Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
PLoS One ; 10(3): e0121634, 2015.
Article in English | MEDLINE | ID: mdl-25803863

ABSTRACT

L-type Ca2+ channel (VGCC) mediated Ca2+ influx in vascular smooth muscle cells (VSMC) contributes to the functional properties of large arteries in arterial stiffening and central blood pressure regulation. How this influx relates to steady-state contractions elicited by α1-adrenoreceptor stimulation and how it is modulated by small variations in resting membrane potential (Vm) of VSMC is not clear yet. Here, we show that α1-adrenoreceptor stimulation of aortic segments of C57Bl6 mice with phenylephrine (PE) causes phasic and tonic contractions. By studying the relationship between Ca2+ mobilisation and isometric tension, it was found that the phasic contraction was due to intracellular Ca2+ release and the tonic contraction determined by Ca2+ influx. The latter component involves both Ca2+ influx via VGCC and via non-selective cation channels (NSCC). Influx via VGCC occurs only within the window voltage range of the channel. Modulation of this window Ca2+ influx by small variations of the VSMC Vm causes substantial effects on the contractile performance of aortic segments. The relative contribution of VGCC and NSCC to the contraction by α1-adrenoceptor stimulation could be manipulated by increasing intracellular Ca2+ release from non-contractile sarcoplasmic reticulum Ca2+ stores. Results of this study point to a complex interactions between α1-adrenoceptor-mediated VSMC contractile performance and Ca2+ release form contractile or non-contractile Ca2+ stores with concomitant Ca2+ influx. Given the importance of VGCC and their blockers in arterial stiffening and hypertension, they further point toward an additional role of NSCC (and NSCC blockers) herein.


Subject(s)
Aorta/physiology , Calcium/metabolism , Models, Biological , Muscle Contraction/physiology , Muscle, Smooth, Vascular/metabolism , Phenylephrine/metabolism , Analysis of Variance , Animals , Calcium Channels, L-Type/metabolism , Membrane Potentials , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/physiology , Myography/methods , Receptors, Adrenergic, alpha-1/metabolism
2.
Eur Heart J ; 36(17): 1049-58, 2015 May 01.
Article in English | MEDLINE | ID: mdl-24553721

ABSTRACT

AIMS: There is a need for animal models of plaque rupture. We previously reported that elastin fragmentation, due to a mutation (C1039G(+/-)) in the fibrillin-1 (Fbn1) gene, promotes atherogenesis and a highly unstable plaque phenotype in apolipoprotein E deficient (ApoE(-/-)) mice on a Western-type diet (WD). Here, we investigated whether plaque rupture occurred in ApoE(-/-)Fbn1(C1039G+/-) mice and was associated with myocardial infarction, stroke, and sudden death. METHODS AND RESULTS: Female ApoE(-/-)Fbn1(C1039G+/-) and ApoE(-/-) mice were fed a WD for up to 35 weeks. Compared to ApoE(-/-) mice, plaques of ApoE(-/-)Fbn1(C1039G+/-) mice showed a threefold increase in necrotic core size, augmented T-cell infiltration, a decreased collagen I content (70 ± 10%), extensive neovascularization, intraplaque haemorrhage, and a significant increase in matrix metalloproteinase-2, -9, -12, and -13 expression or activity. Plaque rupture was observed in 70% of ascending aortas and in 50% of brachiocephalic arteries of ApoE(-/-)Fbn1(C1039G+/-) mice. In ApoE(-/-) mice, plaque rupture was not seen in ascending aortas and only in 10% of brachiocephalic arteries. Seventy percent of ApoE(-/-)Fbn1(C1039G+/-) mice died suddenly, whereas all ApoE(-/-) mice survived. ApoE(-/-)Fbn1(C1039G+/-) mice showed coronary plaques and myocardial infarction (75% of mice). Furthermore, they displayed head tilt, disorientation, and motor disturbances (66% of cases), disturbed cerebral blood flow (73% of cases; MR angiograms) and brain hypoxia (64% of cases), indicative of stroke. CONCLUSIONS: Elastin fragmentation plays a key role in plaque destabilization and rupture. ApoE(-/-)Fbn1(C1039G+/-) mice represent a unique model of acute plaque rupture with human-like complications.


Subject(s)
Death, Sudden/etiology , Elastin/metabolism , Myocardial Infarction/etiology , Plaque, Atherosclerotic/etiology , Stroke/etiology , Animals , Aorta , Apolipoproteins E/deficiency , Biomarkers/metabolism , Brachiocephalic Trunk , Cardiomegaly/etiology , Cardiomegaly/physiopathology , Carotid Artery, Common , Cerebrovascular Circulation/physiology , Diet, Western , Disease Models, Animal , Female , Fibrillin-1 , Fibrillins , Hemorrhage/etiology , Hypoxia, Brain/etiology , Hypoxia, Brain/physiopathology , Mice , Microfilament Proteins/deficiency , Microvessels , Myocardial Infarction/physiopathology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/physiopathology , Nervous System Diseases/etiology , Nervous System Diseases/physiopathology , Plaque, Atherosclerotic/physiopathology , Rupture, Spontaneous/etiology , Rupture, Spontaneous/physiopathology , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology
3.
J Vasc Res ; 50(6): 486-97, 2013.
Article in English | MEDLINE | ID: mdl-24192582

