in hypertension and vaso constriction. These studies support a model where hemoglobin serves as an oxido-reductase, inhibiting NO and promoting higher vessel tone when oxygenated and reducing nitrite to form NO and vasodilate when deoxygenated.
Collapse
Affiliation(s)
- Christine C Helms
- Physics Department, University of Richmond, Richmond, VA, United States
| | - Mark T Gladwin
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Daniel B Kim-Shapiro
- Physics Department, Wake Forest University, Winston-Salem, NC, United States.,Translational Science Center, Wake Forest University, Winston-Salem, NC, United States
| |
Collapse
|
12
|
Exposure of Stored Packed Erythrocytes to Nitric Oxide Prevents Transfusion-associated Pulmonary Hypertension. Anesthesiology 2017; 125:952-963. [PMID: 27517645 DOI: 10.1097/aln.0000000000001294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Transfusion of packed erythrocytes stored for a long duration is associated with increased pulmonary arterial pressure and vascular resistance. Prolonged storage decreases erythrocyte deformability, and older erythrocytes are rapidly removed from the circulation after transfusion. The authors studied whether treating stored packed ovine erythrocytes with NO before transfusion could prevent pulmonary vasoconstriction, enhance erythrocyte deformability, and prolong erythrocyte survival after transfusion. METHODS Ovine leukoreduced packed erythrocytes were treated before transfusion with either NO gas or a short-lived NO donor. Sheep were transfused with autologous packed erythrocytes, which were stored at 4°C for either 2 ("fresh blood") or 40 days ("stored blood"). Pulmonary and systemic hemodynamic parameters were monitored before, during, and after transfusion. Transfused erythrocytes were labeled with biotin to measure their circulating lifespan. Erythrocyte deformability was assessed before and after NO treatment using a microfluidic device. RESULTS NO treatment improved the deformability of stored erythrocytes and increased the number of stored erythrocytes circulating at 1 and 24 h after transfusion. NO treatment prevented transfusion-associated pulmonary hypertension (mean pulmonary arterial pressure at 30 min of 21 ± 1 vs. 15 ± 1 mmHg in control and NO-treated packed erythrocytes, P < 0.0001). Washing stored packed erythrocytes before transfusion did not prevent pulmonary hypertension. CONCLUSIONS NO treatment of stored packed erythrocytes before transfusion oxidizes cell-free oxyhemoglobin to methemoglobin, prevents subsequent NO scavenging in the pulmonary vasculature, and limits pulmonary hypertension. NO treatment increases erythrocyte deformability and erythrocyte survival after transfusion. NO treatment might provide a promising therapeutic approach to prevent pulmonary hypertension and extend erythrocyte survival.
Collapse
|
13
|
Erkens R, Suvorava T, Kramer CM, Diederich LD, Kelm M, Cortese-Krott MM. Modulation of Local and Systemic Heterocellular Communication by Mechanical Forces: A Role of Endothelial Nitric Oxide Synthase. Antioxid Redox Signal 2017; 26:917-935. [PMID: 27927026 PMCID: PMC5455615 DOI: 10.1089/ars.2016.6904] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we discuss the role of nitric oxide (NO) as a key physiological mechanotransducer modulating both local and systemic heterocellular communication and contributing to the integrated (patho)physiology of the cardiovascular system. A deeper understanding of mechanotransduction-mediated local and systemic nodes controlling heterocellular communication between the endothelium, blood cells, and other cell types (e.g., cardiomyocytes) may suggest novel therapeutic strategies for endothelial dysfunction and cardiovascular disease. Recent Advances: Mechanical forces acting on mechanoreceptors on endothelial cells activate the endothelial NO synthase (eNOS) to produce NO. NO participates in (i) abluminal heterocellular communication, inducing vasorelaxation, and thereby regulating vascular tone and blood pressure; (ii) luminal heterocellular communication, inhibiting platelet aggregation, and controlling hemostasis; and (iii) systemic heterocellular communication, contributing to adaptive physiological processes in response to exercise and remote ischemic preconditioning. Interestingly, shear-induced eNOS-dependent activation of vascular heterocellular communication constitutes the molecular basis of all methods applied in the clinical routine for evaluation of endothelial function. Critical Issues and Future Directions: The integrated physiology of heterocellular communication is still not fully understood. Dedicated experimental models are needed to analyze messengers and mechanisms underpinning heterocellular communication in response to physical forces in the cardiovascular system (and elsewhere). Antioxid. Redox Signal. 26, 917-935.
