6
Regulation in mice of protective immunity against Leishmania 459 major Jacques Louis *, Hayo Himmelrich, Carlos Parra-Lopez, Fabienne Tacchini-Cottier and Pascal Launois Resolution of lesions induced by Leishmania major in mice results from the development of Thl responses. Cytokines produced by Th 1 cells activate macrophages to a parasiticidal state. The development of Th2 responses in mice from a few strains underlies susceptibility to infection. Cytokines produced by Th2 cells exacerbate the development of lesions because of their deactivating properties for macrophages. This murine model of infection has provided significant insight into the mechanisms intrinsic to the differentiation of disparate CD4+ T cell subsets in viva in animals from different genetic backgrounds. Addresses WHO Immunology Research and Training Center, Institute of Biochemistry, University of Lausanne, 155 Chemin des Boveresses, CH-1066, Epalinges, Switzerland *e-mail: [email protected] Correspondence: Jacques Louis Current Opinion in Immunology 1998, 10:459-464 http://biomednet.com/elecref/0952791501000459 0 Current Biology Publications ISSN 0952-7915 Abbreviations FasL Fas ligand IFN-y interferon-y IFN-yR interferon-y receptor iNOs inducible nitric oxide synthase IRFl interferon regulatory factor 1 LACK Leishmania-activated C kinase NK natural killer TNF-u tumor necrosis factor CL Introduction hlice from the majority of inbred strains (CS7BL/6, CXH/He, SvlZY/Ev, etc) are resistant to infection with the intracellular pathogen, Lt%A~lanlu majur. In contrast mice from a few strains, such as BALB/c, develop progressive lesions, that never heal, and remain susceptible to rein- fection. This genetically determined resistance and sus- ceptibility to infection with this parasite has been demonstrated to result from the development of specific CD4+ Thl or TM responses, respectively [l]. In this short review, we have summarized recent findings obtained using this murine model of infection pertaining, firstly, to the immune effector mechanisms capable of restricting the growth of L. )tzaJor in resistant mice; sec- ondly, to the downregulation of these protective immune responses in susceptible mice leading to progressive dis- ease; and, thirdly, to the mechanisms mediating differen- tiation of disparate CD4+ T cell subsets in mice from different genetic backgrounds. Acquired protective immune effector mechanisms Healing of lesions induced by L. wajw- requires the induc- tion and expansion of specific CD4+ Thl cells that are restricted by MHC class 11 and produce interferon-y (IFN-y). This cytokine activates the inducible nitric oxide synthase (iNOs) in macrophages leading to the production of reactive nitrogen radicals that is dependent on L-argi- nine and is toxic for intracellular L. /n&r [Z]. The role of IFN-y in the control of infection with L. mt+or was firmly established by results showing that genetically resistant mice with disrupted genes for IFN-)I or IFN-y receptor (IFN-yR) [3,4] failed to resolve their lesions. The importance of the nitric-oxide-dependent pathway of cytotoxicity for the killing of intracellular parasites was demonstrated by results showing that resistant mice, either treated with inhibitors of iXOs or deficient for the gene that encodes iNOs, failed to contain infection [S]. It is well known that L. wu~jiw persists, in low numbers, even in resis- tant mice that have resolved their cutaneous lesions. Reactive nitrogen radicals also play a role in keeping this quiescent status since treatment of resistant CS7BL/6 mice with an inhibitor of iNOs, at a time when cutaneous lesions are resolved, resulted in an expansion of parasites that had been either quiescent or replicating slowly [6]. Recently, using mice genetically deficient for the gene encoding iNOs, it was further shown to control the activity of natur- al killer (NK) cells and the early cytokine response (IFN-7, IL-12 and transforming growth factor /3) during the first 24 hours of infection with L. MU&I. [7’], thereby playing a crit- ical regulatory role on the innate response to this parasite. Even though IFN-U/B induced by L. t~qbr i/l d-o is required for the initial expression of iNOs [7’], mice lack- ing the genes for IFN-yor the IFN-yR, with normal expres- sion of IFN-a/P, are not able to produce nitric oxide in response to various stimuli, suggesting that IFN-y is a cru- cial inducer of the activation of iNOs [8,9]. The cytotoxicity mediated by CD4+ Thl cells & aitro requires a functional Fas (APO-l, CD95) pathway [lo]. In contrast to wild-type CS7BL/6 (resistant) mice, CS7BL/h g/d or /pr mice lacking either a functional Fas or Fas ligand (FasL) are unable to resolve lesions induced by L. wajw in spite of the fact that tile!- develop a CD4+ Thl response and that their macrophages produce normal levels of reac- ti1.e nitrogen radicals in response to IFN-y ill cifro [ll’]. Administration of exogenous recombinant FasL to FasL- deficient CS7BL/6 &f mice resulted in the resolution of lesions, demonstrating the importance of the Fas-FasL pathway in the elimination of parasires: furthermore.

Regulation of protective immunity against Leishmania major in mice

Embed Size (px)

Citation preview

Page 1: Regulation of protective immunity against Leishmania major in mice

Regulation in mice

of protective immunity against Leishmania

459

major

Jacques Louis *, Hayo Himmelrich, Carlos Parra-Lopez, Fabienne Tacchini-Cottier and Pascal Launois

Resolution of lesions induced by Leishmania major in mice

results from the development of Thl responses. Cytokines

produced by Th 1 cells activate macrophages to a

parasiticidal state. The development of Th2 responses in

mice from a few strains underlies susceptibility to infection.

