Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 838
Filter
Add more filters

Publication year range
1.
BMC Vet Res ; 17(1): 274, 2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34391430

ABSTRACT

BACKGROUND: Restrictive cardiomyopathy (RCM) is a common myocardial disease in cats, characterized by diastolic dysfunction and atrial enlargement without myocardial hypertrophy. Especially, endomyocardial form of RCM, one of the subtypes in RCM, is characterized by endocardial fibrosis, endocardial scar bridging the interventricular septum and left ventricular (LV) free wall, and deformation and distortion of the LV. However, it is unclear how the myocardial dysfunction and the endocardial scar contribute to the pathophysiology of RCM disease progression. CASE PRESENTATION: A 3 years and 2 months old, intact male, Domestic shorthaired cat was presented for consultation of cardiac murmur. At the first visit (day 0), the notable abnormal finding was echocardiography-derived chordae tendineae-like structure bridging the interventricular septum and the LV free wall, resulting high-speed blood flow in the left ventricle. Electrocardiography, thoracic radiography and noninvasive blood pressure measurements were normal. No left atrial enlargement was observed, and LV inflow velocity showed an abnormal relaxation pattern. Although there was no abnormality in tissue Doppler imaging-derived myocardial velocity, two-dimensional speckle tracking echocardiography (2D-STE) revealed a decrease in the LV longitudinal strain and an increase in endocardial to epicardial ratio of the LV circumferential strain on day 0. On day 468, obvious left atrium enlargement and smoke like echo in the left atrium were observed. The LV inflow velocity was fused, and the tissue Doppler imaging-derived early-diastolic myocardial velocity of the septal mitral annulus decreased. Regarding 2D-STE, LV circumferential strain was further decreased, and right ventricular strain was additionally decreased. Although the general condition was good, we made a clinical diagnosis of endomyocardial RCM based on the above findings. On day 503, the cat showed the radiographic evidence of pulmonary edema and congestive heart failure signs. CONCLUSIONS: Cats with abnormal LV structure and associated myocardial dysfunction like this case needs careful observation. Additionally, 2D-STE indices may be useful for early detection of myocardial dysfunction in feline RCM.


Subject(s)
Cardiomyopathies/veterinary , Cat Diseases/diagnosis , Endomyocardial Fibrosis/veterinary , Animals , Cardiomyopathies/complications , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Cardiovascular Agents/therapeutic use , Cat Diseases/drug therapy , Cat Diseases/etiology , Cat Diseases/pathology , Cats , Endomyocardial Fibrosis/complications , Endomyocardial Fibrosis/diagnosis , Endomyocardial Fibrosis/pathology , Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/veterinary , Male
2.
Bull Exp Biol Med ; 171(3): 305-311, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34302205

ABSTRACT

We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-ß pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.


Subject(s)
Angiotensin II/pharmacology , Antifibrotic Agents/pharmacology , Bone Morphogenetic Protein 4/genetics , Fibroblasts/drug effects , MAP Kinase Kinase 4/genetics , Vinca Alkaloids/pharmacology , Animals , Animals, Newborn , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/metabolism , Collagen/genetics , Collagen/metabolism , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/metabolism , Models, Biological , Myocardium/metabolism , Myocardium/pathology , Phosphorylation/drug effects , Primary Cell Culture , Rats , Rats, Wistar , Signal Transduction , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/genetics , Smad8 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Am J Physiol Cell Physiol ; 319(5): C781-C796, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32845719

ABSTRACT

Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.


Subject(s)
Cardiomyopathy, Dilated/genetics , Endomyocardial Fibrosis/genetics , Mucocutaneous Lymph Node Syndrome/genetics , Myocardial Infarction/genetics , Myocarditis/genetics , Tenascin/genetics , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Gene Expression Regulation , Humans , Inflammation , Mice , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Tenascin/metabolism , Ventricular Remodeling/genetics , Wound Healing/genetics
4.
Radiographics ; 40(2): 336-353, 2020.
Article in English | MEDLINE | ID: mdl-32004118

ABSTRACT

Endomyocardial fibrosis (EMF) affects approximately 12 million persons worldwide and is an important cause of restrictive cardiomyopathy in the developing world, with the highest prevalence reported in sub-Saharan Africa, South Asia, and South America. EMF is characterized by apical filling with fibrotic tissue of one or both ventricles, often associated with thrombus, calcification, and atrioventricular valve regurgitation, leading to typical symptoms of restrictive heart failure. Transthoracic echocardiography (TTE) is the first-line modality for assessment of EMF, basically owing to its widespread availability. However, in recent years cardiac MRI has emerged as a powerful tool for assessment of cardiac morphology and function, with higher accuracy than TTE, along with the unique advantage of being able to provide comprehensive noninvasive tissue characterization. Delayed enhancement (DE) imaging is the cornerstone of cardiac MRI tissue characterization and allows accurate identification of myocardial fibrosis, conveying valuable additional diagnostic and prognostic information. The typical DE pattern in EMF, described as the "double V" sign, consists of a three-layered pattern of normal myocardium, thickened enhanced endomyocardium, and overlying thrombus at the apex of the affected ventricle; it has excellent correlation with histopathologic findings and plays an important role in differentiating EMF from other cardiomyopathies. Conversely, fibrous tissue deposition quantified using DE imaging, when indexed to body surface area, has been shown to be a strong independent predictor of mortality. The aim of this review is to summarize state-of-the-art applications of cardiac MRI for diagnostic and prognostic assessment of patients with suspected or confirmed EMF. Online supplemental material is available for this article. ©RSNA, 2020.


Subject(s)
Endomyocardial Fibrosis/diagnostic imaging , Endomyocardial Fibrosis/pathology , Magnetic Resonance Imaging/methods , Contrast Media , Humans , Prognosis
5.
J Card Surg ; 35(2): 460-463, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31778572

ABSTRACT

AIMS: The case we report, shows a successful treatment of right ventricle endomyocardial fibrosis. MATERIALS AND METHODS: Surgical therapy by endocardial decortication seems to be beneficial for many patients with advanced disease who are in functional-therapeutic class III or IV. The operative mortality rate is high, but successful surgery has a clear benefit on symptoms and seems to favourably affect survival as well.


Subject(s)
Cardiac Surgical Procedures/methods , Endomyocardial Fibrosis/surgery , Heart Ventricles/surgery , Adult , Echocardiography , Endocardium/pathology , Endomyocardial Fibrosis/diagnostic imaging , Endomyocardial Fibrosis/pathology , Humans , Male , Treatment Outcome
6.
Drug Dev Res ; 81(1): 114-126, 2020 02.
Article in English | MEDLINE | ID: mdl-31605544

ABSTRACT

Aliskiren is an oral antihypertensive medication that acts by directly inhibiting renin. High levels of circulating renin and prorenin activate the pathological signaling pathway of fibrosis. This drug also reduces oxidative stress. Thus, the aim of this systematic review is to analyze experimental studies that show the actions of aliskiren on fibrosis. PubMed and LILACS databases were consulted using the keywords aliskiren and fibrosis within the period between 2005 and 2017. Fifty-three articles were analyzed. In the heart, aliskiren attenuated remodeling, hypertrophy, inflammatory cytokines, collagen deposition, and oxidative stress. In the kidneys, there was a reduction in interstitial fibrosis, the infiltration of inflammatory cells, apoptosis, proteinuria, and in the recruitment of macrophages. In diabetic models, an improvement in the albumin/creatinine relationship and in the insulin pathway in skeletal muscles was observed; aliskiren was beneficial to pancreatic function and glucose tolerance. In the liver, aliskiren reduced fibrosis, steatosis, inflammatory cytokines, and collagen deposition. In the lung and peritoneal tissues, there was a reduction in fibrosis. Many studies have reported on the beneficial effects of aliskiren on endothelial function and arterial rigidity. A reduction in fibrosis in different organs is cited by many authors, which complies with the results found in this review. However, studies diverge on the use of the drug in diabetic patients. Aliskiren has antifibrotic potential in several experimental models, interfering with the levels of fibrogenic cytokines and oxidative stress. Therefore, its use in diseases in which fibrosis plays an important pathophysiological role is suggested.


Subject(s)
Amides/administration & dosage , Endomyocardial Fibrosis/drug therapy , Fumarates/administration & dosage , Nephritis, Interstitial/drug therapy , Amides/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Drug Repositioning , Endomyocardial Fibrosis/immunology , Endomyocardial Fibrosis/pathology , Fibrosis , Fumarates/pharmacology , Humans , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Oxidative Stress/drug effects
7.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32998408

