Está en la página 1de 2

11. Dostlov, A. and Volf, P.

(2012) Leishmania development


in sand ies: parasite-vector interactions overview. Parasit. Social
Vectors 5, 276
molity? Dierenaon?
12. Del Vicario, M. et al. (2016) The spreading of misinformation
online. PNAS 113, 554559
13. Rogers, M. (2012) The role of Leishmania proteophospho-
glycans in sand y transmission and infection of the mam-
malian host. Front. Microbiol. 3, 223
14. Atayde, V. et al. (2015) Exosome secretion by the parasitic
protozoan Leishmania within the sand y midgut. Cell Rep.
13, 957967

Spotlight
Vesicles as Vehicles
for Virulence
Monica R. Mugnier,1,2,*
F. Nina Papavasiliou,1,* and
Danae Schulz1,3,*
Immune
Parasites have long been known modulaon?
to inuence host responses
to infection through the secretion
of virulence factors. Extracellular
vesicles are emerging as impor-
tant mediators of these mani-
pulations, and a new study by
Szempruch et al. suggests they Figure 1. Extracellular Vesicles Could Mediate a Variety of Processes in Trypanosoma brucei
could play a crucial role in host Infection. Szempruch et al. demonstrate that Trypanosoma brucei-generated extracellular vesicles (EVs) have
the ability to transfer protein to other parasites and to host cells [3]. This could be important for modulating the
responses to African trypano- immune response to parasite infection both in the mammal and the y. EVs could also be the mechanism by
some infections. which repellants or other factors important for social motility are transferred between parasites in the y. Finally,
EVs could play a role in quorum sensing that mediates density-dependent differentiation from long slender to
Extracellular vesicles (EVs) are produced stumpy forms within the mammal.

by nearly every cell type, including most


parasites [1]. The transfer of their con-
tents, whether protein, lipid, or nucleic upon the ndings in T. b. gambiense by If functional, these structures might medi-
acid, from one cell to the next can alter identifying parasite membrane nano- ate cell-to-cell communication.
the functioning of the recipient cell, making tubes, which had been described in
these vesicles crucial mediators of cell T. brucei in the 1960s and referred to as One of the most intriguing aspects of EVs
signaling with huge potential as therapeu- plasmanemes at the time [4]. The role of in the context of infectious disease is their
tic and diagnostic tools (Figure 1). EVs plasmanemes was unclear then, but potential to fuse with and alter the function
were recently reported in African trypano- Szempruch and colleagues suggest that of host cells. Szempruch et al. show that,
somes in Trypanosoma brucei gam- these nanotubes/plasmanemes may be indeed, parasite EVs fuse with host eryth-
biense, a form of T. brucei responsible the origin of EVs in African trypanosomes. rocytes, transferring parasite variant sur-
for chronic African sleeping sickness in They microscopically observe the forma- face glycoproteins (VSGs). The transfer of
humans [2]. A new study by Szempruch tion of EVs during the dissociation of VSGs to host erythrocytes has been pre-
et al. identies EVs in an animal infective membrane nanotubes, although it is not viously observed [5], but the mechanism
form of the parasite, Trypanosoma brucei clear if this is the exclusive mechanism of behind it and its functional relevance
brucei [3], which appear very similar to EV formation in T. brucei. Interestingly, remains a mystery. Szempruch et al. dem-
those described in T. b. gambiense, at membrane nanotubes appear to tran- onstrate that fusion of EVs with host eryth-
least morphologically. The authors extend siently interact with neighboring parasites. rocytes affects erythrocyte rigidity and size

