TY - JOUR T1 - Defective heart chamber growth and myofibrillogenesis after knockout of <em>adprhl1</em> gene function by targeted disruption of the ancestral catalytic active site JF - bioRxiv DO - 10.1101/2020.02.13.947424 SP - 2020.02.13.947424 AU - Stuart J. Smith AU - Norma Towers AU - Kim Demetriou AU - Timothy J. Mohun Y1 - 2020/01/01 UR - http://biorxiv.org/content/early/2020/02/27/2020.02.13.947424.abstract N2 - ADP-ribosylhydrolase-like 1 (Adprhl1) is a pseudoenzyme expressed in the developing heart myocardium of all vertebrates. In the amphibian Xenopus laevis, knockdown of the two cardiac Adprhl1 protein species (40 and 23 kDa) causes failure of chamber outgrowth but this has only been demonstrated using antisense morpholinos that interfere with RNA-splicing. Transgenic production of 40 kDa Adprhl1 provides only part rescue of these defects. CRISPR/Cas9 technology now enables targeted mutation of the adprhl1 gene in G0-generation embryos with routine cleavage of all alleles. Testing multiple gRNAs distributed across the locus reveals exonic locations that encode critical amino acids for Adprhl1 function. The gRNA recording the highest frequency of a specific ventricle outgrowth phenotype directs Cas9 cleavage of an exon 6 sequence, where microhomology mediated end-joining biases subsequent DNA repairs towards three small in-frame deletions. Mutant alleles encode discrete loss of 1, 3 or 4 amino acids from a di-arginine (Arg271-Arg272) containing peptide loop at the centre of the ancestral ADP-ribosylhydrolase site. Thus despite lacking catalytic activity, it is the modified (adenosine-ribose) substrate binding cleft of Adprhl1 that fulfils an essential role during heart formation. Mutation results in striking loss of myofibril assembly in ventricle cardiomyocytes. The defects suggest Adprhl1 participation from the earliest stage of cardiac myofibrillogenesis and are consistent with previous MO results and Adprhl1 protein localization to actin filament Z-disc boundaries. A single nucleotide change to the gRNA sequence renders it inactive. Mice lacking Adprhl1 exons 3-4 are normal but production of the smaller ADPRHL1 species is unaffected, providing further evidence that cardiac activity is concentrated at the C-terminal protein portion.Highlights Comparison of adprhl1 morpholinos. Knockdown of the two Xenopus cardiac Adprhl1 protein species (40 and 23 kDa) causes failure of ventricle outgrowth.CRISPR/Cas9 targeted gene mutation of adprhl1 with multiple gRNAs reveals exonic locations that encode critical amino acids for Adprhl1 function.Repair of DSBs at exon 6 yields small in-frame deletions that cause specific ventricle myofibril assembly defects.The deletions disturb a conserved di-arginine containing peptide loop at the centre of the ancestral substrate binding cleft/ADP-ribosylhydrolase site of this pseudoenzyme.Mice lacking Adprhl1 exons 3-4 are normal but production of the smaller ADPRHL1 species is unaffected, providing further evidence that cardiac activity is concentrated at the C-terminal protein portion. ER -