scholarly journals The Role of Gα13 in Integrin β2-Dependent Neutrophil Transendothelial Migration

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1037-1037
Author(s):  
Claire W Chang ◽  
Ni Cheng ◽  
Xiaoping Du

Upon sensing chemoattractant signals, leukocytes migrate towards the chemoattractant. If the signal is extravascular, circulating leukocytes roll on the surface of endothelial cells, firmly adhere and migrate through the endothelial monolayer to the inflammation site. Transendothelial migration of leukocytes requires integrins, particularly β2 family of integrins. We previously demonstrated that the heterotrimeric G protein subunit Gα13 interacts with the cytoplasmic domains of certain integrin β subunits including β2. To understand the role of Gα13 in integrin-dependent neutrophil migration, myeloid-specific Gα13 deficient mice have been generated by cross-breeding lysozyme M-cre mice with Gα13-floxed mice. Neutrophil migration was attenuated in myeloid-specific Gα13 knockout mice in a thioglycolate (3%) induced peritonitis model in vivo, and was also attenuated in transendothelial or mouse ICAM-1-coated transwell migration assays ex vivo. In contrast, endothelial cell/ICAM-independent transwell migration was not affected. Importantly, on an ICAM-coated surface, only the speed of migration of Gα13-deficient neutrophils were attenuated but migration directionality was not affected, suggesting that Gα13 plays an important role in motility rather than chemoattractant sensing. To further determine the role of Gα13-β2 integrin interaction in leukocyte migration, we examined the effect of a peptide, MB2mP6, derived from the Gα13 binding site of integrin β2 subunit. Like Gα13 deficiency, MB2mP6 inhibited integrin-dependent neutrophil transendothelial migration, but had minimal effects on leukocyte transwell migration in the absence of endothelial cell monolayer. Gα13 plays dual role in RhoA regulation. GPCR-activated Gα13 binds to and activates RhoGEFs resulting in RhoA activation, but Gα13 binding to integrin cytoplasmic domain is associated inhibition of RhoA. Interestingly, treatment with MB2mP6 enhanced RhoA activity of neutrophils adherent on ICAM1, suggesting that MB2mP6 mainly affects Gα13-β2 interaction. These data suggest that Gα13-integrin interaction plays an important role in the integrin-dependent transendothelial migration. Thus, Gα13-β2 is important in the immune and inflammatory functions of neutrophils. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 992-992
Author(s):  
Chanchal Sur Chowdhury ◽  
Elizabeth Wareham ◽  
Juying Xu ◽  
Sachin Kumar ◽  
Ashwini S. Hinge ◽  
...  

