You are on page 1of 11

Parasite Immunology, 1997: 19: 29–39

IL-12 eliminates the Th-2 dependent protective immune response of


mice to larval Strongyloides stercoralis

H.L.ROTMAN 1 , S.SCHNYDER-CANDRIAN 1 , P.SCOTT 2 , T.J.NOLAN 2 , G.A.SCHAD 2 & D.ABRAHAM 1

1
Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
2
Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA

SUMMARY INTRODUCTION
The goal of the present study was to determine if immune- Strongyloidiasis, a major worldwide human intestinal infec-
mediated killing of S. stercoralis L3 in mice could be tion, is caused by the nematode Strongyloides stercoralis.
modulated by shifting from a Th-2 to a Th-1 type immune Natural hosts for this organism include humans, nonhuman
response. L3 killing in immunized mice was ablated in primates, and dogs (Schad 1989). Chronic infections are
CD4þ T cell-depleted animals, but not in CD8þ T cell- found in many patients and are thought to be maintained by
depleted or b2-microglobulin-deficient mice. Treatment of an autoinfection process. Massive and uncontrolled auto-
immunized mice with IL-4 or IL-5 neutralizing MoAb sig- infection may result in hyperinfection, which can be fatal in
nificantly reduced the protective effects of vaccination immunocompromised individuals (Genta 1992).
against S. stercoralis, while protective immunity was unim- Mice have been used as a means for studying the
paired in IFN-g knockout mice. Recombinant IL-12 was systematic phase of S. stercoralis infection, as infective
administered to infected mice to switch the immune response larvae migrate to the tissues but do not develop further
from a Th-2 to a Th-1 type response. Protective immunity (Grove, Northern & Heenan 1986). Larvae are found in the
was ablated in immunized mice that received IL-12 therapy. muscles for nine days in S. stercoralis-infected mice before
Eosinophil numbers, eosinophil peroxidase levels, and disappearing spontaneously. This disappearance is asso-
parasite-specific IgG1 levels were lowered in IL-12 treated ciated with inflammatory reactions around dying larvae,
immunized animals, and parasite-specific IgG2a levels were predominantly eosinophilic and histiocytic (Grove, Northern
increased in these animals. The data indicate that eosino- & Heenan 1986). Using live third-stage larvae (L3) for
phils are important as mediators of larval killing, and that immunization, protective immunity to L3 has been demon-
the establishment of Th-2 type immunity results in killing of strated in BALB/cByJ mice (Abraham et al. 1995). Diffusion
infective S. stercoralis L3, while a shift to Th-1 type chambers have been used to assess larval survival in immune
immunity abrogates protective responses. mice, and the only cell type significantly increased in
immunized mice in diffusion chamber fluid and peripheral
Keywords IL-12, Strongyloides stercoralis, nematode, blood was the eosinophil (Abraham et al. 1995). In mice
Th-2 immunity treated with MoAb to eliminate eosinophils, protective
immunity to L3 contained within diffusion chambers is
significantly reduced (Rotman et al. 1996). Human eosinophil
granule products are toxic for host-adapted L3, indicating that
larvae are susceptible to eosinophil-mediated destruction
(Rotman et al. 1996). Humoral immune components also
play an essential role in S. stercoralis larval killing. IgM is
necessary for protection against L3, in that IgM, and no other
isotype, purified from immune sera mediates larval killing in
naive mice (Brigandi et al., 1996). Only IgM binds to the
surface of S. stercoralis L3 in immune serum, while no
Correspondence: D.Abraham
antibody isotypes from control serum bind significantly to
Received: 28 August 1996 the surface of L3 (Abraham et al. 1995). Complement was
Accepted for publication: 17 October 1996 also found to be essential in L3 killing as shown in cobra

q 1997 Blackwell Science Ltd 29


H.L.Rotman et al. Parasite Immunology

venom factor-treated mice, where no larval killing took place (pH 2.7) ad libitum. The animal housing room was tempera-
(Brigandi et al. 1996). ture, humidity, and light cycle controlled.
The importance of Th-2 cytokines such as IL-4 and IL-5
in protective immunity to challenge infection and resistance
Parasites
to primary infection has been demonstrated in several
nematode infections, including Strongyloides venezuelensis S. stercoralis L3 were harvested from the fresh stools of a
(Korenaga et al. 1991), Angiostrongylus cantonensis laboratory dog-infected with the parasite according to
(Sasaki et al. 1993), Onchocerca volvulus (Lange et al. methods previously described (Schad, Hellman & Muncey
1994), Heligmosomoides polygyrus (Urban et al. 1991), and 1984). In brief, faeces were mixed with a small amount of
Trichuris muris (Else & Grencis 1991). IL-5 is essential for water and charcoal and placed in a humidified petri dish at
eosinophil generation and development (Coffman et al. 278C for 5–7 days. The L3 were cleaned of faecal debris by
1989), and IL-4 plays a role in several processes (Tepper mixing with a liquid low-gelling-temperature agar solution
1994), including isotype switching to IgG1 and IgE (Snap- (with a final concentration of 1%) (Sigma Chemical Co., St.
per, Finkelman & Paul 1988), the adhesion and migration of Louis, MO, USA) and allowing the L3 to migrate to the
eosinophils across vascular membranes (Moser, Fehr & surface of the solidified mixture. The larvae were washed
Bruijnzeel 1992), as well as many other effects on B and five times by centrifugation and resuspended in a 1 : 1
T cells, macrophages, and mast cells (Howard et al. 1982, mixture of Iscove’s Modified Dulbecco’s Medium
Snapper & Paul 1987, Trenn et al. 1988, Fernandez-Botran (IMDM) and NCTC-135 (Sigma) with 10 000 U Penicillin,
et al. 1988, Mosmann et al. 1986, McInnes & Rennick 1988, 10 mg Streptomycin, and 10 mg Gentamicin (Sigma) per
Crawford et al. 1987). IL-12, a heterodimeric cytokine 100 ml.
produced primarily by macrophages and other accessory
cell types, can modulate Th-2 responses, favouring devel-
Immunization and challenge
opment and activation of Th-1 cells (Manetti et al. 1993,
Hsieh et al. 1993, Afonso et al. 1994). Five thousand live L3 in IMDM/NCTC medium plus
The objective of the present study was to determine how antibodies were injected subcutaneously posterior to the
the immune response to S. stercoralis infection in mice cervical area of each mouse. Booster immunizations of
could be regulated. Immunized mice depleted of CD4þ T 5000 L3 were administered two weeks after the initial
cells were unable to generate protective immunity to L3, immunization (Abraham et al. 1995).
while depletion of CD8þ T cells had no negative effect on Diffusion chambers were constructed as previously
L3 killing. Inactivation of the Th-2 cytokines IL-4 or IL-5 described (Abraham et al. 1995). Briefly, 14-mm Lucite
resulted in significant reductions in immunity to third-stage rings were covered with 2.0 mm pore-sized polycarbonate
larvae, while depletion of the Th-1 cytokine IFN-g had no Isopore membranes (Millipore, Bedford, MA, USA) or
effect on protective immunity. Mice treated with recombi- 0.1 mm pore-sized Durapore membranes (Millipore).
nant IL-12 to shift the immune response to a Th-1-type Cement for binding the Lucite rings together and to the
response lost their ability to kill challenge L3. Taken Durapore membranes consisted of equal parts of 1,2-
together, these findings indicate that Th-2-type responses dichloroethane (Sigma) and acryloid resin (Rohm and
are essential for the immune-mediated killing of S. stercor- Haas, Philadelphia, PA, USA). Cyanoacrylate adhesive
alis L3 to take place. (Super Glue Corp., Hollis, NY, USA) was used for binding
the Isopore membranes to the Lucite rings. The diffusion
chambers were sterilized by exposure to ethylene oxide gas.
MATERIALS AND METHODS
For challenge infections, 50 S. stercoralis L3, in IMDM/
NCTC medium with antibiotics, as above, were inoculated
Mice
into each diffusion chamber.
Male BALB/cByJ, C57BL/6J, b2 -microglobulin-deficient Immunized mice received challenge infections 21 days
(C57BL/6 background) mice, and IFN-g knockout mice after the initial immunization. Control mice received
(BALB/c background) were purchased from Jackson diffusion chambers at the same time as immunized
Laboratories, Bar Harbor, ME, USA. The mice ranged mice, but had not been previously imunized with larvae.
from six to 12 weeks of age at the beginning of all Animals were anaesthetized with intraperitoneal injections
experiments. Animals were housed in Micro-Isolator of a mixture of 10 mg/ml ketamine hydrochloride (Park-
boxes (Lab Products Inc., Maywood, NJ, USA) and were Advise, Morris Plains, NJ, USA) and 0.02 mg/ml acepro-
fed Autoclaveable Laboratory Rodent Chow (Ralston mazine maleate (Aveco, Fort Dodge, IA, USA). The
Purina, St. Louis, MO, USA), and sterilized acid water anaesthetic solution was administered at 0.1 ml/10 g body

30 q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39


Volume 19, Number 1, January 1997 IL-12 eliminates Th2-type immunity to S. stercoralis

weight. A single diffusion chamber was inserted into the Cytokine treatment
subcutaneous pocket formed in the rear flank of each
Groups of animals were injected i.p. daily for five days on,
animal.
and two days off, with 0.2 mg murine rIL-12 in PBS (specific
activity, 5:6 × 106 U/mg, Genetics Institute, Cambridge,
Recovery and analysis of diffusion chamber contents MA, USA). Treatment started on the day before initial
immunization with live S. stercoralis L3, and continued
All diffusion chambers were recovered one day after for a total of three weeks.
implantation. Mice were anaesthetized with Methoxyflurane
(Pitman-Moore, Inc., Mundelein, IL, USA) and sacrificed
by exsanguination. Blood for cell differential counts was ELISA procedures
collected at the time of death. Diffusion chambers were
Soluble antigen was prepared from S. stercoralis L3. Larvae
removed from the mice and the total number of cells in the
were boiled for ten min in distilled water containing 2%
diffusion chamber fluid was determined. This fluid was
SDS, and the preparation was then centrifuged at 14 000 rpm
centrifuged onto slides using a Cytospin 3 (Shandon Inc.,
at RT for 10 min. Tris buffer, pH 8.8, was added to the
Pittsburg, PA, USA). Slides were stained with DiffQuik
supernatant, to a final concentration of 50 mM. The solution
(Baxter Healthcare Corp., Dade Div., Miami, FL, USA).
was passed through a detergent-removing gel column
Larvae recovered from diffusion chambers were considered
(Pierce, Rockford, IL, USA), and aliquots were collected
live if they were motile.
and used to coat ELISA plates. Protein concentration in the
aliquots was determined by staining with Coomassie Plus
Cytokine depletion protein assay reagent (Pierce, Rockford, IL, USA). The
ELISA was performed by coating each well of the ELISA
The following rat MoAb were used in vivo cytokine deple- plate with 50 ml of antigen at a concentration of 10 mg
tion: anti-mouse IL-5 (TRFK-5) (Coffman et al. 1989) and protein per ml of 50 mM Tris buffer, pH 8.8. The plates
anti-mouse IL-4 (11B11) (Ohara & Paul 1985) (cell lines were incubated at 378C for 2 h. Excess antigen was washed
contributed by Dr R.L.Coffman, DNAX Corp, Palo Alto, away with distilled water. Unoccupied sites in the antigen-
CA, USA). Rat IgG (Sigma Chemical Co., St. Louis, MO, coated wells were blocked using 200 ml of a blocking
USA) was used as the IgG control antibody. Mice that were solution containing 0.25% bovine serum albumin (BSA),
challenged with L3 in diffusion chambers were injected and 0.05% Tween-20 in PBS, pH 7.2 at 378C for 1 h. After
twice i.p. with each MoAb. The first dose was given seven several washes as above, 50 ml of serum samples diluted in
days before challenge, and the second dose at the time of blocking solution were added to each well. Serum-free
challenge. Doses of MoAb were as follows: TRFK-5, 1 mg/ blocking solution served as a negative control. The plates
mouse/dose. The initial dose of 11B11 and the rat control were reincubated at 378C for 2 h and then washed several
Abs was 10 mg/mouse/dose, and the remaining treatments times with distilled water. Secondary and tertiary antibodies
were 5 mg/mouse/dose. were as follows: goat anti-mouse IgG1 (1 : 4000 dilution),
goat anti-mouse IgG2a (1 : 4000 dilution), biotinylated goat
anti-mouse IgM (1 : 4000 dilution), and biotinylated rabbit
Depletion of T cell populations by MoAb treatment
anti-goat IgG (1 : 1000 dilution). All secondary and tertiary
The following rat MoAb were used for in vivo T cell antibodies were from Sigma ImmunoChemicals, and were
depletion: anti-mouse CD4 (GK1.5) (Dialynas et al. 1983, diluted in blocking solution. After each step, the plates were
Wofsy et al. 1985) and anti-mouse CD8 (H35-17.2) (Sar- incubated at 378C for 2 h, and washed with distilled water.
miento, Glasebrook & Fitch 1980). Mice that were chal- Peroxidase conjugated avidin (Cappel, Organon Teknika
lenged with L3 in diffusion chambers were injected i.p. with Corp., Westchester, PA, USA) at a 1 : 4000 dilution in
six doses of anti-CD4 MoAb, or five doses of anti-CD8 blocking solution was added to the plates for 30 min at
MoAb. The first dose of GK1.5 MoAb was given seven 378C. After thorough washing, the enzyme substrate
days, and second dose five days before initial immunization. (ABTS substrate, Kirkegaard and Perry Laboratories, Inc.,
The first dose of H35-17.2 MoAb was given six days before Gaithersburg, MA, USA) was added (75 ml per well). The
initial immunization. All other doses were given at the time enzyme-substrate reaction was allowed to occur at 378C in
of initial immunization, and seven, 14, and 21 days after the dark for 20 min. The optical density of the contents of
initial immunization. Doses of the MoAb were as follows: each well was measured at 410 nm against the blank using a
GK1.5, 1 mg/mouse/dose; and H35-17.2, 0.25 mg/mouse/ Dynatech MR5000 ELISA reader (Dynatech Laboratories,
dose. Inc., Chantilly, VA, USA).

q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39 31


H.L.Rotman et al. Parasite Immunology

Surface staining (Sigma) was added to each well for 30 min. Worms were
placed on slides, and fluorescence was analysed as described
50 ml of undiluted pooled mouse serum was placed with
above.
100–400 live S. stercoralis L3 at 378C for 2 h. Pooled
mouse serum was from 5–7 individual mice. One aliquot
of worms was placed in serum from mice that had been Measurement of eosinophil peroxidase levels
immunized, and the other was placed in serum from
control mice. The worms were then washed five times The method of Strath, Warren & Sanderson 1985, as
in PBS, and were placed in individual wells in a 96-well modified by White et al. 1991 was used for this assay.
round-bottomed ELISA plate. L3 were placed in rat anti- Briefly, 50 ml of diffusion chamber fluid was combined
mouse IgM diluted 1:20 in PBS for one hour at 378C, with 75 ml of a 16 mM o-phenylenediamine solution in
followed by a similar incubation in goat anti-rat FITC 100 nm Tris-HCl, pH 8.0 containing 0.1% Triton X-100
antibodies diluted 1:50 (Sigma). The worms were washed and 0.01% hydrogen peroxide. Polystyrene 96-well micro-
four times in PBS to remove excess antibody, and once in titre plates containing the reactions were placed at 378C
a 1% formalin-PBS solution. Worms were placed on until a color change could be detected, and absorbance at
slides, and fluorescence was analysed using fluorescent 492 nm was measured using a Dynatech Microtiter Plate
microscopy. Reader (Dynatech Laboratories, Inc., Chantilly, VA,
Worms recovered from 0.1 mm diffusion chambers were USA). Horseradish peroxidase (Sigma) was used as a
tested for surface complement binding, 0.1 mm diffusion standard.
chambers, which do not permit cell entry, allowed >90%
of implanted larvae to survive while in the host. Larvae
Statistics
removed from diffusion chambers were fixed in 3%
formalin, aliquoted into 96-well round-bottom plates, All experiments were done a minimum of two times, and the
washed with PBS, and exposed to goat anti-mouse com- tables and graphs presented in this paper are representative
plementary protein 3 (C3) (Cappel, Organon-Teknika examples of experiments performed. Data were analysed
Corp., Durham, NC, USA) at a 1 : 20 dilution for 1 h. when there were two groups by Student’s t-test and when
After washing in PBS, biotin-conjugated rabbit anti-goat there were multiple groups by ANOVA in Systat 5.2 (Systat,
IgG (Sigma) was added to each well for 1 h at dilutions of Inc., Evanston, IL, USA). Probability values of less than
1 : 50. After a final washing in PBS, avidin-FITC (1 : 100) 0.05 were considered significant.

Figure 1 Effect of depleting CD4þ T cells on immune-mediated killing of challenge S. stercoralis larvae. Cells were depleted by MoAb treatment
with GK1.5, at 1 mg/mouse/dose. MoAb was given seven days before, five before, and at the time of initial immunization, and seven, 14 , and 21
days after initial immunization. Mean percentage 6 SD of larval recovery in diffusion chambers. n ¼ 5 mice per group. *P < 0:05: difference
between results in control and immunized mice in like-treated groups.

32 q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39


Volume 19, Number 1, January 1997 IL-12 eliminates Th2-type immunity to S. stercoralis

RESULTS necessary for protective immunity to S. stercoralis L3


(Rotman et al. 1996). Eosinophil numbers in the peripheral
Depletion of CD4þ T cells ablates protective immunity blood and diffusion chambers of immunized mice treated
with anti-CD4 and anti-CD8 were equivalent to the levels
In order to determine which subset of T cells was
found in untreated immunized mice (data not shown).
responsible for generating protective immunity to S.
Eosinophil peroxidase is an indicator of eosinophil numbers
stercoralis L3, CD4þ and CD8þ , T cells were depleted
and activation in tissue (Strath, Warren & Sanderson 1985).
in immunized animals, and larval killing in these animals
Diffusion chamber fluid from chambers implanted in control
was assessed. CD4þ T cells were depleted by treating
and immune mice was analysed for levels of eosinophil
BALB/cByJ mice with the MoAb GK1.5 (Wofsy et al.
peroxidase. Immune mice had eosinophil peroxidase levels
1985). Significantly fewer challenge L3 were recovered
significantly higher than those seen in control mice, in all
from diffusion chambers from immune mice than from
animals tested. In immunized mice which received anti-
control mice. However, protective immunity in immunized
CD4 MoAb, however, eosinophil peroxidase levels dropped
mice was almost completely ablated when mice were
to the levels seen in control mice (Figure 3). Immunized
depleted of their CD4þ T cells (Figure 1). Depletion of
mice which received anti-CD8 MoAb showed no diminish-
CD8þ T cells by MoAb treatment with H35-17.2 did not
ment of eosinophil peroxidase levels when compared to
affect the protective immunity seen in immunized BALB/
untreated immune mice peroxidase levels (Figure 3).
cByJ mice (Figure 2). In addition, immunized b2 -micro-
globulin-deficient mice, which lack CD8þ T cells (Zijlstra
et al. 1990), still developed significant protective immu-
Depletion of IL-4 or IL-5 diminishes protective
nity that was equivalent to that seen in intact immunized
immunity to S. stercoralis L3
mice of the same genetic background (C57/BL6) (Figure 2).
Th-2-type CD4þ cells generate IL-4 and IL-5 (Mosmann et al.
Eosinophil peroxidase levels are diminished in 1986), which are responsible for generating and localizing
anti-CD4 treated mice eosinophils in tissue spaces. BALB/cByJ mice were treated
with MoAbs to eliminate either IL-4 or IL-5. Immunized
Previously, it has been suggested that eosinophils are
mice, untreated with MoAb, had a significant decrease in
parasite survival. Immunized mice treated with either anti-
IL-4 or anti-IL-5 had challenge worm recovery rates that
were not significantly different from those found in control
mice (Figures 4 and 5). The concentration of eosinophils
was elevated in immunized groups of mice that received no
MoAb treatment, but immunized mice which received anti-
IL-5 MoAb had reduced numbers of eosinophils (Figure 4).
No such eosinophil reduction was seen in mice given anti-
IL-4 MoAb, however (Figure 5). Immunized mice treated
with control Ab (rat IgG) had larval survival equivalent to
that seen in untreated immunized mice (data not shown).

IFN-g is not essential for protective immunity


BALB/cByJ and IFN-g knockout mice (BALB/c back-
ground) were immunized as described above. Larvae con-
tained within diffusion chambers were then implanted
Figure 2 Effect of depleting CD8þ T cells on immune-mediated
subcutaneously in both naive and immunized mice, and
killing of challenge S. stercoralis larvae. Cells were depleted by
MoAb treatment with H35-17.2 or by use of b2-microglobulin one day later the diffusion chambers were removed, and
deficient mice. MoAb was given six days before and at the time of larval survival was determined. Larval recoveries were as
initial immunization, and seven, 14, and 21 days after initial follows: Control BALB/cByJ, 57 6 11%; Immune BALB/
immunization, at 1 mg/mouse/dose. Mean percentage 6 SD of larval cByJ, 3 6 4%; Control IFN-g knock-out, 36 6 20%;
recovery in diffusion chambers. n ¼ 5 mice per group. *P < 0:05:
Immune IFN-g-knockout, 3 6 2%. The concentration of
difference between results in control and immunized mice in like-
treated groups. A BALB/cByJ; B BALB/cByJ MoAb depletion; d eosinophils was elevated in all immunized groups of mice
C57B1/6; c C57B1/6 b2 microglobulin deficient. (data not shown).

q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39 33


H.L.Rotman et al. Parasite Immunology

Figure 3 U/ml of eosinophil peroxidase found in diffusion chambers from anti-CD4 or anti-CD8 MoAb treated and untreated BALB/cByJ mice at
the time of larval challenge recovery of S. stercoralis L3 n ¼ 4 or 5 samples per group tested. *P < 0:05: difference between results in control and
immunized mice in like-treated groups. A; control; d immune.

Figure 4 Effect of depleting IL-5 on immune-mediated killing of challenge S. stercoralis larvae. IL-5 was neutralized by MoAb treatment with
TRFK-5, at 1 mg/mouse/dose. MoAb was given at the time of booster immunization, and at the time of parasite challenge. Mean percentage 6 SD
of larval recovery (left) and mean number 6 SD of eosinophils (right) found in diffusion chambers at the time of larval challenge recovery. n ¼ 7
mice per group. *P < 0:05: difference between results in control and immunized mice in like-treated groups. A control; d immune.

34 q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39


Volume 19, Number 1, January 1997 IL-12 eliminates Th2-type immunity to S. stercoralis

Figure 5 Effect of depleting IL-4 on immune-mediated killing of challenge S. stercoralis larvae. IL-4 was neutralized by MoAb treatment with
11B11, at 10 mg/mouse/dose at the time of booster immunization, and at 5 mg/mouse/dose at the time of parasite challenge. Mean percentage
6 SD of larval recovery (left) and mean number 6 SD of eosinophils (right) found in diffusion chambers at the time of larval challenge recovery.
n ¼ 4 mice per group. *P < 0:05: difference between results in control and immunized mice in like-treated groups. A control; d immune.

Recombinant IL-12 therapy abolishes protective levels at a dilution of 1 : 128), and a two-fold decrease in
immunity IgG1 levels (Immune sera dropped to background levels at a
dilution of 1 : 512, while sera from immunized mice treated
The effect of shifting the Th-2 response to S. stercoralis L3
with rIL-12 dropped to background levels at a dilution of
to a Th-1 response by rIL-12 treatment was analysed.
1 : 128), when compared to untreated immune sera, indicating
BALB/cByJ mice were treated with daily injections of
that a shift to a Th-1 type response had occurred. No
rIL-12 during the course of immunization with S. stercoralis
differences were seen between rIL-12 treated and untreated
L3. Immunized mice, untreated with rIL-12, had a signifi-
control sera in regard to IgG1 and IgG2a levels (background
cant decrease in parasite survival. In contrast, immunized
levels were seen in all control sera at a dilution of 1 : 16).
mice treated with rIL-12 had challenge worm recovery rates
As parasite-specific IgM is essential for S. stercoralis L3
that were not significantly different from those found in
killing in this mouse model (Brigandi et al., 1996), IgM
control mice (Figure 6). The concentration of eosinophils
antibody recognition of S. stercoralis L3 surface and soluble
was elevated in immunized groups of mice, with the excep-
antigens was analysed in sera, to determine whether these
tion of those treated with rIL-12 (Figure 6).
responses had been altered by IL-12 treatment. IgM in sera
from rIL-12-treated and untreated immunized mice at dilu-
tions up to 1 : 50 recognized surface antigens of L3, while
The effect of recombinant IL-12 therapy on humoral
IgM in both groups of control sera did not. Parasite-specific
responses
ELISA, using soluble antigen preparation, also showed no
In order to determine whether the Th-2 response had shifted differences in IgM levels between IL-12-treated and
to a Th-1 response in IL-12-treated immunized animals, untreated immune serum groups, as both types of sera
individual mouse sera were tested for parasite-specific anti- dropped to background levels at serum dilutions of
body reactivity using IgG1 and IgG2a-antigen-specific 1 : 20 000.
ELISA. Immune sera from rIL-12-treated animals had an Complement has also been shown to be necessary for
eight-fold increase in IgG2a levels (Immune sera dropped to L3 killing in the mouse model (Brigandi et al. 1996), and
background levels at a dilution of 1 : 16, while sera from so surface complement binding to L3 was analysed in
immunized mice treated with rIL-12 dropped to background IL-12-treated animals. L3 recovered from 0.1 mm diffusion

q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39 35


H.L.Rotman et al. Parasite Immunology

Figure 6 Effect of administering rIL-12 on immune-mediated killing of challenge S. stercoralis larvae. Groups of animals were injected i.p. daily
for five days on, and two days off, with 0.2 mg murine rIL-12 in PBS. Therapy started on the day before initial immunization with live
S. stercoralis L3, and continued for a total of three weeks. Mean percentage 6SD of larval recovery found in diffusion chambers (left) and mean
number 6 SD of eosinophils (right) found at the time of larval challenge recovery. n ¼ 5 mice per group. *P < 0:05: difference between results in
control and immunized mice in like-treated groups. A control; d immune.

chambers implanted in rIL-12-treated and untreated immu- animals with H. polygyrus (Urban et al. 1991), Trichinella
nized mice showed equally strong surface fluorescence at spiralis (Herndon & Kayes 1992), Schistosoma mansoni
24 h post-implantation, whereas L3 recovered from both (Sher et al. 1990), and Nippostrongylus brasiliensis (Coff-
treated and untreated control mice showed background man et al. 1989), has been shown to block the development
levels of surface fluorescence. of eosinophilia in mice but does not prevent the develop-
ment of resistance to these infections.
In addition to IL-5, Th-2 cells produce several other
DISCUSSION
cytokines, including IL-4 (Mosmann et al. 1986). IL-4 has
CD4þ T cells were found to be essential in immune- many immune related functions, including inducing the
mediated killing of S. stercoralis L3 in this model system, production of IgG1 and IgE isotypes (Snapper, Finkelman
while depletion of CD8þ T cells had no effect. CD4þ T cells & Paul 1988). Anti-IgG1 and anti-IgE treatments have
can be divided into Th-1 and Th-2 subsets, based on the been found to have no effect on protective immunity to
cytokines they produce. IL-5, a Th-2 cytokine, has been S. stercoralis L3 (Brigandi et al. 1996), indicating that
shown to be essential for the generation and development of alterations in IgG1 and IgE levels, which are potentially
eosinophils (Coffman et al. 1989). Eosinophils have been affected by IL-4, are not relevant to protective immunity
demonstrated to be an essential component of the protective in this model system. IL-4 also plays a role in the adhesion
immunity to S. stercoralis larvae in mice (Rotman et al. and migration of eosinophils across vascular membranes
1996). In this study, depletion of IL-5 was found to diminish using human endothelial cells (Moser, Fehr & Bruijnzeel
protective immunity to S. stercoralis L3 contained within 1992).
diffusion chambers. Immunized mice had eosinophils Eosinophil numbers found in diffusion chambers and peri-
reduced by treatment with MoAb to IL-5. Immunity to pheral blood are not altered in IL-4 depleted animals. This is
several parasite species has been found to depend on IL-5, in agreement with other studies, in which neutralization of
including O. volvulus (Lange et al. 1994), A. cantonensis IL-4 using MoAb had little effect on the influx of eosino-
(Sasaki et al. 1993), and S. venezuelensis (Korenaga et al. phils during an antigenic challenge (Corry et al. 1996).
1991, 1994). However, administration of anti-IL-5 to However, IL-4 has been shown to have an effect on

36 q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39


Volume 19, Number 1, January 1997 IL-12 eliminates Th2-type immunity to S. stercoralis

eosinophil localization and infiltration in other systems the production of Th-1-associated cytokines. In addition, IL-
(Tepper 1994). The demonstration that IL-4 is required 12 treatment has less of an effect once Th-2 responses have
for protective immunity to S. stercoralis L3, yet has no already become established.
effect on eosinophil numbers at the site of parasite chal- Because IL-12 induces IFN-g production and promotes
lenge, leaves unanswered the mechanism by which protec- differentiation of Th-1 cells (Chan et al. 1991, Germann
tion is diminished. Immunity to H. polygyrus is dependent et al. 1993), it was determined whether IL-12 treatment,
on IL-4, yet IL-4 operates in a mechanism independent of initiated one day prior to initial immunization, would affect
IgE, mast cells, and eosinophils (Urban et al. 1992). Thus, protective immunity to S. stercoralis L3 challenge. Indica-
IL-4 is contributing some essential component to immunity tions that a Th-1-type response was generated in IL-12-
against S. stercoralis which has yet to be identified. IL-4 treated animals comes from the observation that parasite-
has an effect on many other components of the immune specific IgG2a levels increased eight-fold and IgG1 levels
system such as B cell proliferation (Howard et al. 1982), decreased by two-fold, when compared to untreated immu-
IgG2a inhibition (Snapper & Paul 1987), cytotoxic and nized animals. Larval killing was completely ablated in
helper T cell growth and differentiation (Trenn et al. 1988, immunized mice which received IL-12 therapy. Three
Fernandez-Botran et al. 1988), mast cell growth (Mosmann components essential for protective immunity to S. stercor-
et al. 1986), and macrophage cytotoxicity and inhibition alis in mice have been defined: eosinophils (Rotman et al.
(McInnes & Rennick 1988, Crawford et al. 1987), and so it 1996), IgM (Brigandi et al. 1996), and complement (Brigandi
is hypothesized that some other crucial component of the et al. 1996). C3 levels on worms recovered from immune
protective immune response is being altered in anti-IL-4- mice and IgM levels binding to soluble antigen and on the
treated mice. surface of L3 were identical in both IL-12-treated and
In order to measure differences in the number and func- untreated immunized mice. However, the decrease in protec-
tion of eosinophils in this model system, the amount of tive immunity was associated with a decrease in the number
eosinophil peroxidase found within the diffusion chambers of eosinophils found both in diffusion chamber fluid and in
of immunized and control animals was determined. Mea- the peripheral blood. It appears that eosinophils decrease in
surement of eosinophil peroxidase activity has been utilized IL-12-treated animals due to the effects IL-12 has in dimin-
as an assay of eosinophil function and number in biologic ishing Th-2 responses, whether directly or through induced
fluids (White et al. 1991). Mice immunized against S. IFN-g production (Chan et al. 1991, Germann et al. 1993,
stercoralis L3 showed increased EPO levels in diffusion Kobayashi et al. 1989). IL-12 administration has also been
chamber fluid. When immune mice were depleted of CD4þ shown to induce neutropenia, lymphopenia, and anaemia in
T cells, EPO levels in diffusion chambers dropped to back- mice (Brunda 1994, Eng et al. 1995), though eosinophils
ground levels. Depletion of CD8þ T cells had no negative were the only cell types to decrease in treated animals in this
effect on EPO levels. These results further indicate that system. The use of IFN-g knockout mice had no effect on
depleting the T cells responsible for a Th-2-type response in larval survival in immunized mice, indicating that this Th-1
mice results in a lower number of eosinophils. As eosino- cytokine was not essential in generating protective immunity
phils are essential in immune-mediated killing of S. stercor- to S. stercoralis L3.
alis L3, this reduction of eosinophils coincides with loss of These results indicate that the generation of a Th-2 type
L3 killing ability. response is necessary to kill third-stage larvae in S. stercor-
Several studies have demonstrated that reduced eosino- alis-immunized mice. When this response is shifted to a
philia occurs when animals are given exogenous IL-12 Th-1 response by administration of IL-12, protective immu-
during established Th-2 responses (Finkelman et al. 1994, nity is ablated. Eosinophil levels are altered in IL-12-treated
Wynn et al. 1994, Pearlman et al. 1995). However, when animals, while IgM levels and complement activation
rIL-12 is given before a Th-2 response has been established, appear unaltered. The reduction in eosinophils appears be
there is a more complete shift from Th-2 to Th-1 type responsible for the loss of protective immunity seen in this
responses. When given at the beginning of some parasitic model system. These results indicate the importance of
infections, IL-12 can promote protection and prevent pathol- generating the correct T-helper cell response to an infection,
ogy in host animals (Afonso et al. 1994, Finkelman et al. and demonstrate how a shifting to the opposing T cell
1994, Heinzel et al. 1993, Stevenson et al. 1995, Gazzinelli response can completely ablate protective immunity.
et al. 1993, Sypek et al. 1993). These findings all suggest
that IL-12, if given at the beginning of a developing immune
Abbreviations
response, can change the response from one that is char-
acterized by the production of Th-2-associated cytokines, ANOVA, analysis of variance; ELISA, enzyme-linked
along with associated eosinophilia, to one characterized by immunosorbant assay; EPO, eosinophil peroxidase; Ig,

q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39 37


H.L.Rotman et al. Parasite Immunology

immunoglobulin; IMDM, Iscove’s Modified Dulbecco’s Finkelman F.D., Madden K.B., Cheever A.W. et al. (1994) Effects of
Medium; L3, infective third stage larvae; PBS, phosphate interleukin 12 on immune responses and host protection in mice
infected with intestinal nematode parasites. Journal of Experimental
buffered saline; NCTC-135, National Cancer Institute Tissue
Medicine 179, 1563–1572
Culture Medium-135; rIL-12, recombinant interleukin-12. Gazzinelli R.T., Hieny S., Wynn, T.A. et al. (1993) Interleukin 12 is
required for the T-lymphocyte-independent induction of interferon g by
an intracellular parasite and induces resistance in T-cell-deficient hosts.
ACKNOWLEDGEMENTS
Proceedings of the National Academy of Sciences, USA 90, 6115–6119
We thank Ofra Leon and De’Broski Herbert for expert Genta R.M. (1992) Dysregulation of strongyloidiasis: a new hypothesis.
Clinical Microbiological Reviews 5, 345–355
technical assistance, Dr Stanley Wolf and Dr Joseph
Germann T., Gately M.K., Schoenhaut D.S. et al. (1993) Interleukin-
Sypek from the Genetics Institute, Inc. for the kind gift of 12/T cell stimulating factor, a cytokine with multiple effects on T
rIL-12, and Dr Bice Perussia for critical reading of the helper type 1 (Th1) but not on Th2 cells. European Journal of
manuscript. This work was funded in part by NIH grant Immunology 23, 1762–1770
AI22662 and by grants from the Edna McDonnell Clark Grove D.I., Northern C. & Heenan P.J. (1986) Strongyloides stercoralis
infections in the muscles of mice: a model for investigating the
Foundation.
systematic phase of strongyloidiasis. Pathology 18, 72–76
Heinzel F.P., Schoenhaut D.S., Rerko R.M. et al. (1993) Recombinant
interleukin 12 cures mice infected with Leishmania major. Journal of
REFERENCES
Experimental Medicine 177, 1505–1509
Abraham D., Rotman H.L., Haberstroh H.F. et al. (1995) Strongyloides Herndon F.J. & Kayes S.G. (1992) Depletion of eosinophils by anti-IL-5
stercoralis: protective immunity to third-stage larvae in BALB/cByJ monoclonal antibody treatment of mice infected with Trichinella
mice. Experimental Parasitology 80, 297–307 spiralis does not alter parasite burden or immunologic resistance to
Afonso L.C.C., Scharton T.M., Vierira L.Q. et al. (1994) The adjuvant reinfection. Journal of Immunology 149, 3642–3647
effect of interleukin-12 in a vaccine against Leishmania major. Howard M., Farrar J., Hilifiker M. et al. (1982) Identification of a T-cell
Science 263, 235–237 derived B-cell growth factor distinct from interleukin 2. Journal of
Brigandi R.A., Rotman H.L., Yutanawiboonchai W. et al. (1996) Experimental Medicine 155, 914–923
Strongyloides stercoralis: role of antibody and complement in Hsieh C.-S., Macatonia S.E., Tripp C.S. et al. (1993) Development of
immunity to the third stage larvae in BALB/cByJ mice. Experimental Th1 CD4þ T cells through IL-12 produced by Listeria-induced
Parasitology 82, 279–289 macrophages. Science 260, 547–549
Brunda M.J. (1994) Interleukin 12. Journal of Leukocyte Biology 55, Kobayashi M., Fitz L., Ryan M. et al. (1989) Identification and
280–288 purification of natural killer cell stimulatory factor (NKSF), a
Chan S.H., Perussia B., Gupta J.W. et al. (1991) Induction of interferon cytokine with multiple biologic effects on human lymphocytes.
g production by natural killer cell stimulatory factor: characterization Journal of Experimental Medicine 170, 827–845
of the responder cells and synergy with other inducers. Journal of Korenaga M., Hitoshi Y., Yamaguchi N. et al. (1991) The role of
Experimental Medicine 173, 869–879 interleukin-5 in protective immunity to Strongyloides venezuelensis
Coffman R.L., Seymour B.W.P., Hudak, S. et al. (1989) Antibody to infection in mice. Immunology 72, 502–507
interleukin-5 inhibits helminth-induced eosinophilia in mice. Science Korenaga M., Hitoshi Y., Takatsu K. et al. (1994) Regulatory effect of
245, 308–310 anti-interleukin-5 monoclonal antibody on intestinal worm burden
Corry D.N., Folkesson H.G., Warnock M.L. et al. (1996) Interleukin 4, in a primary infection with Strongyloides venezuelensis in mice.
but not interleukin 5 or eosinophils, is required in a murine model of International Journal for Parasitology 24, 951–957
acute airway hyperreactivity. Journal of Experimental Medicine 183, Lange A.M., Yutanawiboonchai W., Scott, P. et al. (1994) IL-4 and IL-
109–117 5-dependent protective immunity to Onchocerca volvulus infective
Crawford R.M., Finbloom D.S., Ohara J. et al. (1987) BSF-1: a new larvae in BALB/cBYJ mice. Journal of Immunology 153, 204–211
macrophage activation factor: B cell stimulatory factor-1 (interleu- Manetti R., Parronchi P., Giudizi M.G. et al. (1993) Natural killer cell
kin-4) activates macrophages for increased tumoricidal activity and stimulatory factor (interleukin 12(IL-12)) induces T helper type 1
expression of Ia antigens. Journal of Immunology 139, 135–141 (Th1)-specific immune responses and inhibits the development of
Dialynas D.P., Quan Z.S., Wall K.A. et al. (1983) Characterization of IL-4-producing Th cells. Journal of Experimental Medicine 177,
the murine T cell surface molecule, designated L3T4, identified by 1199–1204
the monoclonal antibody GK1.5: similarity of L3T4 to the human McInnes A. & Rennick D.M. (1988) Interleukin 4 induces cultured
Leu-3/T4 molecule. Journal of Immunology 131, 2445–2451 monocytes/macrophages to form giant multinucleated cells. Journal
Else K.J. & Grencis R.K. (1991) Cellular immune responses to the murine of Experimental Medicine 167, 598–611
nematode parasite Trichuris muris. I. Differential cytokine production Moser R., Fehr J. & Bruijnzeel P.L.B. (1992) IL-4 controls the selective
during acute or chronic infection. Immunology 72, 508–513 endothelium-driven transmigration of eosinophils from allergic
Eng, V.M., Car B.D., Schnyder B. et al. (1995) The stimulatory effects individuals. Journal of Immunology 149, 1432–1438
of interleukin (IL)-12 on hematopoiesis are antagonized by IL-12- Mosmann T.R., Cherwinski H., Bond M.W. et al. (1986) Two types of
induced interferon g in vivo. Journal of Experimental Medicine 181, murine helper T cell clone. I. Definition according to profiles of
1893–1398 lymphokine activities and secreted proteins. Journal of Immunology
Fernandez-Botran R., Sanders V.M., Mosmann T.R. et al. (1988) 136, 2438–2357
Lymphokine-mediated regulation of the proliferative response of Mosmann T.R., Bond M.W., Coffman R.L. et al. (1986) T cell and mast
clones of T helper 1 and T helper 2 cells. Journal of Experimental cell lines respond to B cell stimulatory factor-1. Proceedings of the
Medicine 168, 543–558 National Academy of Sciences, USA 83, 5654–5658

38 q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39


Volume 19, Number 1, January 1997 IL-12 eliminates Th2-type immunity to S. stercoralis

Ohara J. & Paul W.E. (1985) Production of a monoclonal antibody to Strath M., Warren D.J. & Sanderson C.J. (1985) Detection of eosino-
and molecular characterization of B-cell stimulatory factor-1. Nature phils using an eosinophil peroxidase assay. Its use as an assay for
315, 333–336 eosinophil differentiation factors. Journal of Immunological Methods
Pearlman E., Heinzel F.P., Hazlett Jr, F.E. et al. (1995) IL-12 modula- 83, 209–215
tion of T helper responses to the filarial helminth, Brugia malayi. Sypek J.P., Chung C.L., Mayor S.E.H. et al. (1993) Resolution of
Journal of Immunology 154, 4658–4664 cutaneous leishmaniasis: interleukin 12 initiates a protective T helper
Rotman H.L., Yutanawiboonchai W., Brigandi R.A. et al. (1996) type 1 immune response. Journal of Experimental Medicine 177,
Strongyloides stercoralis: eosinophil dependent immune-mediated 1797-1802
killing of third stage larvae in BALB/cByJ mice. Experimental Tepper R.I. (1994) The eosinophil-mediated antitumor activity of
Parasitology 82, 267–278 interleukin-4. Journal of Allergy and Clinical Immunology 94,
Sarmiento M., Glasebrook A.L. & Fitch F.W. (1980) IgG or IgM 1225–1231
monoclonal antibodies reactive with different determinants on the Trenn G., Takayama H., Hu-Li J. et al. (1988) B cell stimulatory factor
molecular complex bearing Lyt 2 antigen block T cell-mediated 1 (IL-4) enhances the development of cytotoxic T cells from Lyt-2þ ,
cytolysis in the absence of complement. Journal of Immunology 125, resting murine T lymphocytes. Journal of Immunology 140, 1101–
2665–2672 1106
Sasaki O., Sugaya H., Ishida K. et al. (1993) Ablation of eosinophils Urban, Jr. J.F., Katona I.M., Paul W.E. et al. (1991) Interleukin-4 is
with anti-IL-5 antibody enhances the survival of intracranial worms important in protective immunity to a gastrointestinal nematode
of Angiostrongylus cantonensis in the mouse. Parasite Immunology infection in mice. Proceedings of the National Academy of Sciences,
15, 349–354 USA 88, 5513–5517
Schad G.A., Hellman M.E. & Muncey D.W. (1984) Strongyloides Urban, Jr. J.F., Madden K.B., Svetic A. et al. (1992) The importance of
stercoralis: hyperinfection in immunosuppressed dogs. Experimental Th2 cytokines in protective immunity to nematodes. Immunological
Parasitology 57, 287–296 Reviews 127, 205–220
Schad G.A. (1989) Morphology and life history of Strongyloides White S.R, Kulp G.V.P., Spaethe S.M. et al. (1991) A kinetic assay
stercoralis. In Strongyloidiasis: a Major Roundworm Infection of for eosinophil peroxidase activity in eosinophils and eosinophil
Man, ed. D.I. Grove, pp. 85–104, Taylor and Francis conditioned media. Journal of Immunological Methods 44, 257–
Sher A., Coffman R.L., Hieny S. et al. (1990) Ablation of eosinophil 263
and IgE responses with anti-IL-5 or anti-IL4 antibodies fails to affect Wofsy D., Mayes D.C., Woodcock J. et al. (1985) Inhibition of humoral
immunity against Schistosoma mansoni in the mouse. Journal of immunity in vivo by monoclonal antibody to L3T4: studies with
Immunology 145, 3911–3916 soluble antigens in intact mice. Journal of Immunology 135, 1698–
Snapper C.M. & Paul W.E. (1987) Interferon gamma and B cell 1701
stimulatory factor-1 reciprocally regulate Ig isotype production. Wynn T.A., Eltoum I., Oswald I.P. et al. (1994) Endogenous interleukin
Science 236, 944–947 12 (IL-12) regulates granuloma formation induced by eggs of
Snapper C.M., Finkelman F.D. & Paul W.E. (1988) Regulation of IgG1 Schistosoma mansoni and exogenous IL-12 both inhibits and pro-
and IgE production by interleukin 4. Immunological Reviews 102, phylactically immunizes against egg pathology. Journal of Experi-
51–75 mental Medicine 179, 1551–1561
Stevenson M.M., Tam M.F., Wolf S.F. et al. (1995) IL-12-induced Zijlstra M., Bix M., Simister N.E. et al. (1990) b2 -microglobulin
protection against blood-stage Plasmodium chabaudi AS requires deficient mice lack CD4¹ 8þ cytolytic T cells. Nature 344, 742–746.
IFN-g and TNF-a and occurs via a nitric oxide-dependent
mechanism. Journal of Immunology 155, 2545–2556

q 1997 Blackwell Science Ltd, Parasite Immunology, 19, 29–39 39

You might also like