ABSTRACT

AIMS: Insulin resistance, dyslipidemia and hypertension are independent mediators of endothelial dysfunction. It is incompletely defined whether dyslipidemia and hypertension in addition to diabetes mellitus type II (DMII), as seen in the metabolic syndrome (MS), worsen diabetes-induced endothelial dysfunction. Furthermore, it is unclear whether treatment influences endothelial dysfunction similarly in MS and DMII. Therefore, we studied vascular reactivity and the effect of in vivo treatment with angiotensin-converting enzyme inhibition (ACE-I) or hypocaloric diet in LDL receptor- and leptin-deficient (ob/ob), double knockout mice (DKO), featuring MS and in ob/ob mice with DMII. METHODS AND RESULTS: Vascular reactivity was studied in isolated aortic ring segments. Maximum vasorelaxant response to acetylcholine (Ach) was more depressed in DKO than in ob/ob mice, whereas response to bradykinin (BK) was equally attenuated in both genotypes (52 ± 3 and 23 ± 9% reversal of preconstriction induced by 10(-7) M phenylephrine in DKO vs. 76 ± 3 and 23 ± 8% reversal of preconstriction in ob/ob mice, respectively). ACE-I and hypocaloric diet improved ACh-induced vasorelaxation significantly (89 ± 2 and 59 ± 2% reversal of preconstriction in DKO vs. 80 ± 3 and 84 ± 4% in ob/ob mice, respectively), but not the response to BK. CONCLUSION: These results indicate a differential impact of DMII and MS on endothelial function. ACE-I and hypocaloric diet improved ACh-, but not BK-induced vasorelaxation in these mouse models of DMII and MS.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Caloric Restriction , Captopril/pharmacology , Diabetes Mellitus, Type 2/therapy , Dyslipidemias/therapy , Endothelium, Vascular/drug effects , Hypertension/therapy , Nitric Oxide/metabolism , Weight Loss , Animals , Biomarkers/blood , Blood Glucose/metabolism , Combined Modality Therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Dyslipidemias/blood , Dyslipidemias/genetics , Dyslipidemias/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Female , Hypertension/blood , Hypertension/genetics , Hypertension/physiopathology , Leptin/deficiency , Leptin/genetics , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
4.
Thromb Haemost ; 110(4): 785-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23925372

ABSTRACT

Arterial ageing may be associated with a reduction in vasodilation due to increased reactive oxygen species (ROS) production, whereas endothelial cell activation induces procoagulant changes. However, little is known on the effect of ageing on expression of anticoagulant endothelial markers such as endothelial protein C receptor (EPCR). To study age-associated alterations in smooth muscle cell (SMC) and endothelial cell (EC) structure and function, the aorta was isolated from 10-week- and 12- and 24-month-old C57BL/6J mice and analysed for its expression of genes involved in senescence, oxidative stress production, coagulation and matrix remodelling. In addition, vasorelaxation experiments were performed using 10-week- and 24-month-old thoracic aortic ring segments in organ chamber baths. The media thickness of the thoracic aorta progressively increased with age, associated with hypertrophy of vascular SMCs. Basal nitric oxide production and sensitivity to acetylcholine-mediated vasodilation in thoracic aorta rings was reduced with age, whereas no significant differences in ROS production could be demonstrated. Gene expression of tissue factor, EPCR and von Willebrand factor was not affected by ageing of the aorta, whereas that of thrombomodulin was mildly reduced and that of xanthine dehydrogenase, NADPH oxidase 4, tumour necrosis factor-α and vascular cell adhesion molecule-1 significantly enhanced. In conclusion, a reduction in endothelial cell-mediated vasodilation in aged thoracic aortas of C57BL/6J mice was accompanied by a shift towards a pro-inflammatory state of the endothelium.


Subject(s)
Aging/metabolism , Aorta, Thoracic/metabolism , Myocytes, Smooth Muscle/metabolism , Aging/genetics , Aging/immunology , Animals , Aorta, Thoracic/immunology , Blood Coagulation/genetics , Cells, Cultured , Cellular Senescence/genetics , Gene Expression Regulation , Humans , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Organ Culture Techniques , Oxidative Stress/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vasodilation/physiology , Xanthine Dehydrogenase/genetics , Xanthine Dehydrogenase/metabolism
5.
Curr Pharm Des ; 19(33): 5873-82, 2013.
Article in English | MEDLINE | ID: mdl-23438957