Abstract Neutrophils traffic in and out of underlying vascular bed during hematopoiesis and immunosurveillance. However, during inflammatory conditions such as ischemia reperfusion injury or atherosclerosis, excessive neutrophil infiltration into tissue drives disease pathogenesis. Yet, the relationship between neutrophil transmigration and inflammation is ill-defined. Neutrophil extravasation can occur either between two endothelial cells (paracellular) or directly through an endothelial cell body (transcellular). During transcellular migration, neutrophils interact with underlying endothelial cells (EC) via invadosomal structures, which forms a 'pore' into endothelial cell membrane, thus facilitating neutrophil migration through EC body. We have recently reported that deficiency in Rap1b, a member of Ras superfamily of GTPase, enhanced neutrophil transcellular migration, invadosomal structures and metalloproteinase (MMP) release (Kumar et al, JEM, 2014), in a manner dependent on high Akt activity. Further, Rap1-deficiency increased neutrophil recruitment to inflamed lungs and enhanced susceptibility to endotoxin shock, suggesting mode of neutrophil migration may influence inflammatory outcome. Here, to further understand which factors drive neutrophil transcellular migration, we analyzed protein content of Rap1b-/- invadosomal structures during transcellular diapedesis. For this, neutrophils were stimulated in transwell filters of 1µM pore size, with FMLP placed in the lower chamber, allowing only invadosomal protrusions into the pores. After removing the cell body from top of the filter, mass spectrometric analysis was performed on the invadosomal fraction. About 680 proteins were identified in protrusions isolated from WT or Rap1b-/- neutrophils. As expected, majority of them were cytoskeleton and adhesion proteins. Interestingly, Rap1b-/- invadosomal structures contained more enzymes of glycolytic pathways, including HK1, Lactate dehydrogenase A (LDHA) and phosphoglycerate kinase1 (PGK1). Immunofluorescent staining and western blotting confirmed this observation. Importantly, glycolytic enzymes were present at the tip of the protrusions in colocalization with F-actin suggesting site specific glycolytic activity, raising the hypothesis that metabolic remodeling may influence the route of neutrophil migration. LDHA converts pyruvate to lactate and subsequent milieu acidification, which can then cause MMP activation. Consistently, Rap1b-/- neutrophils exhibited increased uptake of glucose analogue (2-NBDG) and concurrent intracellular acidification, as detected by pH sensitive dye. To investigate the importance of LDHA activity during transcellular migration, Rap1b-/- neutrophils were treated with a specific pharmacological inhibitor of LDHA, namely FX11. In vitro, FX11 treatment significantly decreased transcellular migration of Rap1b-/- neutrophils. It also reduced invadosome formation of Rap1b-/- neutrophils within transwell pores, as well as neutrophil acidity and MMP activity. Furthermore, during neutrophil-endothelial cell interactions in vitro, Rap1b-/- neutrophils caused F-actin depolymerization in EC, likely facilitating transcellular passage; this was inhibited by FX11. To examine its effect in vivo, under same inflammatory microenvironment, Rap1b-/- and WT neutrophils were tagged with cell tracker dyes and transferred to recipient mice, treated with FX11 or DMSO control. Ear microvasculature was stimulated with FMLP and labeled with PECAM antibody to visualize EC junctions. Rap1b-/- neutrophils migrated out of vessels at higher frequency than WT cells, which was abrogated by FX11 treatment. Moreover, treatment with FX11 reduced the number of Rap1b-/- neutrophils located away from EC junction (transcellular route), in vivo. These results suggest enhanced local glycolytic metabolism and LDHA activity could act as critical regulators of transcellular migration. Increase in extracellular acidification mediated by LDHA activity, could affect endothelial permeability and alter neutrophil migratory behavior affecting outcome of inflammation. Since milieu acidification plays a major role in ischemic damage to the heart, these findings may be clinically important for our understanding of hyperinflammatory disorders. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1297-1297
Author(s):  
Laura Palau ◽  
Jessica M. Synder ◽  
Traci Beth Topping ◽  
Cathryn Hogarth ◽  
Nina Isoherranen ◽  
...  