ABSTRACT

Despite relevant advances made in therapies for cardiovascular diseases (CVDs), they still represent the first cause of death worldwide. Cardiac fibrosis and excessive extracellular matrix (ECM) remodeling are common end-organ features in diseased hearts, leading to tissue stiffness, impaired myocardial functional, and progression to heart failure. Although fibrosis has been largely recognized to accompany and complicate various CVDs, events and mechanisms driving and governing fibrosis are still not entirely elucidated, and clinical interventions targeting cardiac fibrosis are not yet available. Immune cell types, both from innate and adaptive immunity, are involved not just in the classical response to pathogens, but they take an active part in "sterile" inflammation, in response to ischemia and other forms of injury. In this context, different cell types infiltrate the injured heart and release distinct pro-inflammatory cytokines that initiate the fibrotic response by triggering myofibroblast activation. The complex interplay between immune cells, fibroblasts, and other non-immune/host-derived cells is now considered as the major driving force of cardiac fibrosis. Here, we review and discuss the contribution of inflammatory cells of innate immunity, including neutrophils, macrophages, natural killer cells, eosinophils and mast cells, in modulating the myocardial microenvironment, by orchestrating the fibrogenic process in response to tissue injury. A better understanding of the time frame, sequences of events during immune cells infiltration, and their action in the injured inflammatory heart environment, may provide a rationale to design new and more efficacious therapeutic interventions to reduce cardiac fibrosis.


Subject(s)
Cell Communication/immunology , Endomyocardial Fibrosis/immunology , Immunity, Innate , Myocardial Reperfusion Injury/immunology , Myocardium/immunology , Myofibroblasts/immunology , Adaptive Immunity , Animals , Cytokines/immunology , Cytokines/metabolism , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Extracellular Matrix/chemistry , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Humans , Inflammation , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology
8.
Bull Exp Biol Med ; 168(5): 658-661, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32246376

ABSTRACT

Reduced expression of the key regulator of cardiac metabolism, transcription factor PPARα, in surgical samples of the auricles from patients with coronary heart disease and heart failure was detected by real-time quantitative PCR. These changes indicate reduced activity of this factor and a shift of energy metabolism from oxidative phosphorylation to glycolysis typical of dedifferentiated cells. Electron microscopy revealed dedifferentiated cardiomyocytes with disassembled contractile apparatus and disorganized sarcomeres. In the examined specimens from patients with heart failure, severe myocardial fibrosis was revealed.


Subject(s)
Energy Metabolism/physiology , Heart/physiology , Myocytes, Cardiac/metabolism , PPAR alpha/physiology , Regeneration/physiology , Biopsy , Cell Dedifferentiation/genetics , Coronary Disease/genetics , Coronary Disease/metabolism , Coronary Disease/pathology , Coronary Disease/physiopathology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/physiopathology , Energy Metabolism/genetics , Gene Expression Regulation , Glycolysis/genetics , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Oxidative Phosphorylation , PPAR alpha/genetics , PPAR alpha/metabolism
9.
Bull Exp Biol Med ; 169(4): 549-557, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910392

ABSTRACT

Regenerative potential of multipotent mesenchymal stromal cells from the human umbilical cord (MMSC-UC) in the suspension and spheroid form was revealed during the progression of experimental small focal myocardial infarction in rats. In isoproterenol-induced myocardial infarction, foci of necrosis and inflammatory infiltrate and at later terms fibrosis foci were found mainly in the left ventricle of rat heart. In rats receiving MMSC-UC, destructive changes in the myocardium, fibrous scars, and inflammatory process were less pronounced. MMSC-UC also contributed to normalization of the morphofunctional parameters of the heart. Spheroids exhibited higher efficiency in comparison with cell suspension.


Subject(s)
Endomyocardial Fibrosis/prevention & control , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Myocardial Infarction/therapy , Regeneration/physiology , Spheroids, Cellular/transplantation , Animals , Disease Models, Animal , Endomyocardial Fibrosis/chemically induced , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/physiopathology , Heart Ventricles/pathology , Heart Ventricles/ultrastructure , Humans , Isoproterenol/administration & dosage , Male , Mesenchymal Stem Cells/cytology , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Primary Cell Culture , Rats , Rats, Wistar , Spheroids, Cellular/cytology , Spheroids, Cellular/physiology , Transplantation, Heterologous , Treatment Outcome , Umbilical Cord/cytology , Umbilical Cord/metabolism
10.
J Cell Mol Med ; 23(9): 5859-5867, 2019 09.
Article in English | MEDLINE | ID: mdl-31240820

ABSTRACT

In recent years, the incidence of diabetes has been increasing rapidly, which seriously endangers human health. Diabetic cardiomyopathy, an important cardiovascular complication of diabetes, is characterized by myocardial fibrosis, ventricular remodelling and cardiac dysfunction. It has been documented that mitochondrial dysfunction, oxidative stress, inflammatory response, autophagy, apoptosis, diabetic microangiopathy and myocardial fibrosis are implicated in the pathogenesis of diabetic cardiomyopathy. With the development of molecular biology technology, accumulating evidence demonstrates that non-coding RNAs (ncRNAs) are critically involved in the molecular mechanisms of diabetic cardiomyopathy. In this review, we summarize the pathological roles of three types of ncRNAs (microRNA, long ncRNA and circular RNA) in the progression of diabetic cardiomyopathy, which may provide valuable insights into the pathogenesis of diabetic cardiovascular complications.


Subject(s)
Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , MicroRNAs/genetics , RNA, Circular/genetics , RNA, Long Noncoding/genetics , Autophagy/physiology , Cardiomegaly/pathology , Diabetes Complications/pathology , Diabetes Mellitus/pathology , Endomyocardial Fibrosis/pathology , Humans , Myocytes, Cardiac/metabolism , Oxidative Stress/physiology , Pyroptosis/physiology
11.
Blood ; 130(2): 205-213, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28507082

ABSTRACT

Sickle cell anemia (SCA)-related cardiomyopathy is characterized by diastolic dysfunction and hyperdynamic features. Diastolic dysfunction portends early mortality in SCA. Diastolic dysfunction is associated with microscopic myocardial fibrosis in SCA mice, but the cause of diastolic dysfunction in humans with SCA is unknown. We used cardiac magnetic resonance measurements of extracellular volume fraction (ECV) to discover and quantify diffuse myocardial fibrosis in 25 individuals with SCA (mean age, 23 ± 13 years) and determine the association between diffuse myocardial fibrosis and diastolic dysfunction. ECV was calculated from pre- and post-gadolinium T1 measurements of blood and myocardium, and diastolic function was assessed by echocardiography. ECV was markedly increased in all participants compared with controls (0.44 ± 0.08 vs 0.26 ± 0.02, P < .0001), indicating the presence of diffuse myocardial fibrosis. Seventeen patients (71%) had diastolic abnormalities, and 7 patients (29%) met the definition of diastolic dysfunction. Participants with diastolic dysfunction had higher ECV (0.49 ± 0.07 vs 0.37 ± 0.04, P = .01) and N-terminal pro-brain natriuretic peptide (NT-proBNP; 191 ± 261 vs 33 ± 33 pg/mL, P = .04) but lower hemoglobin (8.4 ± 0.3 vs 10.9 ± 1.4 g/dL, P = .004) compared with participants with normal diastolic function. Participants with the highest ECV values (≥0.40) were more likely to have diastolic dysfunction (P = .003) and increased left atrial volume (57 ± 11 vs 46 ± 12 mL/m2, P = .04) compared with those with ECV <0.4. ECV correlated with hemoglobin (r = -0.46, P = .03) and NT-proBNP (r = 0.62, P = .001). In conclusion, diffuse myocardial fibrosis, determined by ECV, is a common and previously underappreciated feature of SCA that is associated with diastolic dysfunction, anemia, and high NT-proBNP. Diffuse myocardial fibrosis is a novel mechanism that appears to underlie diastolic dysfunction in SCA.


Subject(s)
Anemia, Sickle Cell/physiopathology , Cardiomyopathies/physiopathology , Diastole , Endomyocardial Fibrosis/physiopathology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Adolescent , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/diagnostic imaging , Anemia, Sickle Cell/pathology , Biomarkers/blood , Cardiomyopathies/blood , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/pathology , Child , Echocardiography , Endomyocardial Fibrosis/blood , Endomyocardial Fibrosis/diagnostic imaging , Endomyocardial Fibrosis/pathology , Female , Gene Expression , Hemoglobins/metabolism , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Natriuretic Peptide, Brain/genetics , Peptide Fragments/genetics
12.
BMC Cardiovasc Disord ; 19(1): 302, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31881943

ABSTRACT

BACKGROUND: Endomyocardial fibrosis (EMF) represents the most common cause of restrictive cardiomyopathy worldwide. Despite a high prevalence in tropical regions, it occasionally occurs in patients who have never visited these areas. While researches have proposed various possible triggers for EMF, etiology and pathogenesis remain largely unknown. Diagnosis is based on patient history, heart failure symptoms, and echocardiographic signs of restrictive ventricular filling, atrioventricular valve regurgitation and frequently apical thrombus. Following is a case report of an Austrian patient with EMF who eventually had to undergo a heart transplant. This case report strives to promote awareness for this in non-tropical areas uncommon but nevertheless detrimental disease. CASE PRESENTATION: A 40-year-old woman was presented at our emergency department with chest pain and fever up to 38.1° Celsius. Plasma troponin-T levels and inflammatory markers were slightly elevated, but the echocardiogram was without pathological findings. The patient was hospitalized on the suspicion of acute myocarditis and discharged soon after improvement. Eight months later, she was presented again with chest pain and symptoms of heart failure. The echocardiogram showed normal systolic left ventricular (LV) function with LV wall thickening and severe restrictive mitral regurgitation as well as aortic and tricuspid regurgitation. Coronary angiogram was normal but right heart catheterization showed pulmonary hypertension due to left heart disease. Further diagnostic workup with cardiac magnetic resonance imaging revealed subendocardial late enhancement and apical thrombus formation in the left ventricle compatible with the diagnosis of EMF. A comprehensive diagnostic workup showed no evidence of infection, systemic immunologic or hematological disease, in particular hypereosinophilic syndrome. After a multidisciplinary consideration of several therapeutic options, the patient was listed for heart transplantation. On the waiting list, she deteriorated rapidly due to progressive heart failure and finally underwent a heart transplantation. Histological examination confirmed the diagnosis of EMF. Six years after her heart transplantation, the patient was presented in an excellent clinical condition. CONCLUSIONS: Even in non-tropical regions, the diagnosis of EMF should always be considered in restrictive cardiomyopathy. Knowledge of the distinct phenotype of EMF facilitates diagnosis, but comprehensive workup and therapeutic management remain challenging and require a multidisciplinary approach.


Subject(s)
Endomyocardial Fibrosis/surgery , Heart Failure/surgery , Heart Transplantation , Myocardium/pathology , Adult , Austria , Disease Progression , Endomyocardial Fibrosis/diagnostic imaging , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/physiopathology , Female , Heart Failure/diagnostic imaging , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Recovery of Function , Time Factors , Treatment Outcome , Waiting Lists
13.
J Biol Chem ; 292(18): 7542-7553, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28320863

ABSTRACT

The aim of the study was to characterize the role of Rac1 GTPase for the mineralocorticoid receptor (MR)-mediated pro-fibrotic remodeling. Transgenic mice with cardiac overexpression of constitutively active Rac1 (RacET) develop an age-dependent phenotype with atrial dilatation, fibrosis, and atrial fibrillation. Expression of MR was similar in RacET and WT mice. The expression of 11ß hydroxysteroid dehydrogenase type 2 (11ß-HSD2) was age-dependently up-regulated in the atria and the left ventricles of RacET mice on mRNA and protein levels. Statin treatment inhibiting Rac1 geranylgeranylation reduced 11ß-HSD2 up-regulation. Samples of human left atrial myocardium showed a positive correlation between Rac1 activity and 11ß-HSD2 expression (r = 0.7169). Immunoprecipitation showed enhanced Rac1-bound 11ß-HSD2 relative to Rac1 expression in RacET mice that was diminished with statin treatment. Both basal and phorbol 12-myristate 13-acetate (PMA)-induced NADPH oxidase activity were increased in RacET and correlated positively with 11ß-HSD2 expression (r = 0.788 and r = 0.843, respectively). In cultured H9c2 cardiomyocytes, Rac1 activation with l-buthionine sulfoximine increased; Rac1 inhibition with NSC23766 decreased 11ß-HSD2 mRNA and protein expression. Connective tissue growth factor (CTGF) up-regulation induced by aldosterone was prevented with NSC23766. Cardiomyocyte transfection with 11ß-HSD2 siRNA abolished the aldosterone-induced CTGF up-regulation. Aldosterone-stimulated MR nuclear translocation was blocked by the 11ß-HSD2 inhibitor carbenoxolone. In cardiac fibroblasts, nuclear MR translocation induced by aldosterone was inhibited with NSC23766 and spironolactone. NSC23766 prevented the aldosterone-induced proliferation and migration of cardiac fibroblasts and the up-regulation of CTGF and fibronectin. In conclusion, Rac1 GTPase regulates 11ß-HSD2 expression, MR activation, and MR-mediated pro-fibrotic signaling.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Endomyocardial Fibrosis/enzymology , Fibroblasts/enzymology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Neuropeptides/biosynthesis , Signal Transduction , rac1 GTP-Binding Protein/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aldosterone/pharmacology , Animals , Cell Line , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Endomyocardial Fibrosis/pathology , Fibroblasts/pathology , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation/drug effects , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Mutant Strains , Myocardium/pathology , Myocytes, Cardiac/pathology , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Sulfoxides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , rac1 GTP-Binding Protein/genetics
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 11-23, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28987763

ABSTRACT

Cardiac dysfunction with progressive inflammation and fibrosis is a hallmark of Chagas disease caused by persistent Trypanosoma cruzi infection. Osteopontin (OPN) is a pro-inflammatory cytokine that orchestrates mechanisms controlling cell recruitment and cardiac architecture. Our main goal was to study the role of endogenous OPN as a modulator of myocardial CCL5 chemokine and MMP-2 metalloproteinase, and its pathological impact in a murine model of Chagas heart disease. Wild-type (WT) and OPN-deficient (spp1 -/-) mice were parasite-infected (Brazil strain) for 100days. Both groups developed chronic myocarditis with similar parasite burden and survival rates. However, spp1 -/- infection showed lower heart-to-body ratio (P<0.01) as well as reduced inflammatory pathology (P<0.05), CCL5 expression (P<0.05), myocyte size (P<0.05) and fibrosis (P<0.01) in cardiac tissues. Intense OPN labeling was observed in inflammatory cells recruited to infected heart (P<0.05). Plasma concentration of MMP-2 was higher (P<0.05) in infected WT than in spp1 -/- mice. Coincidently, specific immunostaining revealed increased gelatinase expression (P<0.01) and activity (P<0.05) in the inflamed hearts from T. cruzi WT mice, but not in their spp1 -/- littermates. CCL5 and MMP-2 induction occurred preferentially (P<0.01) in WT heart-invading CD8+ T cells and was mediated via phospho-JNK MAPK signaling. Heart levels of OPN, CCL5 and MMP-2 correlated (P<0.01) with collagen accumulation in the infected WT group only. Endogenous OPN emerges as a key player in the pathogenesis of chronic Chagas heart disease, through the upregulation of myocardial CCL5/MMP-2 expression and activities resulting in pro-inflammatory and pro-hypertrophic events, cardiac remodeling and interstitial fibrosis.


Subject(s)
Atrial Remodeling , Chagas Cardiomyopathy , Chemokine CCL5/metabolism , Matrix Metalloproteinase 2/metabolism , Myocarditis , Osteopontin/physiology , Ventricular Remodeling , Animals , Atrial Remodeling/genetics , Atrial Remodeling/immunology , Cells, Cultured , Chagas Cardiomyopathy/genetics , Chagas Cardiomyopathy/immunology , Chagas Cardiomyopathy/metabolism , Chagas Cardiomyopathy/pathology , Disease Models, Animal , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Osteopontin/genetics , Ventricular Remodeling/genetics , Ventricular Remodeling/immunology
15.
Med Sci Monit ; 24: 4121-4127, 2018 Jun 16.
Article in English | MEDLINE | ID: mdl-29908129

ABSTRACT

BACKGROUND The role of miR-181a in the development of cardiac disease and in particular, myocardial fibrosis following myocardial infarction (MI) remains unknown. The aim of this study was to explore the role of miR-181a in myocardial fibrosis in a rat model of MI and the expression of TGF-ß receptor III (TßRIII). MATERIAL AND METHODS Forty adult male Wistar rats were randomly divided into an MI model group (n=30) and a control group with (n=10). The rat MI model involved ligating the left anterior descending (LAD) coronary artery in the model group; the control group was treated with a sham operation. Cardiac function was assessed using cardiac ultrasound. Myocardial fibroblasts were extracted from the rat hearts and transfected with a miR-mimic or miR-inhibitor, and cell growth was measured using an MTT assay. The level of miR-181a expression was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blots. RESULTS miR-181a expression was significantly increased during the progression of MI (P<0.05). Over-expression of miR-181a was associated with increased deposition of extracellular matrix (ECM) components, collagen I and fibronectin. This effect was reversed with the use of a miR-181a inhibitor (P<0.05). Upregulation of miR-181a suppressed the expression of TGF-ß receptor III (TßRIII) by binding with 3'-UTR. CONCLUSIONS In this rat model of MI, the findings were that miR-181a had a role in the progression of myocardial fibrosis. The findings require further studies to determine whether miR-181a might provide a novel therapeutic target to limit myocardial fibrosis following MI.


Subject(s)
Endomyocardial Fibrosis/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Animals , Cell Proliferation/physiology , Collagen Type I/metabolism , Disease Models, Animal , Disease Progression , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Myocardial Infarction/metabolism , Proteoglycans/biosynthesis , Proteoglycans/genetics , Rats , Rats, Wistar , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics
16.
Med Sci Monit ; 24: 6264-6272, 2018 Sep 08.
Article in English | MEDLINE | ID: mdl-30194718

ABSTRACT

BACKGROUND Myocardial fibrosis is closely related to all types of cardiovascular diseases. Hirudin is widely used in the prevention and treatment of cardiovascular diseases and cancers. In this study, we examined the potential role(s) and mechanism of hirudin in angiotensin II (Ang II)-induced myocardial fibrosis. MATERIAL AND METHODS The viability of myocardial fibroblasts, and reactive oxygen species (ROS) rates were measured respectively using cell counting kit-8 (CCK-8) and flow cytometry. Malondialdehyde (MDA) content, the activities of lactate dehydrogenase (LDH), and superoxide dismutase (SOD) were detected by the respective kits. The mRNA and protein levels of fibrosis-related factors were separately assessed by qRT-PCR and western blot. RESULTS Our data revealed that hirudin suppressed the viability of myocardial fibroblasts, and that it relieved the proliferation induced by Ang II in a dose-dependent manner. We also found that hirudin reduced ROS production, LDH activity, and MDA content; however, it enhanced SOD activity. Moreover, while hirudin significantly downregulated the levels of matrix metalloproteinase-2 (MMP-2), MMP-9, fibronectin (FN), transforming growth factor beta 1 (TGF-ß1), collagen-I (COL-I), and COL-III, it upregulated the expression level of tissue inhibitor of metalloproteinases-2 (TIMP-2). Furthermore, phosphorylated extracellular signal-regulated kinase1/2 (p-ERK1/2) was decreased by hirudin, compared to the Ang-II group. CONCLUSIONS Hirudin depressed Ang II-induced myocardial fibroblasts via inhibiting oxidative stress, regulating fibrosis-related factors, and repressing the ERK1/2 pathway.


Subject(s)
Angiotensin II/pharmacology , Endomyocardial Fibrosis/drug therapy , Hirudins/pharmacology , MAP Kinase Signaling System/drug effects , Animals , Collagen Type I/metabolism , Collagen Type III/metabolism , Cumulus Cells , Endomyocardial Fibrosis/enzymology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Fibronectins/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myocardium/cytology , Myocardium/metabolism , Myofibroblasts/drug effects , Myofibroblasts/enzymology , Myofibroblasts/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Transforming Growth Factor beta1/metabolism
17.
Biochem J ; 474(3): 399-410, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27803247

ABSTRACT

Cardiac fibrosis and chronic inflammation are common complications in type 2 diabetes mellitus (T2D). Since nucleotide oligomerization-binding domain 1 (NOD1), an innate immune receptor, is involved in the pathogenesis of insulin resistance and diabetes outcomes, we sought to investigate its involvement in cardiac fibrosis. Here, we show that selective staining of cardiac fibroblasts from T2D (db/db;db) mice exhibits up-regulation and activation of the NOD1 pathway, resulting in enhanced NF-κB and TGF-ß signalling. Activation of the TGF-ß pathway in cardiac fibroblasts from db mice was prevented after inhibition of NF-κB with BAY-11-7082 (BAY). Moreover, fibrosis progression in db mice was also prevented by BAY treatment. Enhanced TGF-ß signalling and cardiac fibrosis of db mice was dependent, at least in part, on the sequential activation of NOD1 and NF-κB since treatment of db mice with a selective NOD1 agonist induced activation of the TGF-ß pathway, but co-administration of a NOD1 agonist plus BAY, or a NOD1 inhibitor prevented the NOD1-induced fibrosis. Therefore, NOD1 is involved in cardiac fibrosis associated with diabetes, and establishes a new mechanism for the development of heart fibrosis linked to T2D.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endomyocardial Fibrosis/metabolism , Myocardium/metabolism , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Gene Expression Regulation , Humans , Insulin/blood , Insulin Resistance , Mice , Mice, Transgenic , Myocardium/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NIH 3T3 Cells , Nitriles/pharmacology , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Signal Transduction , Sulfones/pharmacology , Transforming Growth Factor beta/agonists , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
18.
Int J Mol Sci ; 19(12)2018 Nov 22.
Article in English | MEDLINE | ID: mdl-30467294

ABSTRACT

Heart failure (HF) is a cardiovascular syndrome characterized by maladaptive changes with an underlying inflammatory mediated pathogenesis. Nevertheless, current therapy is aimed at the heart workload and neurohormonal axis; thus, prognosis remains poor. To continue improving treatment, we rely on murine models for a better understanding of HF pathophysiology. Among them, pressure overload HF (PO-HF) animal models are a common strategy. Development of PO-HF is characterized by monocyte infiltration, which orchestrates a cascade of events leading to sustained inflammation and maladaptive changes. Here, we divide the PO-HF model progression into four phases and describe the inflammatory, structural, and gene expression profiles. This division is relevant due to its similarities with clinical hypertensive heart disease progression to HF. Evidence shows improvement in hemodynamic and other local parameters by altering the inflammatory response in a specific immune response at a specific point of time. Thus, it is relevant to focus on the time-dependent immune response interaction in order to provide more effective therapy. This review summarizes the pathogenesis of PO-HF murine models, highlighting the inflammatory events in a time frame view. By this approach, we expect to provide researchers with a better understanding of the intertwining time-dependent events that occur in PO-HF.