ABSTRACT

The chronic inflammatory nature of atherosclerosis is nowadays widely accepted. Dendritic cells (DCs) are likely to play a crucial role in directing innate and adaptive immunity against altered (self-)antigens, such as oxidized low density lipoproteins (oxLDL). DCs are found in early lesions and their numbers become even higher when the lesion progresses. DCs are most abundant in areas of neovascularization where they are often found near T cells. All stages from precursors to fully mature DCs are present in human plaques. Treatment of atherosclerosis is currently based on reducing risk factors, e.g. by use of statins and beta-blockers. Some of these pharmacological agents also show anti-inflammatory properties and consequently can affect DC function. Yet, many patients remain at risk for acute coronary events, and new therapies to treat atherosclerosis are needed. One therapeutic strategy is based on isolation of patient's DCs that are then pulsed with appropriate antigen(s) ex vivo, e.g. (immunogenic components of) oxLDL or total extract of atherosclerotic plaque tissue, and returned to the blood stream. Other approaches to ensure immune protection include generation of tolerogenic DCs, or using DCs to deplete detrimental Th1 or Th17 cells. However, the future lies in direct targeting of DCs by manipulating functions of different DC subsets. Therefore, it would be useful to isolate plaque-resident DCs to be able to identify unique antigen(s) on their surface. The challenge is to selectively identify regulatory molecules and novel therapies to inhibit DC migration and function during atherogenesis, without affecting normal DC function under physiological conditions.


Subject(s)
Atherosclerosis/drug therapy , Dendritic Cells/drug effects , Adaptive Immunity/drug effects , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Cholesterol/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity, Innate/drug effects , Vaccination/methods
6.
Eur J Pharmacol ; 698(1-3): 404-12, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23099257

ABSTRACT

The aim of the present study is to investigate the effects of TRPV1 and TRPA1 receptor antagonists and their synergism on the visceromotor responses during experimental colitis in rats. Colitis was induced in rats by a TNBS/ethanol enema at day 0 and was assessed at day 3 using endoscopy, histology and a myeloperoxidase assay. The visceromotor response to colorectal distension (10-80 mmHg) was evaluated in conscious rats before (control condition) and 3 days after 2,4,6-trinitrobenzene sulfonic acid (TNBS) administration (colitis condition). At day 3, visceromotor responses were assessed before and after treatment with a TRPV1 (BCTC) or TRPA1 (TCS-5861528) receptor antagonist either alone or in combination and either after intraperitoneal or intrathecal administration. Endoscopy, microscopy and myeloperoxidase activity indicated severe colonic tissue damage 3 days after TNBS administration. Colorectal distension-evoked visceromotor responses demonstrated a 2.9-fold increase during acute colitis (day 3) compared to control conditions. Intraperitoneal and intrathecal administration of BCTC or TCS-5861528 partially reversed the colitis-induced increase in visceromotor responses compared to control conditions (P<0.05). Intraperitoneal blockade of TRPA1 plus TRPV1 further decreased the enhanced visceromotor responses at high distension pressures (40-80 mmHg) compared to blockade of either TRPV1 or TRPA1 alone. This synergistic effect was not seen after combined intrathecal blockade of TRPA1 plus TRPV1. The present study demonstrates that in the rat, TRPV1 and TRPA1 play a pivotal role in visceral hypersensitivity at the peripheral and spinal cord level during acute TNBS colitis. Target interaction, however, is presumably mediated via a peripheral site of action.


Subject(s)
Colitis/physiopathology , Colon/drug effects , Colon/physiopathology , Rectum/drug effects , Rectum/physiopathology , TRPC Cation Channels/metabolism , TRPV Cation Channels/metabolism , Acetanilides/administration & dosage , Acetanilides/pharmacology , Animals , Colitis/metabolism , Colon/metabolism , Drug Synergism , Female , Purines/administration & dosage , Purines/pharmacology , Pyrazines/administration & dosage , Pyrazines/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Wistar , Rectum/metabolism , Reflex/drug effects , TRPA1 Cation Channel , TRPC Cation Channels/antagonists & inhibitors , TRPV Cation Channels/antagonists & inhibitors , Trinitrobenzenesulfonic Acid/administration & dosage , Trinitrobenzenesulfonic Acid/pharmacology
7.
J Cardiovasc Pharmacol ; 61(4): 318-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23249678

ABSTRACT

In the mouse aorta, contractions evoked by the α(1)-adrenoceptor agonist phenylephrine are strongly suppressed by the continuous production of nitric oxide (NO). We investigated whether phenylephrine itself stimulated NO production by activating endothelial α(2)-adrenoceptors. On a prostaglandin F(2α) contraction, the α(2)-adrenoceptor agonist 5-bromo-N-(4,5-dihydro-1H-imidazol-2-yl)-6-quinoxalinamine (UK14304) induced 29.3 ± 7.4% relaxation, which was inhibited by 0.1 µM 2-[(4,5-Dihydro-1H-imidazol-2-yl)methyl]-2,3-dihydro-1-methyl-1H-isoindole (BRL44408) with a pKB' corresponding to α(2)-antagonism. In the presence of NO synthase blockers, UK14304 elicited small contractions above 1 µM that were inhibited by 0.1 µM prazosin, but not influenced by 0.1 µM rauwolscine. At 3 µM or higher concentrations, phenylephrine caused only modest relaxation (up to 7.4 ± 2.3%) of segments constricted with prostaglandin F(2α) in the presence of prazosin, which was abolished with 0.1 µM BRL44408. Furthermore, BRL44408 did not increase contractions induced with 1 µM phenylephrine. These results confirm that α(1)- but not α(2)-adrenoceptors are expressed on aortic smooth muscle cells, whereas endothelial cells only express α(2)-adrenoceptors. Moreover, phenylephrine exerted a very modest relaxing effect through nonspecific stimulation of α(2)-adrenoceptors, but only at concentrations higher than 1 µM. It is concluded that the high basal output of NO in the isolated mouse aorta is not due to stimulation of α-adrenoceptors.


