Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Immunol ; 15: 1360700, 2024.
Article in English | MEDLINE | ID: mdl-38736886

ABSTRACT

Introduction: Myocardial infarction (MI) is a significant contributor to morbidity and mortality worldwide. Many individuals who survive the acute event continue to experience heart failure (HF), with inflammatory and healing processes post-MI playing a pivotal role. Polymorphonuclear neutrophils (PMN) and monocytes infiltrate the infarcted area, where PMN release high amounts of the heme enzyme myeloperoxidase (MPO). MPO has numerous inflammatory properties and MPO plasma levels are correlated with prognosis and severity of MI. While studies have focused on MPO inhibition and controlling PMN infiltration into the infarcted tissue, less is known on MPO's role in monocyte function. Methods and results: Here, we combined human data with mouse and cell studies to examine the role of MPO on monocyte activation and migration. We revealed a correlation between plasma MPO levels and monocyte activation in a patient study. Using a mouse model of MI, we demonstrated that MPO deficiency led to an increase in splenic monocytes and a decrease in cardiac monocytes compared to wildtype mice (WT). In vitro studies further showed that MPO induces monocyte migration, with upregulation of the chemokine receptor CCR2 and upregulation of inflammatory pathways identified as underlying mechanisms. Conclusion: Taken together, we identify MPO as a pro-inflammatory mediator of splenic monocyte recruitment and activation post-MI and provide mechanistic insight for novel therapeutic strategies after ischemic injury.


Subject(s)
Monocytes , Myocardial Infarction , Peroxidase , Animals , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Peroxidase/metabolism , Monocytes/immunology , Monocytes/metabolism , Humans , Mice , Male , Cell Movement , Disease Models, Animal , Mice, Inbred C57BL , Female , Neutrophils/immunology , Neutrophils/metabolism , Mice, Knockout , Receptors, CCR2/metabolism , Middle Aged
2.
Naunyn Schmiedebergs Arch Pharmacol ; 397(4): 2401-2420, 2024 04.
Article in English | MEDLINE | ID: mdl-37843590

ABSTRACT

We have shown that in murine cardiomyopathy caused by overexpression of the ß1-adrenoceptor, Gαi2-deficiency is detrimental. Given the growing evidence for isoform-specific Gαi-functions, we now examined the consequences of Gαi3 deficiency in the same heart-failure model. Mice overexpressing cardiac ß1-adrenoceptors with (ß1-tg) or without Gαi3-expression (ß1-tg/Gαi3-/-) were compared to C57BL/6 wildtypes and global Gαi3-knockouts (Gαi3-/-). The life span of ß1-tg mice was significantly shortened but improved when Gαi3 was lacking (95% CI: 592-655 vs. 644-747 days). At 300 days of age, left-ventricular function and survival rate were similar in all groups. At 550 days of age, ß1-tg but not ß1-tg/Gαi3-/- mice displayed impaired ejection fraction (35 ± 18% vs. 52 ± 16%) compared to wildtype (59 ± 4%) and Gαi3-/- mice (60 ± 5%). Diastolic dysfunction of ß1-tg mice was prevented by Gαi3 deficiency, too. The increase of ANP mRNA levels and ventricular fibrosis observed in ß1-tg hearts was significantly attenuated in ß1-tg/Gαi3-/- mice. Transcript levels of phospholamban, ryanodine receptor 2, and cardiac troponin I were similar in all groups. However, Western blots and phospho-proteomic analyses showed that in ß1-tg, but not ß1-tg/Gαi3-/- ventricles, phospholamban protein was reduced while its phosphorylation increased. Here, we show that in mice overexpressing the cardiac ß1-adrenoceptor, Gαi3 deficiency slows or even prevents cardiomyopathy and increases shortened life span. Previously, we found Gαi2 deficiency to aggravate cardiac dysfunction and mortality in the same heart-failure model. Our findings indicate isoform-specific interventions into Gi-dependent signaling to be promising cardio-protective strategies.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Mice , Cardiomyopathies/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Protein Isoforms/metabolism , Proteomics
3.
Int J Mol Sci ; 24(20)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37894941

ABSTRACT

Vascular diseases like atherosclerosis and abdominal aortic aneurysm (AAA) are common pathologies in the western world, promoting various potentially fatal conditions. Here, we evaluate high-resolution (HR) ultrasound in mouse models of atherosclerosis and AAA as a useful tool for noninvasive monitoring of early vascular changes in vivo. We used Apolipoprotein E-deficient (ApoE-/-) mice as an atherosclerosis model and induced AAA development by the implementation of Angiotensin II-releasing osmotic minipumps. HR ultrasound of the carotid artery or the abdominal aorta was performed to monitor vascular remodeling in vivo. Images were analyzed by speckle tracking algorithms and correlated to histological analyses and subsequent automated collagen quantification. Consistent changes were observed via ultrasound in both models: Global radial strain (GRS) was notably reduced in the AAA model (23.8 ± 2.8% vs. 12.5 ± 2.5%, p = 0.01) and in the atherosclerotic mice (20.6 ± 1.3% vs. 15.8 ± 0.9%, p = 0.02). In mice with AAA, vessel distensibility was significantly reduced, whereas intima-media thickness was increased in atherosclerotic mice. The area and collagen content of the tunica media were increased in diseased arteries of both models as measured by automated image analysis of Picrosirius Red-stained aortic sections. Correlation analysis revealed a strong correlation of multiple parameters, predicting early vascular damage in HR ultrasound and histological examinations. In conclusion, our findings underscore the potential of HR ultrasound in effectively tracing early alterations in arterial wall properties in murine models of atherosclerosis and AAA.


Subject(s)
Aortic Aneurysm, Abdominal , Atherosclerosis , Mice , Animals , Disease Models, Animal , Carotid Intima-Media Thickness , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/pathology , Aorta, Abdominal/diagnostic imaging , Angiotensin II , Collagen , Mice, Inbred C57BL
4.
Basic Res Cardiol ; 118(1): 36, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37656254

ABSTRACT

Cardiotoxicity is a major complication of anthracycline therapy that negatively impacts prognosis. Effective pharmacotherapies for prevention of anthracycline-induced cardiomyopathy (AICM) are currently lacking. Increased plasma levels of the neutrophil-derived enzyme myeloperoxidase (MPO) predict occurrence of AICM in humans. We hypothesized that MPO release causally contributes to AICM. Mice intravenously injected with the anthracycline doxorubicin (DOX) exhibited higher neutrophil counts and MPO levels in the circulation and cardiac tissue compared to saline (NaCl)-treated controls. Neutrophil-like HL-60 cells exhibited increased MPO release upon exposition to DOX. DOX induced extensive nitrosative stress in cardiac tissue alongside with increased carbonylation of sarcomeric proteins in wildtype but not in Mpo-/- mice. Accordingly, co-treatment of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with DOX and MPO aggravated loss of hiPSC-CM-contractility compared to DOX treatment alone. DOX-treated animals exhibited pronounced cardiac apoptosis and inflammation, which was attenuated in MPO-deficient animals. Finally, genetic MPO deficiency and pharmacological MPO inhibition protected mice from the development of AICM. The anticancer efficacy of DOX was unaffected by MPO deficiency. Herein we identify MPO as a critical mediator of AICM. We demonstrate that DOX induces cardiac neutrophil infiltration and release of MPO, which directly impairs cardiac contractility through promoting oxidation of sarcomeric proteins, cardiac inflammation and cardiomyocyte apoptosis. MPO thus emerges as a promising pharmacological target for prevention of AICM.


Subject(s)
Cardiomyopathies , Induced Pluripotent Stem Cells , Peroxidase , Animals , Humans , Mice , Anthracyclines/toxicity , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Doxorubicin/toxicity , Inflammation , Peroxidase/genetics
5.
Cardiovasc Res ; 118(9): 2211-2225, 2022 07 20.
Article in English | MEDLINE | ID: mdl-34324651

ABSTRACT

AIMS: Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the Fibrillin-1 gene. It is associated with formation of thoracic aortic aneurysms that can potentially be a life-threatening condition due to aortic rupture or dissection. Excessive non-canonical transforming growth factor beta signalling, mediated by activation of extracellular signal-regulated kinases 1/2 (ERK1/2), as well as inducible nitric oxide synthase (NOS2)-dependent nitric oxide production, have been identified to drive aortic pathology in MFS through induction of elastin fragmentation and smooth muscle cell apoptosis. Despite promising results in animal studies, specific pharmacological interventions approved for clinical use in patients with MFS-related aortic disease are rare. Nitro-oleic acid (NO2-OA) is an endogenously generated signalling modulator, which is available as an oral compound and has been shown to inhibit ERK1/2 activation and NOS2 expression in different disease models, thereby exerting promising therapeutic effects. In this study, we investigated whether NO2-OA decreases aortic dilation in MFS. METHODS AND RESULTS: Eight-week-old MFS (Fbn1C1041G/+) mice were treated with NO2-OA or vehicle for 4 weeks via subcutaneously implanted osmotic minipumps. Echocardiography indicated progressive ascending aortic dilation and wall stiffening in MFS mice, which was significantly attenuated by NO2-OA treatment. This protective effect was mediated by inhibition of aortic ERK1/2, Smad2 as well as nuclear factor kappa B overactivation and consequent attenuation of elastin fragmentation by matrix metalloproteinase 2, apoptosis, and collagen deposition. Critically, the therapeutic efficacy of NO2-OA in MFS was further emphasized by demonstrating its capability to reduce lethal aortic complications in Fbn1C1041G/+ mice challenged with Angiotensin II. CONCLUSION: NO2-OA distinctly attenuates progression of aortic dilation in MFS via modulation of well-established disease-mediating pathways, thereby meriting further investigation into its application as a therapeutic agent for the treatment of this condition.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Aneurysm , Aortic Diseases , Marfan Syndrome , Animals , Aortic Aneurysm/genetics , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/genetics , Aortic Diseases/pathology , Disease Models, Animal , Elastin/metabolism , Fibrillin-1/genetics , Marfan Syndrome/complications , Marfan Syndrome/drug therapy , Marfan Syndrome/genetics , Matrix Metalloproteinase 2 , Mice , Nitro Compounds , Oleic Acids
6.
Antioxidants (Basel) ; 12(1)2022 Dec 24.
Article in English | MEDLINE | ID: mdl-36670895

ABSTRACT

Myocardial infarction (MI) is a leading cause of morbidity and mortality worldwide. Improved survival has led to an increasing incidence of ischemic cardiomyopathy, making it a major reason for hospitalization in the western world. The inflammatory response in the ischemic myocardium determines the extent of structural remodeling and functional deterioration, with neutrophils (PMN) being a key modulator of the propagation and resolution of inflammation. The heme enzyme myeloperoxidase (MPO) is abundantly expressed in PMN and is an important mediator of their inflammatory capacities. Here, we examine the effects of PMN reduction, MPO deficiency and MPO inhibition in two murine models of MI. Reduction in PMN count resulted in less scar formation and improved cardiac function. Similar results were obtained in genetically MPO deficient mice, suggesting that MPO is a critical factor in PMN-mediated cardiac remodeling. To test our findings in a therapeutic approach, we orally administered the MPO inhibitor AZM198 in the context of MI and could demonstrate improved cardiac function and reduced structural remodeling. Therefore, MPO appears to be a favorable pharmacological target for the prevention of long-term morbidity after MI.

7.
Front Immunol ; 12: 701721, 2021.
Article in English | MEDLINE | ID: mdl-34691017

ABSTRACT

The six-transmembrane protein of prostate 2 (Stamp2) acts as an anti-inflammatory protein in macrophages by protecting from overt inflammatory signaling and Stamp2 deficiency accelerates atherosclerosis in mice. Herein, we describe an unexpected role of Stamp2 in polymorphonuclear neutrophils (PMN) and characterize Stamp2's protective effects in myocardial ischemic injury. In a murine model of ischemia and reperfusion (I/R), echocardiography and histological analyses revealed a pronounced impairment of cardiac function in hearts of Stamp2-deficient- (Stamp2-/- ) mice as compared to wild-type (WT) animals. This difference was driven by aggravated cardiac fibrosis, as augmented fibroblast-to-myofibroblast transdifferentiation was observed which was mediated by activation of the redox-sensitive p38 mitogen-activated protein kinase (p38 MAPK). Furthermore, we observed increased production of reactive oxygen species (ROS) in Stamp2-/- hearts after I/R, which is the likely cause for p38 MAPK activation. Although myocardial macrophage numbers were not affected by Stamp2 deficiency after I/R, augmented myocardial infiltration by polymorphonuclear neutrophils (PMN) was observed, which coincided with enhanced myeloperoxidase (MPO) plasma levels. Primary PMN isolated from Stamp2-/- animals exhibited a proinflammatory phenotype characterized by enhanced nuclear factor (NF)-κB activity and MPO secretion. To prove the critical role of PMN for the observed phenotype after I/R, antibody-mediated PMN depletion was performed in Stamp2-/- mice which reduced deterioration of LV function and adverse structural remodeling to WT levels. These data indicate a novel role of Stamp2 as an anti-inflammatory regulator of PMN and fibroblast-to-myofibroblast transdifferentiation in myocardial I/R injury.


Subject(s)
Heart/physiology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Myocardium/metabolism , Animals , Cardiomyopathies/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , NF-kappa B/metabolism , Neutrophil Activation/physiology , Neutrophils/metabolism , Peroxidase/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Int J Mol Sci ; 22(16)2021 Aug 22.
Article in English | MEDLINE | ID: mdl-34445757

ABSTRACT

Nitro-oleic acid (NO2-OA), a nitric oxide (NO)- and nitrite (NO2-)-derived electrophilic fatty acid metabolite, displays anti-inflammatory and anti-fibrotic signaling actions and therapeutic benefit in murine models of ischemia-reperfusion, atrial fibrillation, and pulmonary hypertension. Muscle LIM protein-deficient mice (Mlp-/-) develop dilated cardiomyopathy (DCM), characterized by impaired left ventricular function and increased ventricular fibrosis at the age of 8 weeks. This study investigated the effects of NO2-OA on cardiac function in Mlp-/- mice both in vivo and in vitro. Mlp-/- mice were treated with NO2-OA or vehicle for 4 weeks via subcutaneous osmotic minipumps. Wildtype (WT) littermates treated with vehicle served as controls. Mlp-/- mice exhibited enhanced TGFß signalling, fibrosis and severely reduced left ventricular systolic function. NO2-OA treatment attenuated interstitial myocardial fibrosis and substantially improved left ventricular systolic function in Mlp-/- mice. In vitro studies of TGFß-stimulated primary cardiac fibroblasts further revealed that the anti-fibrotic effects of NO2-OA rely on its capability to attenuate fibroblast to myofibroblast transdifferentiation by inhibiting phosphorylation of TGFß downstream targets. In conclusion, we demonstrate a substantial therapeutic benefit of NO2-OA in a murine model of DCM, mediated by interfering with endogenously activated TGFß signaling.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Nitro Compounds/therapeutic use , Oleic Acids/therapeutic use , Ventricular Function, Left/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Fibrosis , Heart/drug effects , LIM Domain Proteins/genetics , Mice , Muscle Proteins/genetics , Myocardium/metabolism , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Transforming Growth Factor beta/metabolism
9.
ESC Heart Fail ; 8(1): 729-744, 2021 02.
Article in English | MEDLINE | ID: mdl-33191629

ABSTRACT

AIMS: Reduced physical activity increases the risk of heart failure; however, non-invasive methodologies detecting subclinical changes in myocardial function are not available. We hypothesized that myocardial, left ventricular, systolic strain measurements could capture subtle abnormalities in myocardial function secondary to physical inactivity. METHODS AND RESULTS: In the AGBRESA study, which assessed artificial gravity through centrifugation as potential countermeasure for space travel, 24 healthy persons (eight women) were submitted to 60 day strict -6° head-down-tilt bed rest. Participants were assigned to three groups of eight subjects: a control group, continuous artificial gravity training on a short-arm centrifuge (30 min/day), or intermittent centrifugation (6 × 5 min/day). We assessed cardiac morphology, function, strain, and haemodynamics by cardiac magnetic resonance imaging (MRI) and echocardiography. We observed no differences between groups and, therefore, conducted a pooled analysis. Consistent with deconditioning, resting heart rate (∆8.3 ± 6.3 b.p.m., P < 0.0001), orthostatic heart rate responses (∆22.8 ± 19.7 b.p.m., P < 0.0001), and diastolic blood pressure (∆8.8 ± 6.6 mmHg, P < 0.0001) increased, whereas cardiac output (∆-0.56 ± 0.94 L/min, P = 0.0096) decreased during bed rest. Left ventricular mass index obtained by MRI did not change. Echocardiographic left ventricular, systolic, global longitudinal strain (∆1.8 ± 1.83%, P < 0.0001) decreased, whereas left ventricular, systolic, global MRI circumferential strain increased not significantly (∆-0.68 ± 1.85%, P = 0.0843). MRI values rapidly returned to baseline during recovery. CONCLUSION: Prolonged head-down-tilt bed rest provokes changes in cardiac function, particularly strain measurements, that appear functional rather than mediated through cardiac remodelling. Thus, strain measurements are of limited utility in assessing influences of physical deconditioning or exercise interventions on cardiac function.


Subject(s)
Bed Rest , Gravity, Altered , Blood Pressure , Female , Head-Down Tilt , Heart , Humans
10.
Sci Rep ; 10(1): 15319, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948795

ABSTRACT

Nitro-fatty acids are electrophilic anti-inflammatory mediators which are generated during myocardial ischemic injury. Whether these species exert anti-arrhythmic effects in the acute phase of myocardial ischemia has not been investigated so far. Herein, we demonstrate that pretreatment of mice with 9- and 10-nitro-octadec-9-enoic acid (nitro-oleic acid, NO2-OA) significantly reduced the susceptibility to develop acute ventricular tachycardia (VT). Accordingly, epicardial mapping revealed a markedly enhanced homogeneity in ventricular conduction. NO2-OA treatment of isolated cardiomyocytes lowered the number of spontaneous contractions upon adrenergic isoproterenol stimulation and nearly abolished ryanodine receptor type 2 (RyR2)-dependent sarcoplasmic Ca2+ leak. NO2-OA also significantly reduced RyR2-phosphorylation by inhibition of increased CaMKII activity. Thus, NO2-OA might be a novel pharmacological option for the prevention of VT development.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Catecholamines/pharmacology , Dietary Supplements , Homeostasis/drug effects , Isoproterenol/pharmacology , Male , Mice, Inbred Strains , Myocardial Ischemia/complications , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/prevention & control
11.
Clin Res Cardiol ; 109(11): 1402-1410, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32246250

