Your browser doesn't support javascript.
loading
Semaphorin3f as a cardiomyocyte derived regulator of heart chamber development.
Halabi, Rami; Cechmanek, Paula Bernice; Hehr, Carrie Lynn; McFarlane, Sarah.
Affiliation
  • Halabi R; Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Dr., NW, Calgary, AB, T2N 4N1, Canada.
  • Cechmanek PB; Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Dr., NW, Calgary, AB, T2N 4N1, Canada.
  • Hehr CL; Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Dr., NW, Calgary, AB, T2N 4N1, Canada.
  • McFarlane S; Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, Department of Cell Biology and Anatomy, University of Calgary, 3330 Hospital Dr., NW, Calgary, AB, T2N 4N1, Canada. smcfarla@ucalgary.ca.
Cell Commun Signal ; 20(1): 126, 2022 08 19.
Article in En | MEDLINE | ID: mdl-35986301
ABSTRACT

BACKGROUND:

During development a pool of precursors form a heart with atrial and ventricular chambers that exhibit distinct transcriptional and electrophysiological properties. Normal development of these chambers is essential for full term survival of the fetus, and deviations result in congenital heart defects. The large number of genes that may cause congenital heart defects when mutated, and the genetic variability and penetrance of the ensuing phenotypes, reveals a need to understand the molecular mechanisms that allow for the formation of chamber-specific cardiomyocyte differentiation.

METHODS:

We used in situ hybridization, immunohistochemistry and functional analyses to identify the consequences of the loss of the secreted semaphorin, Sema3fb, in the development of the zebrafish heart by using two sema3fb CRISPR mutant alleles.

RESULTS:

We find that in the developing zebrafish heart sema3fb mRNA is expressed by all cardiomyocytes, whereas mRNA for a known receptor Plexina3 (Plxna3) is expressed preferentially by ventricular cardiomyocytes. In sema3fb CRISPR zebrafish mutants, heart chamber development is impaired; the atria and ventricles of mutants are smaller in size than their wild type siblings, apparently because of differences in cell size and not cell numbers. Analysis of chamber differentiation indicates defects in chamber specific gene expression at the border between the ventricular and atrial chambers, with spillage of ventricular chamber genes into the atrium, and vice versa, and a failure to restrict specialized cardiomyocyte markers to the atrioventricular canal (AVC). The hypoplastic heart chambers are associated with decreased cardiac output and heart edema.

CONCLUSIONS:

Based on our data we propose a model whereby cardiomyocytes secrete a Sema cue that, because of spatially restricted expression of the receptor, signals in a ventricular chamber-specific manner to establish a distinct border between atrial and ventricular chambers that is important to produce a fully functional heart. Video abstract.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Myocytes, Cardiac / Heart Defects, Congenital Limits: Animals Language: En Journal: Cell Commun Signal Year: 2022 Document type: Article Affiliation country: Canadá

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Myocytes, Cardiac / Heart Defects, Congenital Limits: Animals Language: En Journal: Cell Commun Signal Year: 2022 Document type: Article Affiliation country: Canadá