Trends in Parasitology, June 2016, Vol. 32, No. 6 435


and postulate that this results in preferen- for receptor-mediated internalization. parasitemia within the mammal could lead
tial clearance by host myeloid cells [6], This raises interesting questions as to to increased nanotube formation, followed
although this is not tested directly. Con- whether co-infections with T. b. brucei by delivery of SIF via EVs, culminating in
sistent with this model, the intravenous and T. b. rhodesiense could lead to differentiation from long slender to short
injection of parasite EVs into nave mice enhanced ability of T. b. brucei to survive stumpy parasites that are preadapted for
results in a 510% decrease in the number within a primate reservoir. survival within the y midgut. The involve-
of host erythrocytes within 1 h, demon- ment of EVs as carriers of signals for differ-
strating a potential role for parasite EVs In all scenarios tested by Szempruch and entiation to stumpy forms or for social
in infection-associated anemia. The trans- colleagues, the EV-mediated interaction motility is now an open and exciting area
fer of VSGs from the parasite surface to between trypanosomes, or between try- of inquiry.
the recipient erythrocyte raises the possi- panosomes and host cells, leads to the
bility that erythrocytes could become tar- transfer of proteins. Thus, the question This work ties together the mysterious
gets of antibody responses directed remains open as to whether EVs also carry observations of plasmanemes and para-
against parasite VSGs, which might also DNA or RNA cargo, the delivery of which site-to-host transfer of VSGs, while at the
be expected to cause increased erythro- could effectively reprogram the recipient same time suggesting new mechanisms for
cyte clearance, although Stijlemans et al. cell, as happens with Plasmodium falcipa- parasite communication. With these nd-
show preferential clearance of trypano- rum [1]. This type of reprogramming could ings, T. brucei joins the ranks of the numer-
some-altered erythrocytes even in unin- lead to immune modulation in the host, ous parasites that use these unique
fected mice [6]. It remains to be seen which has been demonstrated in a num- structures as mediators of cell-to-cell com-
whether T. brucei EVs fuse with other host ber of other parasitic systems [1]. munication, both between parasites and
cell types, and what, if any, modulatory with their intimately associated hosts [1].
effect they have. Additionally, it remains to be seen whether 1
Laboratory of Lymphocyte Biology, The Rockefeller
T. brucei EVs contain cargo that mediate University, 1230 York Avenue, New York, NY 10065, USA
In addition to the interaction between EVs communication or quorum sensing. While 2The Rockefeller Graduate Program, The Rockefeller
and erythrocytes, Szempruch and col- trypanosomes are single-celled eukar- 3University, 1230 York Avenue, New York, NY 10065, USA
Current address: Department of Biology, Harvey Mudd
leagues demonstrated that African try- yotes, their behavior within their insect College, 301 Platt Bvd, Claremont, CA 91711, USA
panosome EVs were able to transfer and mammalian hosts suggests that they
virulence factors from one parasite to communicate with one another, and the *Correspondence: mmugnier@rockefeller.edu
(M.R. Mugnier), papavasiliou@rockefeller.edu
another. One such factor is the serum means by which this occurs has been a (F.N. Papavasiliou), dschulz@rockefeller.edu (D. Schulz).
resistance-associated (SRA) gene prod- long-standing mystery in the eld. In the http://dx.doi.org/10.1016/j.pt.2016.03.001
uct, which allows Trypanosoma brucei insect form, T. brucei exhibits social motil-
rhodesiense to escape lysis by blocking ity, which has been shown to be important References
1. Coakley, G. et al. (2015) Exosomes and other extracellular
the action of the trypanosome lytic for colonization of the y midgut [8]. Colo- vesicles: the new communicators in parasite infections.
Trends Parasitol. 31, 477489
factor (TLF) found in human blood [7]. nies of migrating parasites repel one
2. Geiger, A. et al. (2010) Exocytosis and protein secretion in
T. b. brucei lacks the SRA gene and is another, but the nature and function of Trypanosoma. BMC Microbiol. 10, 20
thus usually susceptible to lysis by TLF. In the repellants are unknown. Within the 3. Szempruch, A.J. et al. (2016) Extracellular vesicles from
Trypanosoma brucei mediate virulence factor transfer and
an elegant series of experiments, the mammal, T. brucei appears to possess cause host anemia. Cell 164, 246257
authors demonstrated that co-culturing a quorum-sensing mechanism that results 4. Vickerman, K. and Luckins, A.G. (1969) Localization of vari-
T. b. brucei in transwells with either in density-dependent differentiation from able antigens in the surface coat of Trypanosoma brucei
using ferritin conjugated antibody. Nature 224, 11251126
T. b. rhodesiense or with T. b. brucei long slender to short stumpy forms that 5. Rifkin, M.R. and Landsberger, F.R. (1990) Trypanosome
carrying an SRA transgene conferred resis- are transmitted to the y midgut, but variant surface glycoprotein transfer to target membranes:
a model for the pathogenesis of trypanosomiasis. Proc. Natl.
tance to killing by TLF. They went on to the mysterious stumpy induction factor Acad. Sci. U.S.A. 87, 801805
show that resistance to TLF could be con- (SIF) has yet to be identied [9]. Addition- 6. Stijlemans, B. et al. (2015) Development of a pHrodo-based
ferred through the addition of EVs alone, ally, the authors note that stress from assay for the assessment of in vitro and in vivo erythropha-
gocytosis during experimental trypanosomosis. PLoS Negl.
provided that they were harvested from RNAi-mediated depletion of an essential Trop. Dis. 9, e0003561
either SRA transgenic T. b. brucei or from gene or from complement active serum 7. Capewell, P. et al. (2015) A co-evolutionary arms race:
trypanosomes shaping the human genome, humans shap-
T. b. rhodesiense. The authors also dem- increases the frequency of nanotube for- ing the trypanosome genome. Parasitology 142, S108S119
onstrated that SRA and other vesicular mation. Assuming that nanotubes do 8. Imhof, S. and Roditi, I. (2015) The social life of African
membrane proteins from EVs enter the result in an increase in the number of trypanosomes. Trends Parasitol. 31, 490498

endocytic pathway at the agellar pocket Mony, B.M. and Matthews, K.R. (2015) Assembling the
EVs, one could imagine a scenario where 9. components of the quorum sensing pathway in African try-
of the recipient parasite, with no evidence generalized stress induced by high levels of panosomes. Mol. Microbiol. 96, 220232

436 Trends in Parasitology, June 2016, Vol. 32, No. 6

También podría gustarte