Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Med Genet A ; 188(6): 1676-1687, 2022 06.
Article in English | MEDLINE | ID: mdl-35166435

ABSTRACT

The Nexilin F-Actin Binding Protein (Nexilin) encoded by NEXN is a cardiac Z-disc protein important for cardiac function and development in humans, zebrafish, and mice. Heterozygote variants in the human NEXN gene have been reported to cause dilated and hypertrophic cardiomyopathy. Homozygous variants in NEXN cause a lethal form of human fetal cardiomyopathy, only described in two patients before. In a Swedish, four-generation, non-consanguineous family comprising 42 individuals, one female had three consecutive pregnancies with intrauterine fetal deaths caused by a lethal form of dilated cardiomyopathy. Whole-exome sequencing and variant analysis revealed that the affected fetuses were homozygous for a NEXN variant (NM_144573:c.1302del;p.(Ile435Serfs*3)). Moreover, autopsy and histology staining declared that they presented with cardiomegaly and endocardial fibroelastosis. Immunohistochemistry staining for Nexilin in the affected fetuses revealed reduced antibody staining and loss of striation in the heart, supporting loss of Nexilin function. Clinical examination of seven heterozygote carriers confirmed dilated cardiomyopathy (two individuals), other cardiac findings (three individuals), or no cardiac deviations (two individuals), indicating incomplete penetrance or age-dependent expression of dilated cardiomyopathy. RNA sequencing spanning the variant in cDNA blood of heterozygote individuals revealed nonsense-mediated mRNA decay of the mutated transcripts. In the current study, we present the first natural course of the recessively inherited lethal form of human fetal cardiomyopathy caused by loss of Nexilin function. The affected family had uneventful pregnancies until week 23-24, followed by fetal death at week 24-30, characterized by cardiomegaly and endocardial fibroelastosis.


Subject(s)
Cardiomegaly , Endocardial Fibroelastosis , Microfilament Proteins , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Endocardial Fibroelastosis/genetics , Endocardial Fibroelastosis/metabolism , Endocardial Fibroelastosis/pathology , Female , Humans , Immunohistochemistry , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Exome Sequencing
2.
Pediatr Res ; 92(3): 721-728, 2022 09.
Article in English | MEDLINE | ID: mdl-34837068

ABSTRACT

BACKGROUND: Endothelial-to-mesenchymal-transition (EndMT) plays a major role in cardiac fibrosis, including endocardial fibroelastosis but the stimuli are still unknown. We developed an endothelial cell (EC) culture and a whole heart model to test whether mechanical strain triggers TGF-ß-mediated EndMT. METHODS: Isolated ECs were exposed to 10% uniaxial static stretch for 8 h (stretch) and TGF-ß-mediated EndMT was determined using the TGF-ß-inhibitor SB431542 (stretch + TGF-ß-inhibitor), BMP-7 (stretch + BMP-7) or losartan (stretch + losartan), and isolated mature and immature rats were exposed to stretch through a weight on the apex of the left ventricle. Immunohistochemical staining for double-staining with endothelial markers (VE-cadherin, PECAM1) and mesenchymal markers (αSMA) or transcription factors (SLUG/SNAIL) positive nuclei was indicative of EndMT. RESULTS: Stretch-induced EndMT in ECs expressed as double-stained ECs/total ECs (cells: 46 ± 13%; heart: 15.9 ± 2%) compared to controls (cells: 7 ± 2%; heart: 3.1 ± 0.1; p < 0.05), but only immature hearts showed endocardial EndMT. Inhibition of TGF-ß decreased the number of double-stained cells significantly, comparable to controls (cells/heart: control: 7 ± 2%/3.1 ± 0.1%, stretch: 46 ± 13%/15 ± 2%, stretch + BMP-7: 7 ± 2%/2.9 ± 0.1%, stretch + TGF-ß-inhibitor (heart only): 5.2 ± 1.3%, stretch + losartan (heart only): 0.89 ± 0.1%; p < 0.001 versus stretch). CONCLUSIONS: Endocardial EndMT is an age-dependent consequence of increased strain triggered by TGF- ß activation. Local inhibition through either rebalancing TGF-ß/BMP or with losartan was effective to block EndMT. IMPACT: Mechanical strain imposed on the immature LV induces endocardial fibroelastosis (EFE) formation through TGF-ß-mediated activation of endothelial-to-mesenchymal transition (EndMT) in endocardial endothelial cells but has no effect in mature hearts. Local inhibition through either rebalancing the TGF-ß/BMP pathway or with losartan blocks EndMT. Inhibition of endocardial EndMT with clinically applicable treatments may lead to a better outcome for congenital heart defects associated with EFE.