Subject(s)
B-Lymphocytes/immunology , Heart Failure/immunology , Hypertension/immunology , Monocytes/immunology , T-Lymphocytes/immunology , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , Aorta/immunology , Aorta/pathology , B-Lymphocytes/pathology , Cardiomegaly/immunology , Cardiomegaly/pathology , Cell Movement , Constriction, Pathologic/immunology , Constriction, Pathologic/pathology , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Endomyocardial Fibrosis/immunology , Endomyocardial Fibrosis/pathology , Heart Failure/chemically induced , Heart Failure/etiology , Heart Failure/pathology , Humans , Hypertension/complications , Hypertension/pathology , Mice , Monocytes/pathology , T-Lymphocytes/pathology , Time Factors , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/pathology
19.
Int Heart J ; 59(3): 523-530, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29743413

ABSTRACT

To achieve further risk stratification in hypertrophic cardiomyopathy (HCM) patients, we localized and quantified layer-specific LVM fibrosis on MRI in HCM patients using regional layer-specific peak longitudinal strain (PLS) and peak circumferential strain (PCS) in LV myocardium (LVM) on speckle tracking transthoracic echocardiography (TTE). A total of 18 HCM patients (14 males; 58 ± 17 years) underwent 1.5T-MRI and TTE. PLS and PCS in each layer of the LVM (endocardium, epicardium, and whole-layer myocardium) were calculated for 17 AHA-defined lesions. MRI assessment showed that fibrosis was classified as endocardial, epicardial, or whole-layer (= either or both of these). Regional PLS was smaller in fibrotic endocardial lesions than in non-fibrotic endocardial lesions (P = 0.004). To detect LV endocardial lesions with fibrosis, ROC curves of regional PLS revealed an area under the curve (AUC) of 0.609 and a best cut-off point of 13.5%, with sensitivity of 65.3% and specificity of 54.3%. Regional PLS was also smaller in fibrotic epicardial lesions than in non-fibrotic epicardial lesions (P < 0.001). To detect LV epicardial lesions with fibrosis, ROC curves of PLS revealed an AUC of 0.684 and a best cut-off point of 9.5%, with sensitivity of 73.5% and specificity of 55.5%. Using whole-layer myocardium analysis, PLS was smaller in fibrotic lesions than in non-fibrotic lesions (P < 0.001). To detect whole-layer LV lesions with fibrosis, ROC curves of regional PLS revealed an AUC of 0.674 and a best cut-off point of 12.5%, with sensitivity of 79.0% and specificity of 50.7%. There were no significant differences in PCS of LV myocardium (endocardium, epicardium, and whole-layer) between fibrotic and non-fibrotic lesions. Quantitative regional PLS but not PCS in LV endocardium, epicardium, and whole-layer myocardium provides useful non-invasive information for layer-specific localization of fibrosis in HCM patients.


Subject(s)
Cardiomyopathy, Hypertrophic/pathology , Endomyocardial Fibrosis/pathology , Myocardium/pathology , Adult , Aged , Area Under Curve , Cardiomyopathy, Hypertrophic/diagnostic imaging , Echocardiography/methods , Endomyocardial Fibrosis/diagnostic imaging , Female , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , ROC Curve , Risk Assessment , Sensitivity and Specificity
20.
J Proteome Res ; 16(1): 307-318, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27805817

ABSTRACT

Epidemiological data from patients undergoing radiotherapy for thoracic tumors clearly show the damaging effect of ionizing radiation on cardiovascular system. The long-term impairment of heart function and structure after local high-dose irradiation is associated with systemic inflammatory response, contraction impairment, microvascular damage, and cardiac fibrosis. The goal of the present study was to investigate molecular mechanisms involved in this process. C57BL/6J mice received a single X-ray dose of 16 Gy given locally to the heart at the age of 8 weeks. Radiation-induced changes in the heart transcriptome and proteome were investigated 40 weeks after the exposure. The omics data were analyzed by bioinformatics tools and validated by immunoblotting. Integrated network analysis of transcriptomics and proteomics data elucidated the signaling pathways that were similarly affected at gene and protein level. Analysis showed induction of transforming growth factor (TGF) beta signaling but inactivation of peroxisome proliferator-activated receptor (PPAR) alpha signaling in irradiated heart. The putative mediator role of mitogen-activated protein kinase cascade linking PPAR alpha and TGF beta signaling was supported by data from immunoblotting and ELISA. This study indicates that both signaling pathways are involved in radiation-induced heart fibrosis, metabolic disordering, and impaired contractility, a pathophysiological condition that is often observed in patients that received high radiation doses in thorax.


Subject(s)
Endomyocardial Fibrosis/genetics , Heart/radiation effects , PPAR alpha/genetics , Proteome/genetics , Transcriptome , Transforming Growth Factor beta/genetics , Animals , Computational Biology , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/pathology , Gamma Rays/adverse effects , Gene Expression Profiling , Gene Expression Regulation , Heart/physiopathology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , PPAR alpha/metabolism , Proteome/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL