ArticlePDF AvailableLiterature Review

Membrane trafficking and organelle biogenesis in Giardia lamblia: Use it or lose it

Authors:

Abstract and Figures

The secretory transport capacity of Giardia trophozoites is perfectly adapted to the changing environment in the small intestine of the host and is able to deploy essential protective surface coats as well as molecules which act on epithelia. These lumen-dwelling parasites take up nutrients by bulk endocytosis through peripheral vesicles or by receptor-mediated transport. The environmentally-resistant cyst form is quiescent but poised for activation following stomach passage. Its versatility and fidelity notwithstanding, the giardial trafficking systems appear to be the product of a general secondary reduction process geared towards minimization of all components and machineries identified to date. Since membrane transport is directly linked to organelle biogenesis and maintenance, less complexity also means loss of organelle structures and functions. A case in point is the Golgi apparatus which is missing as a steady-state organelle system. Only a few basic Golgi functions have been experimentally demonstrated in trophozoites undergoing encystation. Similarly, mitochondrial remnants have reached a terminally minimized state and appear to be functionally restricted to essential iron-sulfur protein maturation processes. Giardia's minimized organization combined with its genetic tractability provides unique opportunities to study basic principles of secretory transport in an uncluttered cellular environment. Not surprisingly, Giardia is gaining increasing attention as a model for the investigation of gene regulation, organelle biogenesis, and export of simple but highly protective cell wall biopolymers, a hallmark of all perorally transmitted protozoan and metazoan parasites.
Content may be subject to copyright.
Invited Review
Membrane trafficking and organelle biogenesis in Giardia lamblia: Use it or lose it
Carmen Faso, Adrian B. Hehl
Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
article info
Article history:
Received 1 December 2010
Received in revised form 23 December 2010
Accepted 24 December 2010
Available online 4 February 2011
Keywords:
Biopolymer
Encystation
Peripheral vesicle
Mitosome
Antigenic variation
Reductive evolution
Golgi
abstract
The secretory transport capacity of Giardia trophozoites is perfectly adapted to the changing environment
in the small intestine of the host and is able to deploy essential protective surface coats as well as mol-
ecules which act on epithelia. These lumen-dwelling parasites take up nutrients by bulk endocytosis
through peripheral vesicles or by receptor-mediated transport. The environmentally-resistant cyst form
is quiescent but poised for activation following stomach passage. Its versatility and fidelity notwithstand-
ing, the giardial trafficking systems appear to be the product of a general secondary reduction process
geared towards minimization of all components and machineries identified to date. Since membrane
transport is directly linked to organelle biogenesis and maintenance, less complexity also means loss
of organelle structures and functions. A case in point is the Golgi apparatus which is missing as a
steady-state organelle system. Only a few basic Golgi functions have been experimentally demonstrated
in trophozoites undergoing encystation. Similarly, mitochondrial remnants have reached a terminally
minimized state and appear to be functionally restricted to essential iron–sulfur protein maturation pro-
cesses. Giardia’s minimized organization combined with its genetic tractability provides unique opportu-
nities to study basic principles of secretory transport in an uncluttered cellular environment. Not
surprisingly, Giardia is gaining increasing attention as a model for the investigation of gene regulation,
organelle biogenesis, and export of simple but highly protective cell wall biopolymers, a hallmark of
all perorally transmitted protozoan and metazoan parasites.
Ó2011 Australian Society for Parasitology Inc. Published by Elsevier Ltd. All rights reserved.
1. Introduction
Infection with Giardia lamblia (syn. Giardia intestinalis,Giardia
duodenalis) is one of the major causes of diarrheal disease with
an estimated 250 million clinical cases worldwide (WHO, 1997).
As an intestinal lumen-dwelling parasite which can transform into
an environmentally-resistant cyst form, Giardia interfaces with its
environments via secreted proteins and carbohydrates. The most
striking result of secretory activity is a prominent cyst wall, which
is deposited on the surface of encysting cells and whose descrip-
tion dates back to Grassi (1879). The discovery of the variant-spe-
cific surface protein (VSP) coat and antigenic variation pointed to
additional major protein trafficking pathways to the plasma mem-
brane of trophozoites. In addition to classical exocytosis of mem-
brane proteins, there is also evidence for non-conventional
secretion of metabolic enzymes and factors which modulate the re-
sponse of the gut epithelium to infection.
Endocytic uptake and transport of nutrients from the complex
environments in both the natural habitat and in cell culture is
not as well understood. In contrast to the other Diplomonads, the
Giardiinae lack a cytostome organelle with which to handle the
bulk of fluid phase transport. Instead, a unique organellar system
consisting of peripheral vesicles (PVs) which is arrayed just below
the plasma membrane appears to provide an all-in-one solution to
nutrient uptake, digestion and retrograde transport of building
blocks to the interior of the cell (Lanfredi-Rangel et al., 1998). Dur-
ing the past two decades, several research groups have investi-
gated intracellular protein trafficking in greater detail using
rigorous and systematic approaches. This work was initiated by a
report on the transport of cyst wall material (CWM) in encysta-
tion-specific vesicles in differentiating trophozoites (Reiner et al.,
1990). It became clear that despite an intracellular organization
with strongly reduced complexity, Giardia trophozoites have an
efficient membrane trafficking system capable of directing numer-
ous cargo proteins to their correct destination along distinct trans-
port pathways (Marti et al., 2003a). This capability for constitutive
and stage-regulated secretion contrasts with the apparent simplic-
ity of the machinery involved and its molecular underpinnings:
although many conserved elements have been identified, global
analysis of the Giardia genome sequence revealed a significant
and consistent reduction of key components on every level
(Morrison et al., 2007). Most striking is the absence of certain
compartments or even entire organelles such as a Golgi apparatus
with biochemically defined cisternae and an endosomal system
that intersects with it, as well as key protein components such as
0020-7519/$36.00 Ó2011 Australian Society for Parasitology Inc. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.ijpara.2010.12.014
Corresponding author. Tel.: +41 44 635 8526; fax: +41 44 635 89 07.
E-mail address: adrian.hehl@access.uzh.ch (A.B. Hehl).
International Journal for Parasitology 41 (2011) 471–480
Contents lists available at ScienceDirect
International Journal for Parasitology
journal homepage: www.elsevier.com/locate/ijpara
vesicle tethering factors. Combined with the induction of a prom-
inent secretory pathway in encysting cells, this makes Giardia a un-
ique biological model to investigate minimal solutions for
membrane transport.
2. Molecular machineries for trafficking
The description of the giardial membrane-bounded organellar
system relies both on morphology (Lindmark, 1988; Reiner et al.,
1989; McCaffery and Gillin, 1994; Lanfredi-Rangel et al., 1998;
Benchimol, 2004) and detection of common marker proteins
(Reiner et al., 1989; Marti et al., 2003b; Tovar et al., 2003; Touz
et al., 2004; Hernandez et al., 2007; Elias et al., 2008). The use of
heterologous antibodies to detect markers is problematic, how-
ever, due to the significant divergence of many giardial proteins.
This usually requires generation of specific antibodies against the
giardial homologs (Marti et al., 2003b; Elias et al., 2008). Although
the fine structures of the endomembrane system are yet to be de-
fined, the combined data reveals highly simplified organizational
principles on the one hand and highly specialized features on the
other. At morphological and molecular levels only three clearly
identifiable organellar systems have been delineated in trophozo-
ites: an extensive endoplasmic reticulum (ER) which is continuous
with the nuclear envelopes as in other eukaryotes, the PVs and
mitochondrial remnant organelles (mitosomes). An additional set
of prominent organelles, the encystation-specific vesicles (ESVs),
is generated only in encysting trophozoites and disappears again
upon secretion of CWM. Fig. 1 shows a surface-rendered composite
model of four identifiable organellar systems generated from a
three-dimensional reconstruction of a confocal image stack.
Several trafficking pathways to and from these organelles have
been identified and some are characterized in more detail. Current
evidence suggests that the ER (together with ESVs in differentiat-
ing cells) comprises the entire secretory system in Giardia.
Similarly, PVs are the only identifiable organelles involved in endo-
cytic transport. This is in line with the low number of conserved
members of key protein families involved in membrane transport
(see also below).
There is a growing body of evidence indicating a secondary loss
of major compartments and functions associated with membrane
transport in Giardia. However, the reason for this dramatic reduc-
tion is unknown. Nevertheless, Giardia still possesses core machin-
ery for membrane transport with the three well-known ubiquitous
eukaryotic coat complexes (COPII, coatomer (COPI), clathrin) and
two adaptor protein (AP) complexes. Interestingly, COPI and APs
are normally associated with the Golgi apparatus. All but one sub-
unit of the heteroheptameric COPI complex can be identified in the
Giardia genome. In addition, six small GTPases of the Arf and
Arf-like family of small GTPases which are involved in COPI recruit-
ment to membranes (Murtagh et al., 1992; Marti et al., 2003b)
were detected. At least one of the two giardial Arf1 homologs is
sufficiently conserved to rescue a lethal yeast Arf1/2 double mu-
tant (Lee et al., 1992). Giardia has only two complete hetero-tetra-
meric AP complexes corresponding to an AP1 and an AP2/3
equivalent; G. lamblia adaptor protein complex (AP1) is involved
in secretory transport (Touz et al., 2004), whereas the second com-
plex localizes to PVs and functions in endocytosis (Rivero et al.,
2010). The last common eukaryotic ancestor (LCEA) likely
contained three or four AP complexes which evolved early from a
proto F–COP–AP complex by coordinated gene duplication of their
subunits (Schledzewski et al., 1999; Boehm and Bonifacino, 2001;
Marti et al., 2003b) The presence of only two complexes in Giardia
again supports the secondary loss hypothesis. A giardial clathrin
Fig. 1. Surface-rendered composite model showing the compartment distribution and the morphology of the four Giardia organelles. The images were generated from a
three-dimensional reconstruction (volume image) of a confocal image stack using the Imaris software suite (Bitplane, Switzerland). The trophozoite is viewed from the
ventral side. Mitosomes or encystation-specific vesicles (ESVs) are shown together with the nuclei and the endoplasmic reticulum (ER) in A and D, respectively. (B) The
localization of clathrin. Note that clathrin is distributed around peripheral vesicle (PV) organelles and does not label the PVs directly. (C) A combined image with all four
organelle systems. The orientation of the model is indicated at the center of the image; the arrows point to the anterior (up), lateral (left) and dorsal (right) side of the cell.
Mitosome lumen marker: IscU (Giardia Genome Database GL50803_15196); ER membrane marker: protein disulfide isomerase 2 (PDI2) (GL50803_9413); PV region: clathrin
heavy chain (GL50803_9413); ESV lumen marker: cyst wall protein 1 (CWP1) (GL50803_5638); nuclear DNA: DAPI.
472 C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480
heavy chain localizes to the PV region (Fig. 1) and to membranes of
maturing ESVs (Marti et al., 2003b; Gaechter et al., 2008) but a
light chain homolog was not identified. Whether this is the reason
for the lack of clathrin-coated transport intermediates and mem-
brane buds requires further investigation of clathrin coat
recruitment.
Rab (Ras-related in brain) GTPases, SNAREs (soluble N-ethyl-
malemide-sensitive factor attachment protein receptors), and their
effectors are universally conserved molecular machineries which
regulate and promote membrane fusion events and define com-
partment identities by conferring specificity. Due to the specific
interaction with membranes of organelles, their diversity is a good
indicator of the level of compartment complexity in a cell. Organi-
zation of the giardial trafficking pathways and compartment struc-
ture appears to require only seven Rab proteins (Marti et al.,
2003b) in contrast to the large Rab complements of other basal
protozoa such as Trypanosoma brucei (16 Rabs) (Ackers et al.,
2005), or the vastly expanded Rab families in Trichomonas vaginalis
(65 members) (Lal et al., 2005) and Entamoeba histolytica (>90
Rabs) (Saito-Nakano et al., 2005). Rabs consist mostly of a GTPase
domain and are functionally defined by a 30 amino acid domain
at their C-terminus, which includes prenylation sites. Thus, se-
quence constraints are high, which makes it likely that all giardial
Rabs have been identified. Three predicted exocytic (Rab1, 2a/
b)(Langford et al., 2002; Marti et al., 2003b) and one predicted
(recycling) Rab11 homolog (Castillo-Romero et al., 2010) are well
conserved, and cluster robustly with orthologues from other
eukaryotes in phylogenetic analyses. The remaining Rab family
members (F, D, and 32) cannot be assigned to specific Rab sub-
groups (Marti et al., 2003b; Morrison et al., 2007).
Consistent with experimental data indicating a simple compart-
ment structure with few trafficking pathways (Hehl and Marti,
2004), a small number of factors which mediate vesicle docking
and membrane fusion were identified. Members of the SNARE fam-
ily of proteins are highly divergent and therefore more difficult to
classify with confidence. Using BLAST search algorithms only seven
SNARE proteins (Dacks and Doolittle, 2002, 2004; Marti et al.,
2003b; Morrison et al., 2007) were initially identified in the com-
pleted genome. Three putative vesicle-(v-)SNAREs, and four syn-
taxin (target-(t-)SNARE) homologs were identified. Application of
more sensitive methods using Hidden Markov Model (HMM) pro-
files identified 15 (Kloepper et al., 2007) and 17 (Elias et al., 2008)
candidate SNARE family members, respectively. These studies pro-
vide excellent starting points for a detailed analysis of SNARE
function.
Long-range capture of transport vesicles before SNARE-driven
membrane fusion is mediated by large coiled-coil tethering factors
of the golgin family or generally conserved tethering complexes
such as GARP (Golgi-associated retrograde protein), COG (con-
served oligomeric Golgi) and exocyst. Because no Golgi stacks were
identified in Giardia it is not surprising that golgin family proteins
and other Golgi matrix proteins, which act as organizers of this
organelle, cannot be found in the genome database (Marti et al.,
2003b; Morrison et al., 2007). Exceptions are the TRAPPI (transport
protein particle) and the HOPS (homotypic fusion and vacuolar
protein sorting) complexes (Koumandou et al., 2007). Overall, de-
spite the conservation of several key factors and complexes, the
genomic analysis of the machinery mediating intracellular trans-
port shows consistent minimization.
3. Organelles and pathways for secretory transport
Secretory transport plays an essential role in Giardia: every life
cycle stage with the exception of the cyst outside of the host is
known to secrete specific soluble and membrane-bound proteins.
Accumulation of CWM in ESVs, and export beyond the plasma
membrane is the most conspicuous manifestation of secretory
transport in this parasite. Yet, maintenance of the protective VSP
coat alone likely requires a significant investment in secretory re-
sources. Export of both CWM and VSPs occurs simultaneously in
encysting cells, most likely until secretion of CWM. Thereafter,
VSPs are removed from the plasma membrane (McCaffery et al.,
1994; Marti et al., 2003a). These processes require distinct secre-
tory pathways, which do not intersect beyond the ER and are sub-
ject to stage regulation (Marti et al., 2003a). Thus, differentiating
parasites are forced to considerably increase their synthetic capac-
ity. One consequence is the de novo establishment of ESVs, an
additional set of secretory organelles dedicated to maturation
and export of the CWM.
3.1. Endoplasmic reticulum
The giardial ER can be visualized by immunofluorescence
microscopy using antibodies against resident proteins such as heat
shock protein 70/binding immunoglobulin protein (Hsp70/BiP) or
one of the five protein disulfide isomerases (PDIs) (Knodler et al.,
1999; Stefanic et al., 2006; Abodeely et al., 2009). The organelle ex-
tends from the nuclear envelopes through the entire cell body and
has an intricate, bilaterally symmetrical, structure. The very exis-
tence of this archetypical eukaryotic organelle in Giardia had been
called into question (Feely et al., 1990; Meyer, 1994), although ER
membranes had been identified by electron microscopy (Reiner
et al., 1990; McCaffery and Gillin, 1994). Definitive identification
of the giardial ER was achieved using antibodies raised against
the conserved giardial Hsp70/BiP homolog (Gupta et al., 1994;
Lujan et al., 1996b; Soltys et al., 1996). Confocal microscopy with
ER-Tracker™ and electron microscope (EM) tomography in living
trophozoites confirmed the general tubular nature and sub-cellular
distribution of the ER organelle (Abodeely et al., 2009). ER mem-
branes are found throughout the cytoplasm but do not permeate
the space occupied by PVs (Fig. 1). However, EM studies identified
apparent contact points and continuities between the two orga-
nelle systems. This is the basis for a recently postulated endocytic
transport hypothesis (Abodeely et al., 2009).
3.2. Glycosylation and ER quality control
While it appears that Giardia cells possess a conventional ER
with respect to secretory trafficking, some elements of the post-
translational modification machinery are missing entirely. Most
prominently, no calnexin–calreticulin machinery for quality
control of N-glycosylated secreted proteins has been identified
(Samuelson et al., 2005; Banerjee et al., 2007, 2008). This fits with
the absence of conventional GlcNAc
2
Man
9
Glc
3
core glycans linked
to asparagine (Asn) residues of proteins secreted from the ER
(Samuelson et al., 2005). Although complex branched glycans were
postulated (Morelle et al., 2005), rigorous genomic and biochemi-
cal analyses refuted this and revealed that Giardia misses several
nucleotide sugar transporters (Banerjee et al., 2008). In addition,
only one (i.e. Alg7) of the common 12 glycosyltransferases required
for synthesis of the dolichol-PP-GlcNAc
2
Man
9
Glc
3
precursor in the
ER (Samuelson et al., 2005) has been described. Thus, Asn-linked
glycosylation in the giardial ER is limited to the addition of
GlcNAc
1–2
to proteins. Nevertheless, a recent analysis revealed a
sizable N-glycome in Giardia with distinct aspects of stage regula-
tion (Ratner et al., 2008). Co-translational import and folding of se-
creted proteins is supported by a conserved machinery for
translocation (Svard et al., 1999), chaperones, and the five mem-
bers of the PDI family of proteins (Knodler et al., 1999; Morrison
et al., 2007). The latter are unusual in that they have a single thio-
redoxin domain instead of the normal two or three in PDIs of other
C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480 473
eukaryotes. This domain structure appears to be a primary basic
feature and not the product of secondary reduction (Knodler
et al., 1999). Giardial PDIs most likely play a major role in assisting
the folding of the cysteine-rich VSPs, the high cysteine membrane
proteins (HCMPs) (Davids et al., 2006b) and cyst wall proteins
(CWPs).
3.3. Golgi-like organelles and Golgi functions
The earliest electron microscopic examination of cysts and tro-
phozoites revealed a conspicuous absence of organelles, specifi-
cally ER, Golgi and mitochondria (Sheffield and Bjorvat, 1977).
Later claims that a Golgi organelle existed (Gillin et al., 1991; Lujan
et al., 1995a; Lanfredi-Rangel et al., 1999; Dacks et al., 2003) re-
main unconfirmed. The classical Golgi is defined by a series of bio-
chemically distinguishable, dynamic, steady-state compartments
in which most or all secreted proteins are delayed for post-transla-
tional maturation before being sorted and transported to their final
destinations. Whether Golgi cisternae are arranged as easily
detectable ordered stacks with defined cis (receiving) and trans
(exporting) polarities is irrelevant since there are numerous exam-
ples of perfectly functional, delocalized Golgi systems. No known
Giardia organelle which meets these criteria has been identified
to date. Importantly, no classical markers for the Golgi such as
GM130, galactosyl transferases or the trans-Golgi network marker
Rab6 are present in Giardia. This constitutes a strong argument for
the loss of a stacked Golgi which was present in the LCEA (Dacks
and Field, 2007). Nevertheless, post-ER trafficking compartments
in Giardia exist in the form of ESVs of encysting trophozoites which
have many hallmarks of Golgi cisternae but also present clear dif-
ferences (see below). Components of the Golgi-associated COPI
were detected on ESV membranes (Stefanic et al., 2006). However,
antibodies against a giardial COPI subunit also label punctate
structures in trophozoites (Marti et al., 2003b). Indirect evidence
for the existence of post-ER trafficking compartments also derives
from the identification of a putative KDEL-receptor (Lys-Asp-Glu-
Leu) Erd2 homolog (GiardiaDB GL50803_4502) in the Giardia
genome. This protein is likely involved in retrieval of ER resident
proteins such as the chaperone Hsp70/BiP from distal compart-
ments (Stefanic et al., 2006).
4. Secretory transport during growth and differentiation
4.1. Secretion in trophozoites
The transmembrane-anchored VSPs of trophozoites are the ma-
jor surface proteins (Adam et al., 1988). VSP exodomains are re-
leased after cleavage in the conserved C-terminal domain
(Papanastasiou et al., 1996) and become soluble antigens. This re-
quires a constant turnover of VSPs at the surface, independent of
antigenic variation. Thus, synthesis and export of VSPs as well as
additional cysteine-rich, non-variable proteins (Davids et al.,
2006b) targeted to the plasma membrane likely constitute a major
part of the secretory activity of trophozoites. In addition to the
approximately 200 VSPs, more than 500 proteins with a signal se-
quence are currently predicted in the Giardia Genome Database
(www.giardiadb.org). An unknown number of these factors are ex-
ported to the surface or released into the environment by mem-
brane transport. Because Giardia lacks Golgi cisternae and delay
of secreted proteins in post-ER compartments during export has
not been observed (Marti et al., 2003a), secreted proteins are traf-
ficked directly to their final destination after leaving the ER. Cur-
rent data support direct transport of VSPs and likely of other
constitutively secreted soluble and membrane proteins from the
ER to the plasma membrane (PM) (Marti et al., 2003a; Touz
et al., 2003; Hehl and Marti, 2004). Interestingly, export of VSPs
is sensitive to brefeldin A (Lujan et al., 1995a), a fungal metabolite
which inhibits Arf1-dependent recruitment of COPI to Golgi mem-
branes and results in fragmentation of the Golgi organelle in higher
eukaryotes. This observation suggested that VSP transport is
dependent on COPI coat protein complexes, although no corre-
sponding post-ER compartment through which these proteins
transit was identified.
Few targeting signals have been characterized in secreted pro-
teins of trophozoites: short C-terminal sequences directing VSPs
to the PM and an encystation-specific protease (ESCP), (Touz
et al., 2002b) to PVs, respectively. The C-terminal domain of all
VSPs includes an invariable cytoplasmic tail CRGKA (Cys-Arg-Gly-
Lys-Ala), a hydrophobic transmembrane sequence, and a short,
well-conserved region of exodomain. The short cytoplasmic tail
is post-translationally modified by palmitoylation (Papanastasiou
et al., 1997; Touz et al., 2005) and by citrullination of the arginine
residue (Touz et al., 2008). The VSP C-terminal domain is necessary
and sufficient for correct secretion of a heterologous Toxoplasma
gondii SAG1 surface antigen exodomain (Kasper et al., 1984; Tom-
avo et al., 1992) used as a reporter in stably transfected trophozo-
ites (Marti et al., 2002). Removal of the CRGKA cytoplasmic tail
alone or of the entire C-terminal domain resulted in accumulation
of this reporter in the ER (Marti et al., 2003a), suggesting that this
sequence has an essential function in VSP targeting. On the other
hand, a different study showed that an intact VSP transmembrane
and exodomain is sufficient for trafficking to the PM in transgenic
cells (Touz et al., 2003). However, the possibility that the reporter
is co-exported by interaction with endogenous secreted VSPs in
these transgenic cells has not been tested.
ESCP is synthesized as a membrane-anchored pro-protein and
targeted to lysosomes in trophozoites (Touz et al., 2002b, 2003,
2004) by a conserved YXX
U
-type targeting signal YRPI (Tyr-Arg-
Pro-Ile) in its short cytoplasmic tail. Targeting of chimeric reporters
with a ESCP-derived transmembrane domain TM and cytoplasmic
tail and a H7 VSP exodomain to PVs was dependent on this motif
(Touz et al., 2003) and the AP complex 1(Touz et al., 2004). A Gol-
gi-like sorting function has been invoked for ESCP secretion; signif-
icant amounts of epitope-tagged ESCP on the cell surface (Touz
et al., 2003) could also be explained with YXX
U
-mediated retrieval
of ESCP from the PM.
The establishment of axenic culture systems (Keister, 1983) was
a prerequisite for the systematic identification of soluble secretory
products. In vitro (Katelaris et al., 1994; Roxstrom-Lindquist et al.,
2005; Muller et al., 2007) and rodent models (Davids et al., 2006a;
Li et al., 2007) for study of the effects of secreted substances on the
host are also well established. Research on secretion in trophozo-
ites focused on identifying virulence factors, i.e. products which
cause pathology in the host intestine or which interfere with
immunological reactions to giardial infections. No evidence for
the production of secreted toxins was found (Smith et al., 1982),
but a number of secreted proteins have been identified which
could modulate the physiology of host cells on different levels.
Unidentified secreted proteins were implicated in degradation of
barrier function and induction of apoptosis in epithelial cells after
co-cultivation with Giardia (Jimenez et al., 2000; Teoh et al., 2000;
Chin et al., 2002; Rodriguez-Fuentes et al., 2006; Panaro et al.,
2007; Troeger et al., 2007; de Carvalho et al., 2008). Other secretory
products affected the uptake of glucose and phenylalanine in the
intestine of mice (Samra et al., 1988).
Secreted (glyco)proteins appear to modulate certain aspects of
the intestinal immune response and may play a role in trophozoite
survival (Jimenez et al., 2007) although these claims require further
substantiation. In particular, an effect by contamination of serum
glycoproteins which tend to adhere to the trophozoite surface will
have to be excluded. In addition, substantial amounts of soluble VSP
474 C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480
exodomains are released after cleavage in the conserved domain at
the base of the transmembrane anchor (Papanastasiou et al., 1996).
The effect of secreted VSPs, however, is unclear.
In addition to protein secretion via ER and membrane transport,
there are robust data suggesting an important role for non-conven-
tional protein export. Recently it was shown that direct contact be-
tween cultured intestinal epithelial cells and Giardia elicits distinct
changes in chemokine expression (Roxstrom-Lindquist et al.,
2005). Metabolic enzymes secreted by trophozoites (Ringqvist
et al., 2008) are able to alter the microenvironment in co-culture
assays. An example is the proposed depletion of arginine at the
host–parasite interface, resulting in suppression of nitric oxide
(NO) production (Eckmann et al., 2000). The pathway through
which the metabolic enzymes arginine deiminase, ornithine car-
bamoyl transferase and enolase are secreted upon contact with
epithelial cells in vitro is unknown (Ringqvist et al., 2008). These
exported effectors and immunogens (Palm et al., 2003) are cyto-
plasmic proteins, contain no N-terminal signal peptide and are
therefore not detected in vesicular transport pathways. Whether
export occurs through direct unconventional routes or if microve-
sicles such as exosomes (Silverman and Reiner, 2011) are involved
as shown for several prokaryotes and eukaryotes, is an interesting
question requiring further investigation.
Taken together there is solid support for Giardia interference
with epithelial function, presumably via soluble or membrane-
bound secreted proteins. However, these factors and their precise
interaction(s) with host cells await characterization.
4.2. Secretion in encysting cells
The most striking secretion process in the giardial life cycle oc-
curs during stage-differentiation of trophozoites. Secretion of the
CWM is the only known regulated export pathway. In fixed cul-
tured cells neogenesis of ESVs followed by deposition of this extra-
cellular matrix on the surface of differentiated cells is observed by
IFA (Gillin et al., 1987). Several protocols for in vitro encystation
are currently used in laboratories. The two-step method (bile
deprivation, followed by supplementation with porcine bile and
an increase of the pH to 7.85) (Boucher and Gillin, 1990) is the
most popular, but alternative methods such as cholesterol depriva-
tion (Lujan et al., 1996a) or incubation with high concentrations of
bile extract in the medium (Kane et al., 1991) are also very
effective.
Encysting trophozoites can be followed from induction to the
formation of the cyst wall. Although the time from induction to
cyst formation varies considerably between laboratories, this pro-
cess takes at least 14–20 h. Co-translational insertion of the struc-
tural proteins of the cyst wall, CWP1–3 (Lujan et al., 1995b;
Mowatt et al., 1995), into the ER takes place during the first 7h
post-induction (p.i.). The three CWP family members are soluble
proteins with a distinct structure, comprising a central domain
with several leucine-rich repeats (Lujan et al., 1995b). The CWM
biopolymer has a surprisingly low complexity considering how
effectively it acts as a biological barrier on the surface of the cyst.
The three CWPs are paralogous and constitute approximately 40%
of the cyst wall; the rest of this extracellular matrix is made up of a
simple b1-3 linked N-acetylgalactosamine (GalNAc) homopolymer
(Jarroll et al., 1989; Gerwig et al., 2002). A non-VSP type 1 integral
membrane protein termed HCNCp is a member of a large group of
cysteine-rich factors which are potentially secreted stage-specifi-
cally (Davids et al., 2006b). Indeed, while an epitope-tagged variant
of the HCNCp investigated in this study localized to the plasma-
lemma in trophozoites, it was detected in ESVs during encystation
and partially secreted to the cyst wall or the PM – cyst wall inter-
face (Davids et al., 2006b). Recently, another family of soluble cys-
teine-rich proteins which appear to be trafficked via ESVs to the
cyst wall but localize additionally to the cell body have been iden-
tified (Chiu et al., 2010). On the other hand, CWPs are trafficked
exclusively through ESVs and are secreted quantitatively. How-
ever, it is not known when and where in this exocytic pathway
the poly-GalNAc sugar component is synthesized and how it is fi-
nally incorporated into the cyst wall structure. A recent study
showed that the leucine-rich repeat domain of CWP1 has a lectin
activity and binds to the curled fibrils of the GalNAc homopolymer
in the cyst wall (Chatterjee et al., 2010). Taken together with the
sequential secretion of the CWPs (see below) this suggests that
the glycan and at least CWP1 are exported coordinately either in
the same or separate carriers and polymerize after distribution as
an even layer on the surface. Time-lapse microscopy of living cells
showed that the deposited CWM loses its plasticity and transforms
into a rigid shell in 10–30 s (Trepp, F., Spycher, C., Hehl, A.B.,
unpublished data).
The synthesis and trafficking of the CWM components are
stage-specifically regulated and highly coordinated. The assembly
of the cyst wall galactosamine polymer from glucose is mediated
by pathways whose components are upregulated transcriptionally
and allosterically after induction (Macechko et al., 1992; Lujan
et al., 1995a; Das and Gillin, 1996; Van Keulen et al., 1998; Bulik
et al., 2000). CWP1–3 mRNA levels peak at 7 h p.i., and the newly
produced CWPs accumulate at approximately 2 h p.i. in emerging
ESVs (Konrad et al., 2010). CWP export from the ER to ESVs is com-
pleted 8–10 h after induction (Hehl et al., 2000). The fact that ESVs
only contain CWPs and no constitutively secreted proteins strongly
suggests that the former are sorted away from the latter during ER
export (Reiner et al., 1990; Marti et al., 2003a). The first half of the
encystation process is therefore dedicated to synthesis and accu-
mulation of the CWM in the newly established ESVs. This sorting
and export pathway in the absence of a constitutive Golgi appara-
tus has hallmarks of cis Golgi compartment neogenesis. The mem-
brane carriers, if any, for trafficking of the CWM from the ER to
ESVs are currently unknown, but the process appears to be depen-
dent on COPII coat formation and the small GTPase Rab1 (Stefanic
et al., 2009). An alternative scenario invokes lateral segregation of
the CWM into ER sub-domains which eventually differentiate into
ESVs (Lujan and Touz, 2003; Hehl and Marti, 2004). Segregation of
the CWPs to ESVs is determined by dominant signals on CWPs and
occurs at the level of ER export (Marti et al., 2003a). Rather than
defined targeting sequences, entire domains of CWPs, specifically
the leucine-rich repeats (LRRs), are necessary for sorting into ESVs
(Hehl et al., 2000; Marti et al., 2003a; Sun et al., 2003). Thus, sim-
ilar to Golgi cisternae, ESVs are post-ER organelles where cargo is
delayed, presumably for maturation, before regulated secretion.
However, unlike conventional Golgi cisternae, ESVs contain only
one type of cargo, the CWM. However, transient association of COPI
components with ESVs (Marti et al., 2003b), their sensitivity to bre-
feldin A (Lujan et al., 1995a; Marti et al., 2003a), and dependence of
ESV genesis and maturation on giardial Sar1 and Arf1 GTPases,
respectively (Stefanic et al., 2009), support the idea that these
organelles are stage-regulated Golgi-like compartments. The exact
nature of ESVs and their function in regulated secretion in the con-
text of reductive evolution are currently under investigation in sev-
eral laboratories.
After completion of ESV formation CWP1, which is used as a
standard marker for the CWM, is further delayed in ESVs for several
hours, before being secreted to cover the entire cell surface where it
eventually polymerizes. Recent studies revealed that the formation
of an environmentally-resistant cyst wall is more complicated and
requires several tightly synchronized steps (Gottig et al., 2006; Stef-
anic et al., 2009; Konrad et al., 2010). The three CWP family mem-
bers share considerable sequence identity and have a similar
domain structure. CWP2 has an additional 121 residue C-terminal
extension rich in basic amino acids (Lujan et al., 1995b). Proteolytic
C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480 475
processing of this domain is currently the only proteolytic modifi-
cation described for CWPs during maturation of the CWM. In addi-
tion, formation of disulfide (Hehl et al., 2000) and isopeptide
(Davids et al., 2004) bonds between CWPs appears to play a major
role in the export process. Although the evidence clearly implicates
specific cleavage of pro-CWP2 by a cysteine protease, controversy
as to the exact identity of the enzyme remains (Touz et al., 2002a;
DuBois et al., 2008). Previous investigations indicated that the en-
tire C-terminal extension of 13 kDa (Gottig et al., 2006) might
be removed from CWP2. However, the small C-terminal portion of
the native or the transgenic CWP2 had not yet been visualized di-
rectly (Sun et al., 2003). Although processing of CWP2 occurred be-
fore secretion of the CWM, its correlation with expression kinetics
or maturation and morphology of ESVs had not been explored until
recently. Analysis of a CWP2 variant with epitope tags at both ter-
mini showed that cleavage occurred during the maturation stage of
ESVs, before secretion of the CWM (Konrad et al., 2010). Western
blot analysis suggested removal of a short fragment of 5 kDa be-
tween 8 h and 10 h p.i. The C-terminal domain of CWP2 had been
implicated in sorting of all three CWPs from the ER to ESVs (Gottig
et al., 2006). The authors had invoked an export mechanism based
on sequestration to and protrusion from specialized sub-domains of
the ER, rather than conventional ER export. The relation between
ESVs and the ER remains unclear. On the one hand there is a distinct
morphological difference between the two organellar systems as
well as exclusion of cargo and resident factors such as PDIs and
Hsp70/BiP from ESVs. The latter, however, appears to be cycling
through ESVs and may be retrieved by a KDEL receptor protein
(Stefanic et al., 2006). On the other hand, the current data give no
answers to the questions as to how ESVs arise and whether the nas-
cent organelles at least constitute highly specialized ER sub-do-
mains (Touz et al., 2002a; Marti et al., 2003a; Gottig et al., 2006).
In particular, the close spatial proximity of the two organelles and
the vigorous exchange of the CWM between established ESVs raises
the question whether they ever become truly independent before
the secretory process begins (Stefanic et al., 2009).
Dual-tagged CWP2 as well as CWP::GFP chimeras to analyze the
processes during the maturation phase of ESVs revealed a hitherto
unnoticed sorting and partitioning of the CWM (Konrad et al.,
2010). In particular, co-localization studies of CWP1 with CWP3
or with the C-terminus of CWP2 showed that the CWM is parti-
tioned into two biophysically distinct fractions inside maturing
ESVs. CWP3 forms a condensed core structure together with the
small CWP2 C-terminus after processing of pro-CWP2. How the
condensation process is triggered is unknown but an unrelated,
earlier study showed that expression of giardial CWP1 and CWP2
in human embryonic kidney-293 cells led to formation of granules
and secretion to the culture medium (Abdul-Wahid and Faubert,
2004). This suggests that accumulation and condensation is an
inherent property of CWPs. In particular, the basic C-terminal
extension of CWP2 had been implicated in granule formation dur-
ing encystation in this study. Conversely, in encysting Giardia
CWP1 and the mature N-terminal, CWP2 remain in a fluid state un-
til secretion and distribution on the surface of the forming cyst
(Konrad et al., 2010). Thus, the condensed and the fluid fraction
of the CWM contains a mature CWP2 product. Live cell analysis
using GFP-tagged CWP1 variants showed that these cargo compo-
nents are highly motile within an ESV organelle network. These
studies demonstrate that the immobile ESVs are laterally con-
nected via dynamic membrane tubular channels (Stefanic et al.,
2009; Konrad et al., 2010). After partitioning of the fluid and con-
densed fraction of the CWM, the former is sorted away into com-
partments which localize close to the cell periphery. The upshot
is that this fluid material containing CWP1 and the large N-termi-
nal fragment of the processed CWP2 is rapidly and quantitatively
secreted within a few minutes. This first layer of the cyst wall is
laid down at the same time as morphological transformation of
the differentiating cell takes place. The material polymerizes rap-
idly and results in the formation of a structurally resistant cell wall.
The condensed rest of the CWM (CWP3 and the small C-terminal
fragment of CWP2) remains completely in internal compartments
and is secreted slowly over the course of several hours. Although
processing of CWP2 is not required for condensed core formation
and sequential secretion of CWM, it is necessary for correct parti-
tioning of CWP2. Unprocessed CWP2 becomes sequestered in the
condensed cores and is exported only during the second secretion
process resulting in the formation of morphologically normal cysts
which are not water-resistant (Konrad et al., 2010).
Taken together, regulated secretion and cyst wall formation ap-
pears to be significantly more complex than previously thought
and requires several hours from the time the first layer is estab-
lished until the cyst becomes water resistant. Condensed core for-
mation in maturing ESVs and sorting of the CWM for sequential
secretion is strikingly similar to processes at the trans-Golgi net-
work. This lends additional support to the hypothesis that ESVs
are functional Golgi cisternae analogs.
4.3. Secretion in excysting cells
When passing through the low pH environment of the stomach
Giardia cysts are triggered to initiate excystation. Secreted prote-
ases released from the endosome–lysosome PV system play a brief
but essential role in the liberation of the excyzoite (Bernander
et al., 2001) from the protective confines of the cyst wall. Host
and parasite proteases are required for the degradation of the
CWPs whilst a parasite-specific glycohydrolase attacks the GalNAc
fibrils of the cyst wall (Chatterjee et al., 2010). A cathepsin B type
protease (Ward et al., 1997), designated CP2, was identified by
affinity purification with a biotinylated variant of the specific
inhibitor E-64. The inhibitor was also used to determine the sub-
cellular localization of CP2 in PVs or the equivalent of these organ-
elles in cysts. Secretion of the enzyme into the space between the
plasma membrane and the cyst wall is triggered during
excystation.
CWPs are phosphorylated during export (Slavin et al., 2002).
This post-translational modification needs to be reversed for
excystation: inhibition of acid phosphatase activity during the first
phase of in vitro excystation almost completely abolished the pro-
cess (Slavin et al., 2002).
5. Organelles and pathways for endocytic transport
The PVs constitute a very conspicuous organelle system in
transmission EMs of trophozoites. The electron lucent PVs appear
approximately oval-shaped and typically 150 nm long. The
organelles are arrayed as a layer just below the plasma membrane
of the entire dorsal side as well as in the small region at the center
of the ventral disk. In the absence of a cytostome, the PVs are the
only known endocytic organelles (Tai et al., 1993) capable of accu-
mulating fluid phase and membrane-bound molecules. However,
PVs also seem to have lysosomal properties (Feely and Dyer,
1987; Lindmark, 1988; McCaffery and Gillin, 1994), which means
they acidify and mature to become digestive organelles. In addition
to hydrolases, PVs contain cathepsins (Ward et al., 1997; Thirion
et al., 2003). The presence of CWPs in the lumens of PVs (Reiner
et al., 1990) also suggests a role in regulated secretion of CWP.
The dynamics of fluid phase and membrane cargo uptake was
not investigated until recently. Two studies presented data sug-
gesting that PVs periodically open to the environment either via
a channel or by fusion with the PM and take up soluble material
(Gaechter et al., 2008; Abodeely et al., 2009) before closing again.
476 C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480
The reversible, transient communication with the extracellular
space effectively flushes the PV, releasing its contents into the
environment and replacing this volume with extracellular med-
ium. Using fluid phase markers and confocal live-cell imaging it
was possible to determine the dynamics of this environmental
sampling and to quantify endocytic transport. PVs in living and
in chemically fixed cells can also be visualized using Lysotracker™
(Gaechter et al., 2008; Rivero et al., 2010). More importantly, fluo-
rescence recovery after photobleaching (FRAP) demonstrated that
there is no lateral exchange of fluid phase markers between indi-
vidual PVs (Gaechter et al., 2008). Most fluid phase markers re-
mained in PVs until expelled when the organelle again formed a
continuity with the external medium, but others (e.g. casein)
translocated rapidly to the ER or an associated membrane com-
partment termed the tubulo-vesicular network (TVN) (Abodeely
et al., 2009). These data suggest that uptake of soluble material
from the environment into PVs is not selective, in contrast to fur-
ther retrograde transport which allows only certain, as yet unde-
fined substances, to rapidly cross over into the proximal ER
(Abodeely et al., 2009). Most of the fluid phase material taken up
by PVs is likely to require further breakdown after acidification
of the organelle lumen before being transported along endocytic
pathways. A recent study shows that import of low-density lipo-
protein (LDL), which is not present in the gut lumen but abundant
in cell culture medium, is mediated by PVs and dependent on the
predicted AP 2/3 complex (designated as AP2 by the authors)
(Rivero et al., 2010).
PVs may therefore constitute a clever environmental sampling
system which allows the parasite to take up fluid phase material
relatively indiscriminately but safely. Subsequent digestion liber-
ates essential building blocks for transport towards the cell interior
whereas waste material as well as harmful substances are expelled
rapidly.
6. Mitochondrial remnants
Giardia lamblia has long been considered a primitive eukaryote
based on the absence of organelles resembling mitochondria. Based
on the Archezoa hypothesis, this organism was assigned to a loosely
classified group of supposedly amitochondriate lineages that had
not participated in the endosymbiotic event which led to the incor-
poration, maintenance and molecular ‘‘surrender’’ of an
a
-proteo-
bacteria (Cavalier-Smith, 1998). This theory concerning
eukaryotic speciation was strongly challenged by the discovery of
highly degenerate mitochondrion-related organelles in E. histolytica
named mitosomes (Tovar et al., 1999). Shortly thereafter,
G. lamblia homologs of genes IscS and IscU involved in the biosyn-
thesis of Fe–S proteins (an essential function of mitochondria) were
cloned, and their products characterized in terms of sub-cellular
localization in giardial cells (Tovar et al., 2003). Both proteins were
found to accumulate within double membrane-bound organelles
containing no DNA, as previously observed with E. histolytica.
In confocal microscopy images, Giardia mitosomes appear as
100 nm spherical cytoplasmic compartments. An intriguing
aspect of these organelles is their distribution into two distinct
populations, namely an apparently elongated single central mito-
some (CM) organelle and the multiple peripheral mitosomes
(PMs) (Regoes et al., 2005). PMs appear to be distributed randomly
throughout the cell while the CM is invariably found in the basal
body region between the nuclei. A more detailed ultrastructural
analysis by EM revealed that the CM was in fact a tight cluster of
spherical organelles which are indistinguishable from the PMs
(Hehl et al., 2007). This special configuration almost certainly has
a functional significance and was termed the central mitosome
complex (CMC). Upon cell division, only the CMC was shown to
be actively divided and segregated in a cell cycle-dependent man-
ner whilst the PMs appear to be distributed stochastically between
daughter cells (Regoes et al., 2005). The mechanisms which regu-
late mitosomal division and inheritance remain to be elucidated.
In an effort to further characterize giardial mitosomes, a careful
investigation of cellular distribution using specific antibodies
proved that other mitochondrial markers such as the giardial
homologs of chaperonin 60 (Cpn60), mitochondrial Hsp 70
(mtHsp70) and ferredoxin (Fd) were also present in these organ-
elles (Regoes et al., 2005). Taken together, these data indicate that
the absence of bona fide mitochondria in G. lamblia is the result of
secondary loss and not a manifestation of a primary primitive
state.
Interestingly, only giardial iron–sulfur cluster assembly enzyme
(IscU) and Fd were shown to require the presence of amino-termi-
nal targeting presequences, suggesting that mitosomes have main-
tained aspects of both presequence-dependent and independent
import routes (Tovar et al., 2003; Regoes et al., 2005). In the at-
tempt to investigate the molecular machinery mediating protein
import across the double mitosomal membrane, a component of
the inner mitochondrial membrane translocase and the processing
peptidase were identified in a comparative study involving Giardia
mitosomes and T. vaginalis hydrogenosomes (Dolezal et al., 2005).
Combined with the recent identification of a giardial Tom40 sub-
unit of the outer membrane protein import channel (Dagley
et al., 2009), this data supports the hypothesis that mitosomes,
hydrogenosomes and mitochondria represent different forms of
the same fundamental organelle and employ similar translocation
mechanisms for mitosomal protein import (Burri and Keeling,
2007). Further research will be necessary to characterize all com-
ponents of the translocation complexes to comprehend if and
how proteins with and without a canonical targeting presequence
are substrates for the same translocation machinery.
Thus, the identification of mitosomes as mitochondrial rem-
nants further reinforces the notion that extreme lifestyles, fre-
quently encountered in parasitic protozoa such as G. lamblia, may
lead to a substantial reduction in molecular machinery. This in turn
could result in the complete loss of entire organelle types such as
the Golgi apparatus and mitochondria.
7. Conclusions
The secretory transport capacity of Giardia is perfectly adapted
to a changing environment and able to deploy essential protective
surface coats as well as molecules which act on the cells of the host
epithelia. Nevertheless, the giardial trafficking machinery appears
to be the product of a general secondary reduction process that
led to minimization of all components identified to date (Marti
et al., 2003b; Morrison et al., 2007). Despite the low complexity
of the organelles and machinery involved, these diverse protective
surface antigens, as well as secreted proteins, are delivered with
great fidelity (Reiner et al., 1990; McCaffery et al., 1994; Hehl
et al., 2000; Marti et al., 2002, 2003a; Lujan and Touz, 2003; Touz
et al., 2003; Hehl and Marti, 2004; Gottig et al., 2006; Hernandez
et al., 2007; Konrad et al., 2010; Rivero et al., 2010). Of particular
interest are VSPs of trophozoites and the extracellular matrix poly-
mer of cysts, both of which confer environmental resistance in the
respective life cycle stages. Recent and ongoing work in several
laboratories is revealing the basic principles behind the transport
events responsible for the assembly of these key biological barriers.
However, significant work to elucidate the details of these mecha-
nisms, in particular in light of their astonishingly low complexity,
is still needed. For example, it is becoming clear that Giardia could
serve as a model for the investigation of synthesis, transport and
assembly of simple but highly effective biopolymers, a key feature
C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480 477
of all perorally transmitted protozoan and metazoan parasites
(Gottig et al., 2006; Chatterjee et al., 2010; Konrad et al., 2010). An-
other example is the PV system: these organelles appear to be at
the crossroads of endocytic and exocytic transport. Although the
data are still scarce, PVs are emerging as the major sorting station
for the transport of soluble as well as membrane-bound factors in
and out of the cell (Touz et al., 2004; Abodeely et al., 2009; Rivero
et al., 2010). As such, they could provide a protected space for both
final modification and/or activation of secreted molecules on one
hand; on the other hand, PVs can act as an intracellular contain-
ment system. This would allow controlled separation of nutrients
from potentially harmful substances taken up in bulk by constant
sampling of the fluid extracellular environment. As is also the case
for ESVs, a better understanding of the biological role of trafficking
pathways to and from PVs requires elucidation of the ‘‘organelle
cycle’’, i.e. the processes that govern organelle genesis, mainte-
nance and maturation.
Acknowledgements
We apologize to the colleagues whose work we could not cite
for reasons of space. The work in the laboratory of ABH is sup-
ported by the Swiss National Science Foundation (grant
#31003A-125389). We are grateful to all current and past mem-
bers of the laboratory for their contributions, in particular to Dr.
Attila Regoes (University of Zürich, Switzerland) for providing the
composite volume image. We thank the reviewers of this article
for their helpful comments.
References
Abdul-Wahid, A., Faubert, G.M., 2004. Similarity in cyst wall protein (CWP)
trafficking between encysting Giardia duodenalis trophozoites and CWP-
expressing human embryonic kidney-293 cells. Biochem. Biophys. Res.
Commun. 324, 1069–1080.
Abodeely, M., DuBois, K.N., Hehl, A., Stefanic, S., Sajid, M., DeSouza, W., Attias, M.,
Engel, J.C., Hsieh, I., Fetter, R.D., McKerrow, J.H., 2009. A contiguous
compartment functions as endoplasmic reticulum and endosome/lysosome in
Giardia lamblia. Eukaryotic Cell 8, 1665–1676.
Ackers, J.P., Dhir, V., Field, M.C., 2005. A bioinformatic analysis of the RAB genes of
Trypanosoma brucei. Mol. Biochem. Parasitol. 141, 89–97.
Adam, R.D., Aggarwal, A., Lal, A.A., de La Cruz, V.F., McCutchan, T., Nash, T.E., 1988.
Antigenic variation of a cysteine-rich protein in Giardia lamblia. J. Exp. Med. 167,
109–118.
Banerjee, S., Vishwanath, P., Cui, J., Kelleher, D.J., Gilmore, R., Robbins, P.W.,
Samuelson, J., 2007. The evolution of N-glycan-dependent endoplasmic
reticulum quality control factors for glycoprotein folding and degradation.
Proc. Natl. Acad. Sci. USA 104, 11676–11681.
Banerjee, S., Cui, J., Robbins, P.W., Samuelson, J., 2008. Use of Giardia, which appears
to have a single nucleotide-sugar transporter for UDP-GlcNAc, to identify the
UDP-Glc transporter of Entamoeba. Mol. Biochem. Parasitol. 159, 44–53.
Benchimol, M., 2004. The release of secretory vesicle in encysting Giardia lamblia.
FEMS Microbiol. Lett. 235, 81–87.
Bernander, R., Palm, J.E., Svard, S.G., 2001. Genome ploidy in different stages of the
Giardia lamblia life cycle. Cell Microbiol. 3, 55–62.
Boehm, M., Bonifacino, J.S., 2001. Adaptins: the final recount. Mol. Biol. Cell 12,
2907–2920.
Boucher, S.E., Gillin, F.D., 1990. Excystation of in vitro-derived Giardia lamblia cysts.
Infect. Immun. 58, 3516–3522.
Bulik, D.A., van Ophem, P., Manning, J.M., Shen, Z., Newburg, D.S., Jarroll, E.L., 2000.
UDP-N-acetylglucosamine pyrophosphorylase, a key enzyme in encysting
Giardia, is allosterically regulated. J. Biol. Chem. 275, 14722–14728.
Burri, L., Keeling, P.J., 2007. Protein targeting in parasites with cryptic mitochondria.
Int. J. Parasitol. 37, 265–272.
Castillo-Romero, A., Leon-Avila, G., Wang, C.C., Perez Rangel, A., Camacho Nuez, M.,
Garcia Tovar, C., Ayala-Sumuano, J.T., Luna-Arias, J.P., Hernandez, J.M., 2010.
Rab11 and actin cytoskeleton participate in Giardia lamblia encystation, guiding
the specific vesicles to the cyst wall. PLoS Negl. Trop. Dis. 4, e697.
Cavalier-Smith, T., 1998. A revised six-kingdom system of life. Biol. Rev. Cambridge
Philos. Soc. 73, 203–266.
Chatterjee, A., Carpentieri, A., Ratner, D.M., Bullitt, E., Costello, C.E., Robbins, P.W.,
Samuelson, J., 2010. Giardia cyst wall protein 1 is a lectin that binds to curled
fibrils of the GalNAc homopolymer. PLoS Pathogens 6, e1001059.
Chin, A.C., Teoh, D.A., Scott, K.G., Meddings, J.B., Macnaughton, W.K., Buret, A.G.,
2002. Strain-dependent induction of enterocyte apoptosis by Giardia lamblia
disrupts epithelial barrier function in a caspase-3-dependent manner. Infect.
Immun. 70, 3673–3680.
Chiu, P.W., Huang, Y.C., Pan, Y.J., Wang, C.H., Sun, C.H., 2010. A novel family of cyst
proteins with epidermal growth factor repeats in Giardia lamblia. PLoS Negl.
Trop. Dis. 4, e677.
Dacks, J.B., Doolittle, W.F., 2002. Novel syntaxin gene sequences from Giardia,
Trypanosoma and algae: implications for the ancient evolution of the eukaryotic
endomembrane system. J. Cell Sci. 115, 1635–1642.
Dacks, J.B., Davis, L.A., Sjogren, A.M., Andersson, J.O., Roger, A.J., Doolittle, W.F., 2003.
Evidence for Golgi bodies in proposed ‘Golgi-lacking’ lineages. Proc. Biol. Sci.
270 (Suppl. 2), S168–171.
Dacks, J.B., Doolittle, W.F., 2004. Molecular and phylogenetic characterization of
syntaxin genes from parasitic protozoa. Mol. Biochem. Parasitol. 136, 123–136.
Dacks, J.B., Field, M.C., 2007. Evolution of the eukaryotic membrane-trafficking
system: origin, tempo and mode. J. Cell Sci. 120, 2977–2985.
Dagley, M.J., Dolezal, P., Likic, V.A., Smid, O., Purcell, A.W., Buchanan, S.K., Tachezy, J.,
Lithgow, T., 2009. The protein import channel in the outer mitosomal
membrane of Giardia intestinalis. Mol. Biol. Evol. 26, 1941–1947.
Das, S., Gillin, F.D., 1996. Giardia lamblia: increased UDP-N-acetyl-D-glucosamine
and N-acetyl-D-galactosamine transferase activities during encystation. Exp.
Parasitol. 83, 19–29.
Davids, B.J., Mehta, K., Fesus, L., McCaffery, J.M., Gillin, F.D., 2004. Dependence of
Giardia lamblia encystation on novel transglutaminase activity. Mol. Biochem.
Parasitol. 136, 173–180.
Davids, B.J., Palm, J.E., Housley, M.P., Smith, J.R., Andersen, Y.S., Martin, M.G.,
Hendrickson, B.A., Johansen, F.E., Svard, S.G., Gillin, F.D., Eckmann, L., 2006a.
Polymeric immunoglobulin receptor in intestinal immune defense against the
lumen-dwelling protozoan parasite Giardia. J. Immunol. 177, 6281–6290.
Davids, B.J., Reiner, D.S., Birkeland, S.R., Preheim, S.P., Cipriano, M.J., McArthur, A.G.,
Gillin, F.D., 2006b. A new family of giardial cysteine-rich non-VSP protein genes
and a novel cyst protein. PloS One 1, e44.
de Carvalho, T.B., David, E.B., Coradi, S.T., Guimaraes, S., 2008. Protease activity in
extracellular products secreted in vitro by trophozoites of Giardia duodenalis.
Parasitol. Res. 104, 185–190.
Dolezal, P., Smid, O., Rada, P., Zubacova, Z., Bursac, D., Sutak, R., Nebesarova, J.,
Lithgow, T., Tachezy, J., 2005. Giardia mitosomes and trichomonad
hydrogenosomes share a common mode of protein targeting. Proc. Natl. Acad.
Sci. USA 102, 10924–10929.
DuBois, K.N., Abodeely, M., Sakanari, J., Craik, C.S., Lee, M., McKerrow, J.H., Sajid, M.,
2008. Identification of the major cysteine protease of Giardia and its role in
encystation. J. Biol. Chem. 283, 18024–18031.
Eckmann, L., Laurent, F., Langford, T.D., Hetsko, M.L., Smith, J.R., Kagnoff, M.F., Gillin,
F.D., 2000. Nitric oxide production by human intestinal epithelial cells and
competition for arginine as potential determinants of host defense against the
lumen-dwelling pathogen Giardia lamblia. J. Immunol. 164, 1478–1487.
Elias, E.V., Quiroga, R., Gottig, N., Nakanishi, H., Nash, T.E., Neiman, A., Lujan, H.D.,
2008. Characterization of SNAREs determines the absence of a typical Golgi
apparatus in the ancient eukaryote Giardia lamblia. J. Biol. Chem. 283, 35996–
36010.
Feely, D.E., Dyer, J.K., 1987. Localization of acid phosphatase activity in Giardia
lamblia and Giardia muris trophozoites. J. Protozol. 34, 80–83.
Feely, D.E., Holberton, D.V., Erlandsen, S.L., 1990. In: Meyer, E.A. (Ed.), The Biology of
Giardia. Elsevier Science Publishers, New York, pp. 11–49.
Gaechter, V., Schraner, E., Wild, P., Hehl, A.B., 2008. The single dynamin family
protein in the primitive protozoan giardia lamblia is essential for stage
conversion and endocytic transport. Traffic 9, 57–71.
Gerwig, G.J., van Kuik, J.A., Leeflang, B.R., Kamerling, J.P., Vliegenthart, J.F., Karr, C.D.,
Jarroll, E.L., 2002. The Giardia intestinalis filamentous cyst wall contains a novel
beta(1–3)-N-acetyl-D-galactosamine polymer: a structural and conformational
study. Glycobiology 12, 499–505.
Gillin, F.D., Reiner, D.S., Gault, M.J., Douglas, H., Das, S., Wunderlich, A., Sauch, J.F.,
1987. Encystation and expression of cyst antigens by Giardia lamblia in vitro.
Science 235, 1040–1043.
Gillin, F.D., Reiner, D.S., McCaffery, M., 1991. Organelles of protein transport in
Giardia lamblia. Parasitol. Today 7, 113–116.
Gottig, N., Elias, E.V., Quiroga, R., Nores, M.J., Solari, A.J., Touz, M.C., Lujan, H.D., 2006.
Active and passive mechanisms drive secretory granule biogenesis during
differentiation of the intestinal parasite Giardia lamblia. J. Biol. Chem. 281,
18156–18166.
Gupta, R.S., Aitken, K., Falah, M., Singh, B., 1994. Cloning of Giardia lamblia heat
shock protein HSP70 homologs: implications regarding origin of eukaryotic
cells and of endoplasmic reticulum. Proc. Natl. Acad. Sci. USA 91, 2895–2899.
Hehl, A.B., Marti, M., Kohler, P., 2000. Stage-specific expression and targeting of cyst
wall protein-green fluorescent protein chimeras in Giardia. Mol. Biol. Cell 11,
1789–1800.
Hehl, A.B., Marti, M., 2004. Secretory protein trafficking in Giardia intestinalis. Mol.
Microbiol. 53, 19–28.
Hehl, A.B., Regos, A., Schraner, E., Schneider, A., 2007. Bax function in the absence of
mitochondria in the primitive protozoan Giardia lamblia. PloS One 2, e488.
Hernandez, Y., Castillo, C., Roychowdhury, S., Hehl, A., Aley, S.B., Das, S., 2007.
Clathrin-dependent pathways and the cytoskeleton network are involved in
ceramide endocytosis by a parasitic protozoan, Giardia lamblia. Int. J. Parasitol.
37, 21–32.
Jarroll, E.L., Manning, P., Lindmark, D.G., Coggins, J.R., Erlandsen, S.L., 1989. Giardia
cyst wall-specific carbohydrate: evidence for the presence of galactosamine.
Mol. Biochem. Parasitol. 32, 121–131.
478 C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480
Jimenez, J.C., Uzcanga, G., Zambrano, A., Di Prisco, M.C., Lynch, N.R., 2000.
Identification and partial characterization of excretory/secretory products
with proteolytic activity in Giardia intestinalis. J. Parasitol. 86, 859–862.
Jimenez, J.C., Morelle, W., Michalsky, J.C., Dei-Cas, E., 2007. Excreted/secreted
glycoproteins of G. intestinalis play an essential role in the antibody response.
Parasitol. Res. 100, 715–720.
Kane, A.V., Ward, H.D., Keusch, G.T., Pereira, M.E., 1991. In vitro encystation of
Giardia lamblia: large-scale production of in vitro cysts and strain and clone
differences in encystation efficiency. J. Parasitol. 77, 974–981.
Kasper, L.H., Bradley, M.S., Pfefferkorn, E.R., 1984. Identification of stage-specific
sporozoite antigens of Toxoplasma gondii by monoclonal antibodies. J. Immunol.
132, 443–449.
Katelaris, P.H., Naeem, A., Farthing, M.J., 1994. Activity of metronidazole,
azithromycin and three benzimidazoles on Giardia lamblia growth and
attachment to a human intestinal cell line. Aliment Pharm. Therap. 8, 187–192.
Keister, D.B., 1983. Axenic culture of Giardia lamblia in TYI-S-33 medium
supplemented with bile. Trans. R. Soc. Trop. Med. Hyg. 77, 487–488.
Kloepper, T.H., Kienle, C.N., Fasshauer, D., 2007. An elaborate classification of SNARE
proteins sheds light on the conservation of the eukaryotic endomembrane
system. Mol. Biol. Cell 18, 3463–3471.
Knodler, L.A., Noiva, R., Mehta, K., McCaffery, J.M., Aley, S.B., Svard, S.G., Nystul, T.G.,
Reiner, D.S., Silberman, J.D., Gillin, F.D., 1999. Novel protein-disulfide
isomerases from the early-diverging protist Giardia lamblia. J. Biol. Chem. 274,
29805–29811.
Konrad, C., Spycher, C., Hehl, A.B., 2010. Selective condensation drives partitioning
and sequential secretion of cyst wall proteins in differentiating Giardia lamblia.
PLoS Pathogens 6, e1000835.
Koumandou, V.L., Dacks, J.B., Coulson, R.M., Field, M.C., 2007. Control systems for
membrane fusion in the ancestral eukaryote; evolution of tethering complexes
and SM proteins. BMC Evol. Biol. 7, 29.
Lal, K., Field, M.C., Carlton, J.M., Warwicker, J., Hirt, R.P., 2005. Identification of a very
large Rab GTPase family in the parasitic protozoan Trichomonas vaginalis. Mol.
Biochem. Parasitol. 143, 226–235.
Lanfredi-Rangel, A., Attias, M., de Carvalho, T.M., Kattenbach, W.M., De Souza, W.,
1998. The peripheral vesicles of trophozoites of the primitive protozoan Giardia
lamblia may correspond to early and late endosomes and to lysosomes. J. Struct.
Biol. 123, 225–235.
Lanfredi-Rangel, A., Kattenbach, W.M., Diniz Jr., J.A., de Souza, W., 1999.
Trophozoites of Giardia lamblia may have a Golgi-like structure. FEMS
Microbiol. Lett. 181, 245–251.
Langford, T.D., Silberman, J.D., Weiland, M.E., Svard, S.G., McCaffery, J.M., Sogin, M.L.,
Gillin, F.D., 2002. Giardia lamblia: identification and characterization of Rab and
GDI proteins in a genome survey of the ER to Golgi endomembrane system. Exp.
Parasitol. 101, 13–24.
Lee, F.J., Moss, J., Vaughan, M., 1992. Human and Giardia ADP-ribosylation factors
(ARFs) complement ARF function in Saccharomyces cerevisiae. J. Biol. Chem. 267,
24441–24445.
Li, E., Zhao, A., Shea-Donohue, T., Singer, S.M., 2007. Mast cell-mediated changes in
smooth muscle contractility during mouse giardiasis. Infect. Immun. 75, 4514–
4518.
Lindmark, D.G., 1988. Giardia lamblia: localization of hydrolase activities in
lysosome-like organelles of trophozoites. Exp. Parasitol. 65, 141–147.
Lujan, H.D., Marotta, A., Mowatt, M.R., Sciaky, N., Lippincott-Schwartz, J., Nash, T.E.,
1995a. Developmental induction of Golgi structure and function in the
primitive eukaryote Giardia lamblia. J. Biol. Chem. 270, 4612–4618.
Lujan, H.D., Mowatt, M.R., Conrad, J.T., Bowers, B., Nash, T.E., 1995b. Identification of
a novel Giardia lamblia cyst wall protein with leucine-rich repeats. Implications
for secretory granule formation and protein assembly into the cyst wall. J. Biol.
Chem. 270, 29307–29313.
Lujan, H.D., Mowatt, M.R., Byrd, L.G., Nash, T.E., 1996a. Cholesterol starvation
induces differentiation of the intestinal parasite Giardia lamblia. Proc. Natl.
Acad. Sci. USA 93, 7628–7633.
Lujan, H.D., Mowatt, M.R., Conrad, J.T., Nash, T.E., 1996b. Increased expression of the
molecular chaperone BiP/GRP78 during the differentiation of a primitive
eukaryote. Biol. Cell 86 (1), 11–18.
Lujan, H.D., Touz, M.C., 2003. Protein trafficking in Giardia lamblia. Cell Microbiol. 5,
427–434.
Macechko, P.T., Steimle, P.A., Lindmark, D.G., Erlandsen, S.L., Jarroll, E.L., 1992.
Galactosamine-synthesizing enzymes are induced when Giardia encyst. Mol.
Biochem. Parasitol. 56, 301–309.
Marti, M., Li, Y., Kohler, P., Hehl, A.B., 2002. Conformationally correct expression of
membrane-anchored Toxoplasma gondii SAG1 in the primitive protozoan
Giardia duodenalis. Infect. Immun. 70, 1014–1016.
Marti, M., Li, Y., Schraner, E.M., Wild, P., Kohler, P., Hehl, A.B., 2003a. The secretory
apparatus of an ancient eukaryote: protein sorting to separate export pathways
occurs before formation of transient Golgi-like compartments. Mol. Biol. Cell 14,
1433–1447.
Marti, M., Regos, A., Li, Y., Schraner, E.M., Wild, P., Muller, N., Knopf, L.G., Hehl, A.B.,
2003b. An ancestral secretory apparatus in the protozoan parasite Giardia
intestinalis. J. Biol. Chem. 278, 24837–24848.
McCaffery, J.M., Faubert, G.M., Gillin, F.D., 1994. Giardia lamblia: traffic of a
trophozoite variant surface protein and a major cyst wall epitope during
growth, encystation, and antigenic switching. Exp. Parasitol. 79,
236–249.
McCaffery, J.M., Gillin, F.D., 1994. Giardia lamblia: ultrastructural basis of protein
transport during growth and encystation. Exp. Parasitol. 79, 220–235.
Meyer, E.A., 1994. Giardia as an organism. In: Thompson, R.C.A. (Ed.), Giardia: From
Molecules to Disease. CAB International, pp. 3–13.
Morelle, W., Jimenez, J.C., Cieniewski-Bernard, C., Dei-Cas, E., Michalski, J.C., 2005.
Characterization of the N-linked glycans of Giardia intestinalis. Glycobiology 15,
549–559.
Morrison, H.G., McArthur, A.G., Gillin, F.D., Aley, S.B., Adam, R.D., Olsen, G.J., Best,
A.A., Cande, W.Z., Chen, F., Cipriano, M.J., Davids, B.J., Dawson, S.C., Elmendorf,
H.G., Hehl, A.B., Holder, M.E., Huse, S.M., Kim, U.U., Lasek-Nesselquist, E.,
Manning, G., Nigam, A., Nixon, J.E., Palm, D., Passamaneck, N.E., Prabhu, A.,
Reich, C.I., Reiner, D.S., Samuelson, J., Svard, S.G., Sogin, M.L., 2007. Genomic
minimalism in the early diverging intestinal parasite Giardia lamblia. Science
317, 1921–1926.
Mowatt, M.R., Lujan, H.D., Cotten, D.B., Bowers, B., Yee, J., Nash, T.E., Stibbs, H.H.,
1995. Developmentally regulated expression of a Giardia lamblia cyst wall
protein gene. Mol. Microbiol. 15, 955–963.
Muller, J., Sterk, M., Hemphill, A., Muller, N., 2007. Characterization of Giardia
lamblia WB C6 clones resistant to nitazoxanide and to metronidazole. J.
Antimicrob. Chemother. 60, 280–287.
Murtagh Jr., J.J., Mowatt, M.R., Lee, C.M., Lee, F.J., Mishima, K., Nash, T.E., Moss, J.,
Vaughan, M., 1992. Guanine nucleotide-binding proteins in the intestinal
parasite Giardia lamblia. Isolation of a gene encoding an approximately 20-kDa
ADP-ribosylation factor. J. Biol. Chem. 267, 9654–9662.
Palm, J.E., Weiland, M.E., Griffiths, W.J., Ljungstrom, I., Svard, S.G., 2003.
Identification of immunoreactive proteins during acute human giardiasis. J.
Infect. Dis. 187, 1849–1859.
Panaro, M.A., Cianciulli, A., Mitolo, V., Mitolo, C.I., Acquafredda, A., Brandonisio, O.,
Cavallo, P., 2007. Caspase-dependent apoptosis of the HCT-8 epithelial cell line
induced by the parasite Giardia intestinalis. FEMS Immunol. Med. Microbiol. 51,
302–309.
Papanastasiou, P., Hiltpold, A., Bommeli, C., Kohler, P., 1996. The release of the
variant surface protein of Giardia to its soluble isoform is mediated by the
selective cleavage of the conserved carboxy-terminal domain. Biochemistry 35,
10143–10148.
Papanastasiou, P., McConville, M.J., Ralton, J., Kohler, P., 1997. The variant-specific
surface protein of Giardia, VSP4A1, is a glycosylated and palmitoylated protein.
Biochem. J. 322 (Pt 1), 49–56.
Ratner, D.M., Cui, J., Steffen, M., Moore, L.L., Robbins, P.W., Samuelson, J., 2008.
Changes in the N-glycome, glycoproteins with Asn-linked glycans, of Giardia
lamblia with differentiation from trophozoites to cysts. Eukaryotic Cell 7, 1930–
1940.
Regoes, A., Zourmpanou, D., Leon-Avila, G., van der Giezen, M., Tovar, J., Hehl, A.B.,
2005. Protein import, replication, and inheritance of a vestigial mitochondrion.
J. Biol. Chem. 280, 30557–30563.
Reiner, D.S., Douglas, H., Gillin, F.D., 1989. Identification and localization of cyst-
specific antigens of Giardia lamblia. Infect. Immun. 57, 963–968.
Reiner, D.S., McCaffery, M., Gillin, F.D., 1990. Sorting of cyst wall proteins to a
regulated secretory pathway during differentiation of the primitive eukaryote,
Giardia lamblia. Eur. J. Cell Biol. 53, 142–153.
Ringqvist, E., Palm, J.E., Skarin, H., Hehl, A.B., Weiland, M., Davids, B.J., Reiner, D.S.,
Griffiths, W.J., Eckmann, L., Gillin, F.D., Svard, S.G., 2008. Release of metabolic
enzymes by Giardia in response to interaction with intestinal epithelial cells.
Mol. Biochem. Parasitol. 159, 85–91.
Rivero, M.R., Vranych, C.V., Bisbal, M., Maletto, B.A., Ropolo, A.S., Touz, M.C., 2010.
Adaptor protein 2 regulates receptor-mediated endocytosis and cyst formation
in Giardia lamblia. Biochem J. 428, 33–45.
Rodriguez-Fuentes, G.B., Cedillo-Rivera, R., Fonseca-Linan, R., Arguello-Garcia, R.,
Munoz, O., Ortega-Pierres, G., Yepez-Mulia, L., 2006. Giardia duodenalis: analysis
of secreted proteases upon trophozoite–epithelial cell interaction in vitro.
Memorias do Instituto Oswaldo Cruz 101, 693–696.
Roxstrom-Lindquist, K., Ringqvist, E., Palm, D., Svard, S., 2005. Giardia lamblia-
induced changes in gene expression in differentiated Caco-2 human intestinal
epithelial cells. Infect. Immun. 73, 8204–8208.
Saito-Nakano, Y., Loftus, B.J., Hall, N., Nozaki, T., 2005. The diversity of Rab GTPases
in Entamoeba histolytica. Exp. Parasitol. 110, 244–252.
Samra, H.K., Ganguly, N.K., Garg, U.C., Goyal, J., Mahajan, R.C., 1988. Effect of
excretory-secretory products of Giardia lamblia on glucose and phenylalanine
transport in the small intestine of Swiss albino mice. Biochem. Int. 17, 801–812.
Samuelson, J., Banerjee, S., Magnelli, P., Cui, J., Kelleher, D.J., Gilmore, R., Robbins,
P.W., 2005. The diversity of dolichol-linked precursors to Asn-linked glycans
likely results from secondary loss of sets of glycosyltransferases. Proc. Natl.
Acad. Sci. USA 102, 1548–1553.
Schledzewski, K., Brinkmann, H., Mendel, R.R., 1999. Phylogenetic analysis of
components of the eukaryotic vesicle transport system reveals a common origin
of adaptor protein complexes 1, 2, and 3 and the F subcomplex of the coatomer
COPI. J. Mol. Evol. 48, 770–778.
Sheffield, H.G., Bjorvat, B., 1977. Ultrastructure of the cyst of Giardia lamblia. Am. J.
Trop. Med. Hyg. 26, 23–30.
Silverman, J.M., Reiner, N.E., 2011. Exosomes and other microvesicles in infection
biology: organelles with unanticipated phenotypes. Cell Microbiol. 13, 1–9.
Slavin, I., Saura, A., Carranza, P.G., Touz, M.C., Nores, M.J., Lujan, H.D., 2002.
Dephosphorylation of cyst wall proteins by a secreted lysosomal acid
phosphatase is essential for excystation of Giardia lamblia. Mol. Biochem.
Parasitol. 122, 95–98.
Smith, P.D., Gillin, F.D., Spira, W.M., Nash, T.E., 1982. Chronic giardiasis: studies on
drug sensitivity, toxin production, and host immune response.
Gastroenterology 83, 797–803.
C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480 479
Soltys, B.J., Falah, M., Gupta, R.S., 1996. Identification of endoplasmic reticulum in
the primitive eukaryote Giardia lamblia using cryoelectron microscopy and
antibody to Bip. J. Cell Sci. 109 (Pt 7), 1909–1917.
Stefanic, S., Palm, D., Svard, S.G., Hehl, A.B., 2006. Organelle proteomics reveals
cargo maturation mechanisms associated with Golgi-like encystation vesicles in
the early-diverged protozoan Giardia lamblia. J. Biol. Chem. 281, 7595–7604.
Stefanic, S., Morf, L., Kulangara, C., Regos, A., Sonda, S., Schraner, E., Spycher, C., Wild,
P., Hehl, A.B., 2009. Neogenesis and maturation of transient Golgi-like cisternae
in a simple eukaryote. J. Cell Sci. 122, 2846–2856.
Sun, C.H., McCaffery, J.M., Reiner, D.S., Gillin, F.D., 2003. Mining the Giardia lamblia
genome for new cyst wall proteins. J. Biol. Chem. 278, 21701–21708.
Svard, S.G., Rafferty, C., McCaffery, J.M., Smith, M.W., Reiner, D.S., Gillin, F.D., 1999. A
signal recognition particle receptor gene from the early-diverging eukaryote,
Giardia lamblia. Mol. Biochem. Parasitol. 98, 253–264.
Tai, J.H., Ong, S.J., Chang, S.C., Su, H.M., 1993. Giardiavirus enters Giardia lamblia WB
trophozoite via endocytosis. Exp. Parasitol. 76, 165–174.
Teoh, D.A., Kamieniecki, D., Pang, G., Buret, A.G., 2000. Giardia lamblia rearranges F-
actin and alpha-actinin in human colonic and duodenal monolayers and
reduces transepithelial electrical resistance. J. Parasitol. 86, 800–806.
Thirion, J., Wattiaux, R., Jadot, M., 2003. The acid phosphatase positive organelles of
the Giardia lamblia trophozoite contain a membrane bound cathepsin C activity.
Biol. Cell 95 (2), 99–105.
Tomavo, S., Dubremetz, J.F., Schwarz, R.T., 1992. A family of glycolipids from
Toxoplasma gondii. Identification of candidate glycolipid precursor(s) for
Toxoplasma gondii glycosylphosphatidylinositol membrane anchors. J. Biol.
Chem. 267, 11721–11728.
Touz, M.C., Gottig, N., Nash, T.E., Lujan, H.D., 2002a. Identification and
characterization of a novel secretory granule calcium-binding protein from
the early branching eukaryote Giardia lamblia. J. Biol. Chem. 277, 50557–50563.
Touz, M.C., Nores, M.J., Slavin, I., Carmona, C., Conrad, J.T., Mowatt, M.R., Nash, T.E.,
Coronel, C.E., Lujan, H.D., 2002b. The activity of a developmentally regulated
cysteine proteinase is required for cyst wall formation in the primitive
eukaryote Giardia lamblia. J. Biol. Chem. 277, 8474–8481.
Touz, M.C., Lujan, H.D., Hayes, S.F., Nash, T.E., 2003. Sorting of encystation-
specific cysteine protease to lysosome-like peripheral vacuoles in Giardia
lamblia requires a conserved tyrosine-based motif. J. Biol. Chem. 278, 6420–
6426.
Touz, M.C., Kulakova, L., Nash, T.E., 2004. Adaptor protein complex 1 mediates the
transport of lysosomal proteins from a Golgi-like organelle to peripheral
vacuoles in the primitive eukaryote Giardia lamblia. Mol. Biol. Cell 15, 3053–
3060.
Touz, M.C., Conrad, J.T., Nash, T.E., 2005. A novel palmitoyl acyl transferase controls
surface protein palmitoylation and cytotoxicity in Giardia lamblia. Mol.
Microbiol. 58, 999–1011.
Touz, M.C., Ropolo, A.S., Rivero, M.R., Vranych, C.V., Conrad, J.T., Svard, S.G., Nash,
T.E., 2008. Arginine deiminase has multiple regulatory roles in the biology of
Giardia lamblia. J. Cell Sci. 121, 2930–2938.
Tovar, J., Fischer, A., Clark, C.G., 1999. The mitosome, a novel organelle related to
mitochondria in the amitochondrial parasite Entamoeba histolytica. Mol.
Microbiol. 32, 1013–1021.
Tovar, J., Leon-Avila, G., Sanchez, L.B., Sutak, R., Tachezy, J., van der Giezen, M.,
Hernandez, M., Muller, M., Lucocq, J.M., 2003. Mitochondrial remnant
organelles of Giardia function in iron–sulphur protein maturation. Nature 426,
172–176.
Troeger, H., Epple, H.J., Schneider, T., Wahnschaffe, U., Ullrich, R., Burchard, G.D.,
Jelinek, T., Zeitz, M., Fromm, M., Schulzke, J.D., 2007. Effect of chronic Giardia
lamblia infection on epithelial transport and barrier function in human
duodenum. Gut 56, 328–335.
Van Keulen, H., Steimle, P.A., Bulik, D.A., Borowiak, R.K., Jarroll, E.L., 1998. Cloning of
two putative Giardia lamblia glucosamine 6-phosphate isomerase genes only
one of which is transcriptionally activated during encystment. J. Eukaryot.
Microbiol. 45, 637–642.
Ward, W., Alvarado, L., Rawlings, N.D., Engel, J.C., Franklin, C., McKerrow, J.H., 1997.
A primitive enzyme for a primitive cell: the protease required for excystation of
Giardia. Cell 89, 437–444.
480 C. Faso, A.B. Hehl / International Journal for Parasitology 41 (2011) 471–480
... A stacked Golgi is also not observed in Giardia. In this case though many of the normal Golgi functions are greatly reduced and the sorting functions of the Golgi appear to be carried out by the ER [13,53]. However, during encystation the parasite secretes a large amount of cell-wall material via encystation-specific vesicles and these encystation-specific vesicles do exhibit some features reminiscent of the Golgi [54]. ...
... These vacuoles have characteristics of both lysosomes and endosomes, and they appear to be the only endocytic organelle of Giardia [57]. In addition, peripheral vacuoles are also involved in the trafficking of plasma membrane proteins and secreted soluble proteins [13,53]. This trafficking of proteins to the plasma membrane via the peripheral vacuoles is distinct from the trafficking of the major plasma membrane protein called variant surface protein. ...
Article
Full-text available
Virulence in pathogenic protozoa is often tied to secretory processes such as the expression of adhesins on parasite surfaces or the secretion of proteases to assisted in tissue invasion and other proteins to avoid the immune system. This review is a broad overview of the endomembrane systems of pathogenic protozoa with a focus on Giardia, Trichomonas, Entamoeba, kinetoplastids, and apicomplexans. The focus is on unique features of these protozoa and how these features relate to virulence. In general, the basic elements of the endocytic and exocytic pathways are present in all protozoa. Some of these elements, especially the endosomal compartments, have been repurposed by the various species and quite often the repurposing is associated with virulence. The Apicomplexa exhibit the most unique endomembrane systems. This includes unique secretory organelles that play a central role in interactions between parasite and host and are involved in the invasion of host cells. Furthermore, as intracellular parasites, the apicomplexans extensively modify their host cells through the secretion of proteins and other material into the host cell. This includes a unique targeting motif for proteins destined for the host cell. Most notable among the apicomplexans is the malaria parasite, which extensively modifies and exports numerous proteins into the host erythrocyte. These modifications of the host erythrocyte include the formation of unique membranes and structures in the host erythrocyte cytoplasm and on the erythrocyte membrane. The transport of parasite proteins to the host erythrocyte involves several unique mechanisms and components, as well as the generation of compartments within the erythrocyte that participate in extraparasite trafficking.
... Giardia intestinalis, Giardia duodenalis) is a small-intestine protist parasite with a worldwide distribution. Giardia presents a simplified endomembrane system, with only five membranebound compartments, including an extensive ER, and no detectable Golgi apparatus [12][13][14] despite constitutive trafficking of secretory variant surface proteins [15][16][17]. The sole port of entry for fluid-phase material in the Giardia cell are the recently renamed [18] peripheral vacuoles/peripheral endocytic compartments (PVs/PECs), small (ca. ...
Article
Full-text available
Unconventional protein secretion (UPS) plays important roles in cell physiology. In contrast to canonical secretory routes, UPS does not generally require secretory signal sequences and often bypasses secretory compartments such as the ER and the Golgi apparatus. Giardia lamblia is a protist parasite with reduced subcellular complexity which releases several proteins, some of them virulence factors, without canonical secretory signals. This implicates UPS at the parasite–host interface. No dedicated machinery nor mechanism(s) for UPS in Giardia are currently known, although speculations on the involvement of endocytic organelles called PV/PECs, have been put forth. To begin to address the question of whether PV/PECs are implicated in virulence-associated UPS and to define the composition of molecular machinery involved in protein release, we employed affinity purification and mass spectrometry, coupled to microscopy-based subcellular localization and signal correlation quantification to investigate the interactomes of 11 reported unconventionally secreted proteins, all predicted to be cytosolic. A subset of these are associated with PV/PECs. Extended and validated interactomes point to a core PV/PECs-associated UPS machinery, which includes uncharacterized and Giardia-specific coiled-coil proteins and NEK kinases. Finally, a subset of the alpha-giardin protein family was enriched in all PV/PECs-associated protein interactomes, highlighting a previously unappreciated role for these proteins at PV/PECs and in UPS. Taken together, our results provide the first characterization of a virulence-associated UPS protein complex in Giardia lamblia at PV/PECs, suggesting a novel link between these primarily endocytic and feeding organelles and UPS at the parasite–host interface.
... The cellular evolution of the Giardia genus as an obligate parasite adapted to the small intestinal niche of vertebrates is characterized by a reduction in subcellular compartment diversity. Peroxisomes, late endosomes and a permanent stacked Golgi complex have not been detected in Giardia [20]. Two nuclei [21], an extensive endoplasmic reticulum (ER) [22], highly reduced mitochondria-derived organelles-the mitosomes [23]-and peripheral vesicles (PVs) [24] are the only membranebounded organelles with conserved morphology and function documented in the Giardia trophozoite [25][26][27]. ...
Article
Full-text available
Background Giardia lamblia , a parasitic protist of the Metamonada supergroup, has evolved one of the most diverged endocytic compartment systems investigated so far. Peripheral endocytic compartments, currently known as peripheral vesicles or vacuoles (PVs), perform bulk uptake of fluid phase material which is then digested and sorted either to the cell cytosol or back to the extracellular space. Results Here, we present a quantitative morphological characterization of these organelles using volumetric electron microscopy and super-resolution microscopy (SRM). We defined a morphological classification for the heterogenous population of PVs and performed a comparative analysis of PVs and endosome-like organelles in representatives of phylogenetically related taxa, Spironucleus spp. and Tritrichomonas foetus . To investigate the as-yet insufficiently understood connection between PVs and clathrin assemblies in G. lamblia , we further performed an in-depth search for two key elements of the endocytic machinery, clathrin heavy chain (CHC) and clathrin light chain (CLC), across different lineages in Metamonada. Our data point to the loss of a bona fide CLC in the last Fornicata common ancestor (LFCA) with the emergence of a protein analogous to CLC ( Gl ACLC) in the Giardia genus. Finally, the location of clathrin in the various compartments was quantified. Conclusions Taken together, this provides the first comprehensive nanometric view of Giardia ’s endocytic system architecture and sheds light on the evolution of Gl ACLC analogues in the Fornicata supergroup and, specific to Giardia, as a possible adaptation to the formation and maintenance of stable clathrin assemblies at PVs.
... This contrasts with the lack of strong overall reduction in the genome or of metabolic capacities, as noted in the initial report of the draft Helicosporidium genome (Pombert et al., 2014). Nonetheless, many parasites, such as Giardia, apicomplexans, Entamoeba, and kinetoplastids, have reduced their complement of membrane-trafficking machinery (Baum et al., 2008;Faso & Hehl, 2011;Jackson et al., 2016;Klinger et al., 2013;Ralston, 2015;Watanabe et al., 2020). Moreover, specific modifications or reductions in the MTC complement of parasitic lineages have been noted, with the apicomplexans carrying particularly reduced systems (Woo et al., 2015). ...
Article
The chlorophyte algae are a dominant group of photosynthetic eukaryotes. Although many are photoautotrophs, there are also mixotrophs, heterotrophs, and even parasites. The physical characteristics of green algae are also highly diverse, varying greatly in size, shape, and habitat. Given this morphological and trophic diversity, we postulated that diversity may also exist in the protein components controlling intracellular movement of material by vesicular transport. One such set is the multi‐subunit tethering complexes (MTCs) –components regulating cargo delivery. As they span endomembrane organelles and are well‐conserved across eukaryotes, MTCs should be a good proxy for assessing the evolutionary dynamics across the diversity of Chlorophyta. Our results reveal that while green algae carry a generally conserved and unduplicated complement of MTCs, some intriguing variation exists. Notably, we identified incomplete sets of TRAPPII, exocyst, and HOPS/CORVET components in all Mamiellophyceae, and what is more, not a single subunit of Dsl1 was found in Cymbomonas tetramitiformis. As the absence of Dsl1 has been correlated with having unusual peroxisomes, we searched for peroxisome biogenesis machinery, finding very few components in Cymbomonas, suggestive of peroxisome degeneration. Overall, we demonstrate conservation of MTCs across green algae, but with notable taxon‐specific losses suggestive of unusual endomembrane systems.
... In the small intestine, cell density, lipid starvation, and alkaline pH trigger a portion of trophozoites to initiate encystation (5)(6)(7)(8). During the early stage of Giardia encystation, major morphological changes occur that include biogenesis of the Golgi-like encystation-specific vesicles (ESVs), encystation carbohydrate-positive vesicles (ECVs), cell-cycle arrest, and cytoskeleton rearrangement (5,6,9). Giardia's cyst wall is composed of 37% cyst wall proteins (CWPs) and 63% N-acetylgalactosamine (GalNAc) carbohydrate filaments, which are mainly made up of a [D-GalNAc-β(1-3)-D-GalNAc] n homopolymer (10)(11)(12). ...
Article
Full-text available
Significance Transition between life cycle stages is a common feature among parasitic pathogens, and its regulation must be optimized to balance persistence of infection with transmission. The early transcription factors (TFs) regulating commitment to differentiate are totally unknown in Giardia . In this work, we identified Golden2, ARR-B, Psr-1–like protein 1 (GARP)–like protein 4 (GLP4), a previously uncharacterized GARP-like TF, as an early-acting transcriptional repressor that inhibits G1+S cells from entering the encystation pathway. GLP4 is therefore a key regulator controlling the balance between proliferative growth and terminal differentiation into infective cysts.
... Most MVBs have a mean diameter of 400-500 nm (Gruenberg & Stenmark, 2004) and contain small (around 50 nm diameter) round vesicles designated as intraluminal vesicles (ILVs; Williams & Urbé, 2007). Several studies point to the existence of MVBs in G. intestinalis and that the PVs are part of this system, in addition to their functional role as early and late endosomes and lysosomes (Abodeely et al., 2009;Lanfredi-Rangel et al., 1998) and involvement in the parasitic trafficking pathway (Faso & Hell, 2011). ...
Article
Full-text available
Giardia intestinalis has unique characteristics, even in the absence of certain organelles. For instance, Golgi and mitochondria are not found. On the other hand, there is a network of peripheral vacuoles (PVs) and mitosomes. The endoplasmic reticulum (ER), nuclear membrane, peroxisomes, and lipid bodies are present. The peripheral vacuole system seems to play several simultaneous roles. It is involved in the endocytic activity of the trophozoite but also has characteristics of early and late endosomes and even lysosomes, establishing a connection with the ER. Some of the PVs contain small vesicles, acting as multivesicular bodies, including the release of exosomes. The mitosomes are surrounded by two membranes, divide during mitosis, and are distributed throughout the cell. They do not contain DNA, enzymes involved in the citric acid cycle, respiratory chain, or ATP synthesis. However, they contain the iron‐sulfur complex and transporters as TOM and TIM. Some mitosomes are linked to flagellar axonemes through a fibrillar connection. During encystation, two types of larger cytoplasmic vesicles appear. One originating from the ER contains the cyst wall proteins. Another contains carbohydrates. Both migrate to the cell periphery and fuse with plasma membrane secreting their contents to give rise to the cell wall
... During the early encystation phase, the adhesive disc is disassembled [8], the flagella are internalized, the cells round up, and the cyst wall formation starts [6]. In addition, the formation of Encystation Specific 2 of 19 Vesicles (ESVs) occurs, and ESVs transport the three major cyst wall proteins (CWPs [1][2][3] to the cell surface [9]. The CWPs make up most of the cyst wall, together with fibers of N-acetylgalactosamine, a sugar synthesized by enzymes, which are induced early in the encystation process [10]. ...
Article
Full-text available
Giardia intestinalis is an intestinal protozoan parasite that causes diarrheal infections worldwide. A key process to sustain its chain of transmission is the formation of infectious cysts in the encystation process. We combined deep RNAseq of a broad range of encystation timepoints to produce a high-resolution gene expression map of Giardia encystation. This detailed transcriptomic map of encystation confirmed a gradual change of gene expression along the time course of encystation, showing the most significant gene expression changes during late encystation. Few genes are differentially expressed early in encystation, but the major cyst wall proteins CWP-1 and -2 are highly up-regulated already after 3.5 h encystation. Several transcription factors are sequentially up-regulated throughout the process, but many up-regulated genes at 7, 10, and 14 h post-induction of encystation have binding sites in the upstream regions for the Myb2 transcription factor, suggesting that Myb2 is a master regulator of encystation. We observed major changes in gene expression of several meiotic-related genes from 10.5 h of encystation to the cyst stage, and at 17.5 h encystation, there are changes in many different metabolic pathways and protein synthesis. Late encystation, 21 h to cysts, show extensive gene expression changes, most of all in VSP and HCMP genes, which are involved in antigenic variation, and genes involved in chromatin modifications. This high-resolution gene expression map of Giardia encystation will be an important tool in further studies of this important differentiation process.
... The causative agent is the diplomonad Giardia intestinalis. This enteric protist parasite has undergone large genome streamlining and modifications in its typical eukaryotic organelles, particularly in its endomembrane system and the associated trafficking complement [2]. ...
Article
Full-text available
Background Comparing a parasitic lineage to its free-living relatives is a powerful way to understand how that evolutionary transition to parasitism occurred. Giardia intestinalis (Fornicata) is a leading cause of gastrointestinal disease world-wide and is famous for its unusual complement of cellular compartments, such as having peripheral vacuoles instead of typical endosomal compartments. Endocytosis plays an important role in Giardia ’s pathogenesis. Endosomal sorting complexes required for transport (ESCRT) are membrane-deforming proteins associated with the late endosome/multivesicular body (MVB). MVBs are ill-defined in G. intestinalis , and roles for identified ESCRT-related proteins are not fully understood in the context of its unique endocytic system. Furthermore, components thought to be required for full ESCRT functionality have not yet been documented in this species. Results We used genomic and transcriptomic data from several Fornicata species to clarify the evolutionary genome streamlining observed in Giardia, as well as to detect any divergent orthologs of the Fornicata ESCRT subunits. We observed differences in the ESCRT machinery complement between Giardia strains. Microscopy-based investigations of key components of ESCRT machinery such as Gi VPS36 and Gi VPS25 link them to peripheral vacuoles, highlighting these organelles as simplified MVB equivalents. Unexpectedly, we show ESCRT components associated with the endoplasmic reticulum and, for the first time, mitosomes. Finally, we identified the rare ESCRT component CHMP7 in several fornicate representatives, including Giardia and show that contrary to current understanding, CHMP7 evolved from a gene fusion of VPS25 and SNF7 domains, prior to the last eukaryotic common ancestor, over 1.5 billion years ago. Conclusions Our findings show that ESCRT machinery in G. intestinalis is far more varied and complete than previously thought, associates to multiple cellular locations, and presents changes in ESCRT complement which pre-date adoption of a parasitic lifestyle.
... and the classical endo-lysosomal system [24]. Many acidified peripheral vesicles (PVs) have been found near the plasma membrane of Giardia trophozoites and cyst forms which seems to be related to the presence of endosomes and lysosomes [24,26]. Focused-ion-beam scanning electron microscopy showed that PVs contain intraluminal vesicles (ILVs) with a size between 50 and 100 nm and a range of one to seven for trophozoites and one for the cyst forms [27]. ...
Article
In recent years, several parasites have been shown to interact with their hosts through intra- and inter-community communication mechanisms, which were identified to be mediated by extracellular vesicles (EVs) through various uptake mechanisms. EVs are a heterogenous group of nanoparticles (~30–5000 nm) classified into three main types according to their size and biogenesis. EVs contain proteins, lipids, nucleic acids and metabolites from the cell of origin which are essential for genetic exchange, biomarker identification and diagnosis of pathological diseases. As important “forward lines of parasite infectivity”, the parasite-secreted EVs function as information transmitters in the early-stage of host-parasite interaction and subsequent host-cell colonization. For this review, we summarize from the literature the relevance of EVs to the pathogenesis and development of human parasitic protistan diseases such as giardiasis, leishmaniasis, amoebiasis, malaria and Blastocystis-mediated gut pathology. Specific in vitro and in vivo interactions of the parasite-EVs and the host, with the reported cellular and immunological outcomes are discussed in this review. EVs have great potential to be further developed as diagnostic, immunomodulation and therapeutic alternatives to fill the knowledge gaps in the current parasitic diseases discussed in this review. Nanomedicine and vaccine development could be explored, with the utilization and/or modification of the parasitic EVs as novel treatment and prevention strategies.
Article
Bovine trichomonosis (BT), a disease of the bovine urogenital tract, is caused by the protozoan Tritrichomonas foetus (Tf). Tf causes endometritis, infertility, and premature death of the embryo, which generates considerable economic losses. The proteins released can mediate fundamental interactions between the pathogen and the host, triggering factors associated with the symptomatology, immune evasion and pathogenesis characteristic of the species. However, little is known about the profile of the proteins released by Tf. In order to contribute to their knowledge, we performed an isolation protocol and a proteomic profiling of the supernatant (SN) content of six Tf isolates. A total of 662 proteins present in the SN of Tf were detected, out of which 121 were shared by the six isolates, while the remaining 541 were found in at least one of the isolates studied. The comparative analyses using the databases of Tf strain genome K revealed 32.9% of uncharacterized proteins. The bioinformatic analyses showed that the main molecular functions predicted were binding (47.9%) and catalytic activity (38.2%). Additionally, we performed immunodetection assays to evidence the antigenic potential of SN proteins. Interestingly, we observed great ability to detect SN proteins from all six isolates using serum from immunized mice and infected bulls. A complementary mass spectrometry assay allowed us to determine that the proteins that showed the strongest signal intensity in the immunoassays were Grp78 (A0A1J4IZS3) and Ap65 (A0A1J4JSR1). This work represents the first proteomic characterization of Tf SN proteins and their antigenic potential, which might be interesting for the future design of new diagnosis and treatment methods for BT.
Article
Full-text available
Eukaryotic vesicular transport requires the recognition of membranes through specific protein complexes. The heterotetrameric adaptor protein complexes 1, 2, and 3 (AP1/2/3) are composed of two large, one small, and one medium adaptin subunit. We isolated and characterized the cDNA for Arabidopsisγ-adaptin and performed a phylogenetic analysis of all adaptin subunits (proteins) in the context of all known homologous proteins. This analysis revealed (i) that the large subunits of AP1/2/3 are homologous and (ii) two subunits of the heptameric coatomer I (COPI) complex belong to this gene family. In addition, all small subunits and the aminoterminal domain of the medium subunits of the heterotetramers are homologous to each other; this also holds for two corresponding subunits of the COPI complex. AP1/2/3 and a substructure (heterotetrameric, F-COPI subcomplex) of the heptameric COPI had a common ancestral complex (called pre-F-COPI). Since all large and all small/medium subunits share sequence similarity, the ancestor of this complex is inferred to have been a heterodimer composed of one large and one small subunit. The situation encountered today is the result of successive rounds of coordinated gene duplications of both the large and the small/medium subunits, with F-COPI being the first that separated from the ancestral pre-F-COPI.
Article
Full-text available
In preparation for being shed into the environment as infectious cysts, trophozoites of Giardia spp. synthesize and deposit large amounts of extracellular matrix into a resistant extracellular cyst wall. Functional aspects of this developmentally regulated process were investigated by expressing a series of chimeric cyst wall protein 1 (CWP1)-green fluorescent protein (GFP) reporter proteins. It was demonstrated that a short 110 bp 5' flanking region of the CWP1 gene harbors all necessary cis-DNA elements for strictly encystation-specific expression of a reporter during in vitro encystation, whereas sequences in the 3' flanking region are involved in modulation of steady-state levels of its mRNA during encystation. Encysting Giardia expressing CWP1-GFP chimeras showed formation and maturation of labeled dense granule-like vesicles and subsequent incorporation of GFP-tagged protein into the cyst wall, dependent on which domains of CWP1 were included. The N-terminal domain of CWP1 was required for targeting GFP to regulated compartments of the secretory apparatus, whereas a central domain containing leucine-rich repeats mediated association of the chimera with the extracellular cyst wall. We show that analysis of protein transport using GFP-tagged molecules is feasible in an anaerobic organism and provides a useful tool for investigating the organization of primitive eukaryotic vesicular transport.
Article
This report examines the presence of proteolytic activity detected in media collected from in vitro cultures of Giardia intestinalis, and the partial characterization by gelatin-substrate polyacrylamide gel electrophoresis and inhibition studies. Gelatin-substrate polyacrylamide gel electrophoresis revealed 6 bands with proteolytic activity, with estimated molecular weights of 36, 59, 63, 72, 103, and 175 kDa. These bands were not present in the control medium. On the other hand, G. intestinalis trophozoite lysates showed proteolytic bands at 16, 20, 66, 82, 108, and 120 kDa, thus indicating that intracellular proteases could be different from the excretory/secretory (E/S) products. Based on inhibition studies, 2 bands of 59 and 63 kDa were inhibited by iodoacetic acid, indicating the presence of cysteine proteases. Partial inhibition of a band of 36 kDa was found with EDTA, a metal-chelating agent, suggesting the possible presence of metalloproteases. The presence of aspartic and serine proteases were not detected under the assay conditions used. As G. intestinalis E/S may be involved in differentiation mechanisms of the parasite and also be responsible for the mucosal alterations that occur in giardiasis, the characterization of these proteases may facilitate their evaluation as targets in the therapy of the disease.
Article
The mechanisms of epithelial injury in giardiasis remain unknown. The effects of live Giardia lamblia on cellular G-actin, F-actin, α-actinin, and electrical resistance of human intestinal epithelial monolayers were investigated using SCBN and Caco2 cell lines grown on chamber slides or Transwell[Circled Trademark Sign] filter membranes. In separate experiments, some monolayers were also exposed to sonicated trophozoites, some to supernatant from live G. lamblia cultures, and some with or without the Ca2+ channel blocker verapamil. After 2, 24, or 48 hr of coincubation with G. lamblia, monolayers were assessed for cytoskeletal arrangement under fluorescence and confocal laser microscopy, and transepithelial electrical resistance was measured. Exposure to live G. lamblia trophozoites induced localized condensation of F-actin and loss of perijunctional α-actinin while G-actin remained unchanged. Confocal laser microscopy indicated that F-actin rearrangement was not affected by verapamil and was localized within the terminal web area. Coincubation of monolayers with G. lamblia lysates or with spent medium alone similarly rearranged F-actin. Verapamil alone did not alter F-actin. Electrical resistance of SCBN and Caco2 monolayers exposed to G. lamblia was significantly decreased versus controls regardless of whether live or lysed trophozoite samples were used. The results indicate that G. lamblia-induced epithelial injury is associated with F-actin and α-actinin rearrangements in the terminal web area via mechanisms independent of extracellular Ca2+. These alterations are associated with reduced transepithelial electrical resistance and are due at least in part to trophozoite products.
Article
A revised six-kingdom system of life is presented, down to the level of infraphylum. As in my 1983 system Bacteria are treated as a single kingdom, and eukaryotes are divided into only five kingdoms: Protozoa, Animalia, Fungi, Plantae and Chromista. Intermediate high level categories (superkingdom, subkingdom, branch, infrakingdom, superphylum, subphylum and infraphylum) are extensively used to avoid splitting organisms into an excessive number of kingdoms and phyla (60 only being recognized). The two 'zoological' kingdoms, Protozoa and Animalia, are subject to the International Code of Zoological Nomenclature, the kingdom Bacteria to the International Code of Bacteriological Nomenclature, and the three 'botanical' kingdoms (Plantae, Fungi, Chromista) to the International Code of Botanical Nomenclature. Circumscriptions of the kingdoms Bacteria and Plantae remain unchanged since Cavalier-Smith (1981). The kingdom Fungi is expanded by adding Microsporidia, because of protein sequence evidence that these amitochondrial intracellular parasites are related to conventional Fungi, not Protozoa. Fungi are subdivided into four phyla and 20 classes; fungal classification at the rank of subclass and above is comprehensively revised. The kingdoms Protozoa and Animalia are modified in the light of molecular phylogenetic evidence that Myxozoa are actually Animalia, not Protozoa, and that mesozoans are related to bilaterian animals. Animalia are divided into four subkingdoms: Radiata (phyla Porifera, Cnidaria, Placozoa, Ctenophora), Myxozoa, Mesozoa and Bilateria (bilateral animals: all other phyla). Several new higher level groupings are made in the animal kingdom including three new phyla: Acanthognatha (rotifers, acanthocephalans, gastrotrichs, gnathostomulids), Brachiozoa (brachiopods and phoronids) and Lobopoda (onychophorans and tardigrades), so only 23 animal phyla are recognized. Archezoa, here restricted to the phyla Metamonada and Trichozoa, are treated as a subkingdom within Protozoa, as in my 1983 six-kingdom system, not as a separate kingdom. The recently revised phylum Rhizopoda is modified further by adding more flagellates and removing some 'rhizopods' and is therefore renamed Cercozoa. The number of protozoan phyla is reduced by grouping Mycetozoa and Archamoebae (both now infraphyla) as a new subphylum Conosa within the phylum Amoebozoa alongside the subphylum Lobosa, which now includes both the traditional aerobic lobosean amoebae and Multicilia. Haplosporidia and the (formerly microsporidian) metchnikovellids are now both placed within the phylum Sporozoa. These changes make a total of only 13 currently recognized protozoan phyla, which are grouped into two subkingdoms: Archezoa and Neozoa; the latter is modified in circumscription by adding the Discicristata, a new infrakingdom comprising the phyla Percolozoa and Euglenozoa). These changes are discussed in relation to the principles of megasystematics, here defined as systematics that concentrates on the higher levels of classes, phyla, and kingdoms. These principles also make it desirable to rank Archaebacteria as an infrakingdom of the kingdom Bacteria, not as a separate kingdom. Archaebacteria are grouped with the infrakingdom Posibacteria to form a new subkingdom, Unibacteria, comprising all bacteria bounded by a single membrane. The bacterial subkingdom Negibacteria, with separate cytoplasmic and outer membranes, is subdivided into two infrakingdoms: Lipobacteria, which lack lipopolysaccharide and have only phospholipids in the outer membrane, and Glycobacteria, with lipopolysaccharides in the outer leaflet of the outer membrane and phospholipids in its inner leaflet. This primary grouping of the 10 bacterial phyla into subkingdoms is based on the number of cell-envelope membranes, whilst their subdivision into infrakingdoms emphasises their membrane chemistry; definition of the negibacterial phyla, five at least partly photosynthetic, relies chiefly on photosynthetic mechanism and cell-envelope structure and chemistry corroborated by ribosomal RNA phylogeny. The kingdoms Protozoa and Chromista are slightly changed in circumscription by transferring subphylum Opalinata (classes Opalinea, Proteromonadea, Blastocystea cl. nov.) from Protozoa into infrakingdom Heterokonta of the kingdom Chromista. Opalinata are grouped with the subphylum Pseudofungi and the zooflagellate Developayella elegans (in a new subphylum Bigyromonada) to form a new botanical phylum (Bigyra) of heterotrophs with a double ciliary transitional helix, making it necessary to abandon the phylum name Opalozoa, which formerly included Opalinata. The loss of ciliary retronemes in Opalinata is attributed to their evolution of gut commensalism. The nature of the ancestral chromist is discussed in the light of recent phylogenetic evidence.
Article
The WB isolate of Giardia lamblia expresses a cysteine-rich 170-kD surface antigen (CRP170) that undergoes antigenic variation. An (6E7), cytotoxic for isolates expressing CRP170, was used in another study to select antigenic variants from clones of the WB isolate of Giardia. CRP170 was replaced by surface-labeled bands ranging in size from approximately 50 to 170 kD. In this study, mAb 6E7 was used to isolate a 1-kb portion of the CRP170 gene (M2-1) from a lambda gt 11 expression library. The M2-1 clone hybridized to a 5.4-kb transcript from isolates expressing CRP170 but did not hybridize to RNA from antigenic variants. Evidence was found for frequent rearrangements at the CRP170 gene locus. DNA sequencing of the M2-1 clone revealed the presence of long tandem repeats. The putative amino acid sequence of M2-1 reveals a 12% cysteine content, and CRP170 is readily labeled in vivo with cysteine.
Article
The cyst wall ofGiardia lambliais essential for survival of the parasite outside the host.N-acetyl-d-glucosamine (GalNAc) has been reported as a major terminal sugar of cyst wall glycoproteins andN-acetyl-d-galactosamine (GalNAc) as the major sugar of the fibrous insoluble cyst wall fraction. Therefore, we measured UDP-glycosyltransferase activities as the incorporation of [3H]UDP-sugar into an alcohol-insoluble product. We found that during encystation only UDP-GlcNAc and UDP-GalNAc transferase (UDP-GT) activities increased approximately three- to fivefold compared to nonencysting trophozoites. These activities were distributed approximately equally in the pellet and soluble fractions. The apparentKmand Vmaxof UDP-GT in these fractions were similar. The activities from both fractions were dependent on Mn2+; however, the pellet enzymes were also partially activated by other metal ions. Both pUDP-GT and sUDP-GT were inhibited by uridine, UDP, and UDP sugars, but not by GlcNAc or GalNAc. Isolation and analysis of the reaction products suggest that pUDP-GT incorporate GlcNAc and GalNAc into glycoproteins, since the products were proteinase sensitive. In contrast, sUDP-GT products were resistant to proteinase treatment. Hydrolysis of the product of UDP-GlcNAc-T incorporation by trifluoroacetic acid released only glucosamine, while both glucosamine and galactosamine were released from UDP-GalNAc-T products, supporting the presence of an epimerase inGiardiawhich can convert GalNAc to GlcNAc during incorporation. This study suggests that at least two UDP-GT activities are induced during encystation, which are responsible for the transfer of GlcNAc and GalNAc from UDP-GlcNAc or UDP-GalNAc into both proteinase-sensitive and proteinase-resistant components of theGiardiacyst wall.
Article
The release of exosomes and other microvesicles by diverse prokaryotic and eukaryotic cells and organisms was first appreciated early in the 20th century. The functional properties of these organelles, however, have only recently been the focus of rigorous investigation. In this review, we discuss the release of microvesicles of varying complexity by diverse microbial pathogens. This includes vesicle secretion by Gram-negative bacteria, eukaryotic parasites of the kinetoplast lineage and opportunistic fungal pathogens of both the ascomycetes and basidiomycetes lineages. We also discuss vesicle release from mammalian cells brought about as a result of infection with bacteria, viruses and prions. In addition, we review the evidence showing that in their specific microenvironments, release of these organelles from diverse pathogens contributes to pathogenesis. Germane to this and based upon recent findings with Leishmania, we propose a model whereby exosome release by an intracellular pathogen serves as a general mechanism for effector molecule delivery from eukaryotic pathogen to host cell cytosol. These new findings linking exosomes and other microvesicles to infection biology have important implications for understanding the immune response to infection and for the design of research strategies aimed at the development of novel therapeutics and vaccines.