Skip to main content
Erschienen in: Basic Research in Cardiology 4/2020

Open Access 01.07.2020 | Editorial

Interplay of the red blood cell and vascular endothelial nitric oxide synthase system to combat cardiac complications of anemia

verfasst von: Andreas Daiber, Thomas Münzel

Erschienen in: Basic Research in Cardiology | Ausgabe 4/2020

Hinweise
This comment refers to the article available at https://​doi.​org/​10.​1007/​s00395-020-0799-x.

The cardiovascular protective profile of endothelial nitric oxide synthase (eNOS) and nitric oxide (·NO)

The mechanism of cardioprotection described in the clinically important study by Wischmann et al. is based on the assumption of a compensatory enhancement of vascular endothelial nitric oxide synthase (eNOS) activity and nitric oxide (·NO) formation in anemic mice, when red blood cell (RBC) eNOS function and ·NO formation are impaired [28]. The cardioprotective properties of eNOS and ·NO are widely accepted and were extensively reviewed in the past [9, 24] and redefined in recent years [3]. The cardioprotection afforded by ·NO (e.g., from nitrovasodilators such as nitroglycerin [13]) largely depends on the prevention of mitochondrial permeability transition pore (mPTP) opening via S-nitros(yl)ation of the mPTP regulator cyclophilin D during reperfusion [4]. This mechanism reflects a major detrimental process in ischemia/reperfusion (I/R) damage leading to excessive reactive oxygen species (ROS) formation/release as well as onset of apoptotic cell death [7, 21]. Oxidative stress in general plays an important role for development and progression of cardiovascular diseases [19], especially for I/R associated events such as myocardial infarction [9]. Importantly, ·NO can directly reduce I/R-dependent ROS formation by suppression of mitochondrial respiratory complex I activity via S-nitros(yl)ation [5]. Nitric oxide is also implicated in pre-, post- and remote-conditioning, drug- and non-drug-based therapeutic concepts currently discussed for cardioprotection [2, 14, 23].
The general role of ·NO for cardioprotection is also supported by numerous reports on loss of cardioprotective effects of ·NO or ·NO-related therapies upon treatment with the inhibitor of all NOS isoforms, NG-nitro-l-arginine methyl ester (l-NAME) (only citing a few [4, 6, 15, 26]). Also exogenous administration of tetrahydrobiopterin (BH4), an essential cofactor for eNOS function, improved ischemic damage in isolated hearts subjected to I/R [27, 29]. Likewise, cardiac-specific overexpression of GTP-cyclohydrolase-1, the rate-limiting enzyme for tetrahydrobiopterin synthesis, improved ischemic preconditioning [12] and also attenuated post-infarction cardiac remodeling [16], most probably by restoration of tetrahydrobiopterin synthesis and thus by the prevention of eNOS uncoupling [10]. Further support of this concept is provided by genetic models, where eNOS knockout mice showed more pronounced ischemic damage, myocardial fibrosis and impaired left-ventricular end-diastolic volume and ejection fraction, when subjected to myocardial infarction [25]. Of note, genetic deficiency in neuronal nitric oxide synthase (nNOS) or inducible nitric oxide synthase (iNOS) did not show this aggravated ischemic damage; in contrast, iNOS knockout mice were rather protected against ischemic damage [25]. Also, the cardioprotective effects of nitroglycerin upon myocardial infarction were lost in eNOS knockout mice [4]. In addition, a cardiomyocyte-specific overexpression of eNOS largely prevented I/R injury [8]. The proof for the central role of eNOS-derived ·NO to prevent or at least attenuate ischemic heart damage was based on decreased infarct size and cardiac oxidative stress upon coronary artery ligation by therapy with the eNOS enhancer AVE9488, whereas these protective effects were virtually absent in eNOS knockout mice [11]. All these different regulators of eNOS activity, ·NO formation and endothelial function are summarized in the Fig. 1 and have been-reviewed previously [18, 22], and put into context with the novel findings by Wischmann et al. [28].

Essential interplay of red blood cell and vascular eNOS for nitric oxide-mediated cardioprotection against I/R injury in anemia

With the present studies, Wischmann et al. show that cardiovascular protection against I/R damage in response to acute myocardial infarction (AMI) is mainly based on endogenous ·NO formation from either red blood cell (RBC, e.g., by nitrite bioactivation or RBC-eNOS) or vascular/cardiac eNOS [28]. In a mouse model of anemia, the authors demonstrate that AMI-induced mortality is more pronounced due to impaired RBC-derived ·NO formation as also seen with the increase in AMI mortality upon pharmacological eNOS inhibition by l-NAME. The most deleterious outcome was observed when anemic mice were treated with l-NAME and then subjected to AMI. The demonstration that AMI did not induce a severe impairment of cardiac functional parameters in the setting of anemia was attributed to the finding that cardiac and vascular eNOS were upregulated in the anemic mice—this was suggested by the authors as a compensatory mechanism. The impaired circulating ·NO pool in anemic mice could be explained mainly by impaired RBC-derived ·NO formation due to RBC dysfunction as indicated by lower RBC hemoglobin and iron content, whereas cell-free hemoglobin was increased and RBC redox state was impaired (e.g., higher ROS levels and lower reduced glutathione levels). As a proof of concept, the authors show that transfer of RBC from anemic or eNOS knockout mice to wild type mouse hearts subjected to I/R prevented the recovery of cardiac function as compared to the wild type mouse hearts, when healthy wild type RBC were present. This proof-of-concept experiment was also confirmed using RBC from patients with acute coronary syndrome (ACS) with and without anemia. The RBC from ACS patients with anemia caused a significantly worse recovery in wild type mouse hearts that were subjected to I/R as compared to RBC from ACS patients without anemia, further substantiating the proposed concept that RBC-derived ·NO plays a major role for cardioprotection against I/R damage. In line with these observations, patients with chronic severe anemia had substantially increased forearm blood flow (measured by plethysmography) and showed more pronounced decrease in forearm blood flow upon l-NAME administration as compared to  healthy subjects [1], supporting the postulated compensatory activation of vascular eNOS in the state of dysfunctional RBC eNOS.

Implications of eNOS function and nitric oxide bioavailability in patients with ACS and anemia

Thus, in summary, the results of this highly important study demonstrate that moderate blood loss anemia is associated with severe red blood cell dysfunction and increased superoxide production, which may be related at least in part to eNOS uncoupling in RBC leading to a reduction of the ·NO pool. In addition, Wischmann et al. also established with a series of well-designed experiments for the first time that vascular and cardiac eNOS are crucial for the cardiocirculatory adaptation to anemia in particular in the setting of I/R [28]. The presented findings will help to improve therapeutic strategies in the setting of AMI and anemia. Thus, the principle target will not be solely the reduced hemoglobin level, but also the normalization of the reduced ·NO pool in erythrocytes or the enhancement of eNOS activity in the vasculature and the myocardium. This should also be considered for blood transfusion since the duration of storage of RBC in the transfusion department is negatively correlated with flow-mediated dilation measured in anemic subjects after transfusion, indicating that RBC ·NO formation capacity is impaired upon prolonged storage [20]. It remains to be established, whether nitric oxide donor therapy, tetrahydrobiopterin treatment or eNOS enhancement will decrease the cardiovascular risk of patients with anemia.

Acknowledgements

Open Access funding provided by Projekt DEAL. The authors also acknowledge the continuous support by the Foundation Heart of Mainz, the Center for Translational Vascular Biology (CTVB) of the University Medical Center Mainz and the DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany. A.D. is a member of the European COST Action EU-CARDIOPROTECTION (CA16225).

Compliance with ethical standards

Conflict of interest

All authors declare that they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Unsere Produktempfehlungen

Neuer Inhalt

Print-Titel

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Andreadou I, Iliodromitis EK, Rassaf T, Schulz R, Papapetropoulos A, Ferdinandy P (2015) The role of gasotransmitters NO, H2S and CO in myocardial ischaemia/reperfusion injury and cardioprotection by preconditioning, postconditioning and remote conditioning. Br J Pharmacol 172:1587–1606. https://doi.org/10.1111/bph.12811 CrossRefPubMed Andreadou I, Iliodromitis EK, Rassaf T, Schulz R, Papapetropoulos A, Ferdinandy P (2015) The role of gasotransmitters NO, H2S and CO in myocardial ischaemia/reperfusion injury and cardioprotection by preconditioning, postconditioning and remote conditioning. Br J Pharmacol 172:1587–1606. https://​doi.​org/​10.​1111/​bph.​12811 CrossRefPubMed
4.
Zurück zum Zitat Bibli SI, Papapetropoulos A, Iliodromitis EK, Daiber A, Randriamboavonjy V, Steven S, Brouckaert P, Chatzianastasiou A, Kypreos KE, Hausenloy DJ, Fleming I, Andreadou I (2019) Nitroglycerine limits infarct size through S-nitrosation of cyclophilin D: a novel mechanism for an old drug. Cardiovasc Res 115:625–636. https://doi.org/10.1093/cvr/cvy222 CrossRefPubMed Bibli SI, Papapetropoulos A, Iliodromitis EK, Daiber A, Randriamboavonjy V, Steven S, Brouckaert P, Chatzianastasiou A, Kypreos KE, Hausenloy DJ, Fleming I, Andreadou I (2019) Nitroglycerine limits infarct size through S-nitrosation of cyclophilin D: a novel mechanism for an old drug. Cardiovasc Res 115:625–636. https://​doi.​org/​10.​1093/​cvr/​cvy222 CrossRefPubMed
5.
Zurück zum Zitat Chouchani ET, Methner C, Nadtochiy SM, Logan A, Pell VR, Ding S, James AM, Cocheme HM, Reinhold J, Lilley KS, Partridge L, Fearnley IM, Robinson AJ, Hartley RC, Smith RA, Krieg T, Brookes PS, Murphy MP (2013) Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex I. Nat Med 19:753–759. https://doi.org/10.1038/nm.3212 CrossRefPubMedPubMedCentral Chouchani ET, Methner C, Nadtochiy SM, Logan A, Pell VR, Ding S, James AM, Cocheme HM, Reinhold J, Lilley KS, Partridge L, Fearnley IM, Robinson AJ, Hartley RC, Smith RA, Krieg T, Brookes PS, Murphy MP (2013) Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex I. Nat Med 19:753–759. https://​doi.​org/​10.​1038/​nm.​3212 CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Correa F, Buelna-Chontal M, Chagoya V, Garcia-Rivas G, Vigueras RM, Pedraza-Chaverri J, Garcia-Nino WR, Hernandez-Pando R, Leon-Contreras JC, Zazueta C (2015) Inhibition of the nitric oxide/cyclic guanosine monophosphate pathway limited the cardioprotective effect of post-conditioning in hearts with apical myocardial infarction. Eur J Pharmacol 765:472–481. https://doi.org/10.1016/j.ejphar.2015.09.018 CrossRefPubMed Correa F, Buelna-Chontal M, Chagoya V, Garcia-Rivas G, Vigueras RM, Pedraza-Chaverri J, Garcia-Nino WR, Hernandez-Pando R, Leon-Contreras JC, Zazueta C (2015) Inhibition of the nitric oxide/cyclic guanosine monophosphate pathway limited the cardioprotective effect of post-conditioning in hearts with apical myocardial infarction. Eur J Pharmacol 765:472–481. https://​doi.​org/​10.​1016/​j.​ejphar.​2015.​09.​018 CrossRefPubMed
7.
Zurück zum Zitat Di Lisa F, Bernardi P (2006) Mitochondria and ischemia-reperfusion injury of the heart: fixing a hole. Cardiovasc Res 70:191–199CrossRef Di Lisa F, Bernardi P (2006) Mitochondria and ischemia-reperfusion injury of the heart: fixing a hole. Cardiovasc Res 70:191–199CrossRef
10.
Zurück zum Zitat Forstermann U, Munzel T (2006) Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation 113:1708–1714CrossRef Forstermann U, Munzel T (2006) Endothelial nitric oxide synthase in vascular disease: from marvel to menace. Circulation 113:1708–1714CrossRef
20.
Zurück zum Zitat Neuman R, Hayek S, Rahman A, Poole JC, Menon V, Sher S, Newman JL, Karatela S, Polhemus D, Lefer DJ, De Staercke C, Hooper C, Quyyumi AA, Roback JD (2015) Effects of storage-aged red blood cell transfusions on endothelial function in hospitalized patients. Transfusion 55:782–790. https://doi.org/10.1111/trf.12919 CrossRefPubMed Neuman R, Hayek S, Rahman A, Poole JC, Menon V, Sher S, Newman JL, Karatela S, Polhemus D, Lefer DJ, De Staercke C, Hooper C, Quyyumi AA, Roback JD (2015) Effects of storage-aged red blood cell transfusions on endothelial function in hospitalized patients. Transfusion 55:782–790. https://​doi.​org/​10.​1111/​trf.​12919 CrossRefPubMed
27.
28.
Zurück zum Zitat 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 (2020) 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. https://doi.org/10.1007/s00395-020-0799-x 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 (2020) 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. https://​doi.​org/​10.​1007/​s00395-020-0799-x
Metadaten
Titel
Interplay of the red blood cell and vascular endothelial nitric oxide synthase system to combat cardiac complications of anemia
verfasst von
Andreas Daiber
Thomas Münzel
Publikationsdatum
01.07.2020
Verlag
Springer Berlin Heidelberg
Erschienen in
Basic Research in Cardiology / Ausgabe 4/2020
Print ISSN: 0300-8428
Elektronische ISSN: 1435-1803
DOI
https://doi.org/10.1007/s00395-020-0801-7

Weitere Artikel der Ausgabe 4/2020

Basic Research in Cardiology 4/2020 Zur Ausgabe

Bei Herzinsuffizienz muss „Eisenmangel“ neu definiert werden!

16.05.2024 Herzinsuffizienz Nachrichten

Bei chronischer Herzinsuffizienz macht es einem internationalen Expertenteam zufolge wenig Sinn, die Diagnose „Eisenmangel“ am Serumferritin festzumachen. Das Team schlägt vor, sich lieber an die Transferrinsättigung zu halten.

Herzinfarkt mit 85 – trotzdem noch intensive Lipidsenkung?

16.05.2024 Hypercholesterinämie Nachrichten

Profitieren nach einem akuten Myokardinfarkt auch Betroffene über 80 Jahre noch von einer intensiven Lipidsenkung zur Sekundärprävention? Um diese Frage zu beantworten, wurden jetzt Registerdaten aus Frankreich ausgewertet.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

LDL-Cholesterin kann ApoB als Risikomarker nicht ersetzen

16.05.2024 Hypercholesterinämie Nachrichten

Apolipoprotein B (ApoB) ist ein genauer Risikomarker für atherosklerotisch bedingte Erkrankungen. Aber das LDL-Cholesterin doch auch – lohnt sich also die ApoB-Messung überhaupt?

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.