Cytokines produced by Th2 cells exacerbate the

development of lesions because of their deactivating

properties for macrophages. This murine model of infection

has provided significant insight into the mechanisms intrinsic

to the differentiation of disparate CD4+ T cell subsets in viva

in animals from different genetic backgrounds.

Addresses

WHO Immunology Research and Training Center, Institute of

Biochemistry, University of Lausanne, 155 Chemin des Boveresses,

CH-1066, Epalinges, Switzerland

*e-mail: [email protected]

Correspondence: Jacques Louis

Current Opinion in Immunology 1998, 10:459-464

http://biomednet.com/elecref/0952791501000459

0 Current Biology Publications ISSN 0952-7915

Abbreviations

FasL Fas ligand

IFN-y interferon-y

IFN-yR interferon-y receptor

iNOs inducible nitric oxide synthase

IRFl interferon regulatory factor 1

LACK Leishmania-activated C kinase

NK natural killer

TNF-u tumor necrosis factor CL

Introduction hlice from the majority of inbred strains (CS7BL/6,

CXH/He, SvlZY/Ev, etc) are resistant to infection with the

intracellular pathogen, Lt%A~lanlu majur. In contrast mice

from a few strains, such as BALB/c, develop progressive

lesions, that never heal, and remain susceptible to rein-

fection. This genetically determined resistance and sus-

ceptibility to infection with this parasite has been

demonstrated to result from the development of specific

CD4+ Thl or TM responses, respectively [l]. In this

short review, we have summarized recent findings

obtained using this murine model of infection pertaining,

firstly, to the immune effector mechanisms capable of

restricting the growth of L. )tzaJor in resistant mice; sec-

ondly, to the downregulation of these protective immune

responses in susceptible mice leading to progressive dis-

ease; and, thirdly, to the mechanisms mediating differen-

tiation of disparate CD4+ T cell subsets in mice from

different genetic backgrounds.

Acquired protective immune effector mechanisms Healing of lesions induced by L. wajw- requires the induc-

tion and expansion of specific CD4+ Thl cells that are

restricted by MHC class 11 and produce interferon-y

(IFN-y). This cytokine activates the inducible nitric oxide

synthase (iNOs) in macrophages leading to the production

of reactive nitrogen radicals that is dependent on L-argi-

nine and is toxic for intracellular L. /n&r [Z]. The role of

IFN-y in the control of infection with L. mt+or was firmly

established by results showing that genetically resistant

mice with disrupted genes for IFN-)I or IFN-y receptor

(IFN-yR) [3,4] failed to resolve their lesions.

The importance of the nitric-oxide-dependent pathway of

cytotoxicity for the killing of intracellular parasites was

demonstrated by results showing that resistant mice, either

treated with inhibitors of iXOs or deficient for the gene

that encodes iNOs, failed to contain infection [S]. It is well

known that L. wu~jiw persists, in low numbers, even in resis-

tant mice that have resolved their cutaneous lesions.

Reactive nitrogen radicals also play a role in keeping this

quiescent status since treatment of resistant CS7BL/6 mice

with an inhibitor of iNOs, at a time when cutaneous lesions

are resolved, resulted in an expansion of parasites that had

been either quiescent or replicating slowly [6]. Recently,

using mice genetically deficient for the gene encoding

iNOs, it was further shown to control the activity of natur-

al killer (NK) cells and the early cytokine response (IFN-7,

IL-12 and transforming growth factor /3) during the first 24

hours of infection with L. MU&I. [7’], thereby playing a crit-

ical regulatory role on the innate response to this parasite.

Even though IFN-U/B induced by L. t~qbr i/l d-o is

required for the initial expression of iNOs [7’], mice lack-

ing the genes for IFN-yor the IFN-yR, with normal expres-

sion of IFN-a/P, are not able to produce nitric oxide in

response to various stimuli, suggesting that IFN-y is a cru-

cial inducer of the activation of iNOs [8,9].

The cytotoxicity mediated by CD4+ Thl cells & aitro

requires a functional Fas (APO-l, CD95) pathway [lo]. In

contrast to wild-type CS7BL/6 (resistant) mice, CS7BL/h

g/d or /pr mice lacking either a functional Fas or Fas ligand

(FasL) are unable to resolve lesions induced by L. wajw in

spite of the fact that tile!- develop a CD4+ Thl response

and that their macrophages produce normal levels of reac-

ti1.e nitrogen radicals in response to IFN-y ill cifro [ll’].

Administration of exogenous recombinant FasL to FasL-

deficient CS7BL/6 &f mice resulted in the resolution of

lesions, demonstrating the importance of the Fas-FasL

pathway in the elimination of parasires: furthermore.

Page 2: Regulation of protective immunity against Leishmania major in mice

460 Immunity to infection

macrophages infected with L. major up-regulated their sur-

face expression of Fas in response to IFN-y and as a result

became susceptible to apoptotic death induced by CD4+

Thl cells [ll’]. It is thus possible that apoptosis of Fas-

expressing macrophages at the site of infection causes a

limitation in the number of the host cells necessary for par-

asite replication (i.e. macrophages) and thereby increases

the ratio of IFN-y-producing CD4+ Thl cells to infected

macrophages, therefore increasing the efficiency of

macrophage activation to a microbicidal stage.

Acquired immune effector mechanisms underlying susceptibility to infection It is now well accepted that the susceptibility to infection

that characterizes mice of the BALB strains results from

the development of a Th2 response. Recently, a stable

cell-surface molecule, STLZ, has been described, that dis-

tinguishes Th2 from Thl cells [12]. Cytokines produced

by Th2 cells exert a macrophage-deactivating function

[13]. IL-4 has indeed been shown to hamper the activation

of macrophages induced by IFN-y and to suppress the

upregulation of gene expression mediated by IFN-y, par-

ticularly the gene for interferon regulatory factor 1 (IRF-1)

[14’]. Transforming growth factor p, IL-10 and IL-13 are

also able to interfere with the induction of iNOs that is

triggered by IFN-y [13,15].

Innate immune effector mechanisms NK cells have an important role early in infection process-

es since, unlike naive T cells, they respond very rapidly to

stimuli and do not require priming [16,17”]. NK cells pro-

duce a wide range of cytokines such as IFN-y and TNF-cx

which inhibit the growth of pathogens during the initial

stage of infection, thus allowing the host to develop an effi-

cient adaptive immune response. Importantly, these cells

could also play a role in shaping the adaptive response, par-

ticularly by influencing the pathway of differentiation of

CD4* T cells. The effector function of NK cells and their

role in the generation of a Thl response during infection

with L. major has been suggested, by results showing that

depletion of NK cells in resistant mice favours parasite

multiplication in lesions [18]. This treatment also

decreased the IFN-y production normally seen in resistant

mice and increased the IL-4 response. Mice that do not

express the transcription factor IRF-1 have been shown to

be defective in NK cell function [19,20’]. Resistant mice

deficient for the IRF-1 molecule were susceptible to infec-

tion with L. major and mounted an aberrant CD4+ ThZ cell

response, thus indicating that IRF-1 is a factor implicated

in Thl responses [Zl’]; furthermore the successful vacci-

nation of susceptible BALB/c mice with an extract of

L. major and IL-12 required the presence of NK cells [Z!].

In addition to its Thl-promoting effect on CD4+ T cell pre-

cursors, IL-12 activates NK cells to produce IFN-)I. In this

context, it has been proposed that IL-12 also favours Thl

cell development through its capacity to stimulate IFN-y

production by NK cells; however, following infection with

L. major, early IL-12 production and activation of NK cells

are not seen in all strains of resistant mice, rather suggest-

ing that IFN-y production by NK cells is not essential for

Thl cell differentiation [23].

The differentiation of distinct CD4+ T cell subsets in differing mouse strains following infection with Leishmania major Thl and Th2 CD4+ T cells develop from a common naive

CD4+ T cell precursor and several parameters have been

reported to influence the pathway of differentiation of

CD4+ T cell precursors [ 131.

Role of accessory molecules The CD40 molecule and its ligand, as well as CD80 (B7-

1) and CD86 (B7-Z), play a role in the differentiation of

CD4+ T cells after infection with L. major and conse-

quently in the outcome of disease. Resistant mice defi-

cient in either the CD40 or CD40-ligand molecules and

infected with L. major or L. amazonensis fail to mount a

vigourous Thl response and are unable to control infec-

tion [24-261; furthermore, blockade of CDS6 with a spe-

cific antibody ameliorated the infection in BALB/c mice

and inhibited ThZ cell development [27]. It is noteworthy

that BALB/c mice deficient in CD28 remain susceptible

to infection [ZS]. Results from recent elegant experiments

strongly suggest that the interaction between the MHC

class II and the CD4 molecule also plays a role in Th2 cell

development [29”,30”].

The role of cytokines The crucial role of cytokines in directing CD4+ T cell dif-

ferentiation has been established in oitro using naive

CD4+ T cells from mice transgenic for TCR-a and -p

chains. In this system, IL-12 and IFN-y were implicated

in Thl cell maturation whereas IL-4 was implicated in

Th2 cell development [13,31]. Cofactors such as IL-l-a

and IL-18 are also required for the development of a Thl

response [32,33”,34,35].

Studies using neutralizing anti-IL-4 antibodies or mice

with disruption of the gene encoding IL-4 have defined a

critical role for this cytokine in mediating the differentia-

tion of the Th2 subset in vlvo and the failure of BALB/c

mice to control L. major infection 1361. In this context, we

have carefully examined IL-4 mRNA expression in both

genetically susceptible BALB/c and resistant C57BL/6

mice during the first seven days of infection with L. major. Within one day of infection, BALB/c mice exhibited a

peak of IL-4 mRNA expression in the draining lymph

nodes. This returned to baseline levels by 48 hours [37].

From day five, a second wave of IL-4 mRNA was observed

that remained elevated during the entire course of infec-

tion and reflected Th2 cell differentiation. No IL-4 mRNA

expression was seen in resistant mice during the first two

days of infection and although low levels of IL-4 produc-

tion were seen five days after infection they returned to

baseline levels seven days after infection [37]. The early

Page 3: Regulation of protective immunity against Leishmania major in mice

Regulation of protective immunity against Leishmania major in mice Louis et al. 461

IL-4 production in BALB/c mice was not due to activation

of NKl. l+ CD4+ T cells [37] that have been shown to con-

tribute rapidly substantial amounts of IL-4 in &o under

other conditions [38,39]. These NKl.l+ T cells were fur-

ther shown not to play a role in susceptibility to infection

[40,41]; rather, early IL-4 transcription in BALB/c mice

occured in a highly restricted subpopulation of CD4+

T cells that expressed the TCR VP4 and Va8 chains [42”].

The contribution of these cells to the early burst of IL-4

mRNA expression in BALB/c mice in response to infec-

tion was confirmed by results showing that this response

was absent in BALB/c mice rendered selectively deficient

in the Vp4+ CD4+ T cell population [42”]. The

Vp4+ Va8+ CD4+ T cells that produce IL-4 rapidly in

response to L. major are specific for a single antigen of the

parasite called LACK (Leishmanla-activated C kinase;

[42”]) that has been previously identified as an important

antigen recognized by CD4+ T cells in mice infected with

L. major [43].

Clear evidence was obtained that the IL-4, produced by

these Vp4+ VaS+ CD4+ T cells within one day of infection,

plays an essential role in the Th2 differentiation that

occurs in BALB/c mice at a later stage of infection -

among CD4+ T cells no longer restricted to those utilizing

this particular TCR [44’]. Indeed VP4-deficient mice

mounted a polarized Thl response and were fully resistant

to infection [42”]. Thus it appears that a single epitope of

the LACK antigen drives the early IL-4 response by a

restricted population of CD4+ T cells and instructs subse-

quent Th2 differentiation and susceptibility to infection.

This conclusion is in agreement with results from elegant

experiments that have shown that transgenic BALB/c mice

expressing the LACK antigen in the thymus were tolerant

to this antigen and resistant to infection with L. mujor [45].

The essential role of IL-12 in Thl development has been

confirmed in vivo using the murine model of infection with

L. major. Neutralisation of IL-12 in resistant mice allows

the development of a Th2 response [46]. In addition, mice

from a resistant background in which the IL-12 gene had

been deleted develop a Th2 response [47] and suffer from

progressive disease [48]. In complementary studies, exoge-

nous IL-12 given during the first week of infection to sus-

ceptible BALB/c mice resulted in the development of Thl

response and allowed the resolution of lesions [49,50].

Treatment with anti-IL-4 antibodies of resistant mice, that

have been rendered susceptible to infection by treatment

with anti-IL-12 antibodies, allows the resolution of lesions

[46]; this indirectly indicates that one effect of IL-12 is to

reduce IL-4 production. The increased IL-4 mRNA

expression normally observed in draining lymph nodes of

IFN-y-knockout C57BL/6 mice five days after infection

with L. major was indeed readily down-regulated by

exogenous IL-12 [Sl]; this also demonstrates that the

effect is independent of IFN-y. The timing of production

of IL-12 following infection with L. major is still a matter

of debate [S&53]. Results from one study [SZ] have shown

that IL-12 mRNA expression in vivo is delayed up to seven

days after infection, suggesting that the initial steps of

CD4+ T cell differentiation occur in the absence of this

cytokine. This would allow the expression of other genet-

ic tendencies of CD4+ T cell differentiation along a partic-

ular pathway. Downregulation of the IL-12 production that

occurs after ligation of macrophage receptors [54’], partic-

ularly of complement receptor 3, has recently been

demonstrated [SS’]. Since complement receptor 3 is a

potential receptor for the entry of Leishania in

macrophages these results might explain the inhibition of

IL-12 production in macrophages infected with Leishmania

promastigotes [S&53,56].

Recent results obtained using CD4+ T cells transgenic for

TCR-a and -p chains have shown that, in contrast to

BlO.DZ CD4+ T cells, BALB/c CD4+ T cells (that mature

into Th2 effector cells after priming in vitro under neutral

conditions, i.e. in the absence of exogenous cytokines)

rapidly lose their responsiveness to IL-12 during priming

[57,58]. BlO.DZ mice being resistant to infection with

L. major, a model of resistance based on the maintenance

of the IL-12 signaling pathway has been proposed [58,59].

A single locus named Tmp-2 (T-cell modifier phenotype-l)

controls the responsiveness to IL-12 itz vitro and was

mapped to murine chromosome 11, that also contains the

genes for IL-4, IL-S, IL-3 and IRF-1 [60]. The murine IL-

12R is a heterodimer composed of two chains, the IL-

12Rpl and IL-12RP2 [61] which are expressed only in

T cells that have engaged their TCRs [62”]. Extinction of

IL-12 signaling during priming itz vitro of BALB/c CD4+

T cells that are transgenic for TCR-a and -p chains result-

ed from a selective loss of IL-12RP2-chain expression

[62”]. which is necessary for IL-12 signaling through the

Jak/STAT pathway [62”]. The detectable amounts of IL-

4 produced by BALB/c CD4+ T cells during priming under

neutral conditions (i.e. in absence of exogenous cytokines;

[SS]) could inhibit IL-12RP2 chain expression, since addi-

tion of exogenous IL-4 during priming of BlOD2 CD4+

T cells itz vitro was shown to inhibit their responsiveness to

IL-12 [62”].

Within this context we have observed that from 48 hours

and at least up to seven days after infection with L. major,

CD4+ T cells from BALB/c mice become totally unre-

sponsive to IL-12, in contrast to CD4+ T cells from resis-

tant CS7BL/6 mice [44’]. The in vivo induction of this

unresponsive state to IL-12 in the BALB/c CD4+ T cells

resulted from the IL-4 that was produced during the first

day of infection. Recent results from our laboratory have

shown that the expression of the IL-12RP2 chain, which

was induced on CD4+ T cells of both CS7BL/6 and

BALB/c mice within 24 hours of infection with L. major, was readily down-regulated only on CD4+ T cells from

BALB/c mice and occurred as soon as 48 hours after infec-

tion; this effect resulted from the early IL-4 response of

BALB/c mice to infection (H Himmelrich, C Parra-Lopez,

F Tacchini-Cottier, JA Louis, P Louis, unpublished data).

Page 4: Regulation of protective immunity against Leishmania major in mice

462 Immunity to infection

Although IFN-)I was found to be necessary for the matura-

tion of CD4+ T cells transgenic for TCR-a and -p chains

towards the Thl phenotype in several studies (for example

[63]), its requirement has not always been observed

[64,65]. The demonstration that IFN-yis able to rescue the

IL-4-mediated inhibition of IL-1ZRPZ chain expression by

CD4+ T cells differentiating i?z ~itr-o [62”], provides a

rational basis for this discrepancy. Indeed, when IL-4 is

produced during priming ilz z&m, IFN-)I is necessary to

permit the IL-12 signaling required for Thl cell develop-

ment; whereas when IL-4 is not produced during priming,

activated CD4+ T cells maintain expression of the IL-

1ZRPZ chain, allowing Thl cell maturation independently

of IFN-y. The importance of IFN-y in favoring the Thl

response during infection with L. mtGor_ has also been

debated. C57BL/h mice that are deficient in IFN-y devel-

op a Th2 response after L. fnajor infection [3]. In contrast,

SvlZY mice rendered genetically deficient for the binding

chain of the IFN-y receptor develop a Thl response [4].

Inasmuch as IFN-yR-I- mice on a 129/Sv/Ev background,

in contrast to IFN-y-knockout mice on a C57BL/6 back-

ground, do not produce an early IL-4 burst following infec-

‘tion with L. mtiol- (P Launois, JA Louis, unpublished

results), it is possible, in light of the discussion above, that

their CD4+ T cells do not require IFN-y signaling for

maintenance of IL-12RpZ chain expression and IL-12 sig-

naling if/ zVc0.

Finally, it is important to emphasize that resistance and/or

susceptibility to infection with L. mujor appear to be con-

trolled by several genes and thus probably involve several

mechanisms. [Jsing resistant BlO.DZ mice backcrnssed

into susceptible BALB/c mice, six loci located on chromo-

somes 6,7,10,11,15 and 16 were associated with resistance

to L. major infection, but none of them has a major effect

by itself [66”]. Another study using (BALB/c x

C57BL/6)F2 mice also showed a linkage to the HZ region

on the chromosome 17 and to chromosome 9 [67].

Conclusions The murine mode1 of infection with L.. rntior has not

only permitted validation of the two pathways of CD4+

T cell differentiation in eieo but also enabled decipher-

ing of the critical role of CD4+ T cell subsets in the out-

come of infection with an intracellular pathogen. This

mode1 has already been invaluable in recent years for

identifying some molecular mechanisms operating in

viuo in the differentiation of CD4+ T cell subsets in the

periphery. A better definition of these mechanisms is a

prerequesite for the rational development of efficient

immunoprophylactic and immunotherapeutic measures

applicable to humans. Information gained by studying

this disease, which afflicts mainly the developing world,

has no doubt significantly added to our understanding of

critical issues related to the T cell response. It is impor-

tant that this knowledge foments the development of

novel strategies to prevent and treat this and other seri-

ous infectious diseases.

1.

2.

3.

4.

5.

6.

7. .

Acknowledgements &rk from the authors’ laboratory i\ supported by the SHiss National Science Foundation, the European Ilnion and thr \\idd Health Organisation. P Launoi~ is on leave from rhtt Pasteur Inscitut~. Paris.

References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as:

l of special interest l * of outstanding interest

Reiner SL, Locksley RM: The regulation of immunity to Leishmania major. Annu Rev /mmuno/1995, 13:151-177.

Assreuy J, Cunha FQ, Epperlein M, Noronha-Dutra A, O’Donnell CA, Liew FY, Moncada S: Production of nitric oxide and superoxide by activated macrophages and killing of Leishmania major. Eur J lmmunoll994, 24:672-676.

Wang ZE, Reiner SL, Zheng S, Dalton DK, Locksley RM: CD4+ effector cells default to the Th2 pathway in interferon gamma- deficient mice infected with Leishmania major. J Exp Med 1994, 179:1367-1371.

Swihart K, Fruth U, Messmer N, Hug K, Behin R, Huang S, Del Giudice G, Aguet M, Louis JA: Mice from a genetically resistant background lacking the interferon gamma receptor are susceotible to infection with Leishmania major but mount a polariied T helper cell l-type CD4+ T cell response. J Exp Med 1995,181:961-971.

Wei X-Q Charles IG, Smith A, Feng G-J, Huang F-P, Xu D, Muller W, Moncada S, Liew FY: Altered immune responses in mice lacking inductible nitric oxide synthase. Nature 1995, 375:408-41 1,

Stenger S, Donhauser N, ThOrin H, RGllinghoff M, Bogdan C: Reactivation of latent leishmaniasis by inhibition of inducible nitric oxide synthase. J Exp Med 1996, 183:1501-l 514.

Diefenbach A, Schindler H, Donhauser N, Lorenz E, Laskay T, MacMicking J, RBllinghoff M, Gresser I, Bogdan C: Type 1 Interferon (IFNa/P) and type 2 nitric oxide synthase regulate the innate immune response to a protozoan parasite. immunity 1998, 8:77-87. _

This paper shows that genetlc deletion or tunctlonnal InactIvatIon ot type 2 nitric oxide synthase abolishes the natural killer cell response seen one day after infection with Le/shmanis major. Neutralisation of lFNcl@ has a similar effect.

8.

9.

10.

11. .

Huang S, Hendricks W, Althage A, Hemmi S, Bluethmann H, Vilcek J, Zinkernagel RM, Aguet M: Immune response in mice that lack the interferon-y receptor. Science 1993, 259:1742-l 745.

Dalton DK, Pitts-Meek S, Keshav S, Figari IS, Bradley A, Stewart TA: Multiple defects of immune cell function in mice with disrupted interferon gamma gene. Science 1993, 259:1739-l 742.

Yagita H, Hanabuchi S, Asano Y, Tamura T, Nanuchl H, Okumura K: Fas-mediated cytotoxicity: a new immunoregulatory and pathogenic function of Thl CD4+ T cells. lmmunol Rev 1995, 146:223-239.

Conceicao-Silva F. Hahne M. Schrijter M.. Louis J. Tschooo J: The resolut/on of lesibns induced by Leishmania m&or in’mice requires a functional Fas (APO-l, CD95) pathway of cytotoxicity. Eur J lmmunoll998, 28:237-245.

This paper shows for the first time the requirement of a functional Fas/Fas- ligand pathway of cytotoxicity for the resolution by resistant mice of lesions induced by infection with L. major.

12.

13.

14. .

Xu D, Chan WL, Leung BP, Huang FP, Wheeler R, Piedrafita D, Robinson JH, Liew FY: Selective expression of a stable cell surface molecule on type 2 but not on type 1 helper T cells. J Exp Med 1998, 187:787-794.

Abbas AK, Murphy KM, Sher A: Functional diversity of helper T lymphocytes. Nature 1996, 383:787-793.

Ohmori Y, Hamilton TA: IL-4-induced STAT6 suppresses IFN-y- stimulated STATI-dependent transcription in mouse macrophages. J lmmunol 1997, 159:5474-5482.

This paper shows that interferon (IFN)-y-induced expression of the gene for IFN regulatory factor 1 (IRF-1) is suppressed by IL-4.

15. Paludan SR, Lovmand J, Ellermamn-Eriksen S, Mogensen SC: Effect of IL-4 and IL-13 on IFN-gamma induced production of nitric oxide in mouse macrophages infected with herpes simplex virus type 2. FEBS Lett 1997, 414:61-64.

Page 5: Regulation of protective immunity against Leishmania major in mice

Regulation of protective immunity against Leishmania major in mice Louis et al. 463

16. Scott P, Trinchieri G: The role of natural killer cells in host-parasite interactions. Cuff Opin lmmunol 1995, 7:34-40.

17. Lanier LL: Natural killer cells: from no receptors to too many. . . immunity 1997, 6:371-378. This excellent review summarises the current status of knowledge concerning inhibitory receptors on natural killer cells.

18.

19.

20. .

Taki S, Sate T, Ogasawa K, Fukuda T, Sato M, Suzuki G, Mitsuyama M, Shin EH, Taniguchi T, Asano Y: Multistage regulation of Thl-type immune responses by the transcription factor IRF-I. lmmunify 1997, 6:673-679.

This paper shows tor the tlrst time the ,n wtro Importance ot lnterteron regulatory factor 1 (IRF-1) in the regulation of Thl responses.

Sharton TM, Scott P: Natural killer cells are a source of interferon-y that drives differentiation of CD4+ T cell subsets and induces early resistance to L. major in mice. J Exp Med 1993, 178:567-577.

Duncan GS, Mittriicker H-W, Klgi D, Matsuyama T, Mak TW: The transcription factor interferon regulatory factor-l is essential for natural killer cell function in t&o. J Exp Med 1996, 184:2043- 2048.

21. Lohoff M, Ferrick D, MittrScker H-W, Duncan GS, Bischof S, . RBllinghoff M, Mak TW: Interferon regulatory factor-i is required

for a T helper 1 immune response in viva. lmmunify 1998, 6:681_ 689.

This paper shows the in viva importance of IRF-1 in the regulation of Thl responses using the murine model of Leishmania major.

22.

23.

24.

25.

26.

27.

28.

29. . .

Afonso LCC, Sharton TM, Vieira LQ, Wysocka M, Trinchieri G, Scott P: The adjuvant effect of interleukin-12 in a vaccine against Leishmania major. Science 1994, 263:235-237.

Scharton-Kersten T, Afonso LCC, Wysocka M, Trinchieri G, Scott P: IL-12 is required for natural killer cell activation and subsequent T helper I cell development in experimental leishmaniasis. J lmmunol 1995, 154:5320-5330.

Campbell KA, Ovendale PJ, Kennedy MK, Fanslow WC, Reed SG, Maliszewski CR: CD40 ligand is required for protective cell- mediated immunity to Leishmania major. /nununify 1996,4:283-289.

Kamanaka M, Yu P, Yasui T, Yoshida K, Kawabe T, Horii T, Kishimoto T, Kikutani H: Protective role of CD40 in Leishmania major infection at two distinct phases of cell-mediated immunity. immunity 1996, 4:275-261.

Soong L, Xu J-C, Grewa LIS, Kima P, Sun J, Longley BJ, Ruddle NH, McMahon-Pratt D, Flavell RA: Disruption of CD40-CD40 ligand interactions results in an enhanced susceptibility to Leishmania amazonensis infection. immunity 1996, 4:263-273.

Brown JA, Titus RG, Nabavi N, Glimcher LH: Blockade of CD86 ameliorates Leishmania major infection by down regulating the Th2 response. J lnfec Dis 1996, 147:1303-l 308.

Brown DR, Green JM, Moskowitz NH, Davis M, Thompson CB, Reiner SL: Limited role of CD28-mediated signals in T helper subset differentiation. J Exp Med 1996, 184:803-810.

Fowell DJ, Magram J, Turck CW, Killeen N, Locksley RM: Impaired Th2 subset development in the absence of CD4. immunity 1997, 6:559-569.

An interesting study demonstrating the importance of the CD4 molecule in Th2 cell development.

30. Brown DR, Moskowitz NH, Killeen N, Reiner SL: A role of CD4 in peripheral T cell differentiation. J Exp Med 1997, 186:lOi -107.

{is paper demonstrates the importance of the CD4 molecule in Th2 cell development.

31. O’Garra A: Cytokines induce the development of functionally heterogeneous T helper cell subsets. immunity 1998, 8:275-283.

32. Kohno K, Kataoka J, Ohtsuki T, Suemoto Y, Okamoto I, Usui M, lkeda M, Kurimoto M: IFN-7 inducing factor (IGIF) is a costimulatory factor on the activation of Thl but not Th2 cells and exerts its effect independently of IL-12. J lmmunoll997, 158:1541-l 550.

33. Robinson D, Shibuya K, Mui A, Zonin F, Murphy E, Sana T, Hartley SB, . . Menon S, Kastelein R, Bazan F, O’Garra A: IGIF does not drive Thl

development but synergizes with IL-12 for IFN-yproduction and activates IRAK and NF-kB. immunity 1997, 7:571-581.

This paper shows that IL-1 8 does not by itself induce Thl cell development but acts in synergy with IL-1 2, activating the IL-1 -receptor-associated kinase (IRAK) transcription factor.

34.

35.

36.

37.

38.

39.

40.

41.

42. . .

Shibuya K, Robinson D, Zonin F, Hartley SB, Macatonia SE, Somoza C, Hunter CA, Murphy KM, O’Garra A: IL-la and TNF-a are required for IL-12-induced development of Thl cells producing high levels of IFN-yin BALB/c but not C57BL/6 mice. J /mmuno/1998, 160:1708-1716.

Takeda K, Tsutsui H, Yoshimoto T, Adachi 0, Yoshida N, Kishimoto T, Okamura H, Nakanishi K, Akira S: Defective NK cell activity and Thl response in IL-l 8-deficient mice. immunity 1998, 8:383-390.

Kopf M, Brombacher F, KGlher G, Kienzle G, Widmann K-H, Lefrang K, Humborg C, Ledermann B, Solbach W: IL-4 deficient BALB/c mice resist infection with Leishmania major. J Exp Med 1996, 184:l 127-1 136.

Launois P, Ohteki T, Swthart K, MacDonald HR, Louis JA: In susceptible mice, Leishmania major induce very rapid interleukin- 4 production by CD4+ T cells which are NK1.1.. Eur J lmmunol 1995, 25:3298-3307.

Yoshimoto T, Bendelac A, Watson C, Hu-Li J, Paul WE: Role of NKi.i+ T cells in a Th2 response and in immunoglobulin E production. Science 1995, 270:1845-l 847.

Yoshimoto T, Bendelac A, Hu-Li J, Paul WE: Defective IgE production by SJL mice is linked to the absence of CD4+ NKI .l+ T cells that promptly produce interleukin 4. froc Nat/ Acad Sci USA 1995,92:11931-l 1934.

Brown DR, Fowell DJ, Cony DB, Wynn TA, Moskowitz NM, Cheever AW, Locksley RM, Reiner SL: j32-microglobulin-dependent NKi.i+ T cells are not essential for T helper cell 2 immune responses. J Exp Med 1996,184:1295-l 304.

Von der Weid T, Beebe AM, Roopenian DC, Coffman RL: Early production of IL-4 and induction of Th2 responses in the lymph node originate from an MHC class l-independent CD4+ NKl.I- T cell population. J lmmunoll996, 157:4421-4427.

Launois P, Maillard I, Pingel S, Swihart K, Xenarios I, Acha-Orbea H, Diggelmann H, Locksley RM, MacDonald HR, Louis JA: IL-4 rapidly produced by VP Vu8 CD4+ T cells in BALB/c mice infected with Leishmania major instructs Th2 cell development and susceptibility to infection. immunity 1997, 6:541-549.

This paper shows that a highly restricted subpopulation of CD4+ T cells recognising a specific epitope of the Leishmania-activated C kinase (LACK) antigen of L. major produces IL-4 very rapidly in response to infection. The importance of these cells in Th2 cell development and susceptibility to infection is clearly demonstrated.

43. Mougneau E, Altare F, Wakil AE, Zheng S, Coppola T, Wang ZE, Waldmann R, Locksley RM, Glaichenhaus N: Expression cloning of a protective Leishmania antigen. Science 1995, 268:563-566.

44. Launois P, Swihart K, Milon G, Louis JA: Early production of IL-4 in . susceptible mice infected with Leishmania major rapidly induces

IL-1 2 unresponsiveness. J lmmunoll997, 158:3317-3324. This paper shows that, as soon as three days after infection with L. major, CD4+ T cells from BALB/c mice become unresponsive to IL-1 2 and that this unresponsive state results from the early production of IL-4 by Vp4+ Va8+ CD4+ T cells. It is related to reference [58].

45.

46.

47.

48.

49.

50.

Julia V, Rassoulzadegan M, Glaichenhaus N: Resistance to Leishmania major induced by tolerance to a single antigen. Science 1996, 274:421-423.

Heinzel FP, Rerko RM, Ahmed F, Pearlman E: Endogenous IL-1 2 is reauired for control of Th2 cvtokine resoonses caoable of exacerbating Leishmaniasis-in normall; resistantmice. J lmmunol 1995, 155:730-739.

Magram J, Connaughton SE, Warrier RR, Carvajal DM, Wu C-Y, Ferrante J, Stewart C, Sarmiento Y, Faherty DA, Gately MK: IL-12- deficient mice are defective in IFN-yproduction and type 1 cytokine responses. immunity 1996, 4:471-481.

Mattner F, Magram J, Ferrante J. Launois P, Di Padova K, Behin R, Gately MK, Louis JA, Alber G: Genetically resistant mice lacking interleukin-12 are susceptible to infection with Leishmania major and mount a polarized Th2 cell response. Eur J lmmunoll996, 26:1553-l 559.

Heinzel FP, Schoenhaut DS, Rerko RM, Rosser LE, Gately MK: Recombinant interleukin 12 cures mice infected with Leishmania major. J Exp Med 1993, 177:1505-l 509.

Sypek JP, Chung CL, Mayor SEH, Subramanyam SJ, Goldman SJ, Sieburth DS, Wolf SF, Schaub RG: Resolution of cutaneous leishmaniasis: interleukin I2 iniates a protective T helper type 1 immune response. J Exp Med 1993,177:1797-l 802.

Page 6: Regulation of protective immunity against Leishmania major in mice

464 Immunity to infection

51.

52.

53.

54. .

Wang ZE, Zheng S, Cony DB, Dalton DK, Seder RA, Reiner SL, Locksley RM: Interferon gamma-independent effects of interleukin 12 administered during acute or established infection due to Leishmania major. Proc NatI Acad Sci USA 1994, 91 :I 2932-l 2936.

Reiner SL, Zheng S, Wang ZE, Stowring L, Locksley RM: Leishmania promastigotes evade interleukin 12 (IL-1 2) induction by macrophages and stimulate a broad range of cytokines from CD4+ T cells during initiation of infection. J Exp Med 1994, 179:447-456.

Vieira LQ, Hondowicz BD, Afonso LCC, Wysocka M, Trinchieri G, Scott P: Infection with Leishmania major induces interleukin-I 2 production in vivo. lmmunol Lett 1994, 40:157-l 61.

Sutterwala FS, Noel GJ, Clynes R, Mosser DM: Selective suppression of interleukin-I 2 induction after macrophage receptor ligation. J Exp Med 1997, 185:1977-l 985. _ _ ,

This paper shows that llgatlon ot some receptors on the surtace ot macrophages results in impaired IL-1 2 production.

55. Marth T. Kelsall BL: Reaulation of interleukin-12 bv complement 64. . receptor 3 signaling. i&p Med 1997, 185:1987-l-995. This paper could provide a basis for the inhibition of IL-12 production by macrophages infected with Leishmania.

56.

57.

58.

59.

Carrera L, Gazzinelli RT, Badolato R, Hieny S, Mijller W, Kijhn R, Sacks DL: Leishmania promastigotes selectively inhibit interleukin 12 induction in bone marrow-derived macrophages from susceptible and resistant mice. J Exp Med 1996, 183:515-526.

65. Seder RA, Gazzinelli R, Sher A, Paul WE: lnterleukin 12 acts directly on CD4+ T cells to enhance priming for inteferon-yproduction and diminishes interleukin-4 inhibition of such priming. Proc Nat/ Acad SC; USA 1993, 90:10188-l 0192.

Hsieh CS, Macatonia SE, O’Garra A, Murphy KM: T cell genetic background determines default T helper phenotype development in vitro. J Exp Med 1995, 181:713-721.

66. . .

Beebe A, Mauze S, Schork NJ, Coffman RL: Serial backcross mapping of multiple loci associated with resistance to Leishmania major in mice. lmmunify 1997, 6:551-557.

Guler ML, Gorham JD, Hsieh CS, Mackey Al, Steen RG. Dietrich WF, Murphy KM. Genetic susceptibility to Leishmania: IL-1 2 responsiveness in Thl cell development Science 1996, 271:984-967.

This provides an lnterestmg genetlc analysis which has led to the identification of six loci related to resistance to infection with L. major. It is noteworthy that none of these loci, individually, was sufficient for the expression of a resistant phenotype.

Szabo SJ, Jacobson NG, Dighe AS, Giibler U, Murphy KM: 67. Roberts LJ, Badwin TM, Curtis JM, Handman E, Foote SJ: Resistance Developmental commitment to the Th2 lineage by extinction of to Leishmania major is linked to the H2 region on chromosome 17 IL-1 2 signaling. immunity 1995, 2:665-675. and chromosome 9. J Exp Med 1997,185:1705-l 710.

60.

61.

62. . .

Gorham JD, Gi.iler ML, Steen RG, Mackey AA, Daly MJ, Frederick K, Dietrich WF, Murphy KM: Genetic mapping of a murine locus controlling development of T helper l/T helper 2 type responses. Proc Nat/ Acad Sci USA 1996, 93:12467-i 2472.

Presky DH, Yang H, Minetti LJ, Chu AO, Nabavi N, Wu C-Y, Gately M. Gtibler U: A functional interleukin 12 receptor complex is composed of two p-type cytokine receptor subunits. Proc Nat/ Acad Sci USA 1996, 93:14002-l 4007.

Szabo SJ, Dighe AS, Gijbler U, Murphy KM: Regulation of the interleukin (IL)-1 2R 82 subunit expression in developing T helper (Th) 1 and Th2 cells. J Exp Med 1997, 185:817-824.

This paper provides the molecular basis tar the loss ot responsiveness to IL- 12 of CD4+ T cells differentiating towards the Th2 phenotype in vitro.

63. Macatonia SE. Hsieh C-S. Murohv KM. O’Garra A: Dendritic cells and macropheges are requirid ior Thl development of CD4+ T cells from I$ TCR transgenic mice: IL-12 substitution for macrophages’to stimulate IFN-yproduction is IFN-ydependent. Int /mmuno/ 1993,5:1119-l 126.

Seder RA, Paul WE, Davis MM, Fazekas de St Groth B: The presence of interleukin 4 during in vitro priming determines the lymphokine-producing potential of CD4+ T cells from T cell receptor transgenic mice. J Exp Med 1992, 176:1091-l 098.