Subject(s)
Aorta, Thoracic/drug effects , Nitric Oxide/metabolism , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-2/drug effects , Adrenergic alpha-1 Receptor Agonists/administration & dosage , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Aorta, Thoracic/metabolism , Brimonidine Tartrate , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Imidazoles/pharmacology , Isoindoles/administration & dosage , Isoindoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Phenylephrine/administration & dosage , Prazosin/pharmacology , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Receptors, Adrenergic, alpha-2/metabolism , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology , Yohimbine/pharmacology
8.
Autophagy ; 9(3): 386-402, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23242143

ABSTRACT

Transmission electron microscopy (TEM) is an indispensable standard method to monitor macroautophagy in tissue samples. Because TEM is time consuming and not suitable for daily routine, many groups try to identify macroautophagy in tissue by conventional immunohistochemistry. The aim of the present study was to evaluate whether immunohistochemical assessment of macroautophagy-related marker proteins such as LC3, ATG5, CTSD/cathepsin D, BECN1/Beclin 1 or SQSTM1/p62 is feasible and autophagy-specific. For this purpose, livers from starved mice were used as a model because hepatocytes are highly sensitive to autophagy induction. ATG7-deficient mouse livers served as negative control. Our findings indicate that unambiguous immunodetection of LC3 in paraffin-embedded tissue specimens was hampered due to low in situ levels of this protein. Maximum sensitivity could only be obtained using high-quality, isoform-specific antibodies, such as antibody 5F10, in combination with Envision+ signal amplification. Moreover, LC3 stains were optimal in neutral-buffered formalin-fixed tissue, immersed in citrate buffer during antigen retrieval. However, even when using this methodology, LC3 monitoring required overexpression of the protein, e.g., in GFP-LC3 transgenic mice. This was not only the case for the liver but also for other organs including heart, skeletal muscle, kidney and gut. Immunohistochemical detection of the autophagy-related proteins ATG5, CTSD or BECN1 is not recommendable for monitoring autophagy, due to lack of differential gene expression or doubtful specificity. SQSTM1 accumulated in autophagy-deficient liver, thus it is not a useful marker for tissue with autophagic activity. We conclude that TEM remains an indispensable technique for in situ evaluation of macroautophagy, particularly in clinical samples for which genetic manipulation or other in vitro techniques are not feasible.


Subject(s)
Autophagy , Immunohistochemistry/methods , Animals , Autophagy-Related Protein 7 , Liver/pathology , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism
9.
Catheter Cardiovasc Interv ; 81(7): E259-68, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-22887820

ABSTRACT

AIM: To evaluate differences in strut coverage, inflammation and endothelialization between two second-generation polymer-based drug-eluting stents (DES) in an atherosclerotic rabbit double-injury iliac artery model at 28 days follow-up. METHODS AND RESULTS: Rabbits with induced atheroma received bilateral iliac artery stents: everolimus-eluting stent (Xience V EES; Abbott Vascular), zotarolimus-eluting stent (Resolute ZES; Medtronic CardioVascular), or bare-metal stent (BMS; MultiLink Vision; Abbott Vascular). After 28 days, total neointimal coverage examined by scanning electron microscopy was >98% for all three stent types. Neointimal thickness above stent struts was decreased by 50% in Xience V EES (0.06 ± 0.01 mm; P = 0.00001) compared with BMS (0.15 ± 0.03 mm) and Resolute ZES (0.12 ± 0.04 mm). Luminal area was largest for Xience V EES (3.79 ± 0.33 mm(2) ; P = 0.0003 for Xience V EES vs. BMS), followed by Resolute ZES (3.46 ± 0.45 mm(2) ; P = 0.083 for Resolute ZES vs. BMS) and BMS (3.07 ± 0.53 mm(2) ). Percentage area stenosis was smallest for Xience V EES (17.23 ± 3.64%; P = 0.00001), while BMS (30.25 ± 7.48%) and Resolute ZES (30.79 ± 7.15%) did not differ. Endothelial monolayer regrowth was significantly lower in Resolute ZES (65 ± 13%) versus BMS (79 ± 11%; P = 0.004). There was no difference between Xience V EES (74 ± 10%) and BMS. Xience V EES was further associated with a lower number of inflammatory cells surrounding the stent struts (7 ± 2 per strut) in comparison to Resolute ZES (15 ± 6; P = 0.0001) and BMS (17 ± 9; P = 0.0005). CONCLUSION: In this atherosclerotic rabbit model, Xience V EES suppressed neointimal thickening better, with normal endothelial regrowth as compared with BMS, and less strut-induced inflammation.


Subject(s)
Angioplasty, Balloon/instrumentation , Atherosclerosis/therapy , Drug-Eluting Stents , Iliac Artery/injuries , Vascular System Injuries/therapy , Angioplasty, Balloon/adverse effects , Animals , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Cardiovascular Agents/administration & dosage , Cell Proliferation , Disease Models, Animal , Endothelial Cells/ultrastructure , Everolimus , Iliac Artery/diagnostic imaging , Iliac Artery/ultrastructure , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Male , Neointima , Prosthesis Design , Rabbits , Radiography , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Time Factors , Vascular System Injuries/diagnostic imaging , Vascular System Injuries/pathology
10.
Eur J Pharmacol ; 696(1-3): 111-9, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23022329

ABSTRACT

Bioavailability of nitric oxide (NO) is mostly studied in isolated blood vessels. We investigated changes in basal and receptor-stimulated endothelial NO synthase (eNOS) activity after isolation of wild-type and Marfan mouse aorta. Starting 1h after dissection, basal NO release was assessed at hourly intervals by its ability to suppress isometric contractions in aortic segments. Relaxation induced by acetylcholine or α(2)-adrenoceptor agonist 5-bromo-N-(4,5-dihydro-1H-imidazol-2-yl)-6-quinoxalinamine (UK14304) was used to study stimulated NOS activity. One hour after dissection, phenylephrine- or prostaglandin F(2α)-induced force attained only 17 ± 4% or 31 ± 7% of maximum tension in the presence of N(Ω)-nitro-l-arginine-methylesther (l-NAME), and contractions increased to 63 ± 6% and 82 ± 11%, respectively at 5h. In contrast, acetylcholine or UK14304 relaxation curves changed minimally. l-NAME and eNOS-deficiency abolished basal NO production, unlike inhibitors of neuronal (N(Ω)-propyl-l-arginine) or inducible (1400W) NOS. Acetylcholine-induced relaxation was abolished by l-NAME, strongly suppressed by eNOS-deficiency and attenuated by N(Ω)-propyl-l-arginine. In a bioassay based on diethylamine NONOate concentration-response curves the suppression of contractile forces was interpolated into NO equivalents. This showed exponential decay of basal NO, which occurred three times faster in aortas from mice with Marfan syndrome, while acetylcholine-induced relaxation remained unaltered. Immunoblotting showed unchanged eNOS expression, or phosphorylation at Ser1177, Ser617 or Thr495 between 1h and 4h, but Akt phosphorylation declined gradually. The dramatic loss of basal NO release after tissue isolation shows that timing is crucial when studying NO responses. The preservation of receptor-induced relaxation implies differential regulation of basal and stimulated eNOS activity, and phosphoinositide-3-kinase/Akt signalling seems specifically associated with basal eNOS activity.


Subject(s)
Aorta, Thoracic/physiology , Nitric Oxide Synthase Type III/physiology , Nitric Oxide/physiology , Vasodilation/physiology , Animals , Fibrillins , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Vasoconstriction/drug effects , Vasodilation/drug effects
11.
BMC Physiol ; 12: 9, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22943445

ABSTRACT

BACKGROUND: Electrophysiological studies of L-type Ca2+ channels in isolated vascular smooth muscle cells revealed that depolarization of these cells evoked a transient and a time-independent Ca2+ current. The sustained, non-inactivating current occurred at voltages where voltage-dependent activation and inactivation overlapped (voltage window) and its contribution to basal tone or active tension in larger multicellular blood vessel preparations is unknown at present. This study investigated whether window Ca2+ influx affects isometric contraction of multicellular C57Bl6 mouse aortic segments. RESULTS: Intracellular Ca2+ (Cai2+, Fura-2), membrane potential and isometric force were measured in aortic segments, which were clamped at fixed membrane potentials by increasing extracellular K+ concentrations. K+ above 20 mM evoked biphasic contractions, which were not affected by inhibition of IP3- or Ca2+ induced Ca2+ release with 2-aminoethoxydiphenyl borate or ryanodine, respectively, ruling out the contribution of intracellular Ca2+ release. The fast force component paralleled Cai2+ increase, but the slow contraction coincided with Cai2+ decrease. In the absence of extracellular Ca2+, basal tension and Cai2+ declined, and depolarization failed to evoke Cai2+ signals or contraction. Subsequent re-introduction of external Ca2+ elicited only slow contractions, which were now matched by Cai2+ increase. After Cai2+ attained steady-state, isometric force kept increasing due to Ca2+- sensitization of the contractile elements. The slow force responses displayed a bell-shaped voltage-dependence, were suppressed by hyperpolarization with levcromakalim, and enhanced by an agonist of L-type Ca2+ channels (BAY K8644). CONCLUSION: The isometric response of mouse aortic segments to depolarization consists of a fast, transient contraction paralleled by a transient Ca2+ influx via Ca2+ channels which completely inactivate. Ca2+ channels, which did not completely inactivate during the depolarization, initiated a second, sustained phase of contraction, which was matched by a sustained non-inactivating window Ca2+ influx. Together with sensitization, this window L-type Ca2+ influx is a major determinant of basal and active tension of mouse aortic smooth muscle.