Subject(s)
Endocardial Fibroelastosis , Endocardium , Animals , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology , Endocardial Fibroelastosis/metabolism , Endocardium/metabolism , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Losartan/pharmacology , Rats , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
3.
J Mol Med (Berl) ; 99(11): 1623-1638, 2021 11.
Article in English | MEDLINE | ID: mdl-34387706

ABSTRACT

Among neonatal cardiomyopathies, primary endocardial fibroelastosis (pEFE) remains a mysterious disease of the endomyocardium that is poorly genetically characterized, affecting 1/5000 live births and accounting for 25% of the entire pediatric dilated cardiomyopathy (DCM) with a devastating course and grave prognosis. To investigate the potential genetic contribution to pEFE, we performed integrative genomic analysis, using whole exome sequencing (WES) and RNA-seq in a female infant with confirmed pathological diagnosis of pEFE. Within regions of homozygosity in the proband genome, WES analysis revealed novel parent-transmitted homozygous mutations affecting three genes with known roles in cilia assembly or function. Among them, a novel homozygous variant [c.1943delA] of uncertain significance in ALMS1 was prioritized for functional genomic and mechanistic analysis. Loss of function mutations of ALMS1 have been implicated in Alstrom syndrome (AS) [OMIM 203800], a rare recessive ciliopathy that has been associated with cardiomyopathy. The variant of interest results in a frameshift introducing a premature stop codon. RNA-seq of the proband's dermal fibroblasts confirmed the impact of the novel ALMS1 variant on RNA-seq reads and revealed dysregulated cellular signaling and function, including the induction of epithelial mesenchymal transition (EMT) and activation of TGFß signaling. ALMS1 loss enhanced cellular migration in patient fibroblasts as well as neonatal cardiac fibroblasts, while ALMS1-depleted cardiomyocytes exhibited enhanced proliferation activity. Herein, we present the unique pathological features of pEFE compared to DCM and utilize integrated genomic analysis to elucidate the molecular impact of a novel mutation in ALMS1 gene in an AS case. Our report provides insights into pEFE etiology and suggests, for the first time to our knowledge, ciliopathy as a potential underlying mechanism for this poorly understood and incurable form of neonatal cardiomyopathy. KEY MESSAGE: Primary endocardial fibroelastosis (pEFE) is a rare form of neonatal cardiomyopathy that occurs in 1/5000 live births with significant consequences but unknown etiology. Integrated genomics analysis (whole exome sequencing and RNA sequencing) elucidates novel genetic contribution to pEFE etiology. In this case, the cardiac manifestation in Alstrom syndrome is pEFE. To our knowledge, this report provides the first evidence linking ciliopathy to pEFE etiology. Infants with pEFE should be examined for syndromic features of Alstrom syndrome. Our findings lead to a better understanding of the molecular mechanisms of pEFE, paving the way to potential diagnostic and therapeutic applications.


Subject(s)
Alstrom Syndrome , Cardiomyopathies , Ciliopathies , Endocardial Fibroelastosis , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Alstrom Syndrome/pathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Endocardial Fibroelastosis/genetics , Endocardial Fibroelastosis/metabolism , Endocardial Fibroelastosis/pathology , Epithelial-Mesenchymal Transition , Female , Fibroblasts , Humans , Infant , Mutation , Myocardium/metabolism , Myocardium/pathology , Phenotype , RNA-Seq , Transcriptome
4.
Orphanet J Rare Dis ; 12(1): 138, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28793912

ABSTRACT

BACKGROUND: Hypoplastic left heart syndrome (HLHS) covers a spectrum of rare congenital anomalies characterised by a non-apex forming left ventricle and stenosis/atresia of the mitral and aortic valves. Despite many studies, the causes of HLHS remain unclear and there are conflicting views regarding the role of flow, valvar or myocardial abnormalities in its pathogenesis, all of which were proposed prior to the description of the second heart field. Our aim was to re-evaluate the patterns of malformation in HLHS in relation to recognised cardiac progenitor populations, with a view to providing aetiologically useful sub-groupings for genomic studies. RESULTS: We examined 78 hearts previously classified as HLHS, with subtypes based on valve patency, and re-categorised them based on their objective ventricular phenotype. Three distinct subgroups could be identified: slit-like left ventricle (24%); miniaturised left ventricle (6%); and thickened left ventricle with endocardial fibroelastosis (EFE; 70%). Slit-like ventricles were always found in combination with aortic atresia and mitral atresia. Miniaturised left ventricles all had normally formed, though smaller aortic and mitral valves. The remaining group were found to have a range of aortic valve malformations associated with thickened left ventricular walls despite being described as either atresia or stenosis. The degree of myocardial thickening was not correlated to the degree of valvar stenosis. Lineage tracing in mice to investigate the progenitor populations that form the parts of the heart disrupted by HLHS showed that whereas Nkx2-5-Cre labelled myocardial and endothelial cells within the left and right ventricles, Mef2c-AHF-Cre, which labels second heart field-derived cells only, was largely restricted to the endocardium and myocardium of the right ventricle. However, like Nkx2-5-Cre, Mef2c-AHF-Cre lineage cells made a significant contribution to the aortic and mitral valves. In contrast, Wnt1-Cre made a major contribution only to the aortic valve. This suggests that discrete cardiac progenitors might be responsible for the patterns of defects observed in the distinct ventricular sub-groups. CONCLUSIONS: Only the slit-like ventricle grouping was found to map to the current nomenclature: the combination of mitral atresia with aortic atresia. It appears that slit-like and miniature ventricles also form discrete sub-groups. Thus, reclassification of HLHS into subgroups based on ventricular phenotype, might be useful in genetic and developmental studies in investigating the aetiology of this severe malformation syndrome.


Subject(s)
Endocardial Fibroelastosis/metabolism , Endocardial Fibroelastosis/pathology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Hypoplastic Left Heart Syndrome/metabolism , Hypoplastic Left Heart Syndrome/pathology , Animals , Heart Ventricles/metabolism , Heart Ventricles/pathology , Homeobox Protein Nkx-2.5/metabolism , Immunohistochemistry , MEF2 Transcription Factors/metabolism , Mice , Mitral Valve/metabolism , Mitral Valve/pathology , Myocardium/metabolism , Myocardium/pathology
5.
Cell Res ; 27(9): 1157-1177, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28809397

ABSTRACT

Endocardial fibroelastosis (EFE) refers to the thickening of the ventricular endocardium as a result of de novo deposition of subendocardial fibrous tissue layers during neonatal heart development. The origin of EFE fibroblasts is proposed to be postnatal endocardial cells that undergo an aberrant endothelial-to-mesenchymal transition (EndMT). Genetic lineage tracing of endocardial cells with the inducible endocardial Cre line Npr3-CreER and the endothelial cell tracing line Cdh5-CreER on an EFE-like model did not reveal any contribution of neonatal endocardial cells to fibroblasts in the EFE-like tissues. Instead, lineage tracing of embryonic epicardium by Wt1-CreER suggested that epicardium-derived mesenchymal cells (MCs) served as the major source of EFE fibroblasts. By labeling MCs using Sox9-CreER, we confirmed that MCs of the embryonic heart expand and contribute to the majority of neonatal EFE fibroblasts. During this pathological process, TGFß signaling, the key mediator of fibroblasts activation, was highly upregulated in the EFE-like tissues. Targeting TGFß signaling by administration of its antagonist bone morphogenetic protein 7 effectively reduced fibroblast accumulation and tissue fibrosis in the EFE-like model. Our study provides genetic evidence that excessive fibroblasts in the EFE-like tissues mainly originate from the epicardium-derived MCs through epicardial to mesenchymal transition (EpiMT). These EpiMT-derived fibroblasts within the EFE-like tissues could serve as a potential therapeutic target.


Subject(s)
Endocardial Fibroelastosis/pathology , Fibroblasts/pathology , Pericardium/pathology , Animals , Animals, Newborn , Cell Lineage , Disease Models, Animal , Embryo, Mammalian/pathology , Endocardial Fibroelastosis/metabolism , Endocardium/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblasts/metabolism , Heart Transplantation , Integrases/metabolism , Mesoderm , Mice , Models, Biological , SOX9 Transcription Factor/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Basic Res Cardiol ; 111(1): 6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26659360

ABSTRACT

Cardiomyopathy is one of the most common causes of chronic heart failure worldwide. Mutations in the gene encoding nexilin (NEXN) occur in patients with both hypertrophic and dilated cardiomyopathy (DCM); however, little is known about the pathophysiological mechanisms and relevance of NEXN to these disorders. Here, we evaluated the functional role of NEXN using a constitutive Nexn knock-out (KO) mouse model. Heterozygous (Het) mice were inter-crossed to produce wild-type (WT), Het, and homozygous KO mice. At birth, 32, 46, and 22 % of the mice were WT, Het, and KO, respectively, which is close to the expected Mendelian ratio. After postnatal day 6, the survival of the Nexn KO mice decreased dramatically and all of the animals died by day 8. Phenotypic characterizations of the WT and KO mice were performed at postnatal days 1, 2, 4, and 6. At birth, the relative heart weights of the WT and KO mice were similar; however, at day 4, the relative heart weight of the KO group was 2.3-fold higher than of the WT group. In addition, the KO mice developed rapidly progressive cardiomyopathy with left ventricular dilation and wall thinning and decreased cardiac function. At day 6, the KO mice developed a fulminant DCM phenotype characterized by dilated ventricular chambers and systolic dysfunction. At this stage, collagen deposits and some elastin deposits were observed within the left ventricle cavity, which resembles the features of endomyocardial fibroelastosis (EFE). Overall, these results further emphasize the role of NEXN in DCM and suggest a novel role in EFE.


Subject(s)
Cardiomyopathies/metabolism , Endocardial Fibroelastosis/metabolism , Microfilament Proteins/deficiency , Animals , Blotting, Western , Cardiomyopathies/pathology , Disease Models, Animal , Echocardiography , Endocardial Fibroelastosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Polymerase Chain Reaction
7.
Cardiovasc Pathol ; 24(6): 388-94, 2015.
Article in English | MEDLINE | ID: mdl-26363814

ABSTRACT

BACKGROUND: Endocardial fibroelastosis (EFE) is a pathologic condition of abnormal deposition of collagen and elastin within the endocardium of the heart. It is seen in conjunction with a variety of diseases including hypoplastic left heart syndrome and viral endocarditis. While an experimental model using heterotopic heart transplant in rats has been described, we sought to fully describe a mouse model that can be used to further elucidate the potential mechanisms of and treatments for EFE. MATERIALS AND METHODS: The hearts of 2-day-old C57BL/6 mice were transplanted into the abdomen of 7-week-old C57BL/6 mice. At 2 weeks, the hearts were harvested and histologic analysis was performed using hematoxylin and eosin, Masson's trichrome, Russell-Movat's pentachrome, Picrosirius red, Hart's, Verhoeff-Van Gieson, and Weigert's Resorcin-Fuchsin stains. Additionally, one heart was analyzed using transmission electron microscopy (TEM). RESULTS: Specimens demonstrated abnormal accumulation of both collagen and elastin within the endocardium with occasional expansion into the myocardium. Heterogeneity in extracellular matrix deposition was noted in the histologic specimens. In addition, TEM demonstrated the presence of excess collagen within the endocardium. CONCLUSIONS: The heterotopic transplantation of an immature heart into a mouse results in changes consistent with EFE. This model is appropriate to investigate the etiology and treatment of EFE.


Subject(s)
Endocardial Fibroelastosis/pathology , Endocardium/pathology , Heart Transplantation , Age Factors , Animals , Collagen/metabolism , Disease Models, Animal , Elastin/metabolism , Endocardial Fibroelastosis/diagnostic imaging , Endocardial Fibroelastosis/etiology , Endocardial Fibroelastosis/metabolism , Endocardium/diagnostic imaging , Endocardium/metabolism , Endocardium/ultrastructure , Extracellular Matrix/metabolism , Mice, Inbred C57BL , Time Factors , Ultrasonography
8.
J Surg Res ; 182(1): 94-100, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-22938709

ABSTRACT

BACKGROUND: Hypoplastic left heart syndrome (HLHS) is one of the most common severe congenital cardiac anomalies, characterized by a marked hypoplasia of left-sided structures of the heart, which is commonly accompanied by a thick layer of fibroelastic tissue, termed endocardial fibroelastosis (EFE). Because human EFE develops only in fetal or neonatal hearts, and often in association with reduced blood flow, we sought to mimic these conditions by subjecting neonatal and 2-wk-old rat hearts to variations of the heterotopically transplanted heart model with either no intracavitary or normal flow and compare endocardium with human EFE tissue. MATERIALS AND METHODS: Hearts obtained from neonatal and 2-wk-old rats were heterotopically transplanted in young adult Lewis rats in a working (loaded) or nonworking (unloaded) mode. After 2-wk survival, hearts were explanted for histologic analysis by staining for collagen, elastin, and cellular elements. These sections were compared with human EFE tissue from HLHS. RESULTS: EFE, consisting of collagen and elastin with scarce cellular and vascular components, developed only in neonatal unloaded transplanted hearts and displayed the same histopathologic findings as EFE from patients with HLHS. Loaded hearts and 2-wk-old hearts did not show these alterations. CONCLUSIONS: This animal model for EFE will serve as a tool to study the mechanisms of EFE formation, such as fluid forces, in HLHS in a systematic manner. A better understanding of the underlying cause of the EFE formation in HLHS will help to develop novel treatment strategies to better preserve growth of the hypoplastic left ventricle.


Subject(s)
Disease Models, Animal , Endocardial Fibroelastosis/pathology , Heart Transplantation/pathology , Hypoplastic Left Heart Syndrome/pathology , Animals , Animals, Newborn , Collagen/metabolism , Elastin/metabolism , Endocardial Fibroelastosis/metabolism , Hemodynamics/physiology , Humans , Hypoplastic Left Heart Syndrome/metabolism , Hypoplastic Left Heart Syndrome/physiopathology , Rats , Rats, Inbred Lew , Transplantation, Heterotopic
9.
Pediatr Cardiol ; 32(8): 1088-95, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21484220

ABSTRACT

The observation that endocardial fibroelastosis (EFE) can result from an immune response to maternal autoantibody deposition in the fetal myocardium raises the possibility that the fetal immune system may contribute to the pathogenesis of idiopathic EFE and dilated cardiomyopathy (DCM). This study sought to characterize myocardial immune cell presence in fetuses and neonates with idiopathic EFE + DCM, in those with EFE + structural heart disease, and in normal control subjects. Paraffin tissue sections from fetuses identified from the pathology database were stained for B cell, T cell, macrophage, and general hematopoietic cell surface markers. Of the 14 fetuses included in the study, 5 had EFE + DCM, 4 had EFE + structural heart disease, and 5 were normal control fetuses. The EFE + DCM group had fewer B cells than the control group (0.15 vs. 0.44 cells/mm(2); p = 0.005). The EFE + heart disease group had both fewer B cells (0.18 vs. 0.44 cells/mm(2); p = 0.08) and T cells (0.29 vs. 0.80 cells/mm(2); p = 0.04) than the control group. The CD4/CD8 ratio was similar in the EFE + DCM and EFE + heart disease groups (1.0 vs. 0.9; p = 0.17) but higher in the EFE + DCM group than in the control group (0.9 vs. 0.3; p = 0.03). The myocardium of fetuses with EFE contains fewer B and T lymphocytes than normal control fetuses.


Subject(s)
B-Lymphocytes/metabolism , Cardiomyopathy, Dilated/metabolism , Endocardial Fibroelastosis/metabolism , Fetus/metabolism , Myocardium/immunology , Myocardium/metabolism , T-Lymphocytes/metabolism , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/pathology , Case-Control Studies , Dilatation, Pathologic , Endocardial Fibroelastosis/immunology , Humans , Immunohistochemistry , Lymphocyte Count , Myocardium/pathology
10.
J Am Coll Cardiol ; 56(18): 1493-502, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20951326

ABSTRACT

OBJECTIVES: Four variants (K60N, Q128R, G202R, and A592E) in the nebulette gene were identified in patients with dilated cardiomyopathy (DCM) and endocardial fibroelastosis. We sought to determine if these mutations are cardiomyopathy causing. BACKGROUND: Nebulette aligns thin filaments and connects them with the myocardial Z-disk, playing a role in mechanosensation. METHODS: We generated transgenic mice with cardiac-restricted overexpression of human wild-type or mutant nebulette. Chimera and transgenic mice were examined at 4, 6, and 12 months of age by echocardiography and cardiac magnetic resonance imaging. The hearts from embryos and adult mice were assessed by histopathologic, immunohistochemical, ultrastructural, and protein analyses. Rat H9C2 cardiomyoblasts with transient expression of nebulette underwent cyclic mechanical strain. RESULTS: We identified lethal cardiac structural abnormalities in mutant embryonic hearts (K60N and Q128R). Founders of the mutant mouse lines developed DCM with severe heart failure. An irregular localization pattern for nebulette and impaired desmin expression were noted in the proband and chimeric Q128R mice. Mutant G202R and A592E mice exhibited left ventricular dilation and impaired function with specific changes in I-band and Z-disk proteins by 6 months of age. The mutations modulated distribution of nebulette in the sarcomere and Z-disk during stretch of H9C2 cells. CONCLUSIONS: Nebulette is a new susceptibility gene for endocardial fibroelastosis and DCM. Different mutations in nebulette trigger specific mechanisms, converging to a common pathological cascade leading to endocardial fibroelastosis and DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , Carrier Proteins/genetics , Cytoskeletal Proteins/genetics , Endocardial Fibroelastosis/genetics , Mutation/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Cardiomyopathy, Dilated/embryology , Cardiomyopathy, Dilated/metabolism , Carrier Proteins/biosynthesis , Cell Line , Cytoskeletal Proteins/biosynthesis , Endocardial Fibroelastosis/embryology , Endocardial Fibroelastosis/metabolism , Genetic Predisposition to Disease , Humans , LIM Domain Proteins , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Rats
12.
Am J Cardiovasc Pathol ; 3(3): 247-51, 1990.
Article in English | MEDLINE | ID: mdl-2151366

ABSTRACT

The presence and distribution of human atrial natriuretic polypeptide (ANP) were investigated immunohistochemically in the ventricles of hearts of 14 cases with endocardial fibroelastosis and 15 cases with noncardiac disease in children. Paraffin sections of autopsied hearts with endocardial fibroelastosis were stained with polyclonal antibodies against human alpha-ANP. Immunoreactive myocytes were clearly demonstrated in the ventricles of 10 hearts with endocardial fibroelastosis. The distribution of ANP-positive cells was most frequent in the inner one-third of the left ventricle. No ANP immunoreactivity was detected in any heart in cases with noncardiac disease. The left ventricular volume index of hearts with ANP-positive cells was larger than that with ANP-negative cells. The mean diameter of ANP-positive myocytes was greater than that of ANP-negative myocytes. These results suggest that ANP expression in ventricular myocytes is related to severe dilatation of the ventricular cavity and to development of myocardial hypertrophy in endocardial fibroelastosis.


Subject(s)
Atrial Natriuretic Factor/analysis , Endocardial Fibroelastosis/metabolism , Myocardium/chemistry , Child , Endocardial Fibroelastosis/pathology , Humans , Immunoenzyme Techniques , Infant, Newborn , Myocardium/ultrastructure
13.
Arch Pathol Lab Med ; 101(1): 49-54, 1977 Jan.
Article in English | MEDLINE | ID: mdl-576201

ABSTRACT

The average size of elastic fibers in thickened left ventricular endocardium was much larger in four patients with congenital endocardial fibroelastosis (EFE) than in six patients with acquired EFE (secondary to ischemic heart disease in two patients, to prosthetic cardiac valves in three, and to irradiation of the chest in one). Both components of normal elastic tissue (central, amorphous cores, and peripheral microfibrils) were present in endocardial elastic fibers of each patient. Ultrastructural identification of elastic fibers was greatly facilitated by staining with silver tetraphenylporphin sulfonate.


Subject(s)
Endocardial Fibroelastosis/pathology , Endocardium/pathology , Adult , Child , Child, Preschool , Collagen/analysis , Elastic Tissue/analysis , Endocardial Fibroelastosis/etiology , Endocardial Fibroelastosis/metabolism , Endocardium/ultrastructure , Female , Histocytochemistry , Humans , Infant , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...