ABSTRACT

BACKGROUND: Despite all efforts, mortality of out of hospital cardiac arrest (OHCA) remains high. Patients with OHCA due to a primary shockable rhythm typically have a better prognosis. However, outcome worsens if return of spontaneous circulation (ROSC) cannot be achieved quickly. There is insufficient evidence for maximum duration of resuscitation in these patients and it is unclear, which patients profit from transport under ongoing CPR. OBJECTIVE: Investigate predictors for favourable neurologic outcome in OHCA patients with presumed cardiac cause due to refractory shockable rhythm (rSR). METHODS: Retrospective analysis of OHCA patients that presented to a tertiary hospital due to a rSR. RESULTS: One hundred seventy-five OHCA patients with presumed cardiac cause due to rSR were included. Overall hospital mortality was 50% and 83% of initial survivors were discharged with a good neurologic outcome [cerebral performance category (CPC) 1-2]. In patients with a time from cardiac arrest to ROSC of > 45 min, 18% survived to CPC 1-2. Independent predictors for good neurologic outcome were age, lower no-flow time and lower serum lactate levels at hospital arrival. CONCLUSION: In an urban setting, a significant proportion of OHCA patients with rSR can survive to a good neurologic outcome, despite very long time to ROSC.


Subject(s)
Cough/etiology , Out-of-Hospital Cardiac Arrest/therapy , Registries , Cardiopulmonary Resuscitation , Emergency Medical Services , Female , Follow-Up Studies , Germany/epidemiology , Hospital Mortality/trends , Humans , Male , Middle Aged , Out-of-Hospital Cardiac Arrest/complications , Out-of-Hospital Cardiac Arrest/mortality , Prognosis , Retrospective Studies , Time Factors
12.
Lancet Infect Dis ; 19(10): e352-e359, 2019 10.
Article in English | MEDLINE | ID: mdl-31182290

ABSTRACT

In high-income countries, the presentation of tuberculosis is changing, primarily because of migration, and understanding the specific health needs of susceptible populations is becoming increasingly important. Although disseminated tuberculosis is well documented in HIV-positive patients, the disease is poorly described and less expected in HIV-negative individuals. In this Grand Round, we report eight HIV-negative refugees, who presented with extensively disseminated tuberculosis. We discuss the multifactorial causes, such as deprivations during long journeys, precarious living conditions, and the experience of violence, which might add to nutritional factors and chronic disorders, eventually resulting in a state of predisposition to immune deficiency. We also show that disseminated tuberculosis is often difficult to diagnose when pulmonary symptoms are absent. Communication difficulties between refugees and health-care workers are another major hurdle, and every effort should be made to get a valid patient history. This medical history is crucial to guide imaging and other diagnostic procedures to establish a definite diagnosis, which should be confirmed by a positive tuberculosis culture. Because many of these patients are at risk for multidrug-resistant tuberculosis, drug susceptibility testing is imperative to guide therapy. In the absence of treatment guidelines for this entity, clinicians can determine treatment duration according to recommendations provided for extrapulmonary tuberculosis and affected organs. Paradoxical expansion of tuberculous lesions during therapy should be treated with corticosteroids. In many cases, treatment duration must be individualised and might even exceed 12 months.


Subject(s)
Antibiotics, Antitubercular/therapeutic use , HIV Infections , Mycobacterium tuberculosis/genetics , Refugees , Tuberculosis, Pulmonary/diagnostic imaging , Tuberculosis, Pulmonary/drug therapy , Adolescent , Adult , Antibiotics, Antitubercular/administration & dosage , Ethambutol/administration & dosage , Ethambutol/therapeutic use , Fluorine Radioisotopes , Humans , Isoniazid/administration & dosage , Isoniazid/therapeutic use , Male , Microbial Sensitivity Tests , Positron Emission Tomography Computed Tomography/methods , Pyrazinamide/administration & dosage , Pyrazinamide/therapeutic use , Rifampin/administration & dosage , Rifampin/therapeutic use , Treatment Outcome , Tuberculosis, Pulmonary/microbiology , Young Adult
13.
J Histochem Cytochem ; 67(4): 267-274, 2019 04.
Article in English | MEDLINE | ID: mdl-30452870

ABSTRACT

Arterial diseases including abdominal aortic aneurysm and atherosclerosis are biomechanical diseases characterized by significant changes in the structure and strength of the vessel wall. It is now established that local variations in fibrillar collagen and elastin matrix turnover is critical to arterial stiffening and progression of the disease. The collagen content in the aortic wall has nominally been quantified by biochemical assays and immunohistochemical analysis as the total amount because of the difficulty in separating the media and adventitia. In this work, we have developed an algorithm for automatic quantification of layer-specific collagen content from bright-field and polarized microscopic images of histological sections of mouse aorta stained with Picrosirius red (PSR) stain. The images were processed sequentially including separation of layers, erosion, segregation of regions, binarization, and quantification of pixel intensities to obtain collagen content in the media and adventitia separately. We observed that the automated algorithm rapidly and accurately quantified collagen content from a wide range of image quality compared with manual measurements particularly when the medial and adventitial layers overlap. Together, our algorithm will be of significant impact in the rapid, reliable, and accurate analyses of collagen distribution in histological sections of connective tissues.


Subject(s)
Aorta/ultrastructure , Collagen/analysis , Animals , Aorta/diagnostic imaging , Azo Compounds/analysis , Coloring Agents/analysis , Image Processing, Computer-Assisted/methods , Mice, Inbred C57BL , Microscopy, Polarization/methods , Optical Imaging/methods , Staining and Labeling/methods
14.
JCI Insight ; 3(11)2018 06 07.
Article in English | MEDLINE | ID: mdl-29875311

ABSTRACT

Pulmonary arterial hypertension (PAH) remains a disease with limited therapeutic options and dismal prognosis. Despite its etiologic heterogeneity, the underlying unifying pathophysiology is characterized by increased vascular tone and adverse remodeling of the pulmonary circulation. Myeloperoxidase (MPO), an enzyme abundantly expressed in neutrophils, has potent vasoconstrictive and profibrotic properties, thus qualifying as a potential contributor to this disease. Here, we sought to investigate whether MPO is causally linked to the pathophysiology of PAH. Investigation of 2 independent clinical cohorts revealed that MPO plasma levels were elevated in subjects with PAH and predicted adverse outcome. Experimental analyses showed that, upon hypoxia, right ventricular pressure was less increased in Mpo-/- than in WT mice. The hypoxia-induced activation of the Rho-kinase pathway, a critical subcellular signaling pathway yielding vasoconstriction and structural vascular remodeling, was blunted in Mpo-/- mice. Mice subjected to i.v. infusion of MPO revealed activation of Rho-kinase and increased right ventricular pressure, which was prevented by coinfusion of the Rho-kinase inhibitor Y-27632. In the Sugen5416/hypoxia rat model, PAH was attenuated by the MPO inhibitor AZM198. The current data demonstrate a tight mechanistic link between MPO, the activation of Rho-kinase, and adverse pulmonary vascular function, thus pointing toward a potentially novel avenue of treatment.


Subject(s)
Hypertension, Pulmonary/pathology , Hypoxia/pathology , Peroxidase/metabolism , Pulmonary Artery/pathology , rho-Associated Kinases/metabolism , Adult , Amides/administration & dosage , Animals , Cohort Studies , Disease Models, Animal , Female , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/mortality , Hypertension, Pulmonary/physiopathology , Hypoxia/blood , Hypoxia/etiology , Infusions, Intravenous , Kaplan-Meier Estimate , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Peroxidase/administration & dosage , Peroxidase/blood , Pulmonary Artery/physiopathology , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Vascular Remodeling/drug effects , Vascular Remodeling/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology , rho-Associated Kinases/antagonists & inhibitors
15.
Arterioscler Thromb Vasc Biol ; 38(8): 1796-1805, 2018 08.
Article in English | MEDLINE | ID: mdl-29880489

ABSTRACT

Objective- Recruitment of immunologic competent cells to the vessel wall is a crucial step in formation of abdominal aortic aneurysms (AAA). Innate immunity effectors (eg, macrophages), as well as mediators of adaptive immunity (eg, T cells), orchestrate a local vascular inflammatory response. IL-10 (interleukin-10) is an immune-regulatory cytokine with a crucial role in suppression of inflammatory processes. We hypothesized that an increase in systemic IL-10-levels would mitigate AAA progression. Approach and Results- Using a single intravenous injection protocol, we transfected an IL-10 transcribing nonimmunogenic minicircle vector into the Ang II (angiotensin II)-ApoE-/- infusion mouse model of AAA. IL-10 minicircle transfection significantly reduced average aortic diameter measured via ultrasound at day 28 from 166.1±10.8% (control) to 131.0±5.8% (IL-10 transfected). Rates of dissecting AAA were reduced by IL-10 treatment, with an increase in freedom from dissecting AAA from 21.5% to 62.3%. Using flow cytometry of aortic tissue from minicircle IL-10-treated animals, we found a significantly higher percentage of CD4+/CD25+/Foxp3 (forkhead box P3)+ regulatory T cells, with fewer CD8+/GZMB+ (granzyme B) cytotoxic T cells. Furthermore, isolated aortic macrophages produced less TNF-α (tumor necrosis factor-α), more IL-10, and were more likely to be MRC1 (mannose receptor, C type 1)-positive alternatively activated macrophages. These results concurred with gene expression analysis of lipopolysaccharide-stimulated and Ang II-primed human peripheral blood mononuclear cells. Conclusions- Taken together, we provide an effective gene therapy approach to AAA in mice by enhancing antiinflammatory and dampening proinflammatory pathways through minicircle-induced augmentation of systemic IL-10 expression.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Aortic Dissection/prevention & control , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Interleukin-10/biosynthesis , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Aortic Dissection/metabolism , Angiotensin II , Animals , Aorta, Abdominal/immunology , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Cells, Cultured , Dilatation, Pathologic , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Up-Regulation
16.
Nitric Oxide ; 2018 Mar 26.
Article in English | MEDLINE | ID: mdl-29588164

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death and accounts for one third of disease-related mortality worldwide. Dysregulated redox mechanisms, in particular the formation of reactive oxygen species (ROS) play a pivotal pathogenetic role in CVD. Nitro-fatty acids (NO2-FAs) are electrophilic molecules which have a NO2-group bound to one of their olefinic carbons. They are endogenously formed by the reaction of reactive nitrogen species with unsaturated fatty acids. Basal levels of NO2-FAs are in the low nanomolar range and higher concentrations can be encountered under acidic (stomach) and inflammatory (e.g. ischemia/reperfusion) conditions. Dietary intake of polyunsaturated fatty acids in combination with nitrites raises circulating NO2-FAs to a clinically relevant level in mice. NO2-FAs undergo reversible covalent binding to cysteine residues and by virtue of these posttranslational protein modifications act as potent anti-inflammatory signaling mediators via modulation of various critical pathways like nuclear factor E2-related factor 2 (Nrf2)- and peroxisome proliferator-activated receptor γ (PPARγ) activation, nuclear factor-kappa B (NF-κB) inhibition and hem oxygenase-1 (HO-1)- and heat shock protein (HSP) induction. In this review article, we summarize recent findings about the effects and underlying molecular mechanisms of NO2-FAs from a variety of pre-clinical cardiovascular disease models. The described findings suggest the potential of NO2-FAs to emerge as therapeutic agents with a broad range of potential clinical applications for CVD.

17.
Circ Res ; 121(1): 56-70, 2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28404615

ABSTRACT

RATIONALE: Ventricular arrhythmias remain the leading cause of death in patients suffering myocardial ischemia. Myeloperoxidase, a heme enzyme released by polymorphonuclear neutrophils, accumulates within ischemic myocardium and has been linked to adverse left ventricular remodeling. OBJECTIVE: To reveal the role of myeloperoxidase for the development of ventricular arrhythmias. METHODS AND RESULTS: In different murine models of myocardial ischemia, myeloperoxidase deficiency profoundly decreased vulnerability for ventricular tachycardia on programmed right ventricular and burst stimulation and spontaneously as assessed by ECG telemetry after isoproterenol injection. Experiments using CD11b/CD18 integrin-deficient (CD11b-/-) mice and intravenous myeloperoxidase infusion revealed that neutrophil infiltration is a prerequisite for myocardial myeloperoxidase accumulation. Ventricles from myeloperoxidase-deficient (Mpo-/-) mice showed less pronounced slowing and decreased heterogeneity of electric conduction in the peri-infarct zone than wild-type mice. Expression of the redox-sensitive gap junctional protein Cx43 (Connexin 43) was reduced in the peri-infarct area of wild-type compared with Mpo-/- mice. In isolated wild-type cardiomyocytes, Cx43 protein content decreased on myeloperoxidase/H2O2 incubation. Mapping of induced pluripotent stem cell-derived cardiomyocyte networks and in vivo investigations linked Cx43 breakdown to myeloperoxidase-dependent activation of matrix metalloproteinase 7. Moreover, Mpo-/- mice showed decreased ventricular postischemic fibrosis reflecting reduced accumulation of myofibroblasts. Ex vivo, myeloperoxidase was demonstrated to induce fibroblast-to-myofibroblast transdifferentiation by activation of p38 mitogen-activated protein kinases resulting in upregulated collagen generation. In support of our experimental findings, baseline myeloperoxidase plasma levels were independently associated with a history of ventricular arrhythmias, sudden cardiac death, or implantable cardioverter-defibrillator implantation in a cohort of 2622 stable patients with an ejection fraction >35% undergoing elective diagnostic cardiac evaluation. CONCLUSIONS: Myeloperoxidase emerges as a crucial mediator of postischemic myocardial remodeling and may evolve as a novel pharmacological target for secondary disease prevention after myocardial ischemia.


Subject(s)
Arrhythmias, Cardiac/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Peroxidase/deficiency , Ventricular Remodeling/physiology , Animals , Arrhythmias, Cardiac/pathology , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Organ Culture Techniques
19.
Biochem Biophys Res Commun ; 456(1): 294-7, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25450620

ABSTRACT

RATIONALE: Islet-1 positive (Islet-1(+)) cardiac progenitor cells give rise to the right ventricle, atria and outflow tract during murine cardiac development. In the adult heart Islet-1 expression is limited to parasympathetic neurons, few cardiomyocytes, smooth muscle cells, within the proximal aorta and pulmonary artery and sinoatrial node cells. Its role in these cells is unknown. Here we tested the hypothesis that Islet-1(+) cells retain proliferative activity and may therefore play a role in regenerating specialized regions in the heart. METHODS AND RESULTS: DNA synthesis was analyzed by the incorporation of tritiated thymidine ((3)H-thymidine) in Isl-1-nLacZ mice, a transgenic model with an insertion of a nuclear beta-galactosidase in the Islet-1 locus. Mice received daily injections of (3)H-thymidine for 5 days. DNA synthesis was visualized throughout the heart by dipping autoradiography of cryosections. Colocalization of an nLacZ-signal and silver grains would indicate DNA synthesis in Islet-1(+) cells. Whereas Islet(-) non-myocyte nuclei were regularly marked by accumulation of silver grains, colocalization with nLacZ-signals was not detected in >25,000 cells analyzed. CONCLUSIONS: Islet-1(+) cells are quiescent in the adult heart, suggesting that, under normal conditions, even pacemaking cells do not proliferate at higher rates than normal cardiac myocytes.


Subject(s)
DNA Replication , Heart/physiology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Sinoatrial Node/cytology , Stem Cells/cytology , Animals , Autoradiography , Cell Nucleus/metabolism , Cell Proliferation , Heart Ventricles/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thymidine/chemistry , Transgenes , beta-Galactosidase/metabolism
20.
Circ Res ; 110(10): 1303-10, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22427341

ABSTRACT

RATIONALE: The transcription factor Islet-1 is a marker of cardiovascular progenitors during embryogenesis. The isolation of Islet-1-positive (Islet-1(+)) cells from early postnatal hearts suggested that Islet-1 also marks cardiac progenitors in adult life. OBJECTIVE: We investigated the distribution and identity of Islet-1(+) cells in adult murine heart and evaluated whether their number or distribution change with age or after myocardial infarction. METHODS AND RESULTS: Distribution of Islet-1(+) cells in adult heart was investigated using gene targeted mice with nuclear ß-galactosidase inserted into the Islet-1 locus. nLacZ-positive cells were only present in 3 regions of the adult heart: clusters in the interatrial septum and around the pulmonary veins, scattered within the wall of the great vessels, and a strictly delimited cluster between the right atrium and superior vena cava. Islet-1(+) cells in the first type of clusters coexpressed markers for parasympathetic neurons. Positive cells in the great arteries coexpressed smooth muscle actin and myosin heavy chain, indicating a smooth muscle cell identity. Very few Islet-1(+) cells within the outflow tract expressed the cardiomyocyte marker α-actinin. Islet-1(+) cells in the right atrium coexpressed the sinoatrial node pacemaker cell marker HCN4. Cell number and localization remained unchanged between 1 to 18 months of age. Consistently Islet-1 mRNA was detected in human sinoatrial node. Islet-1(+) cells could not be detected in the infarct zone 2 to 28 days after myocardial infarction, aside from 10 questionable cells in 1/13 hearts. CONCLUSIONS: Our results identify Islet-1 as a novel marker of the adult sinoatrial node and do not provide evidence for Islet-1(+) cells to serve as cardiac progenitors.


Subject(s)
LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Sinoatrial Node/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Age Factors , Animals , Aorta/cytology , Aorta/metabolism , Biomarkers/metabolism , Chromogenic Compounds , Galactosides , Indoles , Lac Operon , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/pathology , Myocardium/cytology , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , RNA, Messenger/metabolism , Sinoatrial Node/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...