Stress-induced angiogenesis enormously contributes to both normal development and pathogenesis of various diseases including cancer. Among many stress response pathways implicated in regulation of angiogenesis, the amino acid response (AAR) and the unfolded protein response (UPR) pathways are closely interconnected, as they converge on the common target, eIF2α, which is a key regulator of protein translation. Two kinases, namely Gcn2 (Eif2ak4) and Perk (Eif2ak3), are responsible for transducing signals from AAR and UPR, respectively, to phosphorylation of eIF2α. Even though numerous studies have been performed, this close interconnection between AAR and UPR makes it difficult to clearly distinguish different contributions of these two pathways in regulation of angiogenesis.

In this study, we generated a zebrafish angiogenic model harboring a loss-of-function mutation of the threonyl-tRNA synthetase (tars) gene. Tars belongs to a family of evolutionarily conserved enzymes, aminoacyl-tRNA synthetases (aaRSs), which control the first step of protein translation through coupling specific amino acids with their cognate tRNAs. Deficiencies of several aaRSs in zebrafish have been shown to cause increased branching of blood vessels, and this angiogenic phenotype has roughly been explained by activation of AAR and UPR; however, it is unclear whether both AAR and UPR are required and to what extent they contribute to this process. To address this issue, we first performed RNA-seq analyses of Tars-mutated and control zebrafish embryos, as well as those with knockdown of either Gcn2 or Perk in both genotypes. We found that the AAR target genes are dramatically activated in the Tars-mutants, whereas the genes associated with the three UPR sub-pathways (i.e., Perk-, Ire1- and Atf6-mediated pathways) remain inactive, except for very few genes (e.g., Atf3, Atf4, Asns and Igfbp1) that are shared in both AAR and UPR, thus suggesting activation of AAR, but not UPR, in the Tars-mutants. In support of this notion, knockdown of the AAR-associated kinase Gcn2 in the Tars-mutants largely represses the activated genes, while the Perk knockdown shows very little effect. Nonetheless, in contrast to the apparently dispensable role of Perk in Tars-mutants, knockdown of Perk in control embryos leads to specific gene expression alterations, suggesting that Perk effectively functions in homeostatic states (i.e., controls), but, in the stress condition (i.e., Tars-mutants), its function is largely overwhelmed by activation of the Gcn2-mediated AAR.

To validate these observations, we investigated the angiogenic phenotypes of the zebrafish models upon genetic and pharmacological interference with the AAR and UPR pathways. A transgenic zebrafish line, Tg(flk1:EGFP), was crossed with the Tars-mutants to visualize angiogenesis in vivo. We observed increased branching of blood vessels in the Tars-mutants, which is rescued by tars mRNA but not an enzymatically dead version. Importantly, knockdown of Gcn2 in the Tars-mutants rescues this phenotype. In contrast, knockdown of Perk, or knockdown of two other known eIF2α kinases, Hri (Eif2ak1) or Pkr (Eif2ak2), shows no effect. Furthermore, knockdown of either one of two major factors downstream to eIF2α, namely Atf4 and Vegfα, or inhibition of Vegf receptor with the drug SU5416, also rescue the phenotype. Thus, these results confirm that AAR, but not UPR, is required for the Tars-deficiency-induced angiogenesis.

Taken together, this study demonstrates that, despite being closely interconnected and even sharing a common downstream target, the Gcn2-mediated AAR and the Perk-mediated UPR can be activated independently in different conditions and differentially regulate cellular functions such as angiogenesis. This notion reflects the specificity and efficiency of multiple stress response pathways that are evolved integrally to benefit the organism by ensuring sensing and responding precisely to different types of stresses. This study also provides an example of combining systematic gene expression profiling and phenotypic validations to distinguish activities of such interconnected pathways. Further clarification of the mechanisms shall advance our understanding of how the organisms respond to diverse stresses and how the abnormalities in these regulatory machineries cause cellular stress-related diseases such as cancer, diabetes and immune disorders.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution