Sei sulla pagina 1di 22

MI65CH27-McConville

ARI

ANNUAL
REVIEWS

27 July 2011

9:24

Further

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

Click here for quick links to


Annual Reviews content online,
including:
Other articles in this volume
Top cited articles
Top downloaded articles
Our comprehensive search

Metabolic Pathways Required


for the Intracellular Survival
of Leishmania
Malcolm J. McConville and Thomas Naderer
Department of Biochemistry and Molecular Biology, University of Melbourne,
Bio21 Institute of Molecular Science and Biotechnology, Parkville, Victoria 3010, Australia;
email: malcolmm@unimelb.edu.au, thomas.naderer@monash.edu

Annu. Rev. Microbiol. 2011. 6:54361

Keywords

First published online as a Review in Advance on


June 20, 2011

leishmaniasis, intracellular pathogens, protozoan parasites,


phagolysosome, macrophage, virulence

The Annual Review of Microbiology is online at


micro.annualreviews.org
This articles doi:
10.1146/annurev-micro-090110-102913
c 2011 by Annual Reviews.
Copyright 
All rights reserved
0066-4227/11/1013-0543$20.00

Abstract
Leishmania spp. are sandy-transmitted parasitic protozoa that cause a
spectrum of important diseases and lifelong chronic infections in humans. In the mammalian host, these parasites proliferate within acidied
vacuoles in several phagocytic host cells, including macrophages, dendritic cells, and neutrophils. In this review, we discuss recent progress
that has been made in dening the nutrient composition of the Leishmania parasitophorous vacuole, as well as metabolic pathways required
by these parasites for virulence. Analysis of the virulence phenotype
of Leishmania mutants has been particularly useful in dening carbon
sources and nutrient salvage pathways that are essential for parasite persistence and/or induction of pathology. We also review data suggesting
that intracellular parasite stages modulate metabolic processes in their
host cells in order to generate a more permissive niche.

543

MI65CH27-McConville

ARI

27 July 2011

9:24

Contents

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

INTRODUCTION . . . . . . . . . . . . . . . . . .
THE LEISHMANIA
PARASITOPHOROUS
VACUOLE . . . . . . . . . . . . . . . . . . . . . . .
Nutrient Levels in the
Leishmania PV . . . . . . . . . . . . . . . . . .
LEISHMANIA DIFFERENTIATION
AND METABOLIC
ADAPTATION TO THE
MAMMALIAN HOST . . . . . . . . . . . .
Carbon Metabolism in Intracellular
Amastigotes . . . . . . . . . . . . . . . . . . . .
Metabolic Pathways Required for
Protection Against Oxidative
Stress . . . . . . . . . . . . . . . . . . . . . . . . . .
Glycosylation Pathways Required
for Thermotolerance and
Virulence . . . . . . . . . . . . . . . . . . . . . . .
MODULATION OF HOST
METABOLISM BY
LEISHMANIA . . . . . . . . . . . . . . . . . . . . .
PERSISTENCE VERSUS
PATHOLOGY . . . . . . . . . . . . . . . . . . . .
CONCLUSIONS . . . . . . . . . . . . . . . . . . . .

544

546
547

548
549

551

551

552
553
554

INTRODUCTION

iNOS: inducible
nitric oxide synthase
or NOS2

544

Leishmania spp. comprise an important group


of trypanosomatid parasites that are transmitted by sandy vectors and cause a range of
diseases in humans (72). Depending on the
infecting species and other factors such as host
immunity, these parasites may induce localized,
self-curing, cutaneous lesions (cutaneous leishmaniasis, CL) or disseminate to facial mucosal
tissues or to the liver, spleen, and bone marrow
to cause serious and life-threatening mucocutaneous (ML) and visceral (VL) forms of
leishmaniasis, respectively (72). It is estimated
that 1.5 to 2 million children and adults develop
symptomatic diseases each year, resulting in
more than 70,000 deaths (primarily from VL)
and an infection prevalence of 12 million
people (72). However, these gures are likely
McConville

Naderer

to signicantly underestimate the true disease


burden (7). Only a limited number of drugs
are available for treating severe CL and cases
of ML and VL, although none is optimal
due to toxicity or teratogenicity, expense,
requirement for hospitalization, and/or the
widespread emergence of drug resistance (23).
Over 20 species of Leishmania are transmitted by phlebotomine sandies (Phlebotomus spp.
or Lutzomyia spp.) to humans or alternative
animal hosts. Leishmania develop within the
midgut of the sandy vector as agellated
promastigote stages that transform through
a number of physiological states, culminating
in the nondividing, metacyclic promastigotes
that are preadapted for life in the mammalian
host. Metacyclic promastigotes are injected
into the skin when female sandies take a
blood meal, and are phagocytosed by a variety
of host cells, including neutrophils, dendritic
cells, and macrophages, that are equipped to
clear invading microbes. However, internalized promastigotes differentiate to nonmotile
amastigotes that can replicate within lysosomelike compartments, or parasitophorous vacuoles (PVs), within these cells. In the various
murine models of infection and in humans,
Leishmania evade the innate immune responses
of these host cells, as parasite numbers increase
markedly during the rst few weeks of infection
before the development of acquired immunity
(6). Protective acquired immunity is dependent
on CD4+ and CD8+ T cells with T-helper
type 1 (Th1) cytokine proles (72). The release
of proinammatory cytokines such as gamma
interferon (IFN-), interleukin 12 (IL-12),
and tumor necrosis factor (TNF) leads to the
activation of potent anti-leishmanial activities,
such as induction of inducible NO synthesis
(iNOS) and the oxidative burst in infected
host cells and a decrease in parasitemia (72).
Conversely, induction of T-helper type 2
(Th2) cytokines (IL-4, IL-13) and/or production of the antiinammatory IL-10 can lead
to development of disease and a noncuring
phenotype (97). These immune responses
allow Leishmania parasites to persist in multiple
tissues after spontaneous or treatment-induced

MI65CH27-McConville

ARI

27 July 2011

9:24

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

resolution of clinical disease (97). Persistent


parasites can confer immunity to reinfection
in humans and animals but are also a source of
recrudescence if protective T-cell mechanisms
fail, such as occurs in severe HIV/AIDS (7).
It is generally assumed that macrophages
are the major host cell infected by Leishmania.
However, infected monocytes and monocytederived dendritic cells are often abundant in
inammatory lesions and in draining lymph
nodes, while other host cells in these tissues

(eosinophils, neutrophils, and broblasts) can


also be infected (Figure 1) (12, 27, 70). While
much is known about host immune responses
and the cell biology of Leishmania infection,
we are only just beginning to understand how
amastigotes survive within these hostile niches.
In this review we summarize current information on the nutritional composition of the
Leishmania PV and parasite and host cell
metabolic pathways that are essential for intracellular growth. Much of this knowledge is

Lymph
nodes

Inflammatory
skin lesion

Viscerotropic
species

Hepatic/splenic
granuloma

Promastigote

Neutrophils

Macrophage

mo-DC

Amastigote

Monocyte

Kupffer macrophage

Fibroblasts

Figure 1
Multiple host niches occupied by Leishmania parasites. Metacyclic promastigotes injected into the skin by the
sandy are phagocytosed primarily by neutrophils and resident dendritic cells. Promastigotes released from
apoptotic neutrophils are subsequently phagocytosed by macrophages and monocytes recruited to the site of
infection (blue lines). Dendritic cells derived from recruited monocytes (mo-DC) can be major host cells in
inammatory lesions that develop at the site of the bite or in draining lymph nodes (27, 70). Other host cells,
such as neutrophils and eosinophils, can also harbor parasites. Following resolution of clinical disease,
parasites may persist as a quiescent population in nonprofessional phagocytic cells such as broblasts (12).
Viscerotropic species (Leishmania donovani, L. infantum/chagasi ) can disseminate from dermal tissues to the
liver, spleen, and bone marrow to cause visceral leishmaniasis. The major host cells infected in the liver are
resident Kupffer macrophages (5, 71).
www.annualreviews.org Leishmania Metabolism and Virulence

545

MI65CH27-McConville

ARI

27 July 2011

9:24

derived from analysis of the virulence phenotype of Leishmania mutants that are viable in
culture (as promastigotes) but show reduced
growth and/or survival in macrophages and
mice (63, 76).

THE LEISHMANIA
PARASITOPHOROUS VACUOLE

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

Leishmania engage a variety of receptors (CR3,


CR1, and Fc-R) on the surface of host cells
and are internalized by the conventional phagocytic pathway (4). The newly formed phagosomes subsequently mature as a result of highly

RE

regulated fusion and ssion events with early


and late endosomes and lysosomes (4, 70), resulting in the formation of an acidied (pH 4.7
5.2) PV containing the markers of a mature
phagolysosome (Figure 2). Whereas phagosome maturation may be transiently delayed
by the promastigote stages of some species
(i.e., Leishmania major, L. donovani, but not
L. mexicana) (57, 108, 116), phagosomes containing amastigotes mature at the same rate as
those containing inert latex beads. The mature
PV continues to fuse with late endosomes, as
well as other organelles, such as phagosomes
and autophagolysosomes (96, 114). Primary

Autophagosomes

EE/LE/Lys

eRBC

Markers

Phagosomes

Nutrient
and cation
transporters

Cathepsins
-hexaminidase
Hyaluronidase
Ferritin
GRP86
BiP
Glc6Pase
Chitinase

LAMP
V-H-ATPase
Gp91phox
MHC class II
SLC11a1
MHC class I
Sec61
Sec22

PV
Amastigote

Cargo

Glycoproteins
Endoplasmic
reticulum

Proteoglycans
Lipoproteins

Figure 2
Nutrient acquisition pathways in the Leishmania PV. Leishmania amastigotes occupy individual or spacious,
communal PVs that are acidied (pH 4.75.2) and retain all the markers of the phagolysosome
compartment. This compartment fuses with LEs and Lys, containing macromolecules and micronutrients
internalized via RE and EE. It can also fuse with other phagosomes (containing eRBCs) and
autophagosomes. ER vesicle/membranes can also fuse with primary phagosomes and secondary PVs. Sample
PV membrane and lumenal markers derived from fusion with endolysosome (black), the ER (blue), and the
parasite (red ) are indicated in the box (from References 4, 34, and 80). Abbreviations: EE, early endosome;
ER, endoplasmic reticulum; eRBC, effete red blood cell; Glc6Pase, glucose-6-phosphatase; LE, late
endosome; LAMP, lysosomal acidic membrane protein; Lys, lysosome; PV, parasitophorous vacuole;
RE, recycling endosome; SLC11a1, iron transporter.
546

McConville

Naderer

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

phagosomes and secondary Leishmania PVs


can also fuse with, or receive vesicular trafc
from, the endoplasmic reticulum (ER) (34, 80)
(Figure 2). The mature Leishmania PV is thus
a highly dynamic organelle that is continuously
receiving membrane and lumenal content from
a wide variety of organelles in the secretory and
endolysosomal system (Figure 2).
The composition of the PV may differ
depending on the Leishmania species and
host cell involved. Specically, whereas most
species of Leishmania reside within individual, tight-tting PVs that segregate with
dividing daughter cells, L. mexicana complex
(L. mexicana, L. amazonensis, L. pifanoi ) amastigotes induce large communal vacuoles that can
contain up to 2030 amastigotes (4). These
large PVs contain the same membrane and
lumenal markers as the tight-tting PVs (80,
96) and may represent a specic adaptation by
members of this complex to minimize the toxic
effects of short-lived reactive nitrogen intermediates (RNIs) generated by the host (117, 120).
On the other hand, different host cells differ
markedly in their permissiveness to Leishmania
infection and growth. Although macrophages
appear to be the most permissive (often
containing greater than 10 amastigotes per
cell), other professional and nonprofessional
phagocytic cells (dendritic cells, eosinophils,
and broblasts) generally support fewer parasites (1 to 3 amastigotes per cell) (27, 70).
Neutrophils are the least permissive host cell
for Leishmania growth, and most promastigote
stages internalized by neutrophils are delivered
to the mature lysosome and killed (40) either
as result of induction of potent microbicidal
responses and/or severe nutrient restriction
(95). However, some promastigotes can avoid
or stall neutrophil phagosome maturation and
survive within an intermediate compartment
that is nonlytic and contains markers for the
ER (40).

Nutrient Levels in the Leishmania PV


Nutrient levels in the Leishmania PV are
dependent on the rate of inux of host

macromolecules, the activity of various hydrolases, and the rate of transport of lowmolecular-weight metabolites across the PV
membrane (101). Information on the nutrient
composition of the macrophage PV can be inferred from the known growth requirements of
wild-type parasites as well as the growth phenotype of Leishmania mutants that have additional nutrient requirements (21, 30, 46, 81).
Wild-type Leishmania are auxotrophic for, or
have limited capacity to synthesize de novo, a
wide range of amino acids, purines, vitamins,
lipids, and other metabolites (Figure 3) that
must therefore be available within the PV at sufcient levels to sustain amastigote growth (102,
115, 119). In contrast, parasite mutants lacking enzymes needed for gluconeogenesis (73),
the biosynthesis of specic sugars (37), or myoinositol (43) grow poorly in macrophages, indicating that the PV is generally sugar-poor, as
is the case for other compartments in the endolysosome system (33). As a major site of protein degradation, the PV is thought to be generally rich in amino acids. However, the growth
of intracellular amastigotes can be stimulated by
the addition of exogenous arginine or ornithine
to infected macrophages, indicating that
amastigote growth may be limited by the availability of these amino acids (45, 53). Genetic
disruption of amastigote arginase, the initiating enzyme in arginine catabolism, severely restricts intracellular amastigote growth (38, 89),
conrming that amastigotes are dependent on
the de novo synthesis of polyamines as well as on
salvage of polyamines from the PV. Similarly,
the intracellular growth defect of a L. amazonensis mutant lacking the ferric iron transporter
LIT1 indicates that the PV contains sufcient
heme to supply the porphyrin, but not the iron,
needs of amastigotes (42). Collectively, these
ndings suggest that the Leishmania PVs are nutritionally complex compartments that contain
a range of potential carbon sources and essential
nutrients. In this respect, it is notable that Coxiella burnetii, a bacterial pathogen with complex
nutritional requirements, can also proliferate
long term within Leishmania-conditioned PVs
(114).
www.annualreviews.org Leishmania Metabolism and Virulence

ER: endoplasmic
reticulum
Auxotrophy:
a requirement for a
particular nutrient due
to absence of de novo
pathways for its
production

547

MI65CH27-McConville

ARI

27 July 2011

Glucosamine Mannose

9:24

Glucose

Ribose

Aspartate

Glc6P

Rib6P

Oxaloacetic
acid

Threonine

PM
GlcN6P

Man6P

Succinate

Succinate
PPP

Fru6P

Malate

Malate

Fru1, 6P2

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

Oxaloacetic
acid

Oxaloacetic
acid

Man6P

Succinyl-CoA

Malate
TCA
cycle

Glutamate

Citrate

Triose-P
Man1P

Phosphoenolpyruvate

Pyruvate

AcCoA

Acetate
Succinate

GDP-Man
3-phosphoglycerate
Man n

KG

Succinyl-CoA

Phosphoenolpyruvate

Man n + 1

Alanine

Fatty acids

Mannogen
cycle

Trypanothione

Nucleotides
Sugar nucleotides

Polyamines

Cysteine*
AdoMet

Glycine*
Cysteine*
Sphingolipids

Ovathiol

Isoprenoids
Ergosterol

Ascorbate, biotin,
biopterin, folate,
heme, lipoic acid,
NAD, pantothenate,
pyridoxal, riboflavin,
thiamine

Arginine

Methionine

Serine

Histidine

Leucine

Vitamins, etc.

PM
Purine

Figure 3
Metabolic pathways associated with core metabolism that are required for amastigote survival in macrophages. Asterisks indicate the
presence of de novo pathways of synthesis that are insufcient to sustain normal growth without salvage from the PV/medium. Other
essential amino acids that must be salvaged from the PV include Lys, Ile, Val, Phe, Tyr, and Trp. Several pathways required for
amastigote survival are compartmentalized in glycosomes ( blue) or a single mitochondrion (red ). Abbreviations: AcCoA, acetylcoenzyme A; Mann , mannogen oligosaccharides; PM, plasma membrane; PPP, pentose phosphate pathway; PV, parasitophorous
vacuole; Triose-P, triose phosphate.

LEISHMANIA DIFFERENTIATION
AND METABOLIC ADAPTATION
TO THE MAMMALIAN HOST
Whereas promastigote to amastigote differentiation is associated with marked changes
in morphology (including rounding up and
contraction of the cell body and retraction of
the agellum), it has been more difcult to dene the extent to which intracellular processes
such as metabolism are remodeled. Recent
genome-wide transcriptomic and proteomic

548

McConville

Naderer

studies suggest that transcription (mediated by


polymerase II) and protein synthesis are both
globally downregulated in axenic, in vitro
generated amastigotes (1, 54), suggesting that
intracellular stages enter a slow growth state
with reduced metabolic requirements compared to extracellular promastigotes. However,
transcript-proling studies have consistently
shown that relatively few genes (3%9%) are
modulated at the level of individual mRNAs
during amastigote differentiation (28, 91).
This contrasts with the situation in other

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

trypanosomatids, such as Trypanosoma brucei


and T. cruzi, where extensive posttranscriptional control of mRNA levels occurs during
differentiation and can underpin stage-specic
changes in metabolism (87). Although proteomic analyses of axenic and intracellular
Leishmania amastigotes have identied a
number of key enzymes in central carbon
metabolism that do change during promastigote to amastigote differentiation (84, 93), these
changes are again modest when compared with
those observed in T. brucei or in other model
eukaryotic organisms such as Saccharomyces
cerevisiae under different nutrient and growth
conditions (14). These observations highlight
the potential importance of posttranslational
mechanisms in regulating Leishmania adaptive
responses to stresses encountered in the mammalian host. Indeed, changes in the L. donovani
phosphoproteome have been associated with
adaptive responses to elevated temperature
(66), and a number of protein kinases and
phosphatases have been shown to be required
for thermotolerance and adaptation to nutrient
limitation (59, 75, 118).