Collapse
Affiliation(s)
- Ralf Erkens
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Lukas D Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
14
|
Kuhn V, Diederich L, Keller TCS, Kramer CM, Lückstädt W, Panknin C, Suvorava T, Isakson BE, Kelm M, Cortese-Krott MM. Red Blood Cell Function and Dysfunction: Redox Regulation, Nitric Oxide Metabolism, Anemia. Antioxid Redox Signal 2017; 26:718-742. [PMID: 27889956 PMCID: PMC5421513 DOI: 10.1089/ars.2016.6954] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Recent clinical evidence identified anemia to be correlated with severe complications of cardiovascular disease (CVD) such as bleeding, thromboembolic events, stroke, hypertension, arrhythmias, and inflammation, particularly in elderly patients. The underlying mechanisms of these complications are largely unidentified. Recent Advances: Previously, red blood cells (RBCs) were considered exclusively as transporters of oxygen and nutrients to the tissues. More recent experimental evidence indicates that RBCs are important interorgan communication systems with additional functions, including participation in control of systemic nitric oxide metabolism, redox regulation, blood rheology, and viscosity. In this article, we aim to revise and discuss the potential impact of these noncanonical functions of RBCs and their dysfunction in the cardiovascular system and in anemia. CRITICAL ISSUES The mechanistic links between changes of RBC functional properties and cardiovascular complications related to anemia have not been untangled so far. FUTURE DIRECTIONS To allow a better understanding of the complications associated with anemia in CVD, basic and translational science studies should be focused on identifying the role of noncanonical functions of RBCs in the cardiovascular system and on defining intrinsic and/or systemic dysfunction of RBCs in anemia and its relationship to CVD both in animal models and clinical settings. Antioxid. Redox Signal. 26, 718-742.
Collapse
Affiliation(s)
- Viktoria Kuhn
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Lukas Diederich
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - T C Stevenson Keller
- 2 Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Christian M Kramer
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Wiebke Lückstädt
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Christina Panknin
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Tatsiana Suvorava
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Brant E Isakson
- 2 Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Malte Kelm
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- 1 Cardiovascular Research Laboratory, Division of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
15
|
Cobbold SA, Llinás M, Kirk K. Sequestration and metabolism of host cell arginine by the intraerythrocytic malaria parasite Plasmodium falciparum. Cell Microbiol 2016; 18:820-30. [PMID: 26633083 DOI: 10.1111/cmi.12552] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/14/2015] [Accepted: 11/26/2015] [Indexed: 11/26/2022]
Abstract
Human erythrocytes have an active nitric oxide synthase, which converts arginine into citrulline and nitric oxide (NO). NO serves several important functions, including the maintenance of normal erythrocyte deformability, thereby ensuring efficient passage of the red blood cell through narrow microcapillaries. Here, we show that following invasion by the malaria parasite Plasmodium falciparum the arginine pool in the host erythrocyte compartment is sequestered and metabolized by the parasite. Arginine from the extracellular medium enters the infected cell via endogenous host cell transporters and is taken up by the intracellular parasite by a high-affinity cationic amino acid transporter at the parasite surface. Within the parasite arginine is metabolized into citrulline and ornithine. The uptake and metabolism of arginine by the parasite deprive the erythrocyte of the substrate required for NO production and may contribute to the decreased deformability of infected erythrocytes.
Collapse
Affiliation(s)
- Simon A Cobbold
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA.,Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Manuel Llinás
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA.,Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Kiaran Kirk
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| |
Collapse
|
16
|
Heiss C, Sansone R, Karimi H, Krabbe M, Schuler D, Rodriguez-Mateos A, Kraemer T, Cortese-Krott MM, Kuhnle GGC, Spencer JPE, Schroeter H, Merx MW, Kelm M. Impact of cocoa flavanol intake on age-dependent vascular stiffness in healthy men: a randomized, controlled, double-masked trial. AGE (DORDRECHT, NETHERLANDS) 2015; 37:9794. [PMID: 26013912 PMCID: PMC4444618 DOI: 10.1007/s11357-015-9794-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 05/15/2015] [Indexed: 05/20/2023]
Abstract
Increased vascular stiffness, endothelial dysfunction, and isolated systolic hypertension are hallmarks of vascular aging. Regular cocoa flavanol (CF) intake can improve vascular function in healthy young and elderly at-risk individuals. However, the mechanisms underlying CF bioactivity remain largely unknown. We investigated the effects of CF intake on cardiovascular function in healthy young and elderly individuals without history, signs, or symptoms of cardiovascular disease by applying particular focus on functional endpoints relevant to cardiovascular aging. In a randomized, controlled, double-masked, parallel-group dietary intervention trial, 22 young (<35 years) and 20 elderly (50-80 year) healthy, male non-smokers consumed either a CF-containing drink (450 mg CF) or nutrient-matched, CF-free control drink bi-daily for 14 days. The primary endpoint was endothelial function as measured by flow-mediated vasodilation (FMD). Secondary endpoints included cardiac output, vascular stiffness, conductance of conduit and resistance arteries, and perfusion in the microcirculation. Following 2 weeks of CF intake, FMD improved in young (6.1 ± 0.7 vs. 7.6 ± 0.7 %, p < 0.001) and elderly (4.9 ± 0.6 vs. 6.3 ± 0.9 %, p < 0.001). Secondary outcomes demonstrated in both groups that CF intake decreased pulse wave velocity and lowered total peripheral resistance, and increased arteriolar and microvascular vasodilator capacity, red cell deformability, and diastolic blood pressure, while cardiac output remained affected. In the elderly, baseline systolic blood pressure was elevated, driven by an arterial-stiffness-related augmentation. CF intake decreased aortic augmentation index (-9 %) and thus systolic blood pressure (-7 mmHg; Clinicaltrials.gov: NCT01639781). CF intake reverses age-related burden of cardiovascular risk in healthy elderly, highlighting the potential of dietary flavanols to maintain cardiovascular health.
Collapse
Affiliation(s)
- Christian Heiss
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Moorenstr. 5, 40225, Duesseldorf, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Since the identification of the elusive endothelium-derived relaxing factor as nitric oxide (NO), much attention has been devoted to understanding its physiological effects. NO is a free radical with many roles, and owing to its neutral charge and high diffusion capacity, it appears NO is involved in every mammalian biological system. Most attention has been focused on the NO generating pathways within the endothelium; however, the recent discovery of a NO synthase (NOS)-like enzyme residing in red blood cells (RBC) has increased our understanding of the blood flow and oxygen delivery modulation by RBC. In the present review, pathways of NO generation are summarized, with attention to those residing within RBC. While the bioactivity of RBC-derived NO is still debated due to its generation within proximity of NO scavengers, current theories for NO export from RBC are explored, which are supported by recent findings demonstrating an extracellular response to RBC-derived NO. The importance of NO in the active regulation of RBC deformability is discussed in the context of the subsequent effects on blood fluidity, and the complex interplay between blood rheology and NO are summarized. This review provides a summary of recent advances in understanding the role played by RBC in NO equilibrium and vascular regulation.
Collapse
Affiliation(s)
- Michael J Simmonds
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Queensland, Australia
| | - Jon A Detterich
- Division of Cardiology, Childrens Hospital Los Angeles, Los Angeles, CA, USA
| | - Philippe Connes
- UMR Inserm 1134, Hôpital Ricou, CHU de Pointe à Pitre, Pointe à Pitre, Guadeloupe Institut Universitaire de France, Paris, France Laboratory of Excellence GR-Ex "The red cell: from genesis to death", PRES Sorbonne Paris Cité, Paris, France
| |
Collapse
|
18
|
Nitric oxide synthetic pathway in patients with microvascular angina and its relations with oxidative stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:726539. [PMID: 24864190 PMCID: PMC4016928 DOI: 10.1155/2014/726539] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 03/28/2014] [Accepted: 03/29/2014] [Indexed: 11/17/2022]
Abstract
A decreased nitric oxide (NO) bioavailability and an increased oxidative stress play a pivotal role in different cardiovascular pathologies. As red blood cells (RBCs) participate in NO formation in the bloodstream, the aim of this study was to outline the metabolic profile of L-arginine (Arg)/NO pathway and of oxidative stress status in RBCs and in plasma of patients with microvascular angina (MVA), investigating similarities and differences with respect to coronary artery disease (CAD) patients or healthy controls (Ctrl). Analytes involved in Arg/NO pathway and the ratio of oxidized and reduced forms of glutathione were measured by LC-MS/MS. The arginase and the NO synthase (NOS) expression were evaluated by immunofluorescence staining. RBCs from MVA patients show increased levels of NO synthesis inhibitors, parallel to that found in plasma, and a reduction of NO synthase expression. When summary scores were computed, both patient groups were associated with a positive oxidative score and a negative NO score, with the CAD group located in a more extreme position with respect to Ctrl. This finding points out to an impairment of the capacity of RBCs to produce NO in a pathological condition characterized mostly by alterations at the microvascular bed with no significant coronary stenosis.
Collapse
|
19
|
Cortese-Krott MM, Kelm M. Endothelial nitric oxide synthase in red blood cells: key to a new erythrocrine function? Redox Biol 2014; 2:251-8. [PMID: 24494200 PMCID: PMC3909820 DOI: 10.1016/j.redox.2013.12.027] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 12/21/2013] [Indexed: 02/06/2023] Open
Abstract
Red blood cells (RBC) have been considered almost exclusively as a transporter of metabolic gases and nutrients for the tissues. It is an accepted dogma that RBCs take up and inactivate endothelium-derived NO via rapid reaction with oxyhemoglobin to form methemoglobin and nitrate, thereby limiting NO available for vasodilatation. Yet it has also been shown that RBCs not only act as "NO sinks", but exert an erythrocrine function - i.e an endocrine function of RBC - by synthesizing, transporting and releasing NO metabolic products and ATP, thereby potentially controlling systemic NO bioavailability and vascular tone. Recent work from our and others laboratory demonstrated that human RBCs carry an active type 3, endothelial NO synthase (eNOS), constitutively producing NO under normoxic conditions, the activity of which is compromised in patients with coronary artery disease. In this review we aim to discuss the potential role of red cell eNOS in RBC signaling and function, and to critically revise evidence to this date showing a role of non-endothelial circulating eNOS in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
20
|
Depletion of circulating blood NOS3 increases severity of myocardial infarction and left ventricular dysfunction. Basic Res Cardiol 2013; 109:398. [PMID: 24346018 PMCID: PMC3898535 DOI: 10.1007/s00395-013-0398-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 11/25/2013] [Accepted: 12/06/2013] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) derived from endothelial NO synthase (NOS3) plays a central role in myocardial ischemia/reperfusion (I/R)-injury. Subsets of circulating blood cells, including red blood cells (RBCs), carry a NOS3 and contribute to blood pressure regulation and RBC nitrite/nitrate formation. We hypothesized that the circulating blood born NOS3 also modulates the severity of myocardial infarction in disease models. We cross-transplanted bone marrow in wild-type and NOS3−/− mice with wild-type mice, producing chimeras expressing NOS3 only in vascular endothelium (BC−/EC+) or in both blood cells and vascular endothelium (BC+/EC+). After 60-min closed-chest coronary occlusion followed by 24 h reperfusion, cardiac function, infarct size (IS), NOx levels, RBCs NO formation, RBC deformability, and vascular reactivity were assessed. At baseline, BC−/EC+ chimera had lower nitrite levels in blood plasma (BC−/EC+: 2.13 ± 0.27 μM vs. BC+/EC+ 3.17 ± 0.29 μM; *p < 0.05), reduced DAF FM associated fluorescence within RBCs (BC−/EC+: 538.4 ± 12.8 mean fluorescence intensity (MFI) vs. BC+/EC+: 619.6 ± 6.9 MFI; ***p < 0.001) and impaired erythrocyte deformability (BC−/EC+: 0.33 ± 0.01 elongation index (EI) vs. BC+/EC+: 0.36 ± 0.06 EI; *p < 0.05), while vascular reactivity remained unaffected. Area at risk did not differ, but infarct size was higher in BC−/EC+ (BC−/EC+: 26 ± 3 %; BC+/EC+: 14 ± 2 %; **p < 0.01), resulting in decreased ejection fraction (BC−/EC+ 46 ± 2 % vs. BC+/EC+: 52 ± 2 %; *p < 0.05) and increased end-systolic volume. Application of the NOS inhibitor S-ethylisothiourea hydrobromide was associated with larger infarct size in BC+/EC+, whereas infarct size in BC−/EC+ mice remained unaffected. Reduced infarct size, preserved cardiac function, NO levels in RBC and RBC deformability suggest a modulating role of circulating NOS3 in an acute model of myocardial I/R in chimeric mice.
Collapse
|
21
|
Zhu XY, Li P, Yang YB, Liu ML. Xuezhikang, extract of red yeast rice, improved abnormal hemorheology, suppressed caveolin-1 and increased eNOS expression in atherosclerotic rats. PLoS One 2013; 8:e62731. [PMID: 23675421 PMCID: PMC3651163 DOI: 10.1371/journal.pone.0062731] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Accepted: 03/24/2013] [Indexed: 11/25/2022] Open
Abstract
Background Xuezhikang is the extract of red yeast rice, which has been widely used for the management of atherosclerotic disease, but the molecular basis of its antiatherosclerotic effects has not yet been fully identified. Here we investigated the changes of eNOS in vascular endothelia and RBCs, eNOS regulatory factor Caveolin-1 in endothelia, and hemorheological parameters in atherosclerotic rats to explore the protective effects of Xuezhikang. Methodology/Principal Findings Wistar rats were divided into 4 groups (n = 12/group) group C, controls; group M, high-cholesterol diet (HCD) induced atherosclerotic models; group X, HCD+Xuezhikang; and group L, HCD +Lovastatin. In group X, Xuezhikang inhibited oxidative stress, down-regulated caveolin-1 in aorta wall (P<0.05), up-regulated eNOS expression in vascular endothelia and erythrocytes (P<0.05), increased NOx (nitrite and nitrate) in plasma and cGMP in erythrocyte plasma and aorta wall (P<0.05), increased erythrocyte deformation index (EDI), and decreased whole blood viscosity and plasma viscosity (P<0.05), with the improvement of arterial pathology. Conclusions/Significance Xuezhikang up-regulated eNOS expression in vascular endothelia and RBCs, increased plasma NOx and improved abnormal hemorheology in high cholesterol diet induced atherosclerotic rats. The elevated eNOS/NO and improved hemorheology may be beneficial to atherosclerotic disease.
Collapse
Affiliation(s)
- Xin-Yuan Zhu
- Department of Geriatrics, Peking University First Hospital, Beijing, China
| | | | | | | |
Collapse
|
22
|
Human red blood cells at work: identification and visualization of erythrocytic eNOS activity in health and disease. Blood 2012; 120:4229-37. [PMID: 23007404 DOI: 10.1182/blood-2012-07-442277] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A nitric oxide synthase (NOS)-like activity has been demonstrated in human red blood cells (RBCs), but doubts about its functional significance, isoform identity and disease relevance remain. Using flow cytometry in combination with the nitric oxide (NO)-imaging probe DAF-FM we find that all blood cells form NO intracellularly, with a rank order of monocytes > neutrophils > lymphocytes > RBCs > platelets. The observation of a NO-related fluorescence within RBCs was unexpected given the abundance of the NO-scavenger oxyhemoglobin. Constitutive normoxic NO formation was abolished by NOS inhibition and intracellular NO scavenging, confirmed by laser-scanning microscopy and unequivocally validated by detection of the DAF-FM reaction product with NO using HPLC and LC-MS/MS. Using immunoprecipitation, ESI-MS/MS-based peptide sequencing and enzymatic assay we further demonstrate that human RBCs contain an endothelial NOS (eNOS) that converts L-(3)H-arginine to L-(3)H-citrulline in a Ca(2+)/calmodulin-dependent fashion. Moreover, in patients with coronary artery disease, red cell eNOS expression and activity are both lower than in age-matched healthy individuals and correlate with the degree of endothelial dysfunction. Thus, human RBCs constitutively produce NO under normoxic conditions via an active eNOS isoform, the activity of which is compromised in patients with coronary artery disease.
Collapse
|
23
|
Kanias T, Wang L, Lippert A, Kim-Shapiro DB, Gladwin MT. Red blood cell endothelial nitric oxide synthase does not modulate red blood cell storage hemolysis. Transfusion 2012; 53:981-9. [PMID: 22897637 DOI: 10.1111/j.1537-2995.2012.03850.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The red blood cell (RBC) endothelial nitric oxide synthase (eNOS) has been shown to regulate intrinsic RBC rheologic properties, such as membrane deformability, suggesting that a functional eNOS could be important in RBC viability and function during storage. This study examines the correlation between RBC eNOS deficiency and the propensity of RBCs to hemolyze under selected stress conditions including prolonged hypothermic storage. STUDY DESIGN AND METHODS Fresh or stored RBCs from normal and eNOS knockout (KO) mice or from healthy human volunteers were subjected to selected hemolytic stress conditions including mechanical stress hemolysis, osmotic stress hemolysis, and oxidation stress hemolysis and evaluated during standard storage in CPDA-1 solutions. RESULTS Fresh RBCs from normal and eNOS KO mice demonstrated comparable susceptibility to hemolysis triggered by mechanical stress (mechanical fragility index 6.5 ± 0.5 in eNOS KO vs. 6.4 ± 0.4 for controls; n = 8-9), osmotic stress, and oxidative stress. Additionally, RBCs from both mouse groups exhibited similar hemolytic profile at the end of 14-day hypothermic storage, analogous to 42 days of human RBC storage. Storage of human RBCs (28 days in CPDA-1) in the presence of NOS cofactors (L-arginine and tetrahydro-L-biopterin) or inhibitor (N(5) -[imino(methylamino)methyl]-L-ornithine monoacetate) did not affect cell recovery or hemolytic response to the selected stressors. CONCLUSION These studies suggest that RBC eNOS does not modulate susceptibility to hemolysis in response to selected stress conditions or prolonged hypothermic storage. Other strategies to increase nitric oxide (NO) bioactivity after prolonged storage utilizing NOS-independent pathways such as the nitrate-nitrite-NO pathway may prove a more promising approach.
Collapse
Affiliation(s)
- Tamir Kanias
- Vascular Medicine Institute and the Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|
24
|
Chen SL, Xie Z, Carson PL, Wang X, Guo LJ. In vivo flow speed measurement of capillaries by photoacoustic correlation spectroscopy. OPTICS LETTERS 2011; 36:4017-9. [PMID: 22002371 PMCID: PMC3319062 DOI: 10.1364/ol.36.004017] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We recently proposed photoacoustic correlation spectroscopy (PACS) and demonstrated a proof-of-concept experiment. Here we use the technique for in vivo flow speed measurement in capillaries in a chick embryo model. The photoacoustic microscopy system is used to render high spatial resolution and high sensitivity, enabling sufficient signals from single red blood cells. The probe beam size is calibrated by a blood-mimicking phantom. The results indicate the feasibility of using PACS to study flow speeds in capillaries.
Collapse
Affiliation(s)
- Sung-Liang Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|