4.8 Article

Functions of the COPII gene paralogs SEC23A and SEC23B are interchangeable in vivo

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.1805784115

Keywords

endoplasmic reticulum; vesicular transport proteins; mice; transgenic; SEC23

Funding

  1. National Institute of Health [R01 HL039693, R35-HL135793, P01-HL057346, K08 HL128794, R01 GM053396, R01 HL 124232, R01 GM094231, R01 HL094505, T32 HL007622]
  2. Canadian Institutes of Health Research Foundation [FDN 143301]
  3. MCubed, a research seed-funding program for faculty at the University of Michigan
  4. University of Michigan Cancer Center (NIH) [P30 CA046592]
  5. National Hemophilia Foundation-Shire Clinical Fellowship Award
  6. Parkinson Canada Basic Research Fellowship
  7. American Society of Hematology Scholar Awards

Ask authors/readers for more resources

Approximately one-third of the mammalian proteome is transported from the endoplasmic reticulum-to-Golgi via COPII-coated vesicles. SEC23, a core component of coat protein-complex II (COPII), is encoded by two paralogous genes in vertebrates (Sec23a and Sec23b). In humans, SEC23B deficiency results in congenital dysery-thropoietic anemia type-II (CDAII), while SEC23A deficiency results in a skeletal phenotype (with normal red blood cells). These distinct clinical disorders, together with previous biochemical studies, suggest unique functions for SEC23A and SEC23B. Here we show indistinguishable intracellular protein interactomes for human SEC23A and SEC23B, complementation of yeast Sec23 by both human and murine SEC23A/B, and rescue of the lethality of sec23b deficiency in zebrafish by a sec23a-expressing transgene. We next demonstrate that a Sec23a coding sequence inserted into the murine Sec23b locus completely rescues the lethal SEC23B-deficient pancreatic phenotype. We show that SEC23B is the predominantly expressed paralog in human bone marrow, but not in the mouse, with the reciprocal pattern observed in the pancreas. Taken together, these data demonstrate an equivalent function for SEC23A/B, with evolutionary shifts in the transcription program likely accounting for the distinct phenotypes of SEC23A/B deficiency within and across species, a paradigm potentially applicable to other sets of paralogous genes. These findings also suggest that enhanced erythroid expression of the normal SEC23A gene could offer an effective therapeutic approach for CDAII patients.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available