Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 8(9): e72668, 2013.
Article in English | MEDLINE | ID: mdl-24039792

ABSTRACT

BACKGROUND: Congenital heart block (CHB) is a transplacentally acquired autoimmune disease associated with anti-Ro/SSA and anti-La/SSB maternal autoantibodies and is characterized primarily by atrioventricular (AV) block of the fetal heart. This study aims to investigate whether the T-type calcium channel subunit α1G may be a fetal target of maternal sera autoantibodies in CHB. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate differential mRNA expression of the T-type calcium channel CACNA1G (α1G gene) in the AV junction of human fetal hearts compared to the apex (18-22.6 weeks gestation). Using human fetal hearts (20-22 wks gestation), our immunoprecipitation (IP), Western blot analysis and immunofluorescence (IF) staining results, taken together, demonstrate accessibility of the α1G epitope on the surfaces of cardiomyocytes as well as reactivity of maternal serum from CHB affected pregnancies to the α1G protein. By ELISA we demonstrated maternal sera reactivity to α1G was significantly higher in CHB maternal sera compared to controls, and reactivity was epitope mapped to a peptide designated as p305 (corresponding to aa305-319 of the extracellular loop linking transmembrane segments S5-S6 in α1G repeat I). Maternal sera from CHB affected pregnancies also reacted more weakly to the homologous region (7/15 amino acids conserved) of the α1H channel. Electrophysiology experiments with single-cell patch-clamp also demonstrated effects of CHB maternal sera on T-type current in mouse sinoatrial node (SAN) cells. CONCLUSIONS/SIGNIFICANCE: Taken together, these results indicate that CHB maternal sera antibodies readily target an extracellular epitope of α1G T-type calcium channels in human fetal cardiomyocytes. CHB maternal sera also show reactivity for α1H suggesting that autoantibodies can target multiple fetal targets.


Subject(s)
Autoantibodies/immunology , Calcium Channels, T-Type/immunology , Epitopes/immunology , Heart Block/congenital , Amino Acid Sequence , Animals , Atrioventricular Node/drug effects , Atrioventricular Node/metabolism , Autoantibodies/blood , Autoantigens/immunology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Epitope Mapping , Extracellular Space , Female , Fetal Heart/drug effects , Fetal Heart/immunology , Fetal Heart/metabolism , Gene Expression , Heart Block/genetics , Heart Block/immunology , Humans , Male , Maternal-Fetal Exchange/immunology , Mice , Molecular Sequence Data , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Peptides/immunology , Pregnancy , Rabbits
2.
J Immunol ; 185(6): 3574-82, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20696861

ABSTRACT

Congenital heart block develops in fetuses of anti-Ro52 Ab-positive women. A recurrence rate of 20%, despite the persistence of maternal autoantibodies, indicates that there are additional, yet unidentified, factors critical for development of congenital heart block. In this study, we demonstrate that besides the maternal MHC controlling Ab specificity, fetal MHC-encoded genes influence fetal susceptibility to congenital heart block. Using MHC congenic rat strains, we show that heart block develops in rat pups of three strains carrying MHC haplotype RT1(av1) (DA, PVG.AV1, and LEW.AV1) after maternal Ro52 immunization, but not in LEW rats (RT1(l)). Different anti-Ro52 Ab fine specificities were generated in RT1(av1) versus RT1(l) animals. Maternal and fetal influence was determined in an F(2) cross between LEW.AV1 and LEW strains, which revealed higher susceptibility in RT1(l) than RT1(av1) pups once pathogenic Ro52 Abs were present. This was further confirmed in that RT1(l) pups more frequently developed heart block than RT1(av1) pups after passive transfer of RT1(av1) anti-Ro52 sera. Our findings show that generation of pathogenic Ro52 Abs is restricted by maternal MHC, whereas the fetal MHC locus regulates susceptibility and determines the fetal disease outcome in anti-Ro52-positive pregnancies.


Subject(s)
Atrioventricular Block/genetics , Atrioventricular Block/immunology , Autoantibodies/biosynthesis , Genetic Predisposition to Disease , Histocompatibility Antigens/genetics , Maternal-Fetal Exchange/immunology , Ribonucleoproteins/immunology , Amino Acid Sequence , Animals , Antibody Specificity/genetics , Atrioventricular Block/congenital , Cell Line , Disease Models, Animal , Female , Histocompatibility Antigens/immunology , Maternal-Fetal Exchange/genetics , Molecular Sequence Data , Pregnancy , Rats , Rats, Inbred Lew , Ribonucleoproteins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...