Carbon Metabolism in
Intracellular Amastigotes
From the transcript and proteomic proling
data it is likely that the core pathways in carbon
metabolism outlined in Figure 3 are present in
both promastigote and amastigote stages (100).
In Leishmania and other trypanosomatids the
rst ve enzymes in glycolysis and several enzymes required for succinate fermentation are
compartmentalized in modied peroxisomes
termed glycosomes (65, 81, 99, 100) (Figure 3).
Whereas both developmental stages of Leishmania preferentially utilize glucose or other
sugars when cultured in vitro (41), the extent to
which glycolysis occurs in intracellular amastigotes is unclear, as this stage is also dependent on
gluconeogenesis for intracellular growth (73).
Nonetheless, hexose catabolism is essential
for amastigote survival in macrophages, as
L. mexicana and L. major mutants with defects
in hexose uptake or sugar catabolism are highly

attenuated in vivo (16, 74). Exogenous hexoses


may be needed to sustain essential pathways,
such as the pentose phosphate pathway and
RNA/DNA synthesis, N-glycosylation, and
myo-inositol synthesis (43, 78). Despite growing
in a sugar-poor niche (73), amastigotes accumulate a unique intracellular carbohydrate reserve
material termed mannogen (88). Mannogen
comprises short oligosaccharides of 1-2linked mannose residues that accumulate in
the cytosol to concentrations of 10 mM in
amastigote stages (88, 104) (Figure 3). These
oligosaccharides are degraded under sugarlimiting conditions, and their accumulation
in intracellular stages indicates that rates of
de novo synthesis or uptake of sugars are not
normally limiting for amastigote growth (88).
Leishmania can catabolize a range of sugars in vitro, including mannose, galactose,
(55) and the amino sugars glucosamine and
N-acetylglucosamine (74). Disruption of glucosamine catabolism has a profound effect on
the establishment of L. major infection in
mice and subsequent lesion development, suggesting that amino sugars are a major class
of sugars in the PV (74, 78), most likely
as a result of the internalization and degradation of extracellular matrix proteoglycans
and glycosoaminoglycans by macrophages (48).
Amastigotes may contribute to the nal degradation of glycosoaminoglycan fragments by secreting a chitinase, as overexpression of the
L. mexicana chitinase enhanced intracellular
growth (50).
In addition to hexose, Leishmania amastigotes may use a variety of other carbon sources.
Specically, L. donovani amastigotes upregulate the expression of several enzymes involved
in gluconeogenesis (93), whereas a L. major mutant lacking the gluconeogenic enzyme,
fructose-1,6-bisphosphatase, grows poorly in
macrophages and is unable to induce lesions in
mice (73). Peptides and amino acids generated
by PV proteases are obvious carbon sources
for amastigotes. The Leishmania genomes encode a large number of putative amino acid
permeases, and several of these are expressed
in amastigote stages (44, 105). Leishmania also
www.annualreviews.org Leishmania Metabolism and Virulence

549

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

13 C-isotopologuelabeling study:
an approach for
measuring the activity
of many metabolic
pathways in live
parasites following
metabolic labeling
with 13 C-labeled
substrates and analysis
of labeled intracellular
and extracellular
metabolites by mass
spectrometry or NMR

Axenic amastigotes:
amastigotes stages that
have been generated in
in vitro culture, usually
by subjecting
stationary-phase
promastigote to
elevated temperature
(33 C37 C) and low
pH

550

27 July 2011

9:24

produce an extensive repertoire of lysosomal


proteases that can be upregulated in amastigotes (8). L. mexicana mutants lacking different
classes of cysteine proteinase are attenuated in
vivo, consistent with a role in parasite nutrition
(8). However, direct evidence for a dependency
of amastigotes on amino acid catabolism in vivo
is lacking. Although amino acid uptake is increased in L. mexicana amastigotes (41), recent
13
C-isotopologue-labeling studies suggest that
only a limited number of amino acids, such as
aspartate and alanine, are used as carbon sources
(99).
Fatty acids derived from complex host lipids
are used as an alternative carbon source by
several intracellular pathogens (33). Whereas
fatty acid -oxidation occurs to a negligible
extent in rapidly dividing promastigotes (73),
increased fatty acid -oxidation is evident
in nondividing promastigotes and in axenic
amastigotes (10, 41), and enzymes involved
in fatty acid -oxidation are upregulated in
L. donovani and L. major amastigotes (84,
93). Fatty acid -oxidation would provide
amastigotes with a source of acetyl-CoA for
mitochondrial respiration. However, Leishmania lack the glyoxylate cycle enzymes needed
for use of acetyl-CoA in gluconeogenesis and
are therefore unable to use fatty acids as their
sole carbon source (73). Leishmania amastigotes could obtain lipids from lipoproteins
internalized by infected macrophages (98).
Interestingly, human high-density lipoproteins
contain lytic factors (apoA-1 and Hpr) that are
delivered to the PV and can kill intracellular
promastigote, but not amastigote, stages (98).
Alternatively, lipid exchange may occur across
adhesion zones/tight junctions between the
amastigote plasma membrane and the PV
membrane (76). Host glycosphingolipids
associated with the PV membrane become
intercalated into the amastigote membrane and
contribute to the minimal surface glycocalyx
of this stage (76, 77). Other abundant PV
lipids, such as sphingomyelin, are apparently
internalized by amastigotes and degraded
(123). Intriguingly, a neutral sphingomyelinase
enzyme in L. major was localized to the
McConville

Naderer

mitochondria, suggesting that scavenged sphingomylin is catabolized in the mitochondria in


order to generate ceramide or parasite-specic
lipids, such as inositol phosphoceramides (123).
Amastigotes downregulate their own sphingolipid biosynthetic pathways, suggesting that
this salvage pathway fullls all the sphingolipid
requirements of this stage (122). Lipid salvage
from the PV lumen and membrane may thus
play important roles in both carbon metabolism
and provision of important lipid precursors.
The increased use of both amino acids and
fatty acids as carbon sources by intracellular
amastigotes requires the presence of an active
tricarboxylic acid (TCA) cycle (41). 13 Cisotopologue-labeling studies in L. mexicana
suggest that the TCA cycle is required for the
catapleurotic production of glutamate as well
as ATP synthesis (99). Mitochondrial function
appears to be essential for amastigote viability,
as L. major and L. donovani mutants with defects
in cytochrome c oxidase activity are highly
attenuated or avirulent in mice (29, 113, 124).
Intracellular amastigotes are also killed by the
human antimicrobial peptide histatin 5, which
targets mitochondrial ATP synthesis (58),
while disruption of the mitochondrial neutral
sphingomylinase in L. major leads to increased
thermosensitivity (a hallmark of mitochondrial
dysfunction) and loss of viability in intracellular
amastigote stages (123). Similarly, amastigotes
are acutely sensitive to inhibitors of ergosterol
synthesis, which is required for mitochondrial
function in other eukaryotes (3). Although these
observations are consistent with amastigotes
being dependent on mitochondrial metabolism
for energy and key metabolites, mitochondrial
dysfunction can lead to oxidative stress due to
electron leakage from the respiratory chain.
Furthermore, the single mitochondrion of
Leishmania and other trypanosomatids is
physically tethered to the agellum basal body,
and ongoing mitochondrial metabolism and/or
protein import may also be required for cell
division (113, 124). Thus, the acute sensitivity
of Leishmania amastigotes to mitochondrial
dysfunction may reect the involvement of this
organelle in multiple cellular processes.

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

Amastigotes must salvage other metabolites,


such as purines, from the PV (Figure 3). Many
of the transporters and proteins involved in
purine salvage have been identied and genetically disrupted (13, 56). Whereas disruption
of individual transporters or purine salvage
enzymes has little impact on intracellular
growth, indicating substantial redundancy in
purine salvage, a L. donovani mutant lacking
two key purine phosphoribosyltransferases
(HGPRT and XPRT) was severely attenuated
in vivo (13). Suppressor strains of this mutant
were readily isolated, in which genes encoding
for another phosphoribosyltransferase were
amplied, highlighting the genomic exibility
of these parasites and their capacity to adapt
to nutrient deprivation (13). The surface expression of several nucleobase and nucleoside
transporters is also upregulated when parasites
are starved of purines (19, 82). Although these
observations show that Leishmania can adapt to
low nutrient levels in vivo, the surface expression of most nutrient transporters, with the
notable exception of the LitA iron transporter
(42), either remains constant or is signicantly
downregulated following differentiation of
promastigotes to intracellular amastigotes (31,
90), consistent with the notion that amastigotes
enter a metabolically quiescent state and/or
that the levels of particular nutrients in the PV
are sufcient for growth.

Metabolic Pathways Required for


Protection Against Oxidative Stress
Leishmania are exposed to varying levels of
oxidative stress during the initial innate and
subsequent adaptive immune phases of infection. Gene deletion studies suggest that
the oxidative defenses of these parasites are
nely balanced to cope with these stresses and
that impairment of any of these systems leads
to attenuation of virulence. The major thiol
of Leishmania (and other trypanosomes) is
trypanothione, a conjugate of two glutathione
molecules linked by the polyamine spermidine
(32, 110, 111). Trypanothione has a much
higher capacity to neutralize toxic nitric oxide

than glutathione, possibly accounting for


its existence in pathogenic trypanosomatids
(11), and is required for protection against
oxidative/nitrosative stress in vivo (32, 68,
111). Glutathione and trypanothione are continuously turned over (121), and maintenance
of cellular levels would require high rates of
uptake and/or de novo synthesis of precursor
amino acids, such as arginine, glutamate,
methionine, and serine (102, 119) (Figure 3).
Leishmania amastigotes also accumulate ovothiol, a low-molecular-weight thiol that acts as
a direct oxygen scavenger and is synthesized
from histidine (110). The regeneration of
these thiols is also energetically expensive and
requires continuous production of reductant in
the form of NADPH, which could be supplied
by metabolic pathways such as the pentose
phosphate pathway, a malic enzyme, and the
TCA cycle isocitrate dehydrogenase (30).
A recent genome-wide screen for genes that
conferred resistance to oxidative stress indicated that pteridine reductase 1 (PTR1) may
also play a role in Leishmania oxidative defenses
(79). PTR1 is a well-characterized trypanosomatid enzyme that reduces salvaged biopterin
to dihydrobiopterin and tetrahydrobiopterin.
Deletion of L. major PTR1 enhances the
differentiation of noninfective promastigotes
to infective metacyclic promastigotes, leading
to increased virulence in mice (24). However,
recent studies have shown that loss of PTR1
in several Leishmania species compromises
growth in activated macrophages (67, 79). It
remains to be determined whether the reduced
biopterins generated by PTR1 (or salvaged
from the macrophage) are directly involved in
scavenging reactive oxygen species or whether
their synthesis activates other oxidative defense
mechanisms.

Glycosylation Pathways Required for


Thermotolerance and Virulence
The biosynthesis of the sugar nucleotides
GDP-Man and UDP-GlcNAc is required
for Leishmania thermotolerance and virulence in the mammalian host (36, 78). Both
www.annualreviews.org Leishmania Metabolism and Virulence

551

MI65CH27-McConville

ARI

LPG:
lipophosphoglycan

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

PPG:
proteophosphoglycan

552

27 July 2011

9:24

sugar donors are required for the synthesis


of the paucimannose N-glycans (containing
ve to six mannose residues and two Nacetylglucosamine residues) that are linked to
a variety of cell surface and lysosomal proteins
(64). Leishmania retains a basic ER glycoprotein quality control machinery that includes a
UDP-Glc:glycoprotein
glucosyltransferase,
calreticulin, and glucosidase I, and it is likely
that protein N-glycosylation is required for
the correct folding and/or ER export of one
or more proteins at the elevated temperatures
(33 C37 C) encountered in the mammalian
host (64, 78).
Leishmania promastigotes and amastigotes
synthesize a number of other surface glycolipids and secreted proteophosphoglycans that
contribute to virulence (64). The major surface
macromolecule of promastigote stages is a
complex lipophosphoglycan (LPG) that has
dened functions in both the sandy vector
and in promastigote-initiated infection in the
mammalian host (40, 64, 76, 77, 108, 116).
LPG is essential for the intracellular survival of
L. major and L. donovani promastigotes, but not
L. mexicana promastigotes, possibly reecting
differences in the PV induced by these species
(64, 108). In contrast, amastigotes produce little or no LPG and parasite strains with defects
in LPG biosynthesis retain full virulence when
amastigotes are used to initiate infections (64).
However, amastigotes retain the capacity to
synthesize the structurally related proteophosphoglycans (PPGs), which are secreted into
the PV lumenal space and transported within
vesicular structures in the host cytoplasm (62,
64). The secreted PPGs play an important role
in conditioning the initial host cell response
(92). However, a L. major mutant lacking all
phosphoglycans (LPG and PPG) as a result
of genetic disruption of the Golgi UDP-Gal
transporters exhibited the same delayed lesion
virulence phenotype as mutants lacking LPG
alone (18), indicating that the PPGs and LPGs
have overlapping functions in experimental infections. Intriguingly, L. major and L. donovani
mutants lacking phosphoglycans as a result of
disruption of the Golgi GDP-Man transporter,
McConville

Naderer

LPG2, exhibited a much more severe loss of


virulence phenotype in mice (39, 109). This
transporter can import other sugar nucleotides
into the Golgi apparatus, such as GDP-Fuc,
raising the possibility that additional, and as
yet uncharacterized, glycosylation reactions
occur in the Golgi apparatus that could contribute to amastigote virulence in these species
(103, 112).

MODULATION OF HOST
METABOLISM BY LEISHMANIA
There is increasing evidence that host cell
metabolism can be modulated by the presence
of intracellular pathogens and/or by induction
of specic immune responses in ways that either
promote or retard microbial growth (25). In
particular, activation of macrophages with Th1
cytokines (INF-, TNF, IL-12) leads to the
upregulation of iNOS and the downregulation
of enzymes such as arginase-1 that collectively
reduce arginine and polyamine available for intracellular pathogens. Conversely, activation of
macrophages with Th2 cytokines (IL-4, IL-10,
and IL-13) leads to increased arginase-1 expression at the expense of iNOS (25) and increased
polyamine availability. Leishmania induce the
latter response in susceptible mouse strains,
and parasite proliferation in these mice strongly
correlates with elevated arginase-1 activity (2,
45, 52, 69). Paradoxically, members of the
L. mexicana complex can induce a mild proinammatory response in susceptible mouse
strains (with production of Th1 cytokines such
as INF-) that results in increased surface
expression of the macrophage cationic amino
acid transporter and the bioavailability of
both arginine and ornithine (117). L. mexicana amastigotes may prevent increased NO
synthesis under these conditions by rapidly
depleting host arginine pools via their own
endogenous arginase (38). In contrast, disruption of arginine catabolism in L. major
amastigotes had no effect on host cell NO
production (69). Intriguingly, host arginase
expression increased with increasing parasite
load in L. majorinfected mice, independent

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

of the expression of parasite arginase or the


host immune and cytokine response (69).
Parasite proliferation may lead to the release
of host-derived factors, such as prostaglandins
that activate host arginase expression, further
promoting parasite proliferation (69, 92).
Although Leishmania lack the secretory systems that allow many bacterial pathogens to
export proteins to the host cytoplasm, there
is now compelling evidence that intracellular
stages actively modulate macrophage signaling pathways (9, 51). Two Leishmania proteins
that are normally targeted to the lysosome in
amastigotes, the cysteine protease CBP and a
soluble form of gp63, proteolytically activate
tyrosine-specic phosphatases in the host cell
and degrade transcription factors that mediate host cell responses to INF- (9, 17, 22,
61). Gp63 may also degrade the mammalian
target of rapamycin (mTOR) kinase, leading
to inhibition of protein translation and activation of the amino acid starvation responses in
the host cell (47). The latter responses include
increased lysosomal protein turnover, which
could contribute to the observed increase in
L. major amastigote growth in the presence
of rapamycin (47). These amastigote proteases
could be secreted into the PV lumen and transported across the PV membrane via protein
translocons, such as Sec61 (34). Alternatively,
they could be released in membrane-bound
exosomes and subsequently budded into the
host cytoplasm (107). Another host enzyme activated by intracellular amastigotes is the heme
oxygenase-I, a rate-limiting enzyme in heme
degradation (85). Increased heme degradation
leads to incomplete processing of gp91phox ,
a heme-containing glycoprotein that recruits
components of the NADPH oxidase to the PV
membrane (85). The increased activation of this
protein by L. pifanoi amastigote stages, compared with promastigote stages, is consistent
with the former stage inducing a lower rate of
superoxide production during infection.
Leishmania infection of macrophages also
leads to reduced secretion of the proinammatory cytokine IL-12 (9, 51). This has recently
been linked to Leishmania-induced alterations

in cholesterol levels in the macrophage plasma


membrane (20, 94). Decreased cholesterol
levels lead to the preferential association of
the important costimulatory protein CD40
with signalosome complexes that activate the
Erk1/2 mitogen-activated protein pathway and
IL-10 secretion. As a result, CD40 signaling
via other pathways involving the activation of
p38 mitogen-activated protein and the production of IL-12 is reduced (94). Paradoxically,
macrophages infected with L. mexicana upregulate the expression of many genes required
for de novo sterol biosynthesis (83), possibly
reecting a response to cholesterol depletion.
Alterations in host cell cholesterol homeostasis
may lead to alterations in other signaling
pathways. For example, dyslipidemia is sensed
by the peroxisome proliferator-activated receptors (PPAR-, ) and the liver X receptors (49,
60), both of which increase arginase I and II
expression and/or repression of iNOS synthesis and promote Leishmania infection (15, 35,
106). These studies suggest that subtle changes
in the activities of host metabolic enzymes may
have profound effects on infection outcomes.

Exosomes: small
microvesicles
generated within the
endolysosomal
pathway of eukaryotic
cells and released by
exocytosis

PERSISTENCE VERSUS
PATHOLOGY
An increasing number of Leishmania metabolic
mutants have been shown to establish lowlevel chronic infections without causing
overt pathology. Examples include Leishmania strains with defects in gluconeogenesis
(L. major fbp), iron uptake (L. amazonensis
lit1), purine metabolism (L. major hprt/
xprt), and glycoconjugate synthesis (L. major/
L. donovani lpg2) (13, 39, 42, 73). These mutants characteristically survive in macrophages
but grow slowly and may assume a metabolic
state similar to wild-type parasites that persist
after spontaneous or drug-induced resolution of disease. Analysis of the in vivo host
responses to infection with the L. major
arg mutant indicates that macrophages may
become progressively more permissive as
parasitemia increases, independent of the host
cytokine response (69). Thus, maintenance of a
www.annualreviews.org Leishmania Metabolism and Virulence

553

ARI

27 July 2011

9:24

persistent state may be critically dependent on


keeping parasite numbers below a threshold
level, beyond which rapid expansion of the
parasite population can occur. Other mutants,
including the L. major gcc, arg, and gnd
mutants (69, 74, 102) and the L. mexicana arg
mutant (38), induce lesions after a signicant
delay regardless of whether promastigotes or
amastigotes are used to initiate the infection.
This phenotype might arise if host cells
infected during the initial innate immune
phase are nutritionally more restrictive than
those infected following the development of
an acquired host immune response. In this
respect it is notable that intracellular growth of
L. mexicana is promoted by moderate activation
of macrophages with INF- (86). It is therefore
possible that induction of inammatory lesions
initially facilitates expansion and possibly dissemination of parasites, as has been proposed
to occur in mycobacterial infections (26).

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

CONCLUSIONS
Leishmania spp. are one of the few pathogens to
proliferate in the long term in the phagolysosome of macrophages and other phagocytes.
Recent genomic and biochemical analyses of
the metabolic potential of these parasites, as
well as analysis of the virulence phenotypes
of metabolic mutants, have led to the identication of many metabolic processes that are
essential for intracellular survival and pathogenesis. These studies suggest that intracellular
amastigotes exploit multiple carbon sources
(sugars, amino acids, and lipids) and have

complex transport and salvage pathways for obtaining the large number of essential nutrients
for which they lack pathways of de novo synthesis. Indeed, it is tempting to speculate that
the complex nutritional needs of an ancestral
(monogenetic) trypanosomatid may have been
a key factor in driving these parasites to colonize
this hazardous but comparatively nutrient-rich
intracellular niche (63). Leishmania may further
increase the permissiveness of this compartment by enhancing the fusagenic properties of
the PV with less lytic compartments (such as
the ER), by generating large spacious vacuoles
(in the case of the L. mexicana complex),
and by modulating macrophage signaling
pathways to inhibit microbicidal processes and
increase the availability of essential nutrients.
Residence in such a nutritionally complex
compartment may also confer a high degree
of robustness on Leishmania metabolism. This
is supported by the fact that relatively few
Leishmania mutants lacking nonessential genes
are completely avirulent in vivo, and has important implications for drug development. In
contrast, recent studies have shown that several
Leishmania mutants lacking protein kinases
and phosphatases that might be involved in
regulating parasite metabolism are highly
attenuated or completely avirulent in vivo (59,
75, 118). Elucidating aspects of amastigote
metabolism that are essential for survival and
the delineation of signaling pathways that
regulate these processes should provide further
insights into the molecular basis of Leishmania
pathogenesis and facilitate the development of
new drugs to tackle these devastating diseases.

SUMMARY POINTS
1. Leishmania establish a unique PV compartment in macrophages that is nutritionally complex as a result of continuous fusion and/or membrane transport with other compartments
in the endomembrane system.
2. Obligate intracellular amastigote stages enter a slow growth state. These stages are dependent on the uptake of sugars, particularly amino sugars, as well as the catabolism of
carbon sources, such as amino acids and fatty acids, requiring mitochondrial metabolism.

554

McConville

Naderer

MI65CH27-McConville

ARI

27 July 2011

9:24

3. Other metabolic pathways required for growth are those associated with cellular oxidative
defenses (i.e., trypanothione and biopterin synthesis) and thermotolerance (i.e., protein
N-linked glycosylation).
4. Leishmania amastigotes can modulate metabolic and signaling pathways in their host cell
in order to enhance access to essential nutrients.

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

5. Leishmania metabolic mutants that establish persistent, low-level infections may provide
insights into the metabolic state of persistent wild-type parasites that are responsible for
recrudescence of disease in humans.

FUTURE ISSUES
1. To what extent do amastigotes exist in different growth or physiological states during
preimmune, acute, and chronic stages of infection?
2. What other metabolic pathways are required for intracellular survival and proliferation?
3. How do Leishmania spp. sense and respond to changes in nutrient levels in the host?
4. How does Leishmania infection affect host cell metabolism beyond the few pathways
investigated to date?

DISCLOSURE STATEMENT
The authors are not aware of any afliations, memberships, funding, or nancial holdings that
might be perceived as affecting the objectivity of this review.

ACKNOWLEDGMENTS
We thank Drs. Eleanor Saunders and Julie Ralton for critical reading of the manuscript. We
also thank other members of the McConville group for discussions. We apologize to colleagues
whose references we have not cited due to space constraints or ignorance. Work from the authors
laboratory was supported by the Australian National Health and Medical Research Council. The
present address for Dr. Naderer is Department of Biochemistry and Molecular Biology, Monash
University, Clayton, Victoria, 3800 Australia.

LITERATURE CITED
1. Alcolea PJ, Alonso A, Gomez
MJ, Moreno I, Domnguez M, et al. 2010. Transcriptomics throughout

the life cycle of Leishmania infantum: high down-regulation rate in the amastigote stage. Int J. Parasitol.
40:1497516
2. Al-Mutairi MS, Cadalbert LC, McGachy HA, Shweash M, Schroeder J, et al. 2010. MAP kinase
phosphatase-2 plays a critical role in response to infection by Leishmania mexicana. PLoS Pathog.
6:e1001192
3. Altmann K, Westermann B. 2005. Role of essential genes in mitochondrial morphogenesis in Saccharomyces cerevisiae. Mol. Biol. Cell 16:541017
4. Antoine JC, Prina E, Courret N, Lang T. 2004. Leishmania spp.: on the interactions they establish with
antigen-presenting cells of their mammalian hosts. Adv. Parasitol. 58:168
www.annualreviews.org Leishmania Metabolism and Virulence

555

ARI

27 July 2011

9:24

5. Beattie L, Peltan A, Maroof A, Kirby A, Brown N, et al. 2010. Dynamic imaging of experimental
Leishmania donovaniinduced hepatic granulomas detects Kupffer cell-restricted antigen presentation to
antigen-specic CD8 T cells. PLoS Pathog. 6:e1000805
6. Belkaid Y, Mendez S, Lira R, Kadambi N, Milon G, et al. 2000. A natural model of Leishmania major
infection reveals a prolonged silent phase of parasite amplication in the skin before the onset of lesion
formation and immunity. J. Immunol. 165:96977
7. Bern C, Maguire JH, Alvar J. 2008. Complexities of assessing the disease burden attributable to leishmaniasis. PLoS Negl. Trop. Dis. 2:e313
8. Besteiro S, Williams RA, Coombs GH, Mottram JC. 2007. Protein turnover and differentiation in
Leishmania. Int. J. Parasitol. 37:106375
9. Bhardwaj S, Srivastava N, Sudan R, Saha B. 2010. Leishmania interferes with host cell signaling to devise
a survival strategy. J. Biomed. Biotechnol. 2010:109189
10. Blum JJ. 1990. Effects of culture age and hexoses on fatty acid oxidation by Leishmania major. J. Protozool.
37:50510
11. Bocedi A, Dawood KF, Fabrini R, Federici G, Gradoni L, et al. 2009. Trypanothione efciently intercepts
nitric oxide as a harmless iron complex in trypanosmatid parasites. FASEB J. 24:103542
12. Bogdan C, Donhauser N, Doring R, Rollinghoff M, Diefenbach A, et al. 2000. Fibroblasts as host cells
in latent leishmaniosis. J. Exp. Med. 191:212130
13. Boitz JM, Ullman B. 2010. Amplication of adenine phosphoribosyltransferase suppresses the conditionally lethal growth and virulence phenotype of Leishmania donovani mutants lacking both hypoxanthineguanine and xanthine phosphoribosyltransferases. J. Biol. Chem. 285:1855564
14. Brauer MJ, Huttenhower C, Airoldi EM, Rosenstein R, Matese JC, et al. 2008. Coordination of growth
rate, cell cycle, stress response, and metabolic activity in yeast. Mol. Biol. Cell 19:35267
15. Bruhn KW, Marathe C, Maretti-Mira AC, Nguyen H, Haskell J, et al. 2010. LXR deciency confers
increased protection against visceral Leishmania infection in mice. PLoS Negl. Trop. Dis. 4:e886
16. Burchmore RJ, Rodriguez-Contreras D, McBride K, Merkel P, Barrett MP, et al. 2003. Genetic characterization of glucose transporter function in Leishmania mexicana. Proc. Natl. Acad. Sci. USA 100:39016
17. Cameron P, McGachy A, Anderson M, Paul A, Coombs GH, et al. 2004. Inhibition of lipopolysaccharideinduced macrophage IL-12 production by Leishmania mexicana amastigotes: the role of cysteine peptidases and the NF-kappaB signaling pathway. J. Immunol. 173:3297304
18. Capul AA, Hickerson S, Barron T, Turco SJ, Beverley SM. 2007. Comparisons of mutants lacking the
Golgi UDP-galactose or GDP-mannose transporters establish that phosphoglycans are important for
promastigote but not amastigote virulence in Leishmania major. Infect. Immun. 75:462937
19. Carter NS, Yates PA, Gessford SK, Galagan SR, Landfear SM, et al. 2010. Adaptive responses to purine
starvation in Leishmania donovani. Mol. Microbiol. 78:92107
20. Chakraborty D, Banerjee S, Sen A, Banerjee KK, Das P, et al. 2005. Leishmania donovani affects antigen
presentation of macrophage by disrupting lipid rafts. J. Immunol. 175:321424
21. Chavali AK, Whittemore JD, Eddy JA, Williams KT, Papin JA. 2008. Systems analysis of metabolism
in the pathogenic trypanosomatid Leishmania major. Mol. Syst. Biol. 4:177
22. Contreras I, Gomez MA, Nguyen O, Shio MT, McMaster RW, et al. 2010. Leishmania-induced inactivation of the macrophage transcription factor AP-1 is mediated by the parasite metalloprotease GP63.
PLoS Pathog. 6:e1001148
23. Croft SL, Sundar S, Fairlamb AH. 2006. Drug resistance in leishmaniasis. Clin. Microbiol. Rev. 19:11126
24. Cunningham ML, Titus RG, Turco SJ, Beverley SM. 2001. Regulation of differentiation to the infective
stage of the protozoan parasite Leishmania major by tetrahydrobiopterin. Science 292:28587
25. Das P, Lahiri A, Lahiri A, Chakravortty D. 2010. Modulation of the arginase pathway in the context
of microbial pathogenesis: a metabolic enzyme moonlighting as an immune modulator. PLoS Pathog.
6:e1000899
26. Davis JM, Ramakrishnan L. 2009. The role of the granuloma in expansion and dissemination of early
tuberculous infection. Cell 136:3749
27. De Trez C, Magez S, Akira S, Ryffel B, Carlier Y, et al. 2009. iNOS-producing inammatory dendritic
cells constitute the major infected cell type during the chronic Leishmania major infection phase of
C57BL/6 resistant mice. PLoS Pathog. 5:e1000494

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

556

McConville

Naderer

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

28. Depledge DP, Evans KJ, Ivens AC, Aziz N, Maroof A, et al. 2009. Comparative expression proling of
Leishmania: modulation in gene expression between species and in different host genetic backgrounds.
PLoS Negl. Trop. Dis. 3:e476
29. Dey R, Meneses C, Salotra P, Kamhawi S, Nakhasi HL, et al. 2010. Characterization of a Leishmania
stage-specic mitochondrial membrane protein that enhances the activity of cytochrome c oxidase and
its role in virulence. Mol. Microbiol. 77:399414
30. Doyle MA, MacRae JI, De Souza DP, Saunders EC, McConville MJ, et al. 2009. LeishCyc: a biochemical
pathways database for Leishmania major. BMC Syst. Biol. 3:57
31. Dridi L, Ahmed Ouameur A, Ouellette M. 2010. High afnity S-adenosylmethionine plasma membrane
transporter of Leishmania is a member of the folate biopterin transporter (FBT) family. J. Biol. Chem.
285:1976775
32. Dumas C, Ouellette M, Tovar J, Cunningham ML, Fairlamb AH, et al. 1997. Disruption of the trypanothione reductase gene of Leishmania decreases its ability to survive oxidative stress in macrophages.
EMBO J. 16:259098
33. Eisenreich W, Dandekar T, Heesemann J, Goebel W. 2010. Carbon metabolism of intracellular bacterial
pathogens and possible links to virulence. Nat. Rev. Microbiol. 8:40112
34. Gagnon E, Duclos S, Rondeau C, Chevet E, Cameron PH, et al. 2002. Endoplasmic reticulum-mediated
phagocytosis is a mechanism of entry into macrophages. Cell 110:11931
35. Gallardo-Soler A, Gomez-Nieto
C, Campo ML, Marathe C, Tontonoz P, et al. 2008. Arginase

I induction by modied lipoproteins in macrophages: a peroxisome proliferator-activated receptorgamma/delta-mediated effect that links lipid metabolism and immunity. Mol. Endocrinol. 22:1394402
36. Garami A, Ilg T. 2001. Disruption of mannose activation in Leishmania mexicana: GDP-mannose pyrophosphorylase is required for virulence, but not for viability. EMBO J. 20:365766
37. Garami A, Ilg T. 2001. The role of phosphomannose isomerase in Leishmania mexicana glycoconjugate
synthesis and virulence. J. Biol. Chem. 276:656675
38. Gaur U, Roberts SC, Dalvi RP, Corraliza I, Ullman B, et al. 2007. An effect of parasite-encoded arginase
on the outcome of murine cutaneous leishmaniasis. J. Immunol. 179:844653
39. Gaur U, Showalter M, Hickerson S, Dalvi R, Turco SJ, et al. 2009. Leishmania donovani lacking the
Golgi GDP-Man transporter LPG2 exhibit attenuated virulence in mammalian hosts. Exp. Parasitol.
122:18291
40. Gueirard P, Laplante A, Rondeau C, Milon G, Desjardins M. 2008. Trafcking of Leishmania donovani
promastigotes in non-lytic compartments in neutrophils enables the subsequent transfer of parasites to
macrophages. Cell Microbiol. 10:10011
41. Hart DT, Coombs GH. 1982. Leishmania mexicana: energy metabolism of amastigotes and promastigotes.
Exp. Parasitol. 54:397409
42. Huynh C, Sacks DL, Andrews NW. 2006. A Leishmania amazonensis ZIP family iron transporter is
essential for parasite replication within macrophage phagolysosomes. J. Exp. Med. 203:236375
43. Ilg T. 2002. Generation of myo-inositol-auxotrophic Leishmania mexicana mutants by targeted replacement of the myo-inositol-1-phosphate synthase gene. Mol. Biochem. Parasitol. 120:15156
44. Inbar E, Canepa GE, Carrillo C, Glaser F, Suter Grotemeyer M, et al. 2010. Lysine transporters in
human trypanosomatid pathogens. Amino Acids. In press
45. Iniesta V, Gomez-Nieto LC, Corraliza I. 2001. The inhibition of arginase by N(omega)-hydroxy-Larginine controls the growth of Leishmania inside macrophages. J. Exp. Med. 193:77784
46. Ivens AC, Peacock CS, Worthey EA, Murphy L, Aggarwal G, et al. 2005. The genome of the kinetoplastid
parasite, Leishmania major. Science 309:43624
47. Jaramillo M, Gomez MA, Larsson O, Shio MT, Topisirovic I, et al. 2011. Leishmania repression of host
translation through mTOR cleavage is required for parasite survival and infection. Cell Host Microbe
9:33141
48. Jiang D, Liang J, Noble PW. 2007. Hyaluronan in tissue injury and repair. Annu. Rev. Cell Dev. Biol.
23:43561
49. Joseph SB, Castrillo A, Laftte BA, Mangelsdorf DJ, Tontonoz P. 2003. Reciprocal regulation of inammation and lipid metabolism by liver X receptors. Nat. Med. 9:21319
www.annualreviews.org Leishmania Metabolism and Virulence

557

ARI

27 July 2011

9:24

50. Joshi MB, Rogers ME, Shakarian AM, Yamage M, Al-Harthi SA, et al. 2005. Molecular characterization,
expression, and in vivo analysis of LmexCht1: the chitinase of the human pathogen, Leishmania mexicana.
J. Biol. Chem. 280:384761
51. Kima PE. 2007. The amastigote forms of Leishmania are experts at exploiting host cell processes to
establish infection and persist. Int. J. Parasitol. 37:108796
52. Kropf P, Fuentes JM, Fahnrich E, Arpa L, Herath S, et al. 2005. Arginase and polyamine synthesis are
key factors in the regulation of experimental leishmaniasis in vivo. FASEB J. 19:10002
53. Kropf P, Herath S, Weber V, Modolell M, Muller I. 2003. Factors inuencing Leishmania major infection
in IL-4-decient BALB/c mice. Parasite Immunol. 25:43947
54. Lahav T, Sivam D, Volpin H, Ronen M, Tsigankov P, et al. 2011. Multiple levels of gene regulation
mediate differentiation of the intracellular pathogen Leishmania. FASEB J. 25:51525
55. Lamerz AC, Damerow S, Kleczka B, Wiese M, van Zandbergen G, et al. 2010. Deletion of UDP-glucose
pyrophosphorylase reveals a UDP-glucose independent UDP-galactose salvage pathway in Leishmania
major. Glycobiology 20:87282
56. Landfear SM, Ullman B, Carter NS, Sanchez MA. 2004. Nucleoside and nucleobase transporters in
parasitic protozoa. Eukaryot. Cell 3:24554
57. Lippuner C, Paape D, Paterou A, Brand J, Richardson M, et al. 2009. Real-time imaging of Leishmania mexicana-infected early phagosomes: a study using primary macrophages generated from green
uorescent protein-Rab5 transgenic mice. FASEB J. 23:48391
58. Luque-Ortega JR, vant Hof W, Veerman EC, Saugar JM, Rivas L. 2008. Human antimicrobial peptide
histatin 5 is a cell-penetrating peptide targeting mitochondrial ATP synthesis in Leishmania. FASEB J.
22:181728
59. Madeira da Silva L, Beverley SM. 2010. Expansion of the target of rapamycin (TOR) kinase family and
function in Leishmania shows that TOR3 is required for acidocalcisome biogenesis and animal infectivity.
Proc. Natl. Acad. Sci. USA 107:1196570
60. Marathe C, Bradley MN, Hong C, Lopez F, Ruiz de Galarreta CM, et al. 2006. The arginase II gene is
an anti-inammatory target of liver X receptor in macrophages. J. Biol. Chem. 281:32197206
61. Matte C, Descoteaux A. 2010. Leishmania donovani amastigotes impair gamma interferon-induced
STAT1 nuclear translocation by blocking the interaction between STAT1 and importin-. Infect.
Immun. 78:373643
62. McCall L, Matlashewski G. 2010. Localization and induction of the A2 virulence factor in Leishmania:
evidence that A2 protein is a stress response protein. Mol. Microbiol. 77:51830
63. McConville MJ, de Souza D, Saunders E, Likic VA, Naderer T. 2007. Living in a phagolysosome;
metabolism of Leishmania amastigotes. Trends Parasitol. 23:36875
64. McConville MJ, Mullin KA, Ilgoutz SC, Teasdale RD. 2002. Secretory pathway of trypanosomatid
parasites. Microbiol. Mol. Biol. Rev. 66:12254
65. Michels PA, Bringaud F, Herman M, Hannaert V. 2006. Metabolic functions of glycosomes in trypanosomatids. Biochim. Biophys. Acta 1763:146377
66. Morales MA, Watanabe R, Dacher M, Chafey P, Fortea JO, et al. 2010. Phosphoproteome dynamics
reveal heat-shock protein complexes specic to the Leishmania donovani infectious stage. Proc. Natl. Acad.
Sci. USA 107:838186
67. Moreira W, Leblanc E, Ouellette M. 2009. The role of reduced pterins in resistance to reactive oxygen
and nitrogen intermediates in the protozoan parasite Leishmania. Free Radic. Biol. Med. 46:36775
68. Mukherjee A, Roy G, Guimond C, Ouellette M. 2009. The -glutamylcysteine synthetase gene of
Leishmania is essential and involved in response to oxidants. Mol. Microbiol. 74:91427
69. Muleme HM, Reguera RM, Berard A, Azinwi R, Jia P, et al. 2009. Infection with arginase-decient
Leishmania major reveals a parasite number-dependent and cytokine-independent regulation of host
cellular arginase activity and disease pathogenesis. J. Immunol. 183:806876
70. Muraille E, Gounon P, Cazareth J, Hoebeke J, Lippuner C, et al. 2010. Direct visualization of peptide/MHC complexes at the surface and in the intracellular compartments of cells infected in vivo by
Leishmania major. PLoS Pathog. 6:e1001154
71. Murray HW. 2001. Tissue granuloma structure-function in experimental visceral leishmaniasis. Int. J.
Exp. Pathol. 82:24967

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

558

McConville

Naderer

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

72. Murray HW, Berman JD, Davies CR, Saravia NG. 2005. Advances in leishmaniasis. Lancet 366:156177
73. Naderer T, Ellis MA, Sernee MF, De Souza DP, Curtis J, et al. 2006. Virulence of Leishmania major
in macrophages and mice requires the gluconeogenic enzyme fructose-1,6-bisphosphatase. Proc. Natl.
Acad. Sci. USA 103:55027
74. Naderer T, Heng J, McConville MJ. 2010. Evidence that intracellular stages of Leishmania major utilize
amino sugars as a major carbon source. PLoS Pathogens 6:e1001245
75. Naderer T, Heng, J, McConville MJ. 2011. Calcineurin is required for Leishmania major stress response
pathways and for virulence in the mammalian host. Mol. Microbiol. 80:47180
76. Naderer T, McConville MJ. 2008. The Leishmania-macrophage interaction: a metabolic perspective.
Cell Microbiol. 10:3018
77. Naderer T, Vince JE, McConville MJ. 2004. Surface determinants of Leishmania parasites and their role
in infectivity in the mammalian host. Curr. Mol. Med. 4:64965
78. Naderer T, Wee E, McConville MJ. 2008. Role of hexosamine biosynthesis in Leishmania growth and
virulence. Mol. Microbiol. 69:85869
79. Nare B, Garraway LA, Vickers TJ, Beverley SM. 2009. PTR1-dependent synthesis of tetrahydrobiopterin
contributes to oxidant susceptibility in the trypanosomatid protozoan parasite Leishmania major. Curr.
Genet. 55:28799
80. Ndjamen B, Kang BH, Hatsuzawa K, Kima PE. 2010. Leishmania parasitophorous vacuoles interact continuously with the host cells endoplasmic reticulum; parasitophorous vacuoles are hybrid compartments.
Cell Microbiol. 12:148094
81. Opperdoes FR, Coombs GH. 2007. Metabolism of Leishmania: proven and predicted. Trends Parasitol.
23:14958
82. Ortiz D, Valdes R, Sanchez MA, Hayenga J, Elya C, et al. 2010. Purine restriction induces pronounced
translational upregulation of the NT1 adenosine/pyrimidine nucleoside transporter in Leishmania major.
Mol. Microbiol. 78:10818
83. Osorio y Fortea J, de La Llave E, Regnault B, Coppee JY, Milon G, et al. 2009. Transcriptional signatures
of BALB/c mouse macrophages housing multiplying Leishmania amazonensis amastigotes. BMC Genomics
10:119
84. Paape D, Lippuner C, Schmid M, Ackermann R, Barrios-Llerena ME, et al. 2008. Transgenic, uorescent
Leishmania mexicana allow direct analysis of the proteome of intracellular amastigotes. Mol. Cell Proteomics
7:1688701
85. Pham NK, Mouriz J, Kima PE. 2005. Leishmania pifanoi amastigotes avoid macrophage production of
superoxide by inducing heme degradation. Infect. Immun. 73:832233
86. Qi H, Ji J, Wanasen N, Soong L. 2004. Enhanced replication of Leishmania amazonensis amastigotes in interferon-stimulated murine macrophages: implications for the pathogenesis of cutaneous leishmaniasis.
Infect. Immun. 72:98895
87. Queiroz R, Benz C, Fellenberg K, Hoheisel JD, Clayton C. 2009. Transcriptome analysis of differentiating trypanosomes reveals the existence of multiple post-transcriptional regulons. BMC Genomics
10:495
88. Ralton JE, Naderer T, Piraino HL, Bashtannyk TA, Callaghan JM, et al. 2003. Evidence that intracellular
12 mannan is a virulence factor in Leishmania parasites. J. Biol. Chem. 278:4075763
R, Showalter M, Hickerson S, Beverley SM. 2009. Leishmania major lacking
89. Reguera RM, Balana-Fouce

arginase (ARG) are auxotrophic for polyamines but retain infectivity to susceptible BALB/c mice. Mol.
Biochem. Parasitol. 165:4856
90. Richard D, Leprohon P, Drummelsmith J, Ouellette M. 2004. Growth phase regulation of the main folate
transporter of Leishmania infantum and its role in methotrexate resistance. J. Biol. Chem. 279:54494501
91. Rochette A, Raymond F, Corbeil J, Ouellette M, Papadopoulou B. 2009. Whole-genome comparative
RNA expression proling of axenic and intracellular amastigote forms of Leishmania infantum. Mol.
Biochem. Parasitol. 165:3247
92. Rogers M, Kropf P, Choi BS, Dillon R, Podinovskaia M, et al. 2009. Proteophosphoglycans regurgitated
by Leishmania-infected sand ies target the L-arginine metabolism of host macrophages to promote
parasite survival. PLoS Pathog. 5:e1000555
www.annualreviews.org Leishmania Metabolism and Virulence

559

ARI

27 July 2011

9:24

93. Rosenzweig D, Smith D, Opperdoes F, Stern S, Olafson RW, et al. 2008. Retooling Leishmania
metabolism: from sand y gut to human macrophage. FASEB J. 22:590602
94. Rub A, Dey R, Jadhav M, Kamat R, Chakkaramakkil S, et al. 2009. Cholesterol depletion associated with
Leishmania major infection alters macrophage CD40 signalosome composition and effector function.
Nat. Immunol. 10:27380
95. Rubin-Bejerano I, Fraser I, Grisa P, Fink GR. 2003. Phagocytosis by neutrophils induces an amino
acid deprivation response in Saccharomyces cerevisiae and Candida albicans. Proc. Natl. Acad. Sci. USA
100:1100712
96. Russell DG, Xu S, Chakraborty P. 1992. Intracellular trafcking and the parasitophorous vacuole of
Leishmania mexicana-infected macrophages. J. Cell Sci. 103:1193210
97. Sacks D, Anderson C. 2004. Re-examination of the immunosuppressive mechanisms mediating non-cure
of Leishmania infection in mice. Immunol. Rev. 201:22538
98. Samanovic M, Molina-Portela MP, Chessler AD, Burleigh BA, Raper J. 2009. Trypanosome lytic factor,
an antimicrobial high-density lipoprotein, ameliorates Leishmania infection. PLoS Pathog. 5:e1000276
99. Saunders EC, Chambers JM, Ng W, Naderer T, Ng M, et al. 2011. Isotopologue proling of Leishmania
mexicana promastigotes under glucose replete and starvation conditions. J. Biol. Chem. [Epub ahead of
print]
100. Saunders EC, DE Souza DP, Naderer T, Sernee MF, Ralton JE, et al. 2010. Central carbon metabolism
of Leishmania parasites. Parasitology 137:130313
101. Schroder
B, Wrocklage C, Pan C, Jager R, Kosters
B, et al. 2007. Integral and associated lysosomal

membrane proteins. Trafc 8:167686


102. Scott DA, Hickerson SM, Vickers TJ, Beverley SM. 2008. The role of the mitochondrial glycine cleavage
complex in the metabolism and virulence of the protozoan parasite Leishmania major. J. Biol. Chem.
283:15565
103. Segawa H, Soares RP, Kawakita M, Beverley SM, Turco SJ. 2005. Reconstitution of GDP-mannose
transport activity with puried Leishmania LPG2 protein in liposomes. J. Biol. Chem. 280:202835
104. Sernee MF, Ralton JE, Dinev Z, Khairallah GN, OHair RA, et al. 2006. Leishmania -1,2-mannan is
assembled on a mannose-cyclic phosphate primer. Proc. Natl. Acad. Sci. USA 103:945863
105. Shaked-Mishan P, Suter-Grotemeyer M, Yoel-Almagor T, Holland N, Zilberstein D, et al. 2006. A
novel high-afnity arginine transporter from the human parasitic protozoan Leishmania donovani. Mol.
Microbiol. 60:3038
106. Shamshiev AT, Ampenberger F, Ernst B, Rohrer L, Marsland BJ, et al. 2007. Dyslipidemia inhibits Tolllike receptor-induced activation of CD8alpha-negative dendritic cells and protective Th1 type immunity.
J. Exp. Med. 204:44152
107. Silverman JM, Clos J, deOliveira CC, Shirvani O, Fang Y, et al. 2010. An exosome-based secretion
pathway is responsible for protein export from Leishmania and communication with macrophages. J. Cell
Sci. 123:84252
108. Spath GF, Garraway LA, Turco SJ, Beverley SM. 2003. The role(s) of lipophosphoglycan (LPG) in the
establishment of Leishmania major infections in mammalian hosts. Proc. Natl. Acad. Sci. USA 100:953641
109. Spath GF, Lye LF, Segawa H, Sacks DL, Turco SJ, et al. 2003. Persistence without pathology in
phosphoglycan-decient Leishmania major. Science 301:124143
110. Steenkamp DJ. 2002. Trypanosomal antioxidants and emerging aspects of redox regulation in the trypanosomatids. Antioxid. Redox. Signal. 4:10521
111. Tovar J, Cunningham ML, Smith AC, Croft SL, Fairlamb AH. 1998. Down-regulation of Leishmania
donovani trypanothione reductase by heterologous expression of a trans-dominant mutant homologue:
effect on parasite intracellular survival. Proc. Natl. Acad. Sci. USA 95:531116
112. Turnock DC, Izquierdo L, Ferguson MA. 2007. The de novo synthesis of GDP-fucose is essential for
agellar adhesion and cell growth in Trypanosoma brucei. J. Biol. Chem. 282:2885363
113. Uboldi AD, Lueder FB, Walsh P, Spurck T, McFadden GI, et al. 2006. A mitochondrial protein affects
cell morphology, mitochondrial segregation and virulence in Leishmania. Int. J. Parasitol. 36:1499514
114. Veras PS, Moulia C, Dauguet C, Tunis CT, Thibon M, et al. 1995. Entry and survival of Leishmania
amazonensis amastigotes within phagolysosome-like vacuoles that shelter Coxiella burnetii in Chinese
hamster ovary cells. Infect. Immun. 63:35026

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

560

McConville

Naderer

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

MI65CH27-McConville

ARI

27 July 2011

9:24

115. Vickers TJ, Orsomando G, de la Garza RD, Scott DA, Kang SO, et al. 2006. Biochemical and genetic
analysis of methylenetetrahydrofolate reductase in Leishmania metabolism and virulence. J. Biol. Chem.
281:3815058
116. Vinet AF, Fukuda M, Turco SJ, Descoteaux A. 2009. The Leishmania donovani lipophosphoglycan excludes the vesicular proton-ATPase from phagosomes by impairing the recruitment of synaptotagmin
V. PLoS Pathog. 5:e1000628
117. Wanasen N, MacLeod CL, Ellies LG, Soong L. 2007. L-arginine and cationic amino acid transporter
2B regulate growth and survival of Leishmania amazonensis amastigotes in macrophages. Infect. Immun.
75:280210
118. Wiese M. 2007. Leishmania MAP kinasesfamiliar proteins in an unusual context. Int. J. Parasitol.
37:105362
119. Williams RA, Westrop GD, Coombs GH. 2009. Two pathways for cysteine biosynthesis in Leishmania
major. Biochem. J. 420:45162
120. Wilson J, Huynh C, Kennedy KA, Ward DM, Kaplan J, et al. 2008. Control of parasitophorous vacuole
expansion by LYST/Beige restricts the intracellular growth of Leishmania amazonensis. PLoS Pathog.
4:e1000179
121. Wyllie S, Cunningham ML, Fairlamb AH. 2004. Dual action of antimonial drugs on thiol redox
metabolism in the human pathogen Leishmania donovani. J. Biol. Chem. 279:3992532
122. Zhang K, Hsu FF, Scott DA, Docampo R, Turk J, et al. 2005. Leishmania salvage and remodelling of
host sphingolipids in amastigote survival and acidocalcisome biogenesis. Mol. Microbiol. 55:156678
123. Zhang O, Wilson MC, Xu W, Hsu FF, Turk J, et al. 2009. Degradation of host sphingomyelin is essential
for Leishmania virulence. PLoS Pathog. 5:e1000692
124. Zkova A, Panigrahi AK, Uboldi AD, Dalley RA, Handman E, et al. 2008. Structural and functional
association of Trypanosoma brucei MIX protein with cytochrome c oxidase complex. Eukaryot. Cell 7:1994
2003

www.annualreviews.org Leishmania Metabolism and Virulence

561

MI65-Frontmatter

ARI

11 August 2011

Annual Review of
Microbiology

7:34

Contents

Volume 65, 2011

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

To the Happy Few


Hiroshi Nikaido p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1
Regulation of DnaA Assembly and Activity: Taking Directions from
the Genome
Alan C. Leonard and Julia E. Grimwade p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p19
Regulation of Alternative Sigma Factor Use

Soa Osterberg,
Teresa del Peso-Santos, and Victoria Shingler p p p p p p p p p p p p p p p p p p p p p p p p p p p p37
Fungal Protein Production: Design and Production
of Chimeric Proteins
Peter J. Punt, Anthony Levasseur, Hans Visser, Jan Wery, and Eric Record p p p p p p p p p p p p p57
Structure and Function of MARTX Toxins and Other Large
Repetitive RTX Proteins
Karla J.F. Satchell p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p71
Eukaryotic Picoplankton in Surface Oceans
Ramon Massana p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p91
Life on the Outside: The Rescue of Coxiella burnetii from Its Host Cell
Anders Omsland and Robert A. Heinzen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 111
Molecular Mechanisms of Staphylococcus aureus Iron Acquisition
Neal D. Hammer and Eric P. Skaar p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 129
Protein Quality Control in the Bacterial Periplasm
Melisa Merdanovic, Tim Clausen, Markus Kaiser, Robert Huber,
and Michael Ehrmann p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 149
Prospects for the Future Using Genomics and Proteomics
in Clinical Microbiology
Pierre-Edouard Fournier and Didier Raoult p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 169
The RpoS-Mediated General Stress Response in Escherichia coli
Aurelia Battesti, Nadim Majdalani, and Susan Gottesman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 189
Bacterial Osmoregulation: A Paradigm for the Study
of Cellular Homeostasis
Janet M. Wood p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 215
vi

MI65-Frontmatter

ARI

11 August 2011

7:34

Lipoprotein Sorting in Bacteria


Suguru Okuda and Hajime Tokuda p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 239
Ligand-Binding PAS Domains in a Genomic, Cellular,
and Structural Context
Jonathan T. Henry and Sean Crosson p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 261
How Viruses and Toxins Disassemble to Enter Host Cells
Takamasa Inoue, Paul Moore, and Billy Tsai p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 287

Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org


Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

Turning Hepatitis C into a Real Virus


Catherine L. Murray and Charles M. Rice p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 307
Recombination and DNA Repair in Helicobacter pylori
Marion S. Dorer, Tate H. Sessler, and Nina R. Salama p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 329
Kin Discrimination and Cooperation in Microbes
Joan E. Strassmann, Owen M. Gilbert, and David C. Queller p p p p p p p p p p p p p p p p p p p p p p p p p p 349
Dinoagellate Genome Evolution
Jennifer H. Wisecaver and Jeremiah D. Hackett p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 369
Motility and Chemotaxis in Campylobacter and Helicobacter
Paphavee Lertsethtakarn, Karen M. Ottemann, and David R. Hendrixson p p p p p p p p p p p p 389
The Human Gut Microbiome: Ecology and Recent
Evolutionary Changes
Jens Walter and Ruth Ley p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 411
Approaches to Capturing and Designing Biologically Active Small
Molecules Produced by Uncultured Microbes
Jorn Piel p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 431
Epidemiological Expansion, Structural Studies, and Clinical
Challenges of New -Lactamases from Gram-Negative Bacteria
Karen Bush and Jed F. Fisher p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 455
Gene Regulation in Borrelia burgdorferi
D. Scott Samuels p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 479
Biology of Clostridium difcile: Implications for Epidemiology
and Diagnosis
Karen C. Carroll and John G. Bartlett p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 501
Interactions of the Human Pathogenic Brucella Species
with Their Hosts
Vidya L. Atluri, Mariana N. Xavier, Maarten F. de Jong,
Andreas B. den Hartigh, and Renee M. Tsolis p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 523

Contents

vii

MI65-Frontmatter

ARI

11 August 2011

7:34

Metabolic Pathways Required for the Intracellular Survival


of Leishmania
Malcolm J. McConville and Thomas Naderer p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 543
Capsules of Streptococcus pneumoniae and Other Bacteria: Paradigms for
Polysaccharide Biosynthesis and Regulation
Janet Yother p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 563
Synthetic Poliovirus and Other Designer Viruses: What Have We
Learned from Them?
Eckard Wimmer and Aniko V. Paul p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 583
Annu. Rev. Microbiol. 2011.65:543-561. Downloaded from www.annualreviews.org
Access provided by Universidad de Los Andes - Colombia on 09/01/16. For personal use only.

Regulation of Antigenic Variation in Giardia lamblia


Cesar G. Prucca, Fernando D. Rivero, and Hugo D. Lujan p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 611
Alternative Pathways of Carbon Dioxide Fixation: Insights into the
Early Evolution of Life?
Georg Fuchs p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 631
Index
Cumulative Index of Contributing Authors, Volumes 6165 p p p p p p p p p p p p p p p p p p p p p p p p p p p 659
Errata
An online log of corrections to Annual Review of Microbiology articles may be found at
http://micro.annualreviews.org/

viii

Contents

Potrebbero piacerti anche