Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Lab Anim ; 56(2): 157-164, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34407659

ABSTRACT

Open chest surgery in rodents requires assisted breathing and the most common approach for ventilation is via an endotracheal tube. Even with well-trained operators the endotracheal intubation is technically challenging and may lead to prolonged procedures and endotracheal intubation complications. Nose cone ventilation is a simpler procedure compared to endotracheal intubation and has the potential to improve animal welfare by reducing procedure time and endotracheal intubation associated complications. Rats are obligate nose breathers, and therefore replacing intubation with air supply from a nose cone would be an advantage and a more natural way of breathing. Here, we compared the values for several blood gases, blood pressure and heart rate from rats that were nose cone ventilated with rats that underwent endotracheal intubation at 12 timepoints equally distributed across three surgical stages: baseline, open chest and closed chest. Throughout the monitoring period the hemodynamic and blood gas values for both methods of ventilation were within published, normal ranges for the rat and were biologically equivalent (equivalence test p value ≤ 0.05). Our data showed that nose cone ventilation-maintained blood gases and hemodynamic homeostasis equivalent to endotracheal intubation. Nose cone ventilation can be recommended as an alternative to endotracheal intubation in rat experiments where investigators require airway control.


Subject(s)
Airway Management , Intubation, Intratracheal , Animals , Blood Gas Analysis , Gases , Hemodynamics , Intubation, Intratracheal/adverse effects , Intubation, Intratracheal/methods , Rats
2.
Front Cardiovasc Med ; 8: 753470, 2021.
Article in English | MEDLINE | ID: mdl-34722683

ABSTRACT

Objective: After myocardial infarction (MI), the non-infarcted left ventricle (LV) ensures appropriate contractile function of the heart. Metabolic disturbance in this region greatly exacerbates post-MI heart failure (HF) pathology. This study aimed to provide a comprehensive understanding of the metabolic derangements occurring in the non-infarcted LV that could trigger cardiovascular deterioration. Methods and Results: We used a pig model that progressed into chronic HF over 3 months following MI induction. Integrated gene and metabolite signatures revealed region-specific perturbations in amino acid- and lipid metabolism, insulin signaling and, oxidative stress response. Remote LV, in particular, showed impaired glutamine and arginine metabolism, altered synthesis of lipids, glucose metabolism disorder, and increased insulin resistance. LPIN1, PPP1R3C, PTPN1, CREM, and NR0B2 were identified as the main effectors in metabolism dysregulation in the remote zone and were found differentially expressed also in the myocardium of patients with ischemic and/or dilated cardiomyopathy. In addition, a simultaneous significant decrease in arginine levels and altered PRCP, PTPN1, and ARF6 expression suggest alterations in vascular function in remote area. Conclusions: This study unravels an array of dysregulated genes and metabolites putatively involved in maladaptive metabolic and vascular remodeling in the non-infarcted myocardium and may contribute to the development of more precise therapies to mitigate progression of chronic HF post-MI.

3.
Eur Heart J ; 42(43): 4481-4492, 2021 11 14.
Article in English | MEDLINE | ID: mdl-34297830

ABSTRACT

AIMS: Cardiac injury and remodelling are associated with the rearrangement of cardiac lipids. Glycosphingolipids are membrane lipids that are important for cellular structure and function, and cardiac dysfunction is a characteristic of rare monogenic diseases with defects in glycosphingolipid synthesis and turnover. However, it is not known how cardiac glycosphingolipids regulate cellular processes in the heart. The aim of this study is to determine the role of cardiac glycosphingolipids in heart function. METHODS AND RESULTS: Using human myocardial biopsies, we showed that the glycosphingolipids glucosylceramide and lactosylceramide are present at very low levels in non-ischaemic human heart with normal function and are elevated during remodelling. Similar results were observed in mouse models of cardiac remodelling. We also generated mice with cardiomyocyte-specific deficiency in Ugcg, the gene encoding glucosylceramide synthase (hUgcg-/- mice). In 9- to 10-week-old hUgcg-/- mice, contractile capacity in response to dobutamine stress was reduced. Older hUgcg-/- mice developed severe heart failure and left ventricular dilatation even under baseline conditions and died prematurely. Using RNA-seq and cell culture models, we showed defective endolysosomal retrograde trafficking and autophagy in Ugcg-deficient cardiomyocytes. We also showed that responsiveness to ß-adrenergic stimulation was reduced in cardiomyocytes from hUgcg-/- mice and that Ugcg knockdown suppressed the internalization and trafficking of ß1-adrenergic receptors. CONCLUSIONS: Our findings suggest that cardiac glycosphingolipids are required to maintain ß-adrenergic signalling and contractile capacity in cardiomyocytes and to preserve normal heart function.


Subject(s)
Glucosyltransferases , Myocytes, Cardiac , Animals , Cardiomegaly , Glucosyltransferases/genetics , Mice , Receptors, Adrenergic
4.
Elife ; 102021 05 11.
Article in English | MEDLINE | ID: mdl-33972017

ABSTRACT

Myocardial infarction (MI) promotes a range of systemic effects, many of which are unknown. Here, we investigated the alterations associated with MI progression in heart and other metabolically active tissues (liver, skeletal muscle, and adipose) in a mouse model of MI (induced by ligating the left ascending coronary artery) and sham-operated mice. We performed a genome-wide transcriptomic analysis on tissue samples obtained 6- and 24 hr post MI or sham operation. By generating tissue-specific biological networks, we observed: (1) dysregulation in multiple biological processes (including immune system, mitochondrial dysfunction, fatty-acid beta-oxidation, and RNA and protein processing) across multiple tissues post MI and (2) tissue-specific dysregulation in biological processes in liver and heart post MI. Finally, we validated our findings in two independent MI cohorts. Overall, our integrative analysis highlighted both common and specific biological responses to MI across a range of metabolically active tissues.


The human body is like a state-of-the-art car, where each part must work together with all the others. When a car breaks down, most of the time the problem is not isolated to only one part, as it is an interconnected system. Diseases in the human body can also have systemic effects, so it is important to study their implications throughout the body. Most studies of heart attacks focus on the direct impact on the heart and the cardiovascular system. Learning more about how heart attacks affect rest of the body may help scientists identify heart attacks early or create improved treatments. Arif and Klevstig et al. show that heart attacks affect the metabolism throughout the body. In the experiments, mice underwent a procedure that mimics either a heart attack or a fake procedure. Then, Arif and Klevstig et al. compared the activity of genes in the heart, muscle, liver and fat tissue of the two groups of mice 6- and 24-hours after the operations. This revealed disruptions in the immune system, metabolism and the production of proteins. The experiments also showed that changes in the activity of four important genes are key to these changes. This suggests that this pattern of changes could be used as a way to identify heart attacks. The experiments show that heart attacks have important effects throughout the body, especially on metabolism. These discoveries may help scientists learn more about the underlying biological processes and develop new treatments that prevent the harmful systemic effects of heart attacks and boost recovery.


Subject(s)
Gene Expression Profiling , Heart/physiopathology , Myocardial Infarction/genetics , Transcriptome , Animals , Disease Models, Animal , Genome , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myocardial Infarction/physiopathology , Oxidation-Reduction
6.
J Diabetes Complications ; 35(4): 107881, 2021 04.
Article in English | MEDLINE | ID: mdl-33612386

ABSTRACT

BACKGROUND: Cardiorenal complications are common in patients with dysmetabolism and diabetes. The present study aimed to examine if a nonhuman primate (NHP) model with spontaneously developed metabolic disorder and diabetes develops similar complications to humans, such as proteinuria and cardiac dysfunction at resting condition or diminished cardiac functional reserve following dobutamine stress echocardiography (DSE). METHODS AND RESULTS: A total of 66 dysmetabolic and diabetic cynomolgus (Macaca fascicularis) NHPs were enrolled to select 19 NHPs (MetS) with marked metabolic disorders and diabetes (fasting blood glucose: 178 ±â€¯18 vs. 61 ±â€¯3 mg/dL) accompanied by proteinuria (ACR: 134 ±â€¯34 vs. 1.5 ±â€¯0.4 mg/mmol) compared to 8 normal NHPs (CTRL). Under resting condition, MetS NHPs showed mild left ventricular (LV) diastolic dysfunction (E/A: 1 ±â€¯0.06 vs. 1.5 ±â€¯0.13), but with preserved ejection fraction (EF: 65 ±â€¯2 vs. 71 ±â€¯3%) compared to CTRL. DSE with an intravenous infusion of dobutamine at ascending doses (5, 10, 20, 30 and 40 µg/kg/min, 7 min for each dose) resulted in a dose-dependent increase in cardiac function, however, with a significantly diminished magnitude at the highest dose of dobutamine infusion (40 µg/kg/min) in both diastole (E/A: -12 ±â€¯3 vs. -38 ±â€¯5%) and systole (EF: 25 ±â€¯3 vs. 33 ±â€¯5%) as well as ~42% reduced cardiac output reserve (COR: 63 ±â€¯8 vs. 105 ±â€¯18%, p < 0.02) in the MetS compared to CTRL NHPs. CONCLUSION: These data demonstrate that MetS NHPs with cardiorenal complications: proteinuria, LV diastolic dysfunction and preserved LV systolic function under resting conditions displayed compromised cardiac functional reserve under dobutamine stress. Based on these phenotypes, this NHP model of diabetes with cardiorenal complications can be used as a highly translational model mimic human disease for pharmaceutical research.


