+ All Categories
Home > Documents > Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis)...

Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis)...

Date post: 21-Mar-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
16
81 © The Authors Journal compilation © 2011 Biochemical Society Essays Biochem. (2011) 51, 81–95; doi:10.1042/BSE0510081 6 Intracellular growth and pathogenesis of Leishmania parasites Thomas Naderer and Malcolm J. McConville 1 Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Flemington Rd, Parkville, VIC 3010, Australia Abstract Parasitic protozoa belonging to the genus Leishmania are the cause of a spec‑ trum of diseases in humans, as well as chronic long‑term infections. These parasites exhibit a remarkable capacity to survive and proliferate within the phagolysosome compartment of host macrophages. Studies with defined Leishmania mutants in mouse models of infection have highlighted processes that are required for parasite survival in macrophages. Parasite mutants have been identified that (i) are poorly virulent when the insect (promastigote) stage is used to initiate infection, but retain wild‑type virulence following trans‑ formation to the obligate intracellular amastigote stage, (ii) are highly attenu‑ ated when either promastigotes or amastigotes are used, and (iii) are unable to induce characteristic lesion granulomas, but can persist within macrophages in other tissues. From these analyses it can be concluded that promastigote stages of some species require the surface expression of lipophosphoglycan, but not other surface components. Survival and subsequent proliferation of Leishmania in macrophages requires the activation of heat‑shock responses (involving the up‑regulation and/or phosphorylation of heat‑shock proteins), the presence 1 To whom correspondence should be addressed (email malcolmm@unimelb. edu.au).
Transcript
Page 1: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

81

© The Authors Journal compilation © 2011 Biochemical SocietyEssays Biochem. (2011) 51, 81–95; doi:10.1042/BSE0510081 6

Intracellular growth and pathogenesis of Leishmania parasites

Thomas Naderer and Malcolm J. McConville1

Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Flemington Rd, Parkville, VIC 3010, Australia

Abstract

Parasitic protozoa belonging to the genus Leishmania are the cause of a spec‑trum of diseases in humans, as well as chronic long‑term infections. These parasites exhibit a remarkable capacity to survive and proliferate within the phagolysosome compartment of host macrophages. Studies with defined Leishmania mutants in mouse models of infection have highlighted processes that are required for parasite survival in macrophages. Parasite mutants have been identified that (i) are poorly virulent when the insect (promastigote) stage is used to initiate infection, but retain wild‑type virulence following trans‑formation to the obligate intracellular amastigote stage, (ii) are highly attenu‑ated when either promastigotes or amastigotes are used, and (iii) are unable to induce characteristic lesion granulomas, but can persist within macrophages in other tissues. From these analyses it can be concluded that promastigote stages of some species require the surface expression of lipophosphoglycan, but not other surface components. Survival and subsequent proliferation of Leishmania in macrophages requires the activation of heat‑shock responses (involving the up‑regulation and/or phosphorylation of heat‑shock proteins), the presence

1To whom correspondence should be addressed (email [email protected]).

Page 2: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

82 Essays in Biochemistry volume 51 2011

of oxidative and nitrosative defence mechanisms, and uptake and catabolism of carbon sources (glycoproteins, hexoses and amino acids) and essential nutrients (purines, amino acids and vitamins). Parasite mutants with defects in specific kinase/phosphatase‑dependent signalling pathways are also severely attenuated in amastigote virulence, highlighting the potential importance of post‑ translational regulatory mechanisms in parasite adaptation to this host niche.

Introduction

Leishmania spp. comprise an important group of trypanosomatid parasites that are transmitted by sandfly vectors and cause a range of diseases in humans [1]. Depending on the infecting species and a number of other factors (host immunity etc.), these parasites may induce localized, self‑curing, cutaneous lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious and life‑threatening mucocutaneous and visceral forms of leishmaniasis. Parasites can persist long term in the host, even after apparent resolution of the disease, a factor that may be impor‑tant in the development of long‑term immunity to subsequent infection, but also the cause of re‑occuring infections in immunocompromised individuals. Transmission occurs when an infected sandfly takes a bloodmeal on a human or another animal host, resulting in the injection of pathogenic metacyclic promastigotes into the subdermal layers of the skin. Promastigotes can be phagocytosed by a number of host cell types (neutrophils, dendritic cells and fibroblasts), but disease is propagated primarily within macrophages. Internalized promastigotes are delivered to the mature phagolysosome com‑partment of host macrophages where they differentiate to the non‑motile amastigote stage. Depending on the Leishmania species, amastigotes can reside within individual tight‑fitting phagolysosomes or within large communal phagolysosomes (Figure 1). Propagation of intracellular amastigotes to other macrophages occurs following cell division or lysis of the host cell. Leishmania are stealthy pathogens that either avoid or actively suppress macrophage microbicidal processes. Although much is known about the Leishmania–macro phage interactions, we are only just beginning to understand how amastigotes adapt to life in this hostile host niche [2,3]. In recent years, an increasing number of Leishmania mutants have been generated that can be cultured as promastigotes in vitro, but show reduced levels of growth and sur‑vival in macro phages and mice. These mutants have revealed processes that are required for Leishmania pathogenesis, as well as insights into the nature of the intracellular niches occupied by these parasites (Table 1). Although it is often difficult to define whether loss of virulence is due to impaired promas‑tigote differentiation or amastigote survival in macrophages, the generation of novel molecular tools promises to distinguish between the different processes in the future [4]. In the present chapter, we summarize some of the key pro‑cesses that are required for parasite survival in mammalian macrophages, tissue tropism and pathogenesis in susceptible hosts.

Page 3: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 83

Table 1. Genes required for Leishmania virulence in the BALB/c model of infectionRefer to [2,3] or the text for original references. ADS, alkyldihydroxyacetonephosphate

synthase; ATG, autophagy‑specific gene; CP, cysteine protease; DPMS, dolichol‑phosphate

mannose synthase; FBP, fructose‑1,6‑bisphosphatase; GCC, glycine cleavage complex;

GFAT, glutamine:fructose‑6‑phosphate amidotransferase; GT, glucose transporter; HGPRT,

hypoxanthine‑guanine phosphoribosyltransferase; INO1, inositol‑1‑phosphate synthase; Lch,

L. chagasi; Ldn, L. donovani; LitA, Leishmania iron transporter A; Lmj, L. major; Lmx, L. mexicana;

PMI, phosphomannose isomerase; PMM, phosphomannomutase; PPG, proteophosphoglycan;

SPT, serine palmitoyltransferase; TR, trypanothione reduction; VPS, vacuolar protein sorting;

XPRT, xanthine phosphoribosyltransferase.

Infectivity

Gene Defect Species Promastigote* Amastigote†

Surface components and lipids

LPG1 LPG Lmj/Ldn ‑ ++

Lmx ++ +++

LPG2 LPG/PPG Lmj/Ldn − ++

Lmx ++ +++

(Continued)

Figure 1. The intracellular life style of Leishmania parasitesLeishmania amastigotes can reside within single tight‑fitting phagolysosomes (left‑hand panel) or within large communal phagolysosomes (right‑hand panel) in infected macrophages. In these images, macro‑phages were infected with L. major (left‑hand panel) or L. mexicana (right‑hand panel) and stained for LAMP1 (lysosome‑associated membrane protein1; green). Macrophage and parasite nuclei were visu‑alized with the DNA dye propidium iodide (red), which also labels the mitochondrial genome local‑ized in the kinetoplast. The white outline indicates the plasma membrane of the infected macrophage.

Page 4: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

84 Essays in Biochemistry volume 51 2011

Table 1. (continued)

Gene Defect Species Promastigote* Amastigote†

Surface components and lipids (continued)

LPG4/5 LPG/PPG Lmj + ++

DPMS GPI/LPG Lmx + ++

SPT Sphingolipids Lmj + ++

ADS Ether lipid Lmj + ++

Heat shock

LPG3 Golgi chaperone Lmj − ++

A2 ER chaperone Ldn + +

HSP100 Cytoplasmic chaperone Lmj + +

Oxidative stress

TR+/− Trypanothione

reduction

Ldn + +

SODg+/− SOD Lch + +

Metabolism

FBP Gluconeogenesis Lmj + −P

GT1/2/3 Hexose uptake Lmx − −

LitA Iron uptake Lam + −P

INO1 Inositol synthesis Lmx − −

GMP Mannosylation Lmx/Lmj − −

PMM Mannosylation Lmx − −

PMI Mannose synthesis Lmx − +

GCC‑P Glycine cleavage Lmj ++ ++

MIX+/− Mitochondrial

respiration

Lmj − −

GFAT Hexosamine synthesis Lmj − +++

HGPRT/XPRT Purine salvage Ldn − ++

Membrane trafficking

AP1σ/μ Endocytosis Lmx − −

AP3 Acidocalcisome

biogenesis

Lmj + +

Protein degradation

CpB Lysosome degradation Lmx − +

CpA Lysosome degradation Lmx + +

VPS4/ATG4 Autophagy Lmx − ?

Signalling

MPK1 MAPK Lmx + −

MPK7 MAPK Ldn d +O

MKK4 MAPKK Lmx − +++

TOR3 TOR pathway Lmj + −

PTP1 Tyrosine phosphatase Ldn + −

*Promastigote infectivity: ++, promastigote stages differentiate to amastigote and grow

normally in macrophages; +, promastigote stages differentiate to amastigotes in macrophages

Page 5: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 85

but do not proliferate; −, promastigote stages are poorly virulent in macrophages (can lead to

delayed lesion development if amastigotes stages retain virulence); d, activity deleterious.

†Amastigote infectivity: +++, amastigotes induce normal pathology with the same kinetics as

wild‑type parasites; ++, amastigotes induce normal pathology with delayed kinetics (often,

but not always, associated with a defect in promastigote virulence); +, amastigotes induce

attenuated pathology; −P, amastigotes fail to cause pathology, but persist (in lymph nodes, skin

or liver); −, parasites are completely cleared in BALB/c mice; +O, overexpression leads to

reduced amastigote survival in BALB/c mice.

Genes required for parasite survival in macrophages

Cell‑surface glycolipidsPromastigote stages are coated with a densely packed surface glycocalyx that protects these stages from host hydrolases and serum complement com‑ponents in the insect bloodmeal and digestive tract. Leishmania mutants lacking specific surface components were among the first class of mutants to be generated and have helped to define the functions of different surface components in the insect vector and the mammalian host. The major com‑ponent of the promastigote surface glycocalyx is a complex lipoglycoconjugate termed LPG (lipophosphoglycan). LPG comprises a long phosphoglycan chain (made of oligosaccharide–phosphate repeat units) that is anchored to the plasma membrane via a novel GPI (glycosylphosphatidylinositol) anchor [2]. Leishmania mutants lacking LPG are unable to colonize their sandfly vec‑tor, primarily because they cannot bind to the mid‑gut wall of the digestive tract and are rapidly cleared with the remains of the bloodmeal. Leishmania major and Leishmania donovani mutants lacking LPG are also poorly virulent in mammalian macrophages and exhibit a characteristic delayed lesion phe‑notype in susceptible BALB/c mice. Studies by Spath et al. [5] have suggested that the attenuated virulence of these mutants primarily reflects their increased susceptibility to ROS (reactive oxygen species) transiently generated during macrophage invasion. Although other functions for LPG have been proposed (as a ligand for macrophage receptors, inhibition of phagolysosome matura‑tion and inhibition of host cell signalling pathways), these functions appear to be less important or redundant [5]. Remarkably, the surface glycocalyx of LPG is completely lost following promastigote differentiation to amastigotes, suggesting that LPG is not required for amastigote virulence [6]. Indeed, pro‑mastigote stages of Leishmania LPG− mutants that manage to differentiate to amastigotes are as virulent as wild‑type parasites, explaining why these mutants characteristically show a delay of several weeks before inducing normal lesions in susceptible mice (Table 1). Interestingly, LPG does not appear to be required for virulence in members of the Leishmania mexicana

Page 6: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

86 Essays in Biochemistry volume 51 2011

complex [7]. These parasites may be intrinsically more resistant to oxidative stress. Alternatively, these species induce quite a different vacuolar structure in the host macrophage (Figure 1) and may thus be exposed to lower levels of ROS during invasion.

Promastigotes express a number of other surface molecules that contri‑bute to the surface glycocalyx, but do not appear to be essential for survival in macrophages. These include a number of GPI‑anchored proteins and a highly abundant family of glycolipids, termed GIPLs (glycosylphosphati‑dylinositol lipids), that are structurally related to the protein and LPG GPI anchors [2]. GIPLs are abundant plasma membrane components in both promastigote and amastigote stages, and the absence of a detectable loss‑of‑virulence phenotype in an L. major GIPL mutant was surprising. It is likely that these parasite glyco lipids are functionally replaced by host glycosphingolipids that accumulate in the plasma membrane of intracel‑lular amastigotes [2]. How amastigotes acquire these host lipids is unclear. One possibility is that phagolysosome membrane lipids are flipped across tight junctions that form between the posterior end of the amastigote and the vacuolar membrane (Figure 2). The acquisition of host glycolipids may protect the parasite membrane from lysosomal hydrolases. There is also evidence that scavenged host lipids can be used to synthesize para‑site lipids. Specifically, the L. major ∆spt mutant with a defect in de novo sphingolipid biosynthesis is able to infect and induce lesions in mice, des‑pite displaying severe growth defects when cultivated in sphingolipid‑free medium in vitro [8]. Amastigote stages of this mutant synthesize complex parasite‑specific sphingolipids, such as inositol‑phosphoceramide, sug‑gesting that scavenged sphingosine bases are internalized to the amas‑tigote Golgi apparatus and are re‑used by lipid biosynthetic enzymes in this organelle [8]. Collectively, these studies suggest that amastigote lipid uptake is important for pathogenesis, conferring ‘host‑like’ characteristics to the parasite plasma membrane, as well as providing a source of pre‑cursors for complex lipid biosynthesis.

The heat‑shock response pathwayLeishmania thermotolerance is essential for survival at the elevated tempera‑tures (33–37°C) encountered in mammalian hosts. Elevated temperature is also one of the key environmental cues, together with acidification, that induces promastigote‑to‑amastigote differentiation in vitro [9,10]. Recent studies have suggested that HSPs (heat‑shock proteins) play a key role in co‑ordinating responses to heat shock and differentiation. Interestingly, in contrast with the situation in other organisms, the major HSP fami‑lies of Leishmania, including HSP70, HSP90 and STI1 (stress‑inducible protein 1)/HOP (HSP organizer protein), are constitutively expressed in both life cycle stages and only marginally up‑regulated during heat shock [11]. However, these abundant cytoplasmic proteins are extensively

Page 7: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 87

phosphorylated and assemble into large protein complexes following pro‑mastigote‑to‑amastigote differentiation [11]. From detailed proteomic and genetic studies, it has been suggested that phosphorylation may control the interaction of HSP proteins with client proteins involved in regulating a variety of cellular processes [11]. Recruitment to HSP complexes may directly regulate the activities of these client proteins and/or their suscepti‑bility to proteosomal degradation. Consistent with this proposal, pharma‑cological inhibition of L. donovani HSP90 with geldanamycin or radicicol results in spontaneous amastigote differentiation under promastigote culture conditions and increased expression of other protein chaperones [12]. The Leishmania heat‑shock response may thus be regulated by heat‑induced kinases instead of heat‑shock transcriptional factors, as in other eukaryo‑tes. The identity of these kinases and the precise signals that lead to their activation have yet to be identified. Finally, it is worth noting that the expression of some stress proteins, including HSP100, a mitochondrial

Figure 2. The phagolysosome compartmentLeishmania must be able to scavenge several important nutrients from its host cell. A range of macromolecules and metabolites are thought to be delivered to the phagolysosome com‑partment of infected macrophages by different membrane transport pathways (endocytosis, pinocytosis and autophagy). Macromolecules may be degraded by lysosomal hydrolases and/or internalized by amastigotes and degraded in their lysosomes. Some nutrients (i.e. arginine) may also be directly imported from the host cytoplasm. Depletion of arginine may lead to reduced NO synthesis. Intracellular amastigotes can also scavenge a wide range of host lipids, including abundant glycosphingolipids, from the phagolysosomal membrane (see inset). On the other hand, Leishmania contains mechanisms ensuring survival and growth under different stresses encountered in the macrophage that include high temperature, acidic pH and oxida‑tive radicals.

Page 8: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

88 Essays in Biochemistry volume 51 2011

chaperone, and the ER (endoplasmic reticulum) A2 stress protein, are up‑regulated during promastigote‑to‑amastigote differentiation [13]. Most of these proteins (i.e. HSP100 and A2) are not essential for promastigote growth, but are required for survival in the mammalian host [13].

Protection against oxidative stressIntracellular stages are exposed to oxidative and nitrosative stresses as a result of the production of ROS and NOS (nitrosative species) by the macrophage. Metabolic reactions within the parasite, such as mitochondrial respiration, may also lead to oxidative stress. As expected, partial or complete deletion of any of the oxidative defence mechanisms of Leishmania leads to a dramatic loss of virulence. For example, the major thiol of Leishmania (and other trypanosomes) is trypanothione, a conjugate of two glutathione molecules linked by the polyamine spermidine [14–16]. Trypanothione is linked to many processes performed by glutathione in other organisms, such as the metabolism of peroxides, biosynthesis of deoxyribonucleotides, detoxifica‑tion of methylglyoxal and detoxification of zenobiotics. Although enzymes involved in the synthesis and recycling of oxidized trypanothione are essential, Leishmania mutants with single allele deletions in trypanothione reductase are avirulent in macrophages [14,15]. Significantly, this enzyme is the major target of front‑line anti‑leishmanial drugs, such as trivalent and pentavalent antimony [16]. Similarly, deletion of a single allele of a glycosomal Leishmania chagasi iron SOD (superoxide dismutase) results in an attenuated virulence phenotype [17]. Collectively, these observations suggest that the oxidative defence mech‑anisms of Leishmania, while comprehensive, can be overwhelmed if part of the network is inhibited.

Amastigote metabolism and nutrient scavengingAlthough the metabolism of in vitro cultivated promastigotes has been exten‑sively studied, comparatively little is known about the metabolism of the intracellular amastigote stages. Transcriptomic analyses of promastigote and intracellular amastigotes stages indicate that most genes are constitutively expressed in all life cycle stages [18]. Of the few genes that are differentially regulated, most are down‑regulated in amastigote stages, suggesting that this stage may enter a growth‑restricted state [19]. In view of this transcriptional hardwiring, it is likely that stage‑specific changes in metabolism are regu‑lated by changes in protein expression and post‑translational modifications. Indeed, proteomic analyses have revealed co‑ordinated stage‑specific changes in the level of expression of enzymes involved in a number of key pathways of carbon‑source utilization, as well as alterations in the abundance and distri‑bution of different post‑translational modifications [20–22]. Analyses of the virulene phenotype of Leishmania metabolic mutants have allowed further dissection of metabolic pathways that are important for intracellular survival (Table 1). For example, disruption of glucose uptake in L. mexicana results

Page 9: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 89

in loss of virulence in macrophages and susceptible BALB/c mice, suggest‑ing that glucose uptake is essential for virulence [23]. However, levels of free sugars in the macrophage endo‑lysosomal system are thought to be low, sug‑gesting that glucose will not be a major carbon source for amastigotes. Indeed, a L. major mutant with a defect in gluconeogenesis is highly attenuated in macrophages and mice, confirming that sugar levels in vivo are insufficient to sustain amastigote growth [24]. Metabolomic analyses of L. mexicana amas‑tigotes suggest that this stage is capable of gluconeogenesis, but that the flux through this pathway is insufficient for growth in the absence of exogenous glucose (E.  Saunders, T. Naderer and M. McConville, unpublished work). Amastigotes may therefore need to salvage sugars for specific pathways of carbohydrate metabolism, such as the pentose phosphate pathway (supplying essential NADPH‑reducing equivalents), protein N‑glycosylation (required for thermotolerance) and/or synthesis of the major intracellular reserve carbohydrate mannogen [25,26].

The phagolysosome is a major site of protein degradation, raising the possibility that amino acids are an important carbon source for intracellu‑lar amastigotes (Figure 2). Leishmania are also auxotrophic for many amino acids, supporting the notion that this compartment contains a rich supply of amino acids. Amino acids may be taken up directly by amino acid permeases in the amastigote plasma membrane. Alternatively, amastigotes may internalize host proteins and degrade them in enlarged lysosomal compartments, termed megasomes. Amastigote cysteine proteases are required for virulence, suggest‑ing that parasite lysosomal degradation is important for amastigote growth [27]. The possibility that amastigotes depend on amino acid catabolism as a major source of energy is further supported by the finding that mitochondrial metabolism is essential for intracellular survival. In particular, mitochondrial respiratory chain complexes, such as the COX (cytochrome c oxidase com‑plex) have been shown to contain novel protein subunits that are essential for amastigote virulence [28,29]. Intriguingly, amino acid uptake by intracellular amastigotes may affect host cell responses. For example, arginine uptake and catabolism by intracellular amastigotes may lead to depletion of arginine levels in the host cell and inhibition of key microbicidal processes such as NO pro‑duction (Figure 2) [30,31].

Leishmania amastigotes express all of the enzymes involved in fatty acid β‑oxidation, suggesting that lipids derived from internalized lipoproteins or the phagolysosome membrane could be used for energy or as carbon sources [32]. However, Leishmania lacks the metabolic capability to utilize fatty acids as a major carbon source, and direct evidence that fatty acids are used for ener‑gy generation in vivo has yet to be obtained [24]. Other nutrients scavenged by amastigotes include purines, vitamins and haem. Some of these metabolite salvage systems have been well characterized and shown to be essential for intracellular growth. Not surprisingly, many of these pathways exhibit a high degree of redundancy. For example, purine salvage is initiated by a number

Page 10: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

90 Essays in Biochemistry volume 51 2011

of surface and secreted nucleotidases and a redundant network of nucleotide synthases. Interestingly, genetic disruption of the purine salvage network in L. donovani resulted in the rapid selection of suppressor strains during animal infections [33]. The latter finding highlights the genomic flexibility of these parasites and their capacity to adapt to the loss of a particular metabolic path‑way as a result of gene amplification.

Host sensing and signal transduction pathwaysAll microbial pathogens must sense and respond to changes in their environ‑ment. Leishmania express a large number of protein kinases and phosphatases, some of which appear to be involved in parasite adaptations to life in the mam‑malian host [34]. Many of these proteins share homology with well character‑ized signalling kinases/phosphatases involved in regulating cellular responses to stress and nutritional responses in other eukaryotes. However, Leishmania and other trypanosomatids lack most of the cell surface receptors that initiate these signalling cascades (i.e. G‑protein‑coupled receptors), as well as the major down‑stream effectors (i.e. transcription factors) [34], suggesting that signalling path‑ways in these parasites will differ substantially from those in other eukaryotes.

The most intensively studied signalling kinases in Leishmania to date are the MAPKs (mitogen‑activated protein kinases). Leishmania is predicted to contain 23 MAPKKKs (MAPK kinase kinases), seven MAPKKs (MAPK kinases) and 15 MAPKs, considerably more than in other eukaryotes [35]. Wiese [35] has shown that a number of these kinases may have important roles in regulating differentiation, as well as stress responses important for survival in the mammalian host. MPK1 (mitogen‑activated protein kinase 1) was discovered serendipitously and shown to be essential for survival in BALB/c mice, but not for promastigote growth in culture [35a]. On the other hand, MPK4 is essential for both developmental stages, whereas MPK7 and MPKK4 are selectively expressed in promastigote stages and reduce amastig‑ote viability when overexpressed in this stage [36]. The deleterious effect of MPK7 expression on amastigote growth was dependent on MAPK activity and associated with reduced protein translation and growth arrest. It is poss‑ible that MPK7 promotes parasite growth but causes parasite death when con‑ditions are not optimal for rapid proliferation.

The TOR (target of rapamycin) serine/threonine‑specific kinase is a master regulator of many processes in other eukaryotic cells, including cell growth, energy balance and metabolism. Leishmania express at least three putative TOR kinases (compared with one in mammals and two in yeast). L. major TOR1 and TOR2 appear to be essential for promastigote growth [37]. In contrast, loss of TOR3 has little effect on promastigote growth, but is essential for infectivity in macrophages and mice [37]. Parasite mutants lacking TOR3 are more sensitive to glucose starvation than wild‑type para‑sites, providing a possible explanation for the loss of virulence in mice (see amastigote metabolism). Loss of TOR3 also leads to defects in the biogenesis

Page 11: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 91

of acidocalcisomes, an organelle involved in the storage of polyphosphates, divalent cations (Ca2+) and amino acids in many protists. Other Leishmania mutants with defects in acidocalcisome biogenesis [27] are attenuated in viru‑lence, providing an additional explanation for why TOR3 is required for intra‑cellular survival.

Leishmania also express a large number of as yet uncharacterized ser‑ine/threonine‑specific protein phosphatases [38]. On the other hand, a non‑receptor PTPase (protein tyrosine phosphatase), PTP1, has been shown to be essential for virulence of L. donovani amastigotes in BALB/c mice, but not for growth of promastigotes or their differentiation to amastigotes in vitro [39]. Intriguingly, the Trypanosoma brucei PTP1 is required for sens‑ing extracellular nutrients and stage differentiation [40]. The T. brucei PTP1 appears to act on a serine/threonine‑specific phosphatase located in modified peroxisomes (glycosomes), providing a direct link between the plasma mem‑brane sensor and potential intracellular targets. A related pathway is predicted to occur in Leishmania [38] and may play a role in regulating amastigote res‑ponses to altered nutrient levels in the phagolysosome. Collectively, these studies highlight both the importance of phosphorylation during parasite colo‑nization of the mammalian host, but also the potential opportunities to exploit these pathways in developing new anti‑parasite therapeutics.

Genes required for lesion formation

Leishmania infections in susceptible mouse strains and in many human infections are commonly associated with the development of large lesions or granulomas at the site of infection or in the liver. The formation of a lesion granuloma is driven by the host immune response and the massive recruit‑ment of macrophages and lymphocytes to the site of infection. A number of Leishmania mutants have been generated that are able to persist long term in circulating macrophages (in lymph nodes, etc.), but do not induce the formation of granulomatous lesions. These persistent mutants include Leishmania strains with defects in gluconeogenesis (L. major ∆FBP), iron uptake (Leishmania amazonensis ∆Lit1), serine biosynthesis (L. major ∆GCS) and lysosome cysteine proteases (L. mexicana ∆CPB). All of these mutants have a defect in their capacity to scavenge various nutrients and may grow more slowly than wild‑type parasites in vivo. As a result they may fail to reach a threshold parasite load needed to induce a robust immune response and granuloma formation. Alternatively, nutrient levels in granuloma macrophages may be more restrictive than in other macrophage populations, preventing the establishment of a core of infected macrophages.

Genes involved in induction of visceral disease

Although a number of factors are thought to determine the outcome of Leishmania infections, the identity of the infecting species is a key factor

Page 12: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

92 Essays in Biochemistry volume 51 2011

in determining whether infections remain localized in the skin or dissemi‑nate to other tissues to cause mucocutaneous (infections of the mucosa) or visceral (infections of the liver, spleen and bone marrow) leishmaniasis. Comparative genome analysis of species associated with cutaneous (L. major), mucocutaneous (Leishmania braziliensis) and visceral disease (Leishmania infantum) have revealed a limited number of genes that are selectively expressed in L.  infantum and L. donovani, but not in the other species [41]. These include the A2 protein, a well characterized marker of amastigote stages in visceralizing species. A2 has recently been localized to the ER and shown to interact with the major ER protein chaperone BiP (immunoglobulin heavy‑chain‑binding protein) [42]. A2 expression is induced by heat shock and chemical inducers of protein misfolding, suggesting that it may bolster the amastigote heat‑shock response and allow these species to tolerate the elevated temperatures in the liver, as well as the fever induced by visceral infections. Supporting this notion, overexpression of the A2 protein in L. major increases the capacity of this species to metastasize and survive in the liver and spleen of mice. A number of other L.  infantum/L. donovani pro‑teins have been shown to increase the metastatic potential of L. major when ectopically expressed in this species [41,43]. One of these proteins (a puta‑tive regulator of membrane transport) is normally expressed in L. major, but at much lower levels than in L. donovani. Species‑specific differences in disease pathology may therefore not reflect the unique expression of a limited number of specific proteins, but rather differences in protein expression of a large number of shared proteins [41].

Conclusions

Leishmania parasites proliferate within the mature phagolysosome com‑partment of macrophages, a hostile host niche occupied by a very few other pathogens. Analysis of the virulence phenotype of Leishmania mutants has provided insights into the properties of the phagolysosome and para‑sites factors required for virulence. Not surprisingly, proteins required for thermotolerance, protection against oxidative stress and salvage of essential carbon sources and nutrients have been shown to be important for pathogenesis. Remarkably, many of these processes may be regulated post‑translationally, providing opportunities for development of new thera‑peutics. Major questions that remain to be answered are (i) the physiological state of amastigotes at different stages of infection, (ii) the identity of the signalling pathways that allow these parasites to adapt to changes in their intracellular niches during acute and chronic stages of infection, and (iii) processes that allow some species to metastasize to non‑cutaneous tissues and cause life‑threatening disease.

Page 13: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 93

Summary

• Infections of the mammalian host are initiated by sandfly‑transmitted promastigote stages. The LPG surface coat of this stage is required dur‑ing the initial stages of infection of some Leishmania species.

• Genes involved in thermotolerance (particularly HSPs), protection against oxidative stress (trypanothione synthesis and metabolism, SODs) and metabolism (nutrient salvage, hexose and amino acid catabolism) are required for survival in macrophages and susceptible animal models.

• Many of these processes are regulated post‑translationally. In particular, HSPs are phosphorylated in a stage‑specific manner.

• Leishmania express a large number of kinases and phosphatases that are likely to interact with novel upstream and downstream effectors, and to be key virulence determinants.

We thank other members of the McConville group for discussions and apologize to colleagues whose references we have not cited due to space constraints. Work from M.J.M.’s laboratory was supported by the Australian National Health and Medical Research Council.

References1. Stuart, K., Brun, R., Croft, S., Fairlamb, A., Gurtler, R.E., McKerrow, J., Reed, S. and Tarleton,

R. (2008) Kinetoplastids: related protozoan pathogens, different diseases. J. Clin. Invest. 118, 1301–1310

2. Naderer, T. and McConville, M.J. (2008) The Leishmania–macrophage interaction: a metabolic perspective. Cell. Microbiol. 10, 301–308

3. McConville, M.J., de Souza, D., Saunders, E., Likic, V.A. and Naderer, T. (2007) Living in a phagolysosome; metabolism of Leishmania amastigotes. Trends Parasitol. 23, 368–375

4. Madeira da Silva, L., Owens, K.L., Murta, S.M. and Beverley, S.M. (2009) Regulated expression of the Leishmania major surface virulence factor lipophosphoglycan using conditionally destabilized fusion proteins. Proc. Natl. Acad. Sci. U.S.A. 106, 7583–7588

5. Spath, G.F., Garraway, L.A., Turco, S.J. and Beverley, S.M. (2003) The role(s) of lipophosphoglycan (LPG) in the establishment of Leishmania major infections in mammalian hosts. Proc. Natl. Acad. Sci. U.S.A. 100, 9536–9541

6. McConville, M.J. and Blackwell, J.M. (1991) Developmental changes in the glycosylated phosphatidylinositols of Leishmania donovani. Characterization of the promastigote and amastigote glycolipids. J. Biol. Chem. 266, 15170–15179

7. Ilg, T. (2000) Lipophosphoglycan is not required for infection of macrophages or mice by Leishmania mexicana. EMBO J. 19, 1953–1962

8. Zhang, K. and Beverley, S.M. (2009) Phospholipid and sphingolipid metabolism in Leishmania. Mol. Biochem. Parasitol. 170, 55–64

9. Zilberstein, D. and Shapira, M. (1994) The role of pH and temperature in the development of Leishmania parasites. Annu. Rev. Microbiol. 48, 449–470

10. Alcolea, P.J., Alonso, A., Gomez, M.J., Sanchez‑Gorostiaga, A., Moreno‑Paz, M., Gonzalez‑Pastor, E., Torano, A., Parro, V. and Larraga, V. (2010) Temperature increase prevails over acidification in gene expression modulation of amastigote differentiation in Leishmania infantum. BMC Genomics 11, 31

Page 14: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

94 Essays in Biochemistry volume 51 2011

11. Morales, M.A., Watanabe, R., Dacher, M., Chafey, P., Osorio y Fortea, J., Scott, D.A., Beverley, S.M., Ommen, G., Clos, J., Hem, S. et al. (2010) Phosphoproteome dynamics reveal heat‑shock protein complexes specific to the Leishmania donovani infectious stage. Proc. Natl. Acad. Sci. U.S.A. 107, 8381–8386

12. Wiesgigl, M. and Clos, J. (2001) Heat shock protein 90 homeostasis controls stage differentiation in Leishmania donovani. Mol. Biol. Cell 12, 3307–3316

13. Hubel, A., Krobitsch, S., Horauf, A. and Clos, J. (1997) Leishmania major Hsp100 is required chiefly in the mammalian stage of the parasite. Mol. Cell. Biol. 17, 5987–5995

14. Dumas, C., Ouellette, M., Tovar, J., Cunningham, M.L., Fairlamb, A.H., Tamar, S., Olivier, M. and Papadopoulou, B. (1997) Disruption of the trypanothione reductase gene of Leishmania decreases its ability to survive oxidative stress in macrophages. EMBO J. 16, 2590–2598

15. Tovar, J., Cunningham, M.L., Smith, A.C., Croft, S.L. and Fairlamb, A.H. (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. U.S.A. 95, 5311–5316

16. Wyllie, S., Cunningham, M.L. and Fairlamb, A.H. (2004) Dual action of antimonial drugs on thiol redox metabolism in the human pathogen Leishmania donovani. J. Biol. Chem. 279, 39925–39932

17. Plewes, K.A., Barr, S.D. and Gedamu, L. (2003) Iron superoxide dismutases targeted to the glycosomes of Leishmania chagasi are important for survival. Infect. Immun. 71, 5910–5920

18. Depledge, D.P., Evans, K.J., Ivens, A.C., Aziz, N., Maroof, A., Kaye, P.M. and Smith, D.F. (2009) Comparative expression profiling of Leishmania: modulation in gene expression between species and in different host genetic backgrounds. PLoS Negl. Trop. Dis. 3, e476

19. Alcolea, P.J., Alonso, A., Gomez, M.J., Moreno, I., Dominguez, M., Parro, V. and Larraga, V. (2010) Transcriptomics throughout the life cycle of Leishmania infantum: high down‑regulation rate in the amastigote stage. Int. J. Parasitol. 40, 1497–1516

20. Rosenzweig, D., Smith, D., Opperdoes, F., Stern, S., Olafson, R.W. and Zilberstein, D. (2007) Retooling Leishmania metabolism: from sand fly gut to human macrophage. FASEB J. 22, 590–602

21. Rosenzweig, D., Smith, D., Myler, P.J., Olafson, R.W. and Zilberstein, D. (2008) Post‑translational modification of cellular proteins during Leishmania donovani differentiation. Proteomics 8, 1843–1850

22. Paape, D., Lippuner, C., Schmid, M., Ackermann, R., Barrios‑Llerena, M.E., Zimny‑Arndt, U., Brinkmann, V., Arndt, B., Pleissner, K.P., Jungblut, P.R. and Aebischer, T. (2008) Transgenic, fluorescent Leishmania mexicana allow direct analysis of the proteome of intracellular amastigotes. Mol. Cell. Proteomics 7, 1688–1701

23. Burchmore, R.J., Rodriguez‑Contreras, D., McBride, K., Merkel, P., Barrett, M.P., Modi, G., Sacks, D. and Landfear, S.M. (2003) Genetic characterization of glucose transporter function in Leishmania mexicana. Proc. Natl. Acad. Sci. U.S.A. 100, 3901–3906

24. Naderer, T., Ellis, M.A., Sernee, M.F., De Souza, D.P., Curtis, J., Handman, E. and McConville, M.J. (2006) Virulence of Leishmania major in macrophages and mice requires the gluconeogenic enzyme fructose‑1,6‑bisphosphatase. Proc. Natl. Acad. Sci. U. S.A. 103, 5502–5507

25. Sernee, M.F., Ralton, J.E., Dinev, Z., Khairallah, G.N., O’Hair, R.A., Williams, S.J. and McConville, M.J. (2006) Leishmania β‑1,2‑mannan is assembled on a mannose‑cyclic phosphate primer. Proc. Natl. Acad. Sci. U.S.A. 103, 9458–9463

26. Naderer, T., Wee, E. and McConville, M.J. (2008) Role of hexosamine biosynthesis in Leishmania growth and virulence. Mol. Microbiol. 69, 858–869

27. Besteiro, S., Tonn, D., Tetley, L., Coombs, G.H. and Mottram, J.C. (2008) The AP3 adaptor is involved in the transport of membrane proteins to acidocalcisomes of Leishmania. J. Cell Sci. 121, 561–570

28. Zikova, A., Panigrahi, A.K., Uboldi, A.D., Dalley, R.A., Handman, E. and Stuart, K. (2008) Structural and functional association of Trypanosoma brucei MIX protein with cytochrome c oxidase complex. Eukaryot. Cell 7, 1994–2003

29. Dey, R., Meneses, C., Salotra, P., Kamhawi, S., Nakhasi, H.L. and Duncan, R. (2010) Characterization of a Leishmania stage‑specific mitochondrial membrane protein that enhances the activity of cytochrome c oxidase and its role in virulence. Mol. Microbiol. 77, 399–414

Page 15: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

© The Authors Journal compilation © 2011 Biochemical Society

T. Naderer and M.J. McConville 95

30. Gaur, U., Roberts, S.C., Dalvi, R.P., Corraliza, I., Ullman, B. and Wilson, M.E. (2007) An effect of parasite‑encoded arginase on the outcome of murine cutaneous leishmaniasis. J. Immunol. 179, 8446–8453

31. Muleme, H.M., Reguera, R.M., Berard, A., Azinwi, R., Jia, P., Okwor, I.B., Beverley, S. and Uzonna, J.E. (2009) Infection with arginase‑deficient Leishmania major reveals a parasite number‑dependent and cytokine‑independent regulation of host cellular arginase activity and disease pathogenesis. J. Immunol. 183, 8068–8076

32. Hart, D.T. and Coombs, G.H. (1982) Leishmania mexicana: energy metabolism of amastigotes and promastigotes. Exp. Parasitol. 54, 397–409

33. Boitz, J.M. and Ullman, B. (2010) Amplification of adenine phosphoribosyltransferase suppresses the conditionally lethal growth and virulence phenotype of Leishmania donovani mutants lacking both hypoxanthine‑guanine and xanthine phosphoribosyltransferases. J. Biol. Chem. 285, 18555–18564

34. Parsons, M., Worthey, E.A., Ward, P.N. and Mottram, J.C. (2005) Comparative analysis of the kinomes of three pathogenic trypanosomatids: Leishmania major, Trypanosoma brucei and Trypanosoma cruzi. BMC Genomics 6, 127

35. Wiese, M. (2007) Leishmania MAP kinases – familiar proteins in an unusual context. Int. J. Parasitol. 37, 1053–1062

35a. Wiese, M. (1998) A mitogen‑activated protein (MAP) kinase homologue of Leishmania mexicana is essential for parasite survival in the infected host. EMBO J. 17, 2619–2628

36. Morales, M.A., Pescher, P. and Spath, G.F. (2010) Leishmania major MPK7 protein kinase activity inhibits intracellular growth of the pathogenic amastigote stage. Eukaryot. Cell 9, 22–30

37. Madeira da Silva, L. and Beverley, S.M. (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. U.S.A. 107, 11965–11970

38. Szoor, B. (2010) Trypanosomatid protein phosphatases. Mol. Biochem. Parasitol. 173, 53–6339. Nascimento, M., Zhang, W.W., Ghosh, A., Houston, D.R., Berghuis, A.M., Olivier, M. and

Matlashewski, G. (2006) Identification and characterization of a protein‑tyrosine phosphatase in Leishmania: involvement in virulence. J. Biol. Chem. 281, 36257–36268

40. Szoor, B., Ruberto, I., Burchmore, R. and Matthews, K.R. (2010) A novel phosphatase cascade regulates differentiation in Trypanosoma brucei via a glycosomal signaling pathway. Genes Dev. 24, 1306–1316

41. Zhang, W.W., Peacock, C.S. and Matlashewski, G. (2008) A genomic‑based approach combining in vivo selection in mice to identify a novel virulence gene in Leishmania. PLoS Negl. Trop. Dis. 2, e248

42. McCall, L.I. and Matlashewski, G. (2010) Localization and induction of the A2 virulence factor in Leishmania: evidence that A2 is a stress response protein. Mol. Microbiol. 77, 518–530

43. Zhang, W.W. and Matlashewski, G. (2010) Screening Leishmania donovani‑specific genes required for visceral infection. Mol. Microbiol. 77, 505–517

Page 16: Intracellular growth and pathogenesis of Leishmania parasites · lesions (cutaneous leishmaniasis) or disseminate to facial mucosal tissues or the liver and spleen to cause serious

Recommended