Subject(s)
Aorta/physiology , Calcium/metabolism , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Animals , Aorta/metabolism , Calcium Channels, L-Type/metabolism , Fura-2/metabolism , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Potassium/metabolism
12.
PLoS One ; 7(8): e43357, 2012.
Article in English | MEDLINE | ID: mdl-22916248

ABSTRACT

BACKGROUND: Dendritic cells (DCs), professional antigen-presenting cells with the unique ability to initiate primary T-cell responses, are present in atherosclerotic lesions where they are exposed to oxidative stress that generates cytotoxic reactive oxygen species (ROS). A large body of evidence indicates that cell death is a major modulating factor of atherogenesis. We examined antioxidant defence systems of human monocyte-derived (mo)DCs and monocytes in response to oxidative stress. METHODS: Oxidative stress was induced by addition of tertiary-butylhydroperoxide (tert-BHP, 30 min). Cellular responses were evaluated using flow cytometry and confocal live cell imaging (both using 5-(and-6)-chloromethyl-2,7-dichlorodihydrofluorescein diacetate, CM-H(2)DCFDA). Viability was assessed by the neutral red assay. Total RNA was extracted for a PCR profiler array. Five genes were selected for confirmation by Taqman gene expression assays, and by immunoblotting or immunohistochemistry for protein levels. RESULTS: Tert-BHP increased CM-H(2)DCFDA fluorescence and caused cell death. Interestingly, all processes occurred more slowly in moDCs than in monocytes. The mRNA profiler array showed more than 2-fold differential expression of 32 oxidative stress-related genes in unstimulated moDCs, including peroxiredoxin-2 (PRDX2), an enzyme reducing hydrogen peroxide and lipid peroxides. PRDX2 upregulation was confirmed by Taqman assays, immunoblotting and immunohistochemistry. Silencing PRDX2 in moDCs by means of siRNA significantly increased CM-DCF fluorescence and cell death upon tert-BHP-stimulation. CONCLUSIONS: Our results indicate that moDCs exhibit higher intracellular antioxidant capacities, making them better equipped to resist oxidative stress than monocytes. Upregulation of PRDX2 is involved in the neutralization of ROS in moDCs. Taken together, this points to better survival skills of DCs in oxidative stress environments, such as atherosclerotic plaques.


Subject(s)
Dendritic Cells/metabolism , Monocytes/cytology , Oxidative Stress/physiology , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism
13.
Parasitology ; 139(11): 1429-37, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22776404

ABSTRACT

Leishmania parasites are able to survive in the macrophage, one of the most hostile environments of the vertebrate host. The present study investigated how Leishmania infection influences these host cell defence mechanisms. Macrophages were infected with antimony-susceptible and -resistant Leishmania strains. Free radical production in Leishmania-infected macrophages was measured by electron paramagnetic resonance. Apoptosis was detected with fluorescence microscopy using Annexin-V FITC labelling and with Western blotting to detect caspase-3 cleavage. Independent of their drug susceptibility profile or species background, all studied Leishmania strains induced a similar increase in free radical production in macrophages. O2 ●- production was significantly elevated during phagocytosis of the stationary phase promastigotes. Conversely, NO levels increased later in the infection and none of the strains induced capsase-3 cleavage. Leishmania donovani infection led to phosphatidylserine externalization only in RAW 264.7 cells. After an initial burst of O2 ●- during phagocytosis of promastigotes, amastigotes protect themselves by decreasing the O2 ●- production to the basal level. An increased NO production was observed 6 h after infection. Finally, induction of cell death is probably not essential in the survival of the parasite within the macrophage.


Subject(s)
Apoptosis/immunology , Leishmaniasis/immunology , Macrophages/immunology , Macrophages/parasitology , Oxidative Stress/immunology , Animals , Cell Line, Tumor , Macrophages/cytology , Mice , Nitric Oxide/metabolism , Superoxides/metabolism
14.
Basic Res Cardiol ; 107(3): 269, 2012 May.
Article in English | MEDLINE | ID: mdl-22543675

ABSTRACT

Atherosclerotic plaques tend to rupture as a consequence of a weakened fibrous cap, particularly in the shoulder regions where most macrophages reside. Macrophages express Toll-like receptors to recognize pathogens and eliminate intracellular pathogens by inducing autophagy. Because Toll-like receptor 7 (TLR7) is thought to be expressed in macrophages but not in smooth muscle cells (SMCs), we investigated whether induction of macrophage autophagic death by TLR7 ligand imiquimod can affect the composition of atherosclerotic plaques in favor of their stability. Immunohistochemical staining of human carotid plaques as well as Western blotting of cultured macrophages and SMCs confirmed that TLR7 was expressed in macrophages, but not in SMCs. In vitro experiments showed that only TLR7 expressing cells underwent imiquimod-induced cell death, which was characterized by autophagosome formation. Imiquimod-treated macrophages activated nuclear factor-κB (NF-κB) and released pro-inflammatory cytokines and chemokines. This effect was inhibited by the glucocorticoid dexamethasone. Imiquimod-induced cytokine release was significantly decreased in autophagy-deficient macrophages because these cells died by necrosis at an accelerated pace. Local in vivo administration of imiquimod to established atherosclerotic lesions in rabbit carotid arteries induced macrophage autophagy without induction of cell death, and triggered cytokine production, upregulation of vascular adhesion molecule-1, infiltration of T-lymphocytes, accumulation of macrophages and enlargement of plaque area. Treatment with dexamethasone suppressed these pro-inflammatory effects in vivo. SMCs and endothelial cells in imiquimod-treated plaques were not affected. In conclusion, imiquimod induces macrophage autophagy in atherosclerotic plaques, but stimulates plaque progression through cytokine release and enhanced infiltration of inflammatory cells.


Subject(s)
Aminoquinolines/pharmacology , Autophagy/drug effects , Inflammation/chemically induced , Macrophages/drug effects , Membrane Glycoproteins/agonists , Plaque, Atherosclerotic/drug therapy , Toll-Like Receptor 7/agonists , Aminoquinolines/toxicity , Animals , Anti-Inflammatory Agents/pharmacology , Blotting, Western , Cytokines/metabolism , Dexamethasone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Imiquimod , Immunohistochemistry , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/pathology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Necrosis , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Rabbits , Time Factors , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , U937 Cells
15.
Arterioscler Thromb Vasc Biol ; 32(5): 1228-35, 2012 May.
Article in English | MEDLINE | ID: mdl-22345170

ABSTRACT

OBJECTIVE: Stent-based delivery of the mammalian target of rapamycin (mTOR) inhibitor everolimus is a promising strategy for the treatment of coronary artery disease. We studied potential adverse effects associated with mTOR inhibition. METHODS AND RESULTS: Macrophages in culture were either treated with everolimus or starved to inhibit mTOR. Everolimus led to inhibition of protein translation, activation of p38 MAPK, and the release of proinflammatory cytokines (eg, IL-6, TNFα) and chemokines (eg, MCP1, Rantes) before induction of autophagic death. These effects were also observed with rapamycin, but not after starvation. Everolimus-induced cytokine release was similar in macrophages lacking the essential autophagy gene Atg7 but was inhibited when macrophages were cotreated with p38 MAPK inhibitor SB202190 or the glucocorticoid clobetasol. Combined stent-based delivery of clobetasol and everolimus in rabbit plaques downregulated TNFα expression as compared with everolimus-treated plaques but did not affect the ability of everolimus to induce macrophage clearance. CONCLUSIONS: mTOR inhibition by everolimus triggers cytokine release in macrophages through inhibition of protein translation and p38 activation. These findings provide a rationale for combined local treatment of atherosclerotic plaques with everolimus and an anti-inflammatory agent.


Subject(s)
Coronary Artery Disease/surgery , Cytokines/biosynthesis , Drug-Eluting Stents , Glucocorticoids/pharmacology , Macrophages/metabolism , Sirolimus/analogs & derivatives , Animals , Autophagy/drug effects , Blotting, Western , Cell Survival , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Everolimus , Humans , Immunohistochemistry , Macrophages/drug effects , Macrophages/ultrastructure , Mice , Microscopy, Electron, Transmission , Prosthesis Design , Rabbits , Sirolimus/pharmacology
17.
Mediators Inflamm ; 2011: 941396, 2011.
Article in English | MEDLINE | ID: mdl-21976788

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic inflammatory disease with atherosclerotic plaques containing inflammatory infiltrates predominantly consisting of monocytes/macrophages and activated T cells. More recent is the implication of dendritic cells (DCs) in the disease. Since DCs were demonstrated in human arteries in 1995, numerous studies in humans suggest a role for these professional antigen-presenting cells in atherosclerosis. AIM: This paper focuses on the observations made in blood and arteries of patients with atherosclerosis. In principal, flow cytometric analyses show that circulating myeloid (m) and plasmacytoid (p) DCs are diminished in coronary artery disease, while immunohistochemical studies describe increased intimal DC counts with evolving plaque stages. Moreover, mDCs and pDCs appear to behave differently in atherosclerosis. Yet, the origin of plaque DCs and their relationship with blood DCs are unknown. Therefore, several explanations for the observed changes are postulated. In addition, the technical challenges and discrepancies in the research field are discussed. FUTURE: Future studies in humans, in combination with experimental animal studies will unravel mechanisms leading to altered blood and plaque DCs in atherosclerosis. As DCs are crucial for inducing but also dampening immune responses, understanding their life cycle, trafficking and function in atherosclerosis will determine potential use of DCs in antiatherogenic therapies.


Subject(s)
Atherosclerosis/immunology , Dendritic Cells/immunology , Plaque, Atherosclerotic/immunology , Animals , Dendritic Cells/cytology , Flow Cytometry , Humans , Plaque, Atherosclerotic/metabolism
18.
Arterioscler Thromb Vasc Biol ; 31(11): 2552-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21799179

ABSTRACT

OBJECTIVE: The goal of this study was to examine the functional relationship between aging endothelium and thrombogenicity in a mouse model of premature aging. METHODS AND RESULTS: Coagulation tests and factors, blood cell counts, aorta endothelial function, aorta gene expression, and FeCl(3)-induced thrombosis in mesenteric blood vessels were analyzed in 10- to 30-week-old brain and muscle ARNT-like protein-1 (Bmal1)-deficient (knockout [KO]) mice and wild-type littermates. Ten-week-old KO mice manifested shortened prothrombin times (9.7 versus 11.3 seconds in wild-type) and elevated plasma fibrinogen (264 versus 172 mg/dL). At 30 weeks, factor VII (198% versus 149%), and platelet counts (2049 versus 1354 K/µL) were increased in KO mice. Gene deficiency reduced the vasoactive nitric oxide production at 10 and 30 weeks and tended to reduce and increase the protein expression of thrombomodulin and von Willebrand factor, respectively, with aging. Shortened venular and arteriolar occlusion times on FeCl(3)-induced injury in 10-week-old KO mice confirmed higher thrombogenicity, culminating in priapism, observed in 60% of 25- to 30-week-old KO males. CONCLUSION: Endothelial dysfunction and a hypercoagulable state cause early arterial and venous thrombogenicity in Bmal1 KO mice. With aging, progressive endothelial dysfunction, rising platelet counts, and high factor VII further enhance thrombogenicity, provoking priapism.


Subject(s)
ARNTL Transcription Factors/deficiency , Aging, Premature/metabolism , Aging, Premature/physiopathology , Disease Progression , Thrombosis/metabolism , Thrombosis/physiopathology , ARNTL Transcription Factors/genetics , Aging/physiology , Animals , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Fibrinogen/metabolism , Male , Mice , Mice, Knockout , Platelet Count , Prothrombin Time , Thrombomodulin/metabolism , von Willebrand Factor/metabolism
19.
Pathology ; 43(3): 239-47, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21436634

ABSTRACT

BACKGROUND: Previously we demonstrated decreased blood myeloid (m) and plasmacytoid (p) dendritic cell (DC) counts in atherosclerotic patients. Therefore, we examined whether DCs, in particular DC precursors, accumulate in human plaques. METHODS: Blood DC antigen (BDCA)-1, CD11c (mDCs), BDCA-2, CD123 (pDCs), langerin, fascin, S-100 (immature/mature DCs), and CD1a and CD83 (mature DCs) were investigated by immunohistochemistry of carotid arteries obtained by endarterectomy (EAS, frozen n = 11, fixed n = 11) or autopsy (fixed, n = 87). RESULTS: Fascin and S-100 required formaldehyde fixation, other markers needed cryo-preservation. BDCA-1, BDCA-2, langerin, and S-100 appeared specific for intimal DCs, unlike CD123 and fascin (staining endothelial cells), CD11c and CD1a (staining monocytes, foam cells) or CD83 (staining lymphocytes). BDCA-1 and BDCA-2 cells were detected in EAS, preferentially near microvessels. S-100 cells increased successively from intimal thickening, via pathological intimal thickening, fibrous cap atheroma and finally complicated plaques. Fascin cells followed the same pattern, but were more abundant. However, in lesions containing microvessels (complicated plaques, plaque shoulders and most EAS) this was partly explained by fascin positive endothelial cells. Even complicated plaques contained relatively few mature CD83 DCs. CONCLUSIONS: Accumulation of BDCA-1 and BDCA-2 around neovessels showed that mDCs and pDCs are recruited to advanced plaques, which is in line with the previously described decline of circulating blood DCs in patients with coronary artery disease. Unexpectedly, several DC markers yielded false positive signals. Hence, some accounts on numbers, trafficking and activation of DCs in atherosclerotic plaques may require re-evaluation.


Subject(s)
Atherosclerosis/pathology , Dendritic Cells/pathology , Plaque, Atherosclerotic/pathology , Antigens, CD1 , Antigens, Surface/metabolism , Atherosclerosis/metabolism , Biomarkers/metabolism , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Arteries/surgery , Dendritic Cells/metabolism , Endarterectomy, Carotid , Fluorescent Antibody Technique, Indirect , Glycoproteins , Humans , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Plaque, Atherosclerotic/metabolism , Receptors, Immunologic/metabolism
20.
Atherosclerosis ; 216(1): 54-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21316678

ABSTRACT

OBJECTIVE: Recent evidence suggests that amyloid precursor protein (APP) is overexpressed in atherosclerosis-prone regions of mouse aorta. We therefore investigated in the present study whether APP has a role in the progression and composition of atherosclerotic plaques. METHODS AND RESULTS: Apolipoprotein E-deficient (apoE(-/-)) mice were crossbred with animals lacking APP (APP(-/-)). After 16 weeks on a Western-type diet, apoE(-/-) and APP(-/-)/apoE(-/-) mice showed similar cholesterol levels. However, atherosclerotic plaque size was significantly reduced in the distal thoracic aorta (90% reduction) and abdominal aorta (75% reduction) of APP(-/-)/apoE(-/-) mice as compared to apoE(-/-). Plaques at the level of the aortic valves were not different in size, but showed a more stable phenotype in APP(-/-)/apoE(-/-) mice, as indicated by a reduced macrophage content, an increased amount of collagen and a thicker fibrous cap. CONCLUSION: Our findings provide evidence that lack of APP attenuates atherogenesis and leads to plaque stability.


Subject(s)
Amyloid beta-Protein Precursor/deficiency , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Plaque, Atherosclerotic/prevention & control , Amyloid beta-Protein Precursor/genetics , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Valve/metabolism , Aortic Valve/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/blood , Collagen/metabolism , Disease Models, Animal , Fibrosis , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Rupture, Spontaneous
SELECTION OF CITATIONS
SEARCH DETAIL
...