Subject(s)
Proteinuria , Ventricular Dysfunction, Left , Animals , Cardiac Output , Diabetes Mellitus , Dobutamine/pharmacology , Macaca fascicularis , Proteinuria/complications , Stroke Volume , Ventricular Dysfunction, Left/complications
7.
Expert Opin Drug Saf ; 19(3): 281-294, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32064957

ABSTRACT

Introduction: Drug-induced myocardial dysfunction is an important safety concern during drug development. Oncology compounds can cause myocardial dysfunction, leading to decreased left ventricular ejection fraction and heart failure via several mechanisms. Cardiovascular imaging has a major role in the early detection and monitoring of cardiotoxicity. Echocardiography is the method of choice because of its widespread availability, low cost, and absence of radiation exposure. Cardiac magnetic resonance imaging can provide better reliability, reproducibility, and accuracy in the detection of drug-induced myocardial dysfunction. In addition, it enables assessment of myocardial edema, fibrosis, and necrosis. Cardiac serologic biomarkers such as troponins and B-type natriuretic peptides are used in combination with imaging during drug development. This article provides a general overview of each imaging modality and practical guidance for early detection and monitoring of cardiotoxicity.Areas covered: Cardiovascular imaging modalities and cardiac biomarkers for monitoring of cardiac function and early detection of drug-induced myocardial dysfunction in drug development.Expert opinion: Some new drugs especially in the oncology field, can cause myocardial dysfunction. Depending on the strength of pre-clinical or clinical data, CV imaging modalities and cardiac biomarkers play an important role in the early detection and mitigation plans for such drugs during their development.


Subject(s)
Antineoplastic Agents/adverse effects , Biomarkers/blood , Cardiotoxicity/diagnostic imaging , Drug Development/methods , Echocardiography/methods , Magnetic Resonance Imaging/methods , Animals , Cardiotoxicity/blood , Early Diagnosis , Humans
8.
Sci Rep ; 8(1): 17509, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504800

ABSTRACT

Capable of mediating efficient transfection and protein production without eliciting innate immune responses, chemically modified mRNA holds great potential to produce paracrine factors at a physiologically beneficial level, in a spatiotemporally controlled manner, and with low toxicity. Although highly promising in cardiovascular medicine and wound healing, effects of this emerging therapeutic on the microvasculature and its bioactivity in disease settings remain poorly understood. Here, we longitudinally and comprehensively characterize microvascular responses to AZD8601, a modified mRNA encoding vascular endothelial growth factor A (VEGF-A), in vivo. Using multi-parametric photoacoustic microscopy, we show that intradermal injection of AZD8601 formulated in a biocompatible vehicle results in pronounced, sustained and dose-dependent vasodilation, blood flow upregulation, and neovessel formation, in striking contrast to those induced by recombinant human VEGF-A protein, a non-translatable variant of AZD8601, and citrate/saline vehicle. Moreover, we evaluate the bioactivity of AZD8601 in a mouse model of diabetic wound healing in vivo. Using a boron nanoparticle-based tissue oxygen sensor, we show that sequential dosing of AZD8601 improves vascularization and tissue oxygenation of the wound bed, leading to accelerated re-epithelialization during the early phase of diabetic wound healing.


Subject(s)
Diabetic Angiopathies/etiology , Diabetic Angiopathies/pathology , Microvessels/metabolism , RNA, Messenger/genetics , Vascular Endothelial Growth Factor A/genetics , Wound Healing/genetics , Animals , Diabetic Angiopathies/diagnostic imaging , Disease Models, Animal , Humans , Mice , Microvessels/drug effects , Myocytes, Smooth Muscle/metabolism , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/genetics , Oxygen Consumption , Time-Lapse Imaging , Wound Healing/drug effects
9.
ACS Med Chem Lett ; 9(7): 600-605, 2018 Jul 12.
Article in English | MEDLINE | ID: mdl-30034586

ABSTRACT

A lead generation campaign identified indole-based sPLA2-X inhibitors with a promising selectivity profile against other sPLA2 isoforms. Further optimization of sPLA2 selectivity and metabolic stability resulted in the design of (-)-17, a novel, potent, and selective sPLA2-X inhibitor with an exquisite pharmacokinetic profile characterized by high absorption and low clearance, and low toxicological risk. Compound (-)-17 was tested in an ApoE-/- murine model of atherosclerosis to evaluate the effect of reversible, pharmacological sPLA2-X inhibition on atherosclerosis development. Despite being well tolerated and achieving adequate systemic exposure of mechanistic relevance, (-)-17 did not significantly affect circulating lipid and lipoprotein biomarkers and had no effect on coronary function or histological markers of atherosclerosis.

