Abstract

The immunomodulatory receptor Siglec-3/CD33 influences risk for late-onset Alzheimer's disease (LOAD), an apparently human-specific post-reproductive disease. CD33 generates two splice variants: a full-length CD33M transcript produced primarily by the "LOAD-risk" allele and a shorter CD33m isoform lacking the sialic acid-binding domain produced primarily from the "LOAD-protective" allele. An SNP that modulates CD33 splicing to favor CD33m is associated with enhanced microglial activity. Individuals expressing more protective isoform accumulate less brain β-amyloid and have a lower LOAD risk. How the CD33m isoform increases β-amyloid clearance remains unknown. We report that the protection by the CD33m isoform may not be conferred by what it does but, rather, from what it cannot do. Analysis of blood neutrophils and monocytes and a microglial cell line revealed that unlike CD33M, the CD33m isoform does not localize to cell surfaces; instead, it accumulates in peroxisomes. Cell stimulation and activation did not mobilize CD33m to the surface. Thus, the CD33m isoform may neither interact directly with amyloid plaques nor engage in cell-surface signaling. Rather, production and localization of CD33m in peroxisomes is a way of diminishing the amount of CD33M and enhancing β-amyloid clearance. We confirmed intracellular localization by generating a CD33m-specific monoclonal antibody. Of note, CD33 is the only Siglec with a peroxisome-targeting sequence, and this motif emerged by convergent evolution in toothed whales, the only other mammals with a prolonged post-reproductive lifespan. The CD33 allele that protects post-reproductive individuals from LOAD may have evolved by adaptive loss-of-function, an example of the less-is-more hypothesis.

Highlights

  • The immunomodulatory receptor Siglec-3/CD33 influences risk for late-onset Alzheimer’s disease (LOAD), an apparently human-specific post-reproductive disease

  • To determine whether CD33 is expressed at the cell surface or inside the cell, we probed neutrophils using an antibody (HIM3-4) that reacts with C2-set domain of CD33

  • Because CD33 expression is relevant for a role of microglia in Alzheimer’s dementia, we chose a human microglia cell line CHME-5 to determine the status of CD33

Read more

Summary

Results and discussion

Previous studies reported very low expression of CD33 on mature granulocytes [6, 23]. It has previously been shown that a CD33M mutant (one where the cysteine of the V-set domain is mutated) stays inside the cell and cannot traffic to the cell surface [16] In pursuing this question, we could not get consistent and reproducible Western blots for CD33m despite trying various lysis protocols for neutrophils and CHME-5 cells such as with Nonidet P-40 buffer, RIPA (radioimmune precipitation assay buffer) buffer, 1% SDS lysis, freezethaw, and 1% SDS lysisϩsonication. We found no significant change in the cell surface expression of CD33 (Fig. 5A) This shows that the retention of CD33M on the cell surface and the intracellular localization of CD33m are not a result of their respective ability to bind sialic acids and be retained by extracellular ligands. Using different anti-CD33 antibodies (CD33M and CD33mϩM) we found there is partial mobilization of CD33 to the surface, and this fraction is CD33m (Fig. 5B, top and bottom left panel)

Phylogenetic origins of the putative peroxisome trafficking motif
Conclusions and perspectives
Type of mutation
Disease improved
Cells and antibodies
Immunofluorescence and confocal microscopy
Full Text
Published version (Free)

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