Abstract

Tapasin plays an important role in the quality control of major histocompatibility complex (MHC) class I assembly, but its precise function in this process remains controversial. Whether tapasin participates in the assembly of HLA-G has not been studied. HLA-G, an MHC class Ib molecule that binds a more restricted set of peptides than class Ia molecules, is a particularly interesting molecule, because during assembly, it recycles between the endoplasmic reticulum (ER) and the cis-Golgi until it is loaded with a high affinity peptide. We have taken advantage of this unusual trafficking property of HLA-G and its requirement for high affinity peptides to demonstrate that a critical function of tapasin is to transform class I molecules into a high affinity, peptide-receptive form. In the absence of tapasin, HLA-G molecules cannot bind high affinity peptides, and an abundant supply of peptides cannot overcome the tapasin requirement for high affinity peptide loading. The addition of tapasin renders HLA-G molecules capable of loading high affinity peptides and of transporting to the surface, suggesting that tapasin is a prerequisite for the binding of high-affinity ligands. Interestingly, the "tapasin-dependent" HLA-G molecules are not empty in the absence of tapasin but are in fact associated with suboptimal peptides and continue to recycle between the ER and the cis-Golgi. Together with the finding that empty HLA-G heterodimers are strictly retained in the ER and degraded, our data suggest that MHC class I molecules bind any available peptides to avoid ER-mediated degradation and that the peptides are in turn replaced by higher affinity peptides with the aid of tapasin.

Highlights

  • The HLA-G/E114Q mutant, in which residue 114 was replaced by the neutrally charged glutamine, fell between HLA-G wild type and HLA-G/E114H in the spectrum of tapasin dependence. These results indicate that, like HLA class Ia molecules, the tapasin dependence of HLA-G is influenced by the nature of the amino acid at position 114

  • We addressed the question of whether tapasin is involved in the assembly and trafficking of HLA-G

  • Our results demonstrate that tapasin is not required for suboptimal peptide binding but is critical for loading of high affinity peptides onto HLA-G

Read more

Summary

A Critical Function of Tapasin in Quality Control of HLA-G

HLA-G, an MHC class Ib molecule, is expressed primarily in trophoblast cells and has limited polymorphism [2]. HLA-G is a interesting molecule in protein trafficking, because it recycles between the ER and the cis-Golgi until it is loaded with a high affinity peptide [19]. This feature makes it possible to estimate whether HLA-G molecules are loaded with high affinity or low affinity peptides. Because HLA-G binds a restricted set of peptides [23, 24] and recycles between the ER and cis-Golgi, it is an attractive model for investigating the roles of individual components of the ER peptide-loading complex in the assembly of functional MHC class I molecules. We propose that a critical function of tapasin is to transform the peptide-binding groove of HLA-G into a high affinity, peptidereceptive form, which could promote the replacement of low affinity peptides with high affinity peptides

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.