10.
J Card Fail ; 24(7): 470-478, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29802896

ABSTRACT

BACKGROUND: Oxytocin (Oxt) and its receptor (Oxtr) gene system has been implicated in cardiomyogenesis and cardioprotection; however, effects of chronic activation of Oxtr are not known. We generated and investigated transgenic (TG) mice that overexpress Oxtr specifically in the heart. METHODS AND RESULTS: Cardiac-specific overexpression of Oxtr was obtained by having the α-major histocompatibility complex promoter drive the mouse Oxtr gene (α-Mhc-Oxtr). Left ventricular (LV) function and remodeling were assessed by magnetic resonance imaging and echocardiography. In α-Mhc-Oxtr TG mice, LV ejection fraction was severely compromised at 14 weeks of age compared with wild-type (WT) littermates (25 ± 6% vs 63 ± 3%; P < .001). LV end-diastolic volume was larger in the TG mice (103 ± 6 µL vs 67 ± 5 µL; P < .001). α-Mhc-Oxtr TG animals displayed cardiac fibrosis, atrial thrombus, and increased expression of pro-fibrogenic genes. Mortality of α-Mhc-Oxtr TG animals was 45% compared with 0% (P < .0001) of WT littermates by 20 weeks of age. Most cardiomyocytes of α-Mhc-Oxtr TG animals but not WT littermates (68.0 ± 12.1% vs 5.6 ± 2.4%; P = .008) were positive in staining for nuclear factor of activated T cells (NFAT). To study if thrombin inhibitor prevents thrombus formation, a cohort of 7-week-old α-Mhc-Oxtr TG mice were treated for 12 weeks with AZD0837, a potent thrombin inhibitor. Treatment with AZD0837 reduced thrombus formation (P < .05) and tended to attenuate fibrosis and increase survival. CONCLUSIONS: Cardiac-specific overexpression of Oxtr had negative consequences on LV function and survival in mice. The present findings necessitate further studies to investigate potential adverse effects of chronic Oxt administration. We provide a possible mechanism of Oxtr overexpression leading to heart failure by nuclear factor of activated T cell signaling. The recapitulation of human heart failure and the beneficial effects of the antithrombin inhibitor render the α-Mhc-Oxtr TG mice a promising tool in drug discovery for heart failure.


Subject(s)
Cardiomyopathies/genetics , Gene Expression Regulation , Myocardium/metabolism , RNA/genetics , Receptors, Oxytocin/genetics , Animals , Cardiomyopathies/diagnosis , Cardiomyopathies/metabolism , Disease Models, Animal , Echocardiography , Magnetic Resonance Imaging, Cine , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Real-Time Polymerase Chain Reaction , Receptors, Oxytocin/biosynthesis
11.
Sci Rep ; 7(1): 17434, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234030

ABSTRACT

Sepsis induced cardiac dysfunction (SIC) is a severe complication to sepsis which significantly worsens patient outcomes. It is known that bacteria have the capacity to release outer membrane vesicles (OMVs), which are nano-sized bilayered vesicles composed of lipids and proteins, that can induce a fatal inflammatory response. The aim of this study was to determine whether OMVs from a uropathogenic Escherichia coli strain can induce cardiac dysfunction, and to elucidate any mechanisms involved. OMVs induced irregular Ca2+ oscillations with a decreased frequency in cardiomyocytes through recordings of intracellular Ca2+ dynamics. Mice were intraperitoneally injected with bacteria-free OMVs, which resulted in increased concentration of pro-inflammatory cytokine levels in blood. Cytokines were increased in heart lysates, and OMVs could be detected in the heart after OMVs injection. Troponin T was significantly increased in blood, and echocardiography showed increased heart wall thickness as well as increased heart rate. This study shows that E. coli OMVs induce cardiac injury in vitro and in vivo, in the absence of bacteria, and may be a causative microbial signal in SIC. The role of OMVs in clinical disease warrant further studies, as bacterial OMVs in addition to live bacteria may be good therapeutic targets to control sepsis.


Subject(s)
Cell-Derived Microparticles/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Heart Diseases/immunology , Sepsis/immunology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Calcium/metabolism , Cell Line , Cell Survival/physiology , Cell-Derived Microparticles/drug effects , Cytokines/metabolism , Escherichia coli Infections/diagnostic imaging , Escherichia coli Infections/drug therapy , Escherichia coli Infections/pathology , Heart Diseases/diagnostic imaging , Heart Diseases/drug therapy , Heart Diseases/pathology , Lipopolysaccharides , Mice, Inbred C57BL , Myocardium/immunology , Myocardium/pathology , Polymyxin B/pharmacology , Random Allocation , Sepsis/diagnostic imaging , Sepsis/drug therapy , Sepsis/pathology , Troponin T/blood
12.
Circ Res ; 116(4): 642-52, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25556206

ABSTRACT

RATIONALE: In human genetic studies a single nucleotide polymorphism within the salt-inducible kinase 1 (SIK1) gene was associated with hypertension. Lower SIK1 activity in vascular smooth muscle cells (VSMCs) leads to decreased sodium-potassium ATPase activity, which associates with increased vascular tone. Also, SIK1 participates in a negative feedback mechanism on the transforming growth factor-ß1 signaling and downregulation of SIK1 induces the expression of extracellular matrix remodeling genes. OBJECTIVE: To evaluate whether reduced expression/activity of SIK1 alone or in combination with elevated salt intake could modify the structure and function of the vasculature, leading to higher blood pressure. METHODS AND RESULTS: SIK1 knockout (sik1(-/-)) and wild-type (sik1(+/+)) mice were challenged to a normal- or chronic high-salt intake (1% NaCl). Under normal-salt conditions, the sik1(-/-) mice showed increased collagen deposition in the aorta but similar blood pressure compared with the sik1(+/+) mice. During high-salt intake, the sik1(+/+) mice exhibited an increase in SIK1 expression in the VSMCs layer of the aorta, whereas the sik1(-/-) mice exhibited upregulated transforming growth factor-ß1 signaling and increased expression of endothelin-1 and genes involved in VSMC contraction, higher systolic blood pressure, and signs of cardiac hypertrophy. In vitro knockdown of SIK1 induced upregulation of collagen in aortic adventitial fibroblasts and enhanced the expression of contractile markers and of endothelin-1 in VSMCs. CONCLUSIONS: Vascular SIK1 activation might represent a novel mechanism involved in the prevention of high blood pressure development triggered by high-salt intake through the modulation of the contractile phenotype of VSMCs via transforming growth factor-ß1-signaling inhibition.


Subject(s)
Aorta/enzymology , Arterial Pressure , Hypertension/enzymology , Protein Serine-Threonine Kinases/deficiency , Vascular Remodeling , Adventitia/enzymology , Adventitia/pathology , Animals , Aorta/pathology , Aorta/physiopathology , Cells, Cultured , Collagen/metabolism , Endothelin-1/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , Genotype , Humans , Hypertension/etiology , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Mice, Knockout , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Natriuresis , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Signal Transduction , Sodium Chloride, Dietary , Sympathetic Nervous System/physiopathology , Transfection , Transforming Growth Factor beta1/metabolism , Vasoconstriction
13.
Eur Heart J Cardiovasc Imaging ; 15(2): 152-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23832950

ABSTRACT

AIMS: Stress-induced cardiomyopathy (SIC) is an important differential diagnosis to acute myocardial infarction (AMI) that is associated with significant morbidity and mortality. The typical hallmark of SIC is left-ventricular apical akinesia but preserved function in basal segments. Catecholamines are postulated to play an important role in SIC but the precise pathophysiology is incompletely understood. Whether myocardial perfusion of the affected segments is impaired in SIC has been debated and remains unknown. METHODS AND RESULTS: Myocardial contrast echocardiography (MCE) was used to study regional myocardial perfusion in a rat model of SIC. Twelve rats received 50 mg/kg isoproterenol (ISO) i.p. and were continuously monitored by MCE. Apical and basal perfusion were estimated and expressed as a ratio at baseline, 5, 10, 20, 30, 40, 50, 60, 70, 80, and 90 min post-ISO. The rats developed typical apical ballooning after 43 ± 9 min post-ISO injection. The ratio of apical:basal perfusion was close to 1.00 at all time-points and never dropped below 0.89 (95% CI never extended below 0.73). Light and electron microcoscopical investigation revealed no structural damage of myocardial vessels. CONCLUSION: Apical perfusion is not impaired in the early phase of SIC in this rat model.


