04.11.2014 Views

trans

trans

trans

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

GENETICS OF ANTIGENIC VARIATION 101<br />

structure having an intrinsic suppressive effect<br />

on <strong>trans</strong>cription, perhaps because telomeres<br />

are sequestered in particular regions of the<br />

nucleus (specifically at the nuclear envelope<br />

in S. cerevisiae). However, it is unclear what<br />

would allow just one telomere to escape a suppressive<br />

structure. TPE has been demonstrated<br />

in T. brucei, by inserting reporter cassettes into<br />

different genomic environments, but it is not<br />

clear how much of a role it has in regulating ES<br />

<strong>trans</strong>cription from the telomere-distal ES promoter.<br />

TPE could play a role in suppressing <strong>trans</strong>cription<br />

from telomere-proximal metacyclic<br />

ES promoters.<br />

The use of trypanosome lines that have been<br />

highly adapted to laboratory use has received<br />

criticism from some investigators, on the basis<br />

that these lines have been selected for virulence<br />

and VSG stability, which may be incompatible<br />

with, and have led to loss or attenuation of<br />

mechanisms involved in, rapid VSG switching.<br />

On the other hand, most studies of VSG switching<br />

would not have been successful in rapidly<br />

switching lines. Because of this issue, suggestions<br />

about the relative roles of different switching<br />

mechanisms should be not be considered<br />

to be written in stone, at this stage of our knowledge.<br />

The existence of specific VSG recombination<br />

mechanisms is suggested by the extremely<br />

conserved sequences flanking the VSG conversion<br />

cassette, which form the boundaries<br />

of the gene conversion in most cases in which<br />

these have been determined. Again, there are<br />

exceptions to this statement, and one must<br />

always carefully examine the experimental conditions<br />

used in different studies before making<br />

direct comparisons of the outcome and<br />

interpretation.<br />

When metacyclic trypanosomes enter the<br />

bloodstream, they continue to express metacyclic<br />

VSGs for several cycles of replication.<br />

Then the metacyclic ESs shut down and a<br />

bloodstream ES takes over. It has been<br />

suggested that the early bloodstream infection<br />

is a time of rapid VSG switching. Both logic and<br />

experimental evidence suggest that gene conversion<br />

is likely to be the major mechanism for<br />

VSG expression, because there are so many<br />

silent VSGs and only a few ESs. However, most<br />

mechanistic studies of VSG switching have so<br />

far focused on ES switching, probably because<br />

it is more accessible to experimental investigation.<br />

One interesting question is why multiple<br />

ESs exist? One possibility is that it is not only<br />

the VSG that needs to be exchanged on the trypanosome<br />

surface. Several ESAG-encoded proteins<br />

are predicted to be present on the cell<br />

surface (Table 5.1). Perhaps these proteins have<br />

specific functions that make them less tolerant<br />

of variation, or perhaps they are less exposed to<br />

the immune response, so the existence of only a<br />

few variants, encoded in different ESs, is sufficient<br />

for their survival and function. This may<br />

be fortunate because the existence of two large<br />

families of variant proteins, whose switching<br />

had to be coordinated by simultaneous recombination<br />

events, would pose a major problem<br />

for any cell. The function of only two ESAGs is<br />

known: the proteins encoded by ESAG6 and<br />

ESAG7 form a heterodimeric receptor for <strong>trans</strong>ferrin,<br />

whose uptake is essential for the survival<br />

of bloodstream trypanosomes. Several studies<br />

have shown that the <strong>trans</strong>ferrin receptors<br />

encoded by different ESs have different affinities<br />

for <strong>trans</strong>ferrins from different host species.<br />

It has been suggested that this variation in<br />

<strong>trans</strong>ferrin receptor affinity, perhaps coupled<br />

to other receptor interactions, could allow<br />

T. brucei to optimize its growth in different hosts,<br />

and this would determine which ES would be<br />

favored in any particular host. Although this is<br />

an interesting hypothesis, it remains a hypothesis.<br />

One interesting point about the <strong>trans</strong>ferrin<br />

receptor is that it is encoded by the most promoter-proximal<br />

ESAGs, and these are <strong>trans</strong>cribed<br />

simultaneously at a low level from many<br />

MOLECULAR BIOLOGY

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!