Abstract Hematopoietic stem cells (HSCs) reside in a complex microenvironment that enforces the balance between self-renewal and differentiation. The exact physiologic mechanisms by which the niche controls HSC fate remain elusive. Retinoic acid (RA) is a powerful morphogen that controls stem cell behavior across a variety of systems. In bone marrow (BM), mesenchymal stroma cells (MSCs) express cytochrome P450 (CYP)26 enzymes and can inactivate endogenous and pharmacological retinoids (Ghiaur G et al PNAS 2013, Su M et al. PlosOne 2015, Alonso S et al. 2016). Stromal CYP26 activity is required to maintain human HSCs ex vivo (Ghiaur G et al PNAS 2013). Here we set to study the role of CYP26 in HSC homeostasis in vivo. For this, we induced CYP26 knockout via injection of Tamoxifen in ROSA26CreERT CYP26A1loxP/loxPCYP26B1loxP/loxPmice (CYP26KO) and ROSA26CreERT wildtype mice (CTR). After 5 daily tamoxifen injections, the knockout was confirmed at DNA and RNA level in multiple tissues of CYP26KO mice. Within 4 weeks, CYP26KO mice showed profound leukocytosis (5.97 ±0.37 vs. 21.12 ±3.81 k/mm3, n=4, p<0.01 CTR vs. CYP26KO) with neutrophilia and monocytosis compared to CTR mice. By 6 weeks, they experience profound weight loss, became moribund and had to be sacrificed. At that time, they had massive splenomegaly and lymphadenopathy as well as brittle/pale bones compared to CTR (Figure A). Histological analysis revealed presence of extramedullary hematopoiesis with a predominantly myeloid infiltrate in the spleen (confirmed by flow cytometry analysis) and lymph nodes but also multiple clusters megakaryocytes (Figure B). The mice also displayed decreased BM cellularity (11.25 ±1.01 vs. 5.62 ±0.36 x106/femur, n=5, p<0.01, CTR vs. CYP26KO), increased frequency of CFU-C (44.33 ±3.72 vs. 69.83 ±8.10 CFU/25000 BM mononuclear cells (MNCs), n=10, p=0.02, CTR vs. CYP26KO) with a myeloid bias (26.76±3.89 vs. 44.57 ±3.21 %CFU-GM/G/M, n=10, p<0.01, CTR vs. CYP26KO). When cellularity was taken into account, total CFU-C per femur was comparable between the two groups. The mice also had increased frequency of LSK cells in the BM (0.26 ±0.12 vs 0.73 ±0.18 % LSK of MNCs, n=2, CTR vs. CYP26KO) and an increased frequency of circulating CFU-C in the peripheral blood (37 ±4.04 vs. 179.5 ±43.06 per 200 ml of blood, n=4, p=0.01, CTR vs. CYP26KO). On histological analysis, the BM is dominated by a myeloid infiltrate and shows a striking decrease in radial bone diameter (Figure C,D). MSCs derived from CYP26KO mice have lower levels of CXCL12 and SCF and have impaired osteoblast differentiation potential compared to MSCs derived from control mice (Figure E). These findings were confirmed with ex vivo generated CYP26KO MSCs via retroviral mediated Cre recombination of CYP26A1loxP/loxPCYP26B1loxP/loxPstroma cells. More so, preliminary studies suggest that the hematopoietic phenotype observed depends on cell extrinsic presence of CYP26 activity as transplantation of CYP26A1loxP/loxPCYP26B1loxP/loxP BM cells into wildtype BoyJ recipients did not reproduce the phenotype upon injection with Tamoxifen of the recipient mice in spite of 100% donor derived hematopoiesis. Alternatively, transplant of wildtype cells into CYP26A1loxP/loxPCYP26B1loxP/loxPrecipients, did reproduce the phenotype after injection of Tamoxifen. In conclusion, we show here that dysregulated RA homeostasis in the BM impairs MSCs function and result in egress of hematopoiesis to extramedullary sites. These results come to complement data from RARγKO (Walkley CR et al Cell 2007) and SMRTmRIDmice models (Hong S-H et al PNAS 2013) and suggest a pivotal role of RA signaling in pathogenesis of myeloproliferative neoplasm and myelofibrosis. To what extent these findings correlate with human primary myelofibrosis is currently under investigation. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


1993 ◽  
Vol 264 (4) ◽  
pp. F715-F721 ◽  
Author(s):  
X. Wu ◽  
J. Pippin ◽  
J. B. Lefkowith

Nephrotoxic nephritis (NTN), a model of autoimmune glomerulonephritis, is characterized by glomerular inflammation, which results in both proteinuria and an increase in eicosanoid production. In light of the ability of CD18 integrins to participate in leukocyte adherence (and thereby migration), we examined the role of the integrin CD11b/CD18 in NTN using OX42, a monoclonal antibody directed against rat CD11b. Administration of OX42 30 min before induction of NTN decreased proteinuria (by 50%) but did not affect the number of leukocytes found in the glomerulus or the accompanying increase in glomerular eicosanoid production. Administration of OX42 16 h before disease induction led to a more substantial decrease in proteinuria (80%) and, in contrast to 30 min pretreatment, decreased the number of neutrophils found in the glomerulus and the accompanying increase in glomerular eicosanoid production (both by 50%). OX42 pretreatment had no effect on the number of macrophages found in glomeruli. Circulating leukocytes from animals treated with OX42 in vivo showed saturating surface levels of antibody by fluorescence-activated cell sorting (FACS) analysis and normal upregulation of CD11b by pharmacological activation. Sixteen hours after in vivo injection of OX42, 50% more peripheral leukocytes were labeled relative to control leukocytes labeled with OX42 ex vivo. Glomerular leukocytes in NTN exhibited upregulated expression of CD11b relative to peripheral leukocytes. These data show that CD11b/CD18 may participate in the acute expression of glomerular damage in NTN in a fashion not wholly dependent on blocking neutrophil migration into glomeruli. Blockade of surface receptors (as opposed to inhibition of upregulation) is sufficient to obtain this effect.


PLoS ONE ◽  
2014 ◽  
Vol 9 (8) ◽  
pp. e103505 ◽  
Author(s):  
Rit Vatsyayan ◽  
Hema Kothari ◽  
Nigel Mackman ◽  
Usha R. Pendurthi ◽  
L. Vijaya Mohan Rao

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 436-436
Author(s):  
Claire W Chang ◽  
Ni Cheng ◽  
Randal Skidgel ◽  
Yanyan Bai ◽  
Xiaoping Du

Abstract Transendothelial migration of neutrophils requires chemoattractant signals and also integrin family of adhesion receptors, particularly the β 2 family of integrins, including Mac-1 and LFA-1. Signals transmitted by G protein-coupled receptors (GPCR) for chemoattractants and cytokines induces inside-out signaling activating the ligand binding function of integrins. Conversely ligand binding to integrins stimulates outside-in signaling, leading to cell spreading, retraction and migration. The heterotrimeric G protein subunit, Gα13, is important for GPCR signaling leading to RhoA activation but also binds to integrins, including β2 integrins to stimulate outside-in signaling. To study the roles of Gα13 in neutrophil migration, we tested the effect of Gα13 knockout on transendothelial migration of neutrophils stimulated by chemoattractant fMIVIL. We demonstrate that transendothelial migration of Gα13 knockout neutrophils was significantly but partially reduced as compared with wild type mice. Transendothelial migration of Gα13 knockout neutrophils is similar to wild type neutrophil migration neutralized with an anti-Mac1 (anti-αm) antibody, and was not further inhibited by the anti-Mac1 antibody, suggesting that transendothelial migration mediated by integrin αmβ2 was predominantly Gα13-dependent. Interestingly, either anti-β2 antibody or anti-LFA1 (anti-αL) antibody appeared to inhibit transendothelial migration of not only wild type neutrophils, but also to a degree, Gα13-knockout neutrophils, suggesting a minor LFA1-dependent but Gα13-independent component of transendothelial migration in addition to the Gα13-dependent transendothlial migration. Furthermore, even though the fibrinogen and ICAM-1 are both β2 ligands, we show that more neutrophils migrated through ICAM-1-coated transwells than fibrinogen-coated transwells, and only ICAM-1-mediated neutrophil migration is Gα13 dependent, suggesting that Gα13-dependent neutrophil migration is selective for certain β2 integrin ligand (ICAM-1). Importantly, Gα13 knockout selectively inhibited the velocity of neutrophil migration on integrin ligand ICAM-1, but had no effect on the directionality of neutrophil migration which requires GPCR-dependent chemoattactant signaling. To understand whether and how Gα13 regulate integrin signaling, we show that Gα13 knockout did not affect the static adhesion of neutrophils to ICAM1, but significantly inhibited neutrophil spreading on ICAM-1. Furthermore, Gα13 bound to β2 integrins in neutrophils adherent on ICAM-1, and this binding was inhibited by the ExE motif peptide MB2mP6 derived from the Gα13 binding site of β2 integrin cytoplasmic domain. MB2mP6 also inhibited transendothelial cell migration similarly as Gα13 knockout. These data suggest that Gα13 plays an important role in promoting β2-integrin dependent neutrophil transendothelial migration mainly by mediating integrin outside-in signaling. Consistent with previous findings of the role of Gα13-dependent outside-in signaling in negative regulation of RhoA in other integrin subtypes, both Gα13 knockout and MB2mP6 abolished the transient inhibition in RhoA during adhesion of neutrophils on ICAM-1. These data suggest that Gα13 mediates outside-in signaling and transient inhibition of RhoA, and thus promotes neutrophil spreading and migration on integrin ligands. To test the role of Gα13 in neutrophil migration in vivo, we showed that neutrophil infiltration in vivo was reduced in leukocyte-selective Gα13 knockout mice using both thioglycolate-induced peritoneal neutrophil infiltration model and LPS-induced neutrophil lung infiltration model in vivo. Furthermore, MB2mP6 inhibited neutrophil infiltration in cardiac tissues in the cardiac ischemia-reperfusion injury model in mice. These data suggest that Gα13-integrin interaction plays an essential role in the integrin-dependent transendothelial migration and is likely to be important in neutrophils' immune and inflammatory functions. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 313 (5) ◽  
pp. H974-H987 ◽  
Author(s):  
Eynav Kliger ◽  
Batya Kristal ◽  
Galina Shapiro ◽  
Judith Chezar ◽  
Shifra Sela

Increased counts and priming of peripheral polymorphonuclear leukocytes (PMNLs) are associated with future or ongoing atherosclerosis; however, the role of PMNLs in enhancing monocyte transendothelial migration is still unclear. Our aims were to examine endothelial and monocyte activation, transmigration, and posttransmigration activation induced ex vivo by in vivo primed PMNLs and the effect of antioxidants on the activation. A unique ex vivo coculture system of three cell types was developed in this study, enabling interactions among the following: primary human umbilical vein endothelial cells (HUVECs), monocytes (THP-1 cell line), and in vivo primed PMNLs from hemodialysis (HD) patients and healthy control (HC) subjects. The interactions among these cells were examined, and an intervention with superoxide dismutase and catalase was performed. Preexposed HUVECs to HD/HC PMNLs showed a significant monocyte transmigration yield, 120–170% above HCs. Monocyte exposure to HD PMNLs induced pre- and posttransmigration activation. When the three cell types were cocultivated at the same time, monocyte chemoattractant protein-1 protein levels released from HUVECs, and activation markers on HUVECs [CD54 and chemokine (C-X3-C motif) ligand 1] and monocytes [chemokine (C-X3-C) receptor 1 and chemokine (C-C motif) receptor 2] were increased. Monocyte transmigration yield decreased to 70% (compared with HC subjects) due to adherence and accumulation of monocytes to HUVECs. When superoxide dismutase and catalase were used, reduced HUVEC and monocyte activation markers brought the transmigration yields to control levels and abolished accumulation of monocytes, emphasizing the role of superoxide in this process. We conclude that peripheral primed PMNLs play a pivotal role in enhancing monocyte transendotelial migration, the hallmark of the atherosclerotic process. Primed PMNLs can be used as a mediator and a biomarker of atherosclerosis even before plaque formation.NEW & NOTEWORTHY Primed polymorphonuclear leukocytes are key mediators in monocyte transendothelial migration, a new understanding of the initiation of endothelial dysfunction and monocyte activation, transmigration, and accumulation in the subendothelial layer.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3441-3441
Author(s):  
Jamie A Moore ◽  
Jayna J Mistry ◽  
Charlotte Hellmich ◽  
Rebecca H Horton ◽  
Edyta Wojtowicz ◽  
...  

Abstract The bone marrow (BM) microenvironment regulates acute myeloid leukemia (AML) initiation, proliferation and chemotherapy resistance. Following cancer cell death, a growing body of evidence suggests an important role for uncleared apoptotic debris in regulating the immunologic response to, and growth of, solid tumors. LC3-associated phagocytosis (LAP) maintains tissue homeostasis by regulating immune responses, such as tumor immunity. Here we investigate the role of LAP in macrophage within the BM microenvironment of AML. We find that depletion of BM macrophages via clodronate liposomes increased AML growth in-vivo. We show that LAP is an important pathway in BM macrophage to process dead and dying cells in the AML microenvironment. We used two syngeneic leukemia models (HOXA9/Meis1 and MN1) to investigate the role of LAP on AML proliferation. AML cells were injected into LAP deficient (Atg16L1 E230-) and wild-type (Atg16L1 E230+) mice. Targeted inhibition of LAP leads to accumulation of apoptotic cells (AC) and apoptotic bodies (AB) in the tumor microenvironment resulting in accelerated leukemia growth and decreased animal survival. Mechanistically, we show, via cytokine arrays and gene analysis, that the phagocytosis of AML derived AB via LAP in BM macrophage resulted in STING pathway activation in the phagocytic cells. Furthermore, through inhibition of STING using H-151 STING inhibitor, we show that STING activation in vivo supressed leukemia growth. STING activation can lead to a type I IFN response and to recruitment of cytotoxic T-cells. We saw no increase in CD8 + T-cell numbers or activation, however, via ex vivo analysis found that STING activation is required for phagocytic functions in macrophages. Next, we found that leukemic AB can induce a STING response in BM derived macrophages and that leukemic AB have increased mitochondria content that are processed by macrophages. Moreover, we identify that mitochondrial damage associated molecular patterns (DAMPs) from leukemic AB are processed by BM macrophages via LAP. Additionally, the depletion of mitochondrial DNA (mtDNA) in AML derived AB identified that the mtDNA from leukemic AB is responsible for the induction of STING signalling in BM macrophages. In summary, we report that LAP in BM macrophage of apoptotic debris in the AML microenvironment suppresses leukemic growth, through mechanisms stimulated by AML apoptotic bodies which contain mtDNA in the BM microenvironment. This process is mediated by the activation of the STING pathway. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2012 ◽  
Vol 82 (3) ◽  
pp. 228-232 ◽  
Author(s):  
Mauro Serafini ◽  
Giuseppa Morabito

Dietary polyphenols have been shown to scavenge free radicals, modulating cellular redox transcription factors in different in vitro and ex vivo models. Dietary intervention studies have shown that consumption of plant foods modulates plasma Non-Enzymatic Antioxidant Capacity (NEAC), a biomarker of the endogenous antioxidant network, in human subjects. However, the identification of the molecules responsible for this effect are yet to be obtained and evidences of an antioxidant in vivo action of polyphenols are conflicting. There is a clear discrepancy between polyphenols (PP) concentration in body fluids and the extent of increase of plasma NEAC. The low degree of absorption and the extensive metabolism of PP within the body have raised questions about their contribution to the endogenous antioxidant network. This work will discuss the role of polyphenols from galenic preparation, food extracts, and selected dietary sources as modulators of plasma NEAC in humans.


1988 ◽  
Vol 08 (02) ◽  
pp. 90-99 ◽  
Author(s):  
H. Schröder ◽  
K. Schrör

ZusammenfassungOrganische Nitrate unterschiedlicher chemischer Struktur sowie Nitroprussidnatrium und Molsidomin (bzw. ihre biologisch aktiven Metaboliten) können die (primäre) Aggregation und Sekretion von Humanthrombozyten in vitro und ex vivo hemmen. Eine solche Wirkung wird für Molsidomin (SIN-1) und Nitroprussidnatrium in vitro in Konzentrationen beobachtet, die in der gleichen Größenordnung liegen wie die vasodilatierenden Effekte der Substanzen. Dagegen sind für eine direkte Antiplättchenwirkung organischer Nitrate (Glyzeryltrinitrat, Isosorbiddinitr at, Isosorbidmononitrate, Teopranitol) in vitro Konzentrationen erforderlich, die ca. 100- bis 1000fach höher sind als die Plasmaspiegel der Substanzen nach therapeutischer Dosierung bzw. die Konzentrationen, die isolierte Gefäßstreifen relaxieren. Als gemeinsamer Wirkungsmechanismus der direkten thrombozy-tenfunktionshemmenden und gefäßerweiternden Wirkung all dieser Substanzen kann heute eine Stickoxid-(NO)-vermittelte Stimulation der cGMP-Bildung angenommen werden, das aus organischen Nitraten als »Pro-drug« entsteht. Die Freisetzung von NO, eines »endothelial cell-derived relaxing factors« (EDRF) aus Nitroprussidnatrium und SIN-1 erfolgt spontan. Dagegen erfordert die Freisetzung von NO aus organischen Nitraten einen enzymatischen Stoffwechselweg, der in isolierten Thrombozyten nicht vorhanden ist. Eine Antiplättchenwirkung organischer Nitrate in vivo bzw. ex vivo wird daher über die Stimulation eines endothelialen, thrombozyteninhibitorischen Faktors erklärt. Hierbei sind Prostazyklin sowie ein bisher unbekannter Endothel-zellfaktor neben einer synergistischen Wirkung organischer Nitrate mit endogenem Prostazyklin in Diskussion. Eine thrombozytenfunktionshemmen-de Wirkung organischer Nitrate könnte in Kombination mit ihren hämody-namischen Effekten auch für die an-tianginöse Wirkung in der Klinik bedeutsam sein, insbesondere zur Verhinderung vasospastischer Zustände bei der instabilen Angina pectoris.


Sign in / Sign up

Export Citation Format

Share Document