Subject(s)
Coronary Circulation/physiology , Echocardiography/methods , Takotsubo Cardiomyopathy/diagnostic imaging , Animals , Contrast Media , Disease Models, Animal , Image Processing, Computer-Assisted , Isoproterenol/pharmacology , Rats, Sprague-Dawley , Reproducibility of Results , Takotsubo Cardiomyopathy/physiopathology
14.
PLoS One ; 8(11): e80330, 2013.
Article in English | MEDLINE | ID: mdl-24324556

ABSTRACT

Adiponectin has been shown to have beneficial cardiovascular effects and to signal through the adiponectin receptors, AdipoR1 and AdipoR2. The original aim of this study was to investigate the effect of combined AdipoR1 and AdipoR2 deficiency (AdipoR1(-/-)AdipoR2(-/-)) on atherosclerosis. However, we made the interesting observation that AdipoR1(-/-) AdipoR2(-/-) leads to embryonic lethality demonstrating the critical importance of the adiponectin signalling system during development. We then investigated the effect of AdipoR2-ablation on the progression of atherosclerosis in apolipoprotein E deficient (ApoE(-/-)) mice. AdipoR2(-/-)ApoE(-/-) mice fed an atherogenic diet had decreased plaque area in the brachiocephalic artery compared with AdipoR2(+/+) ApoE(-/-) littermate controls as visualized in vivo using an ultrasound biomicroscope and confirmed by histological analyses. The decreased plaque area in the brachiocephalic artery could not be explained by plasma cholesterol levels or inflammatory status. However, accumulation of neutral lipids was decreased in peritoneal macrophages from AdipoR2(-/-)ApoE(-/-) mice after incubation with oxidized LDL. This effect was associated with lower CD36 and higher ABCA1 mRNA levels in peritoneal macrophages from AdipoR2(-/-)ApoE(-/-) mice compared with AdipoR2(+/+)ApoE(-/-) controls after incubation with oxidized LDL. In summary, we show that adiponectin receptors are crucial during embryonic development and that AdipoR2-deficiency slows down the progression of atherosclerosis in the brachiocephalic artery of ApoE-deficient mice.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Receptors, Adiponectin/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Blotting, Western , Body Weight/genetics , Body Weight/physiology , Female , Genotype , Heterozygote , Macrophages/metabolism , Male , Mice , Mice, Knockout , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics
15.
J Cardiovasc Transl Res ; 6(5): 715-28, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23877202

ABSTRACT

Myocardial ischemia is recognized as an important mechanism increasing the risk for cardiovascular events in both symptomatic and asymptomatic patients. In addition to obstructive coronary diseases, systemic inflammation, macro- and microvascular function are additional important mechanisms contributing to the ischemic myocardium. Accumulating evidence indicates that coronary flow reserve (CFR) is a quantitative measurement of ischemia including integrated information on structure and function of the coronary artery at all levels. Not surprisingly, CFR has been shown to confer strong prognostic value for hard cardiovascular (CV) events in a number of relevant patient cohorts. Using high-resolution imaging, it is now possible to study coronary arteries from mouse to man. Therefore, CFR may be an important translational tool to risk-stratify patients and to perform both preclinical and clinical proof-of-concept studies before investing in large-scale outcome trials, thus improving the translational value for novel CV targets.


Subject(s)
Coronary Artery Disease/physiopathology , Coronary Vessels/physiopathology , Fractional Flow Reserve, Myocardial , Animals , Cardiovascular Agents/therapeutic use , Coronary Artery Disease/diagnosis , Coronary Artery Disease/drug therapy , Coronary Vessels/drug effects , Diagnostic Imaging/methods , Disease Models, Animal , Fractional Flow Reserve, Myocardial/drug effects , Hemodynamics , Humans , Mice , Predictive Value of Tests , Prognosis , Severity of Illness Index
16.
Int J Mol Med ; 29(3): 331-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22160183

ABSTRACT

Diet is a significant modifiable risk factor for cardiovascular disease and high fish intake has been associated with vascular health in population studies. However, intervention studies have been inconclusive. In this study, male low-density lipoprotein receptor-deficient mice were given 16-week high fat/high sucrose diets, supplemented with either minced herring fillets or minced beef. The diets were matched in total fat and cholesterol content; taurine content and fatty acid composition was analysed. Body weights were recorded throughout the study; plasma lipids were analysed at week 8 and 16. Body composition and adipocyte size were evaluated at study end. Atherosclerosis was evaluated at week 12 (ultrasound) and at termination (en face histology). Herring-fed mice had a higher proportion of long-chain n-3 polyunsaturated fatty acids in the hepatic triacylglycerides (TAG) and phospholipid fractions. The herring-fed mice had increased body weight (P=0.007), and reduced epididymal adipocyte size (P=0.009), despite similar food intake and body composition as the beef-fed mice. The herring-fed mice had lower plasma TAG and very-low-density lipoprotein (VLDL)-cholesterol concentrations throughout the study (TAG; P=0.0012 and 0.004, VLDL-cholesterol; P=0.006 and 0.041, week 8 and 16, respectively). At week 16, the herring-fed had higher plasma concentrations of HDL-cholesterol (P=0.004) and less atherosclerotic lesions in the aortic arch (P=0.007) compared with the beef-fed mice. In conclusion, dietary herring in comparison to beef markedly improved vascular health in this mouse model, suggesting that herring provides an added value beyond its content of macronutrients.


Subject(s)
Atherosclerosis/diet therapy , Fishes , Lipids/blood , Receptors, LDL/genetics , Adipocytes/cytology , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Body Weight , Cell Size , Diet, High-Fat , Lipid Metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , Mice, Obese , Receptors, LDL/deficiency
17.
Am J Physiol Heart Circ Physiol ; 297(3): H1078-86, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19617410

ABSTRACT

Previous studies have shown that exposure to chronic hypoxia protects against myocardial infarction, but little is known about the cellular and molecular mechanisms involved. Here we observed that chronic hypoxia for 3 wk resulted in improved survival of mice (from 64% to 83%), reduced infarction size (from 45 +/- 4% to 32 +/- 4%, P < 0.05), increased cardiac ejection fraction (from 19 +/- 4% to 35 +/- 5%, P < 0.05), coronary flow velocity under adenosine-induced hyperemia (from 58 +/- 2 to 75 +/- 5 cm/s, P < 0.05), myocardial capillary density (from 3,772 +/- 162 to 4,760 +/- 197 capillaries/mm(2), P < 0.01), and arteriolar density (from 8.04 +/- 0.76 to 10.34 +/- 0.69 arterioles/mm(2), P < 0.05) 3 wk after myocardial infarction. With two-dimensional gel electrophoresis, we identified that protein disulfide isomerase (PDI) was highly upregulated in hypoxic myocardial capillary endothelial cells. The loss of PDI function in endothelial cells by small interfering RNA significantly increased the number of apoptotic cells (by 3.4-fold at hypoxia, P < 0.01) and reduced migration (by 52% at hypoxia, P < 0.001) and adhesion to collagen I (by 42% at hypoxia, P < 0.01). In addition, the specific inhibition of PDI by PDI small interfering RNA (by 46%, P < 0.01) and bacitracin (by 72%, P < 0.001) reduced the formation of tubular structures by endothelial cells. Our data indicate that chronic hypoxic exposure improves coronary blood flow and protects the myocardium against infarction. These beneficial effects may be partly explained by the increased endothelial expression of PDI, which protects cells against apoptosis and increases cellular migration, adhesion, and tubular formation. The increased PDI expression in endothelial cells may be a novel mechanism to protect the myocardium against myocardial ischemic diseases.


Subject(s)
Coronary Circulation/physiology , Endothelial Cells/enzymology , Hypoxia/metabolism , Hypoxia/physiopathology , Neovascularization, Physiologic/physiology , Protein Disulfide-Isomerases/metabolism , Animals , Apoptosis/physiology , Arterioles/physiology , Body Weight , Capillaries/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Chronic Disease , Endothelial Cells/cytology , Hemoglobins/metabolism , Humans , Hypoxia/pathology , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Organ Size , Protein Disulfide-Isomerases/genetics , RNA, Small Interfering , Umbilical Veins/cytology , Up-Regulation/physiology
18.
Ultrasound Med Biol ; 34(7): 1053-62, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18313201

ABSTRACT

For an accurate estimate of volumetric coronary flow reserve (CFR) using Doppler-assessed flow velocity measurement, it is important to take into consideration potential diameter change during coronary hyperemia. Using ultrasound techniques, left coronary artery (LCA) flow velocity and LCA lumen diameter (LCA(D)) were measured simultaneously for the first time to measure coronary flow during baseline and adenosine-induced hyperemic condition in isoflurane-anesthetized C57BL/6 (n = 38) and in old apolipoprotein E-gene deficient (ApoE(-/-)) mice (n = 44) mice. LCA(D) increased significantly and to a similar extent during adenosine infusion in both groups (3.7 +/- 1.1 %, p < 0.003 for C57BL/6; 4.2 +/- 0.9 %, p < 0.00003 for ApoE(-/-)). Yet, a positive correlation was still found between velocity-based coronary flow velocity reserve (CFVR) and volumetric CFR in both strains (R(2) = 0.77, p < 0.001 for C57BL/6; R(2) = 0.80, p < 0.001 for ApoE(-/-)). Coronary reserve was higher in C57BL/6 mice than in ApoE(-/-) mice (CFR 1.93 +/- 0.17 vs. 1.47 +/- 0.07, p < 0.05; CFVR 1.73 +/- 0.13 vs. 1.28 +/- 0.07, p < 0.01). Thus, ultrasound techniques can be used to measure volumetric flow in the LCA and flow-based CFR measurements of intact, living mice. The positive correlation between CFR and CFVR, together with the lower method variability of the latter, makes CFVR a more robust protocol for assessing mouse in-vivo coronary artery function. Therefore, the CFVR protocol will probably work well in most settings.


Subject(s)
Coronary Vessels/physiology , Vasodilation/physiology , Adenosine , Animals , Apolipoproteins E/deficiency , Atherosclerosis/diagnostic imaging , Atherosclerosis/physiopathology , Blood Flow Velocity , Coronary Vessels/anatomy & histology , Coronary Vessels/diagnostic imaging , Coronary Vessels/physiopathology , Hyperemia/diagnostic imaging , Hyperemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Acoustic , Species Specificity , Vasodilator Agents
19.
Atherosclerosis ; 190(2): 313-20, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16677654

ABSTRACT

There are increasing needs to develop imaging techniques to study in vivo vascular morphology and function in various mouse models of atherosclerosis. Using ultrasound biomicroscopy (UBM), we developed and validated a new imaging protocol to follow lesion progression in atherosclerotic mice. ApoE and LDL receptor double knockout mice (DKO) with various degree of atherosclerosis and normal control mice were imaged at the level of the ascending aorta using UBM. Average plaque thickness, as well as plaque area were delineated in the short-axis images, and were subsequently compared with histological measurements. We showed that plaque area at this vascular site was closely correlated to total plaque burden from en face measurement (p<0.0001). UBM-measured plaque thickness and area correlated with indices for histology measures from the same vascular region (p<0.0001 respective p<0.0001). Furthermore, in 16 DKO mice aged from 32 to 35 weeks, UBM showed significantly weekly increases of IMT in the ascending aorta from 0.106+/-0.108 mm at 32 weeks of age to 0.256+/-0.345 mm at 35 weeks of age (p=0.0002). In conclusion, this novel imaging protocol provides us with a non-invasive, accurate and inexpensive way to follow lesion progression in mice in vivo.


Subject(s)
Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/pathology , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Aging , Animals , Aorta, Thoracic/growth & development , Apolipoproteins E/deficiency , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/pathology , Disease Models, Animal , Image Processing, Computer-Assisted , Mice , Mice, Knockout , Receptors, LDL/deficiency , Reproducibility of Results , Sensitivity and Specificity , Ultrasonography
20.
Arterioscler Thromb Vasc Biol ; 26(5): 1126-31, 2006 May.
Article in English | MEDLINE | ID: mdl-16514080

ABSTRACT

BACKGROUND: We aimed to establish a completely noninvasive technique to assess coronary artery atherosclerosis in living mice using proximal to middle left coronary artery (LCA) velocity ratio as assessed with color Doppler echocardiography (CDE). METHODS AND RESULTS: Three groups of apolipoprotein E/low-density lipoprotein receptor double-knockout (apoE/LDLr dko) mice 10, 40, and 80 weeks of age and 3 additional age-matched groups of C57BL/6 mice were examined under anesthesia. Coronary flow velocity in proximal (Vprox) and middle part (Vmid) of LCA was measured using CDE. A 40-MHz ultrasound biomicroscope (UBM) was used to visualize lumen and outer vessel diameter in the proximal LCA. Flow velocity in the proximal LCA increased significantly with age and remained constant in the middle part in the apoE/LDLr dko mice, whereas velocities at both the sites remained unchanged in C57 mice. CDE-assessed flow velocity ratio (Vprox/Vmid) increased significantly with age in apoE/LDLr dko mice (P=0.0055) and correlated significantly to percentage wall thickness, as assessed by UBM (P=0.0044; r=0.65) and histology (P=0.0002; r=0.78). Wall thickness increased with age in the apoE/LDLr dko mice as measured with UBM (P=0.0093; r=0.49), which was also confirmed with histology (P<0.0001; r=0.73). CONCLUSIONS: CDE and UBM are useful noninvasive tools to quantify mouse coronary artery atherosclerosis in vivo.


Subject(s)
Coronary Artery Disease/diagnosis , Coronary Circulation , Echocardiography, Doppler, Color , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/physiology , Blood Flow Velocity , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/etiology , Coronary Artery Disease/physiopathology , Coronary Vessels/pathology , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Acoustic , Receptors, LDL/deficiency , Receptors, LDL/physiology , Vascular Resistance
SELECTION OF CITATIONS
SEARCH DETAIL
...