Skip to main content
Erschienen in: BMC Immunology 1/2012

Open Access 01.12.2012 | Research article

Intermittent preventive treatment with sulfadoxine-pyrimethamine does not modify plasma cytokines and chemokines or intracellular cytokine responses to Plasmodium falciparum in Mozambican Children

verfasst von: Diana Quelhas, Laura Puyol, Llorenç Quintó, Tacilta Nhampossa, Elisa Serra-Casas, Eusébio Macete, Pedro Aide, Sergi Sanz, John J Aponte, Denise L Doolan, Pedro L Alonso, Clara Menéndez, Carlota Dobaño

Erschienen in: BMC Immunology | Ausgabe 1/2012

Abstract

Background

Cytokines and chemokines are key mediators of anti-malarial immunity. We evaluated whether Intermittent Preventive Treatment in infants with Sulfadoxine-Pyrimethamine (IPTi-SP) had an effect on the acquisition of these cellular immune responses in Mozambican children. Multiple cytokines and chemokines were quantified in plasma by luminex, and antigen-specific cytokine production in whole blood was determined by intracellular cytokine staining and flow cytometry, at ages 5, 9, 12 and 24 months.

Results

IPTi-SP did not significantly affect the proportion of CD3+ cells producing IFN-γ, IL-4 or IL-10. Overall, plasma cytokine or chemokine concentrations did not differ between treatment groups. Th1 and pro-inflammatory responses were higher than Th2 and anti-inflammatory responses, respectively, and IFN-γ:IL-4 ratios were higher for placebo than for SP recipients. Levels of cytokines and chemokines varied according to age, declining from 5 to 9 months. Plasma concentrations of IL-10, IL-12 and IL-13 were associated with current infection or prior malaria episodes. Higher frequencies of IFN-γ and IL-10 producing CD3+ cells and elevated IL-10, IFN-γ, MCP-1 and IL-13 in plasma were individually associated with increased malaria incidence, at different time points. When all markers were analyzed together, only higher IL-17 at 12 months was associated with lower incidence of malaria up to 24 months.

Conclusions

Our work has confirmed that IPTi-SP does not negatively affect the development of cellular immune response during early childhood. This study has also provided new insights as to how these cytokine responses are acquired upon age and exposure to P. falciparum, as well as their associations with malaria susceptibility.

Trial Registration

ClinicalTrials.gov: NCT00209795
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2172-13-5) contains supplementary material, which is available to authorized users.

Authors' contributions

PLA, CM, EM and PA carried out the IPTi clinical trial. PLA, CM and CD conceived, designed and coordinated the study. DQ, TN, ES and CD participated in the sample collection and processing. CD and DLD designed and supervised the immunoassays. DQ and LP carried out the immunoassays. LQ, SS and JJA performed the statistical analyses. DQ and CD drafted the first version of the manuscript. All authors read and approved the final manuscript.
Abkürzungen
APC
(allophycocyanin)
CD
(cluster of differentiation)
CI
(confidence interval)
CV
(coefficient of variation)
DTP/OPV/Hep B
(diphteria+tetanus+pertussis/oral polio vaccine/hepatitis B)
EDTA
(ethylenediaminetetraacetic acid)
EPI
(expanded program on immunization)
FITC
(fluorescein isothiocyanate)
FSC
(forward scatter)
G-CSF
(granulocyte colony stimulating factor)
GM-CSF
(granulocyte macrophage colony stimulating factor)
GM
(geometric means)
HIV
(human immunodeficiency virus)
ICS
(intracellular cytokine staining)
Ig
(immunoglobulin)
IL
(interleukin)
IFN
(interferon)
IPTi
(Intermittent Preventive Treatment in infants)
IRR
(incidence rate ratio)
MCP
(monocyte chemotactic protein)
MCAF
(monocyte chemotactic and activating factor)
MIP
(macrophage inflammatory protein)
P. falciparum (Plasmodium falciparum
)
PBS
(phosphate buffered saline)
PCR
(polymerase chain reaction)
PE
(phycoerythrin)
PerCP
(Peridinin chlorophyll protein)
PYAR
(person years at risk)
SSC
(side scatter)
SP
(sulfadoxine-pyrimethamine)
TGF
(transforming growth factor)
Th
(T helper)
TNF
(tumor necrosis factor)
WBA
(whole blood assay).

Background

In 2009 there were an estimated 68,925,435 cases of malaria in the African region (78% of worldwide estimates) and 111,885 malaria deaths (91% of worldwide estimates) [1]. These figures demonstrate that Plasmodium falciparum malaria remains a major threat to the health of Africans, in particular children under 5 years of age. In malaria endemic areas, older children and adults develop immunity to severe morbidity and death, though remaining susceptible to infection [2, 3]. Immunoglobulin passive transfer studies in humans suggest that antibodies are key mediators of naturally acquired immunity [4]. More recent data [5] suggest that cellular immunity also plays an important role in the protection against P. falciparum disease in humans [6].
Cytokines and chemokines are considered key mediators of anti-malarial immunity. For example, the production of cytokines and chemokines by activated leukocytes, including CD8+ and CD4+ T cells, mediate mechanisms such as phagocytosis and killing of parasitized erythrocytes [7] and inhibition of parasite growth and development inside hepatocytes [8].
It has been also shown that cytokines play an important role in the immunopathology of malaria and many field studies have described an association of specific cytokines with severity of disease, in particular IL-2 [9], IL-12 [10, 11], IFN-γ [9, 12], IL-1β [13], IL-6 [14, 15], TNF [9, 1322], IL-4 [2325], IL-10 [9, 12, 20, 26, 27], MIP-1β [28], and TGF-β [29].
There is less field data available on the relevance of individual cytokines in naturally acquired immunity to malaria, e.g. IL-12 [3032], IFN-γ [30, 3236], TNF [30, 34, 37], or IL-10 [38]. Previous studies have investigated individual or a few cytokines but it is more likely that a network of cytokine and chemokines determines protection or susceptibility from P. falciparum infections [39]. Most studies of cytokine responses have been cross-sectionals done after the onset of symptoms and at the initial stages of clinical illness [4043]. Only a few studies have assessed longitudinally the evolution of cellular immune responses to infection, and those have been mostly in adult populations [30, 44, 45]. Furthermore, while a number of studies have evaluated newborn cytokine responses in cord blood [4654], very few have measured prospectively cellular immune responses to P. falciparum in infants [55] or children [30, 44, 56].
In the context of a randomized, placebo-controlled trial of intermittent preventive treatment in infants with sulfadoxine-pyrimethamine (IPTi-SP) in Mozambique [57], we have previously shown that this intervention had no impact on the antibody responses to P. falciparum erythrocytic stage antigens and to variant surface antigens in Mozambican infants, nor on the capacity of antibodies to inhibit parasite growth [58, 59]. Only a handful of studies have evaluated the impact of any malaria control interventions on the development of cellular immune responses. In one study, continuous chemoprophylaxis for 3 years in Gambian children resulted in higher lymphoproliferative responses and IFN-γ production [60], and there was no clinical rebound of malaria one year after termination of prophylaxis (despite a decrease in anti-malarial antibody levels). In another study, permethrin-treated bednets showed a significant impact on percent lean body mass in Kenyan school children, thought to be due to decreased production of pro-inflammatory cytokines TNF, IL-1 and IL-6 in this group [61]. However, no study has yet assessed the impact on the child's cellular immunity of IPTi-SP, a safe and efficacious malaria control strategy consisting in the administration of a full dose of this antimalarial drug within the Expanded Program on Immunization (EPI) [62].
In the current study, we have determined prospectively the effect of IPTi-SP on levels of P. falciparum antigen-specific intracellular cytokines using flow cytometry in children up to 2 years of age. To partially overcome the difficulty to study this age group because of limited blood volumes, we used whole blood assays (WBA) instead of isolated peripheral blood mononuclear cells for measuring antigen-specific responses by intracellular cytokine staining (ICS). This approach also provides an environment more similar to that existing in vivo, and it is faster and more economical [53, 6365]. In addition, we have used luminex-based microsphere suspension array methods to measure multiple cytokines and chemokines free in small volumes of plasma [54, 66]. Finally, we assessed the factors influencing the levels of cytokines and chemokines at different time points and the associations between these and the incidence of malaria in the first and second years of life. This type of work has not been as comprehensively conducted in this age group with a longitudinal design in previous field studies.

Methods

Study area

The study was conducted at the Centro de Investigação em Saúde da Manhiça, Manhiça District, southern Mozambique. This area is characterized by a perennial malaria transmission mostly due to Plasmodium falciparum. Anopheles funestus is the main vector, and the estimated average number of infective mosquito bites per person per year was 38 (IDRC and INDEPTH network 2002). For children in this area, SP showed a combined (early and late) therapeutic efficacy rate of 83%, with an in vivo parasitological sensitivity of 78.6% at day 14 [67].

Study design

The efficacy study was an individually randomized, placebo-controlled trial [57]. Infants were recruited from those attending the EPI clinic to receive dose 2 of DTP/OPV/Hep B between September 2002 and February 2004. Treatment with SP or placebo was administered at 3, 4, 9 months of age alongside the routine vaccinations. Cross-sectional visits were scheduled at 5, 9, 12 and 24 months of age. For the ancillary immunological studies within IPTi, we included the last 501 children recruited in the main trial. At each cross-sectional visit, capillary blood collected by fingerprick was placed into EDTA microtainers (1 ml), to obtain plasma for extracellular cytokines, and into heparin microtainers (0.5 ml) for intracellular cytokine determinations. All immunological assays were performed by personnel who were blinded to the children's study group. Clinical surveillance for malaria morbidity was done through passive case detection. Written informed consent was obtained from all parents or guardians and ethical approval for the protocol was obtained from the ethics review committees of Mozambique and the Hospital Clinic, Barcelona, Spain. The trial registration number is NCT00209795 http://​clinicaltrials.​gov.
For intracellular cytokine staining (ICS) assays, among those recruited, children with the following criteria were selected: (i) having received all 3 doses of SP or placebo, (ii) having a full set of months 5, 9 and 12 blood samples or the least visits missing, and (iii) having an equal representation of SP- and placebo-recipients. Among those, 258 samples were analyzed from month 5, 208 samples from month 9 and 144 samples from month 12. Once month 24 samples were collected, 143 that had the previous set were analyzed.
For luminex assays, those children who came for all 4 visits or the least visits missing were prioritized, and plasmas were selected for analysis when there were sufficient volumes available: 229 children samples from month 5, 221 from month 9, 225 from month 12, and 158 from month 24.

Multiplex flow cytometric assay to measure cytokines and chemokines in plasma

Plasma cytokines and chemokines IL-2, IL-12 (p70), IFN-γ, IL-1β, IL-6, TNF, IL-4, IL-5, IL-8, IL-13, IL-10, IL-7, IL-17, G-CSF, GM-CSF, MCP-1 (MCAF), MIP-1β were determined using a Bio-plex 100 System (Bio-Rad, CA, USA) and following manufacturer's instructions. According to supplier availability, two types of cytokine detection kits were used (10-plex and 17-plex). Most samples (all except those from 26 children) were tested using the 17-plex kit; the first 27 children (visits 5, 9, 12 months) were tested using the 10-plex kit (which did not contain IL-1β, IL-5, IL-7, IL-8, IL-12, IL-13 IL-17, G-CSF, GM-CSF).
Assays were conducted in 96-well Millipore multiscreen filter plates (Millipore, MA, USA), running in duplicate 8 standards and 40 plasmas per plate. Plasma samples previously stored at -80°C were thawed in a water bath at 37°C, and centrifuged at 1,000 g at 4°C for 10 min. Antigen-coated luminex beads were incubated with patient plasma, in conjunction with manufacturer's reagents. Plasma samples were diluted in Bio-Plex human sample diluent as 1:3 volumes (30 μl sample + 90 μl diluent). To each sample well, the following were added in a step wise manner: 50 μl premixed beads (1×), 50 μl of diluted standard or sample, and 25 μl Bio-Plex detection antibody (1×), and 50 μl streptavidin-PE preparation (1×). The mixture was incubated for 30 min in the dark, at room temperature, on a rotating platform. The wells were then washed 3 times to remove unbound excess serum using a vacuum filtration system and a final incubation was done with streptavidin-PE for 10 min at room temperature while shaking at 300 rpm. Beads were finally resuspended in 125 μl assay buffer, and shaken on a microplate shaker at 1,100 rpm at room temperature for 30 sec immediately before reading the plate on the Luminex 100, in accordance with manufacturer's instructions. Calibration was done using the CAL2 high RP1 target value. Concentrations of unknown cytokines were calculated by plotting unknowns against a 5-parameter logistic regression standard curve of 8 points that determine the limits of detection for each cytokine, expressed as pg/ml. The coefficient of variation (%CV, reproducibility between duplicate values) needed to be < 20% to accept the value for analysis. Results were analyzed using the Bio-Rad Bioplex Manager Software v4.0.

Intracellular cytokine staining to measure P. falciparum-specific responses

CD3+ specific intracellular cytokines IFN-γ, IL-4 and IL-10 were measured by ICS. Heparinized whole blood was aliquoted into two tubes to test in parallel the stimulated and the non-stimulated (negative/background control) samples. These were respectively incubated with 20 μl extract of P. falciparum schizont lysate at 1 × 108schizonts/ml or 20 μl uninfected erythrocyte extract, both in the presence of 1 μg/ml co-stimulatory antibodies to CD28 (clone CD28.2) and CD49d (clone 9F10) (BD Biosciences, San Jose, CA) for 16 h at 37°C and 5% CO2. To block cytokine secretion, 1 μM Brefeldin A (GolgiPlug™, Pharmingen, San Diego, CA) was added during the final 4 h of incubation. To arrest activation and avoid loss of activated cells by adhesion to polystyrene tubes, 2 mM EDTA/PBS solution was added for 15 min at room temperature, and tubes were vigorously vortexed before and after the incubation. In order to lyse erythocytes and fix lymphocytes, 5 ml 1× FACS Lysing solution were added. These were vortexed, incubated for 10 min at room temperature, and immediately placed at -80°C. Frozen samples were thawed in batches at a later time for parallel processing and staining, hereby avoiding loss of function or increased background staining. Thawing was done by briefly placing samples in a 37°C water bath and immediately washing off lysing solution using 7 ml Cell Wash buffer. Samples with clumping/debris of dead cells were previously filtered through 70 μm cell sieve filters (Cell Strainer BD Falcon), and pellet was resuspended with a pipette in 2 ml permeabilizing solution before vortexing for 10 min at RT. Prior to staining, frozen cells were washed with 8 ml wash buffer and centrifuged at 1,500 rpm for 10 min at room temperature. Supernatant was decanted, residual pellet was resuspended in 500 μl wash buffer, and 100 μl of each sample were transferred to 96 well round bottom microtiter plates. Plates were centrifuged at 2,000 rpm for 5 min, then flicked to discard the supernatants and finally pellets were resuspended by vortexing before adding the staining antibodies. Samples were stained with a pool of fluorescence-conjugated antibodies: 5 μl CD3 PerCP (clone SK7), 7 μl IFN-γ FITC (clone 25723.11), 3 μl IL-4 PE (clone 8D4-8), and 4 μl IL-10 APC (clone JES3-19F1) (BD Biosciences, San Jose, CA). After incubating at room temperature in the dark for 30 min, samples were washed twice and supernatants discarded. Samples were brought up to a total volume of 300 μl by adding Cell Wash buffer before acquisition. Stained samples were acquired on a 4-color FACS Calibur (BD Biosciences) and 50,000-100,000 total events were collected. Isotype controls were included to evaluate autofluorescence and to assist in setting the quadrants. The data were analyzed with CELLQuest software (BD Biosciences). Lymphocytes were gated using FSC vs. SSC plots and then SSC vs. CD3+ T cells plots were used to isolate CD3+ cells (Figure 1). We analyzed samples in which > 1,000 CD3+ events could be acquired, and in which defined lymphocyte and CD3+ cell populations could be identified, thus excluding those with compromised cell recovery that could confound the readings. Each intracellular cytokine was assessed within its respective fluorescence channel (PerCP vs. FITC or PE or APC). Non-stimulated control samples were used to establish the threshold quadrants of background responses that were applied to quantify % of positive cytokine-producing cells in paired antigen-stimulated samples; results were expressed as the percentage of cytokine (IL-4, IL-10, IFN-γ)-producing CD3+ cells. Samples were considered positive if there was a true difference in response proportions (95% confidence interval [CI]) between the response to test antigens (stimulated) and the background responses to the non-stimulated samples: if the CI was entirely below 0.05%, the response was negative; if the CI was entirely above 0.05%, the response was positive; if the CI overlapped with 0.05%, the response was indeterminate.

Definitions and statistical methods

Malaria infection was defined as the presence of asexual P. falciparum parasites of any density in a blood smear measured by microscopy and/or by PCR [59]. A clinical malaria episode was defined as a positive blood smear plus an axillary temperature of 37.5°C or history of fever within the prior 24 h. The sensitivity and specificity of these definitions are 100% and 84%, respectively, in infants and 100% and 79.4% in children 1-4 years old [68]. To estimate the incidences, the time at risk was calculated as the number of person years at risk (PYAR; episodes per 365.25 days) since the beginning of the time at risk until the end of follow-up, migration, death or withdrawn consent, whichever occurred first. Children did not contribute to the time at risk or the clinical malaria cases during the lag periods (an arbitrary lag of 21 days was applied after a case of clinical malaria) to account for the effective prophylactic time after antimalarial drug treatment. Months were defined as 30.4 days.
A Shapiro-Wilk normality test revealed a non-normal distribution of data, therefore all analyses were carried out in log scale (Ln) to normalise the data, and averages were expressed as Geometric Means (GM). A Pearson correlation coefficient was used to measure the degree of correlation among cytokines. Correlations were considered "low" if Rho < 0.33, "moderate" if Rho was between 0.33 and 0.66, or "high" if Rho > 0.66. Ratios between cytokines were obtained by dividing the values from each child, transforming that value to its natural log, then calculating the mean for the total number of children, and finally transforming that value to its exponential (GM). T-test and chi-square or fisher exact test were used to compare mean levels or prevalence (tertiles of distribution) respectively, between IPTi treatment groups. Unadjusted and adjusted linear regression models were also used to estimate the effect of IPTi-SP on the cytokine levels at each visit.
To identify what variables were independently associated with cytokine responses (intervention group [SP or placebo], age at visit, neighborhood of residence, current infection and the occurrence of previous clinical episodes [yes/no]), linear regression models with random intercept were estimated, assuming normal distribution of residuals. Selection of the variables for the final model was done through a stepwise procedure, where the criterion for including or excluding a variable was having a p-value ≤ 0.05 or ≥ 0.10 respectively from the Likelihood ratio test.
To evaluate the relationship between each of the individual cytokines at ages 5 and 12 months and the incidence of clinical malaria episodes from 5 to 12 or from 12 to 24 months, respectively, unadjusted and adjusted negative binomial regression models were estimated using a stepwise procedure. In the study of multiple episodes, the probabilities that the criteria to apply Poisson regressions are met are very low, and the phenomenon of over dispersion can occur, resulting in underestimation of CI. The negative binomial regression can estimate part of the variance that Poisson regression cannot identify, and it is exactly the same when there is no over dispersion. Cytokine responses were treated as continuous values, or categorized by tertiles (cut-offs in Additional file 1, Table S1). The incidence rate ratio (IRR) of children with cytokine levels in the highest tertile against those in the lowest tertile was estimated, as well as the IRR per 2-fold increase in the value of cytokines. Co-variates included were: sex, neighborhood of residence, treatment group and previous malaria episodes.
Two additional separate negative binomial regression models were estimated including (i) all the cytokine responses together and (ii) all the cytokine together plus antibody variables assessed in our previous studies (IgG against whole parasite and variant antigens on the surface of infected erythrocytes, IgG [total or isotypes] and IgM against the merozoite antigens AMA-1, EBA-175 and MSP-119) [58, 59] to identify what immunological variables were independently associated with the subsequent incidence of clinical malaria.
Data analysis was performed using Stata 11 for Windows (Stata Corporation, College Station, TX). Statistical significance was defined as a P of < 0.05. Crude P values reported in the manuscript were interpreted for internal coherence and consistency of results and biological plausibility. In addition, P values were adjusted statistically for multiple comparisons by the Bonferroni correction (P values not reported). Most of the associations were no longer statistically significant (P ≥ 0.05) once adjusted for multiple comparisons, except for a few associations which remained significant (indicated in the text and tables).

Results

The clinical and parasitological characteristics of the children included in this study are reported in Additional file 2, Table S2.

Intracellular and plasma cytokine responses in relation to IPTi-SP

The administration of IPTi-SP did not significantly affect the proportion of CD3+ cells producing intracellular IFN-γ, IL-4 or IL-10 in peripheral whole blood samples stimulated ex-vivo with P. falciparum extract antigens, at any of the time points, neither by unadjusted analysis nor by adjusting for previous episodes of clinical malaria (Table 1).
Table 1
Comparison of levels of intracellular (P. falciparum-specific) and plasma cytokines and chemokines between IPTi treatment groups for each cross-sectional visit.
   
Unadjusted geometric mean
Unadjusted linear regression*
Adjusted linear regression*,
   
Placebo
SP
P value
 
P value
 
P value
Cytokines and chemokines
Age (months)
GM
95% CI
GM
95% CI
 
Proportional difference§
 
Proportional difference
 
Intracellular (% producing lymphocytes)
IFN-γ
5
0.17
0.13 - 0.23
0.13
0.10 - 0.18
0.210
0.77
0.207
0.80
0.293
  
9
0.05
0.04 - 0.07
0.05
0.04 - 0.07
0.994
1.00
0.994
1.01
0.965
  
12
0.07
0.05 - 0.11
0.05
0.03 - 0.08
0.348
0.75
0.344
0.76
0.363
  
24
0.05
0.04 - 0.08
0.04
0.03 - 0.06
0.395
0.80
0.391
0.82
0.433
 
IL-4
5
0.96
0.81 - 1.12
0.94
0.80 - 1.10
0.864
0.98
0.863
1.00
0.995
  
9
0.87
0.77 - 0.99
0.82
0.71 - 0.95
0.544
0.94
0.542
0.94
0.546
  
12
0.80
0.64 - 0.99
0.91
0.71 - 1.17
0.427
1.14
0.422
1.13
0.453
  
24
0.86
0.70 - 1.04
0.82
0.65 - 1.04
0.814
0.96
0.812
0.97
0.848
 
IL-10
5
0.37
0.30 - 0.45
0.36
0.31 - 0.43
0.933
0.99
0.933
1.01
0.922
  
9
0.15
0.12 - 0.19
0.18
0.15 - 0.21
0.285
1.16
0.282
1.15
0.294
  
12
0.23
0.17 - 0.31
0.22
0.17 - 0.31
0.886
0.97
0.885
0.96
0.852
  
24
0.32
0.25 - 0.42
0.35
0.27 - 0.45
0.667
1.08
0.664
1.07
0.695
Plasma (pg/ml)
IL-2
5
16.17
12.31 - 21.24
17.25
13.04 - 22.82
0.742
1.07
0.741
1.12
0.569
  
9
12.99
9.48 - 17.79
12.17
9.35 - 15.83
0.752
0.94
0.750
0.93
0.742
  
12
8.56
6.34 - 11.56
7.17
5.22 - 9.86
0.424
0.84
0.422
0.84
0.438
  
24
7.09
5.08 - 9.90
5.36
3.83 - 7.49
0.241
0.76
0.237
0.76
0.254
 
IL-12
5
2.36
1.80 - 3.10
1.87
1.34 - 2.61
0.280
0.79
0.277
0.79
0.284
  
9
2.37
1.74 - 3.22
1.54
1.21 - 1.96
0.030
0.65
0.030
0.66
0.030
  
12
1.43
1.14 - 1.78
1.31
1.08 - 1.58
0.550
0.92
0.548
0.93
0.596
  
24
2.35
1.83 - 3.01
2.22
1.74 - 2.83
0.747
0.95
0.745
0.97
0.844
 
IFN-γ
5
156.50
105.27-232.66
147.8
98.96 - 220.93
0.842
0.94
0.841
0.97
0.928
  
9
145.93
93.78 - 227.08
115.0
75.84 - 174.44
0.437
0.79
0.435
0.79
0.434
  
12
77.61
51.44 - 117.10
55.47
35.17 - 87.47
0.280
0.71
0.278
0.71
0.267
  
24
128.29
85.71 - 192.03
79.89
51.45 - 124.06
0.116
0.62
0.113
0.62
0.106
 
IL-1β
5
35.03
25.11 - 48.87
41.15
28.66 - 59.09
0.516
1.17
0.514
1.16
0.547
  
9
11.61
8.50 - 15.86
11.45
8.46 - 15.50
0.950
0.99
0.950
0.99
0.957
  
12
13.53
9.70 - 18.86
13.98
10.54 - 18.55
0.880
1.03
0.879
1.04
0.869
  
24
20.05
13.99 - 28.73
17.64
12.12 - 25.69
0.625
0.88
0.622
0.86
0.566
 
IL-6
5
99.22
72.21 - 136.34
127.6
97.52 - 166.95
0.231
1.29
0.228
1.27
0.254
  
9
59.24
43.36 - 80.95
56.47
41.93 - 76.05
0.825
0.95
0.824
0.95
0.810
  
12
76.28
54.51 - 106.75
70.96
51.65 - 97.48
0.756
0.93
0.755
0.93
0.772
  
24
82.64
55.45 - 123.14
80.68
54.88 - 118.62
0.931
0.98
0.931
0.97
0.907
 
TNF
5
53.64
38.35 - 75.02
54.50
40.40 - 73.51
0.944
1.02
0.943
1.00
0.991
  
9
31.91
22.10 - 46.06
27.30
19.11 - 39.00
0.546
0.86
0.544
0.85
0.534
  
12
21.72
15.16 - 31.12
20.49
14.29 - 29.37
0.820
0.94
0.819
0.95
0.832
  
24
12.36
7.72 - 19.81
9.86
6.58 - 14.76
0.468
0.80
0.464
0.78
0.430
 
IL-4
5
3.95
2.83 - 5.52
5.64
4.18 - 7.62
0.117
1.43
0.115
1.49
0.078
  
9
2.96
1.99 - 4.39
3.64
2.69 - 4.93
0.403
1.23
0.401
1.23
0.406
  
12
2.49
1.81 - 3.44
2.65
1.93 - 3.64
0.785
1.06
0.783
1.08
0.747
  
24
1.41
0.98 - 2.04
1.44
1.09 - 1.90
0.939
1.02
0.938
1.01
0.972
 
IL-5
5
2.65
2.14 - 3.28
2.66
2.16 - 3.29
0.966
1.01
0.966
1.03
0.833
  
9
2.57
1.97 - 3.35
2.16
1.73 - 2.69
0.310
0.84
0.307
0.84
0.313
  
12
2.05
1.68 - 2.50
1.62
1.32 - 2.00
0.109
0.79
0.106
0.80
0.116
  
24
2.15
1.87 - 2.46
1.97
1.75 - 2.22
0.349
0.92
0.346
0.92
0.351
 
IL-13
5
0.81
0.66 - 0.99
0.78
0.61 - 0.99
0.811
0.96
0.810
0.97
0.858
  
9
0.71
0.54 - 0.92
0.57
0.46 - 0.71
0.212
0.81
0.209
0.81
0.213
  
12
0.56
0.45 - 0.69
0.41
0.32 - 0.52
0.052
0.73
0.050
0.74
0.068
  
24
1.36
1.11 - 1.67
1.36
1.11 - 1.67
0.993
1.00
0.993
1.01
0.930
 
IL-10
5
3.77
3.01 - 4.72
4.20
3.32 - 5.32
0.510
1.11
0.508
1.14
0.431
  
9
3.94
3.00 - 5.16
3.56
2.94 - 4.30
0.540
0.90
0.538
0.89
0.483
  
12
2.74
2.21 - 3.40
2.45
1.99 - 3.01
0.451
0.89
0.449
0.91
0.536
  
24
2.53
2.00 - 3.19
2.40
1.80 - 3.20
0.781
0.95
0.779
1.00
0.997
 
IL-7
5
3.79
3.13 - 4.59
3.37
2.80 - 4.04
0.379
0.89
0.376
0.91
0.465
  
9
3.98
3.13 - 5.05
3.20
2.71 - 3.79
0.144
0.81
0.142
0.81
0.144
  
12
3.01
2.55 - 3.54
2.33
1.96 - 2.79
0.038
0.78
0.037
0.78
0.043
  
24
2.41
2.11 - 2.75
2.29
2.02 - 2.59
0.560
0.95
0.556
0.94
0.519
 
IL-17
5
4.73
3.47 - 6.45
4.77
3.70 - 6.16
0.968
1.01
0.968
0.99
0.950
  
9
3.72
2.86 - 4.84
2.87
2.22 - 3.72
0.167
0.77
0.164
0.78
0.169
  
12
3.01
2.32 - 3.91
2.93
2.29 - 3.76
0.887
0.97
0.886
0.98
0.890
  
24
5.56
4.37 - 7.08
4.28
3.26 - 5.61
0.152
0.77
0.149
0.76
0.134
 
G-CSF
5
17.84
14.28 - 22.29
16.05
12.87 - 20.01
0.502
0.90
0.499
0.91
0.540
  
9
13.85
10.91 - 17.57
12.32
9.98 - 15.19
0.464
0.89
0.462
0.89
0.467
  
12
10.48
8.17 - 13.44
11.11
9.18 - 13.44
0.712
1.06
0.710
1.07
0.647
  
24
27.96
23.58 - 33.16
22.21
19.14 - 25.76
0.044
0.79
0.042
0.79
0.043
 
GM-CSF
5
18.29
11.87 - 28.19
19.95
13.03 - 30.54
0.777
1.09
0.776
1.12
0.712
  
9
18.34
11.82 - 28.46
12.78
8.34 - 19.59
0.243
0.70
0.240
0.70
0.243
  
12
10.62
7.05 - 16.00
8.10
5.37 - 12.24
0.356
0.76
0.353
0.77
0.363
  
24
10.91
7.05 - 16.87
8.77
6.23 - 12.35
0.433
0.80
0.429
0.81
0.437
 
MCP-1
5
43.17
36.47 - 51.10
46.18
38.41 - 55.51
0.594
1.07
0.592
1.08
0.547
  
9
41.14
33.21 - 50.95
41.49
35.24 - 48.85
0.949
1.01
0.949
1.01
0.969
  
12
39.93
33.47 - 47.62
32.71
27.32 - 39.16
0.118
0.82
0.116
0.82
0.122
  
24
56.52
48.17 - 66.32
50.18
41.82 - 60.22
0.330
0.89
0.326
0.89
0.332
 
MIP-1β
5
565.32
474.10 - 674.10
693.5
603.63 - 796.78
0.070
1.23
0.069
1.18
0.131
  
9
402.91
342.05 - 474.60
456.4
387.36 - 537.80
0.287
1.13
0.285
1.13
0.291
  
12
487.27
413.65 - 573.99
583.6
482.50 - 705.98
0.157
1.20
0.155
1.20
0.153
  
24
618.41
515.94 - 741.22
593.8
494.69 - 712.89
0.754
0.96
0.752
0.96
0.770
 
IL-8
5
200.40
148.28 - 270.82
238.2
183.74 - 308.79
0.389
1.19
0.386
1.15
0.489
  
9
126.87
99.27 - 162.15
139.2
108.84 - 178.11
0.597
1.10
0.596
1.10
0.592
  
12
183.34
143.44 - 234.33
228.4
174.16 - 299.56
0.236
1.25
0.234
1.24
0.244
  
24
208.38
150.71 - 288.13
170.5
123.01 - 236.25
0.386
0.82
0.382
0.81
0.345
The three analyses methods are presented and P values correspond to each one of these.
* Linear regression
Adjusted by previous episodes of clinical malaria
T test
§ Proportional difference in cytokine/chemokine concentrations in SP-recipients in relation to placebo-recipients
Also, in general, no significant differences were found in plasma cytokine or chemokine responses between treatment groups. There were three isolated exceptions (IL-12 at 9 months, IL-7 at 12 months, G-CSF at 24 months) in which cases cytokine/chemokine concentrations were consistently higher in children who received placebo (Table 1), but associations were no longer statistically significant once adjusted for multiple comparisons. The complete raw data depicting levels of each cytokine and chemokine studied per age and treatment group is shown in Additional file 3, Figure S1.

Factors associated with individual cytokine responses

Cytokine/chemokine levels varied according to the age of the child. All of them showed a decline from 5 to 9 months of age. Thereafter, some cytokines continued decreasing gradually throughout all measurements (plasma IL-2, TNF, IL-4, IL-10), others reached their highest levels at 24 months (plasma IL-12, IL-13, IL-17, MCP-1, G-CSF), and the remaining varied without any specific pattern (Table 2; Additional file 3, Figure S1).
Table 2
Intracellular and plasma cytokines and chemokines by age, analyzed by multivariate random-effect models estimated by stepwise procedure and adjusted by previous episodes, current infection, IPTi treatment, gender and neighborhood of residence.
Cytokines and chemokines
Age (months)
Proportional difference
95% CI
P value*
Overall P value
Intracellular
IFN-γ
5
1
-
-
< 0.0001
  
9
0.36
0.26; 0.48
< 0.0001
 
  
12
0.41
0.29; 0.57
< 0.0001
 
  
24
0.32
0.23; 0.45
< 0.0001
 
 
IL-10
5
1
-
-
< 0.0001
  
9
0.45
0.37; 0.55
< 0.0001
 
  
12
0.63
0.50; 0.78
< 0.0001
 
  
24
0.92
0.74; 1.15
0.4700
 
Plasma
IL-2
5
1
-
-
< 0.0001
  
9
0.74
0.58; 0.95
0.0193
 
  
12
0.47
0.37; 0.60
< 0.0001
 
  
24
0.38
0.29; 0.51
< 0.0001
 
 
IL-12
5
1
-
-
0.0001
  
9
0.87
0.69; 1.10
0.2428
 
  
12
0.63
0.50; 0.79
0.0001
 
  
24
1.02
0.79; 1.31
0.9070
 
 
IFN-γ
5
1
-
-
< 0.0001
  
9
0.84
0.59; 1.21
0.3510
 
  
12
0.43
0.30; 0.61
< 0.0001
 
  
24
0.69
0.47; 1.03
0.0703
 
 
IL-1β
5
1
-
-
< 0.0001
  
9
0.30
0.22; 0.40
< 0.0001
 
  
12
0.36
0.27; 0.48
< 0.0001
 
  
24
0.51
0.38; 0.70
< 0.0001
 
 
IL-6
5
1
-
-
0.0001
  
9
0.51
0.38; 0.68
< 0.0001
 
  
12
0.65
0.49; 0.87
0.0032
 
  
24
0.75
0.55; 1.03
0.0806
 
 
TNF
5
1
-
-
< 0.0001
  
9
0.54
0.39; 0.73
0.0001
 
  
12
0.39
0.29; 0.53
< 0.0001
 
  
24
0.21
0.15; 0.30
< 0.0001
 
 
IL-4
5
1
-
-
< 0.0001
  
9
0.69
0.53; 0.90
0.0073
 
  
12
0.54
0.41; 0.71
< 0.0001
 
  
24
0.31
0.23; 0.41
< 0.0001
 
 
IL-5
5
1
-
-
0.0001
  
9
0.88
0.75; 1.04
0.1450
 
  
12
0.68
0.58; 0.81
< 0.0001
 
  
24
0.77
0.65; 0.93
0.0056
 
 
IL-13
5
1
-
-
< 0.0001
  
9
0.77
0.64; 0.92
0.0045
 
  
12
0.58
0.49; 0.70
< 0.0001
 
  
24
1.59
1.30; 1.94
< 0.0001
 
 
IL-10
5
1
-
-
< 0.0001
  
9
0.85
0.71; 1.03
0.0941
 
  
12
0.60
0.50; 0.73
< 0.0001
 
  
24
0.53
0.43; 0.66
< 0.0001
 
 
IL-7
5
1
-
-
< 0.0001
  
9
0.99
0.86; 1.15
0.9244
 
  
12
0.74
0.64; 0.86
< 0.0001
 
  
24
0.66
0.56; 0.77
< 0.0001
 
 
IL-17
5
1
-
-
< 0.0001
  
9
0.68
0.54; 0.85
0.0009
 
  
12
0.63
0.50; 0.79
0.0001
 
  
24
1.05
0.82; 1.35
0.6774
 
 
G-CSF
5
1
-
-
< 0.0001
  
9
0.77
0.64; 0.91
0.0032
 
  
12
0.64
0.53; 0.76
< 0.0001
 
  
24
1.50
1.24; 1.81
< 0.0001
 
 
GM-CSF
5
1
-
-
< 0.0001
  
9
0.78
0.56; 1.09
0.1525
 
  
12
0.49
0.35; 0.68
< 0.0001
 
  
24
0.52
0.36; 0.75
0.0004
 
 
MCP-1
5
1
-
-
< 0.0001
  
9
0.92
0.80; 1.06
0.2724
 
  
12
0.80
0.70; 0.93
0.0025
 
  
24
1.22
1.04; 1.42
0.0156
 
 
MIP-1β
5
1
-
-
< 0.0001
  
9
0.68
0.58; 0.80
< 0.0001
 
  
12
0.85
0.73; 1.00
0.0437
 
  
24
0.98
0.82; 1.16
0.7803
 
 
IL-8
5
1
-
-
0.0003
  
9
0.60
0.47; 0.77
0.0001
 
  
12
0.94
0.73; 1.20
0.6171
 
  
24
0.89
0.68; 1.17
0.3991
 
Intracellular IL-4 is not included (null model). The first P value corresponds to the comparison in between ages and the second corresponds to the overall comparison.
* Each age in relation to 5 months
In addition to age, plasma IL-10 levels were also associated with having had previous episodes of clinical malaria (proportional difference = 1.45 [1.14-1.86], P = 0.0028) and having current infection (proportional difference = 1.80 [1.41-2.29], P < 0.0001). IL-12 and IL-13 were also associated with having had previous malaria episodes (proportional difference = 1.35 [1.02-1.77], P = 0.0340; proportional difference = 1.32 [1.04-1.67], P = 0.0234). None of the other factors analyzed in the multivariate analyses (gender, neighborhood of residence, treatment) had any significant association with the cytokines assessed here.

Correlations and ratios between cytokines

We observed no correlation between the magnitude of IL-4, IL-10 or IFN-γ responses measured intracellularly upon antigen-stimulation and plasma concentrations (data not shown). A few correlations were found between plasma cytokines when analyzed against each other (Additional file 4, Figure S2). Concentrations of Th1 (IL-2, IL-12, IFN-γ) and Th2 (IL-4, IL-5, IL-13) cytokines correlated moderately within themselves, and both these two types showed a moderate/high correlation in between them (Th1 vs Th2). Pro-inflammatory (IL-1β, IL-6, TNF, IL-17) and anti-inflammatory/regulatory (IL-10, IL-7) cytokines correlated moderately within themselves, and both these showed a moderate/low correlation between them (pro- vs anti-inflammatory). Chemokines G-CSF and GM-CSF both correlated moderately/highly to Th1 and Th2 type cytokines, whereas MCP-1 correlated with most anti-inflammatory cytokines and MIP-1 β and IL-8 correlated with most pro-inflammatory cytokines.
We examined whether there was a transition from Th2 to Th1 [55] or from anti-inflammatory to pro-inflammatory cytokines with age in early infancy by analyzing the ratios. Overall, Th1 responses were higher than Th2 responses over the 2 years of life (Additional file 3, Figure S1), and the ratio of IFN-γ:IL-4 increased from 5 to 24 months (Table 3). When data was stratified by treatment, IFN-γ:IL-4 ratios were higher for placebo than for SP recipients at all ages (Table 3), particularly at age 24 months coinciding with the highest prevalence of P. falciparum infection (7.7% at 5 months, 13.5% at 9 months, 8% at 12 months and 26.6% at 24 months). Overall, the pro-inflammatory responses were higher than anti-inflammatory responses over the 2 years of life (Additional file 3, Figure S1), and the ratio of TNF:IL-10 decreased from 5 to 24 months (Table 3).
Table 3
Ratios between prototype Th1:Th2 and pro-: anti-inflammatory cytokines.
  
5 months
9 months
12 months
24 months
  
Ratio
95% CI
Ratio
95% CI
Ratio
95% CI
Ratio
95% CI
 
Overall
32
31.73 - 32.27
39
38.73 - 39.27
25
24.69 - 25.31
71
70.75 - 71.25
IFN-γ:IL-4
SP
26
25.64 - 26.36
32
31.62 - 32.38
21
20.61 - 21.39
56
55.62 - 56.38
 
Placebo
40
39.62 - 40.38
49
48.62 - 49.38
31
30.54 - 31.46
91
90.68 - 91.32
 
Overall
14
13.80 - 14.20
8
7.79 - 8.21
8
7.78 - 8.22
4
3.69 - 4.31
TNF:IL-10
SP
13
12.71 - 13.29
8
7.73 - 8.27
8
7.70 - 8.30
4
3.58 - 4.42
 
Placebo
14
13.73 - 14.27
8
7.69 - 8.31
8
7.67 - 8.33
5
4.55 - 5.45

Individual cytokines and incidence of malaria

Elevated cytokine responses, both antigen-specific and non-specific, were not associated with a reduction in the subsequent incidence of clinical malaria in unadjusted or adjusted analyses (Table 4). In some cases there was an association between higher cytokine levels and higher malaria incidence and this was manifested at certain age intervals (5-12 months or 12-24 months). In particular, intracellular IFN-γ and IL-10 production by CD3+ cells was associated with higher incidence of malaria during the first year of age but not in the second year (Table 4). Adjusting for previous episodes did not affect these results. After Bonferroni correction for multiple comparisons, a non significant trend association was still observed (Table 4).
Table 4
Intracellular and plasma cytokines and chemokines and their association with incidence of malaria during the first and second years of age.
  
5 - 12 months
12 - 24 months
  
Tertiles*
Two-fold increment
Tertiles
Two-fold increment
  
Unadjusted
Adjusted
Unadjusted
Adjusted
Unadjusted
Adjusted
Unadjusted
Adjusted
Cytokines and chemokines
IRR
95% CI
P value§
IRR
95% CI
P value
IRR
95% CI
P value
IRR
95% CI
P value
IRR
95% CI
P value
IRR
95% CI
P value
IRR
95% CI
P value
IRR
95% CI
P value
 
IFN-γ
3.24
1.13 - 9.33
0.052
4.62
1.58 - 13.47
0.004
1.21
1.03 - 1.43
0.021
1.26
1.07 - 1.49
0.003
0.95
0.38 - 2.39
0.635
0.68
0.32 - 1.45
0.533
0.97
0.84 - 1.11
0.639
0.96
0.85 - 1.08
0.5226
Intracellular
IL-4
1.49
0.64 - 3.48
0.645
1.31
0.62 - 2.77
0.783
1.19
0.90 - 1.58
0.212
1.17
0.91 - 1.49
0.213
0.53
0.20 - 1.37
0.404
0.78
0.35 - 1.72
0.816
0.81
0.60 - 1.10
0.178
0.95
0.75 - 1.20
0.6635
 
IL-10
3.11
1.12 - 8.67
0.086
3.40
1.30 - 8.89
0.033
1.32
1.04 - 1.67
0.022
1.38
1.11 - 1.71
0.004
0.97
0.38 - 2.47
0.989
1.39
0.68 - 2.82
0.239
0.98
0.80 - 1.20
0.833
0.96
0.82 - 1.12
0.5904
 
IL-2
2.71
0.94 - 7.83
0.131
2.13
0.83 - 5.42
0.219
1.19
0.97 - 1.45
0.083
1.11
0.95 - 1.29
0.200
1.54
0.66 - 3.59
0.423
1.98
1.01 - 3.90
0.055
1.12
0.98 - 1.29
0.090
1.11
1.00 - 1.24
0.051
 
IL-12
1.38
0.56 - 3.41
0.309
1.53
0.71 - 3.30
0.080
1.10
0.91 - 1.34
0.330
1.06
0.90 - 1.25
0.453
1.63
0.63 - 4.21
0.528
1.87
0.91 - 3.82
0.223
1.09
0.88 - 1.35
0.422
1.10
0.92 - 1.32
0.266
 
IFN-γ
0.92
0.37 - 2.32
0.929
0.99
0.44 - 2.21
0.378
1.00
0.89 - 1.13
0.992
0.98
0.89 - 1.08
0.706
1.28
0.56 - 2.93
0.836
3.60
1.79 - 7.23
0.001
1.05
0.95 - 1.16
0.316
1.16
1.07 - 1.26
0.0004
 
IL-1β
1.42
0.43 - 4.73
0.654
1.24
0.41 - 3.71
0.653
1.00
0.84 - 1.19
0.975
1.02
0.87 - 1.20
0.813
1.52
0.63 - 3.68
0.050
2.11
1.11 - 4.01
0.009
1.11
0.94 - 1.31
0.194
1.18
1.04 - 1.33
0.011
 
IL-6
1.03
0.40 - 2.69
0.967
1.05
0.45 - 2.45
0.967
1.00
0.85 - 1.18
0.979
1.02
0.88 - 1.19
0.747
1.52
0.67 - 3.46
0.609
1.86
0.95 - 3.63
0.184
1.10
0.95 - 1.26
0.194
1.16
1.03 - 1.30
0.013
Plasma
TNF
1.53
0.52 - 4.48
0.716
2.08
0.80 - 5.43
0.116
1.13
0.95 - 1.34
0.166
1.12
0.97 - 1.30
0.117
1.00
0.43 - 2.33
0.926
1.37
0.68 - 2.77
0.580
1.05
0.94 - 1.19
0.390
1.06
0.96 - 1.16
0.243
 
IL-4
1.28
0.48 - 3.41
0.506
1.21
0.53 - 2.78
0.516
1.08
0.94 - 1.24
0.253
1.07
0.95 - 1.20
0.271
1.83
0.82 - 4.07
0.168
2.26
1.19 - 4.27
0.041
1.11
0.98 - 1.25
0.093
1.16
1.05 - 1.29
0.004
 
IL-5
1.67
0.58 - 4.80
0.618
1.74
0.69 - 4.37
0.485
1.13
0.87 - 1.48
0.347
1.05
0.84 - 1.32
0.652
1.90
0.77 - 4.69
0.329
2.80
1.43 - 5.49
0.001
1.15
0.89 - 1.49
0.289
1.39
1.13 - 1.70
0.002
 
IL-13
1.35
0.46 - 3.92
0.527
1.70
0.66 - 4.37
0.114
1.22
0.92 - 1.62
0.157
1.19
0.94 - 1.50
0.132
3.33
1.21 - 9.15
0.038
3.26
1.45 - 7.32
0.016
1.42
1.11 - 1.81
0.005
1.32
1.07 - 1.62
0.006
 
IL-10
3.36
1.27 - 8.87
0.035
3.06
1.21 - 7.76
0.025
1.28
1.04 - 1.57
0.016
1.17
0.99 - 1.38
0.059
3.61
1.60 - 8.13
0.009
2.30
1.19 - 4.44
0.045
1.43
1.16 - 1.75
0.0004
1.25
1.06 - 1.47
0.006
 
IL-7
1.77
0.62 - 5.04
0.529
1.41
0.56 - 3.52
0.594
1.26
0.92 - 1.73
0.137
1.10
0.85 - 1.43
0.456
1.56
0.62 - 3.96
0.242
2.16
1.09 - 4.30
0.029
1.20
0.88 - 1.65
0.245
1.34
1.04 - 1.72
0.019
 
IL-17
0.67
0.25 - 1.83
0.622
0.76
0.33 - 1.74
0.551
1.01
0.83 - 1.23
0.914
1.04
0.88 - 1.22
0.681
0.78
0.30 - 2.00
0.055
0.80
0.36 - 1.77
0.278
0.97
0.79 - 1.20
0.785
0.92
0.78 - 1.09
0.342
 
G-CSF
1.83
0.64 - 5.27
0.439
2.42
0.95 - 6.20
0.074
1.25
0.95 - 1.64
0.102
1.21
0.96 - 1.53
0.085
2.41
0.90 - 6.46
0.207
3.10
1.46 - 6.56
0.013
1.25
1.00 - 1.55
0.048
1.35
1.13 - 1.63
0.001
 
GM-CSF
1.19
0.41 - 3.42
0.945
1.19
0.48 - 2.97
0.490
1.05
0.92 - 1.21
0.475
1.04
0.93 - 1.16
0.517
1.63
0.64 - 4.10
0.187
2.41
1.16 - 5.05
0.011
1.11
0.99 - 1.26
0.077
1.13
1.03 - 1.24
0.010
 
MCP-1
3.01
1.18 - 7.63
0.064
3.16
1.37 - 7.25
0.015
1.64
1.21 - 2.23
0.001
1.66
1.29 - 2.15
< 0.001
3.07
1.34 - 7.06
0.008
2.85
1.43 - 5.69
0.007
1.31
1.02 - 1.69
0.038
1.36
1.11 - 1.67
0.003
 
MIP-1β
0.52
0.20 - 1.35
0.113
0.83
0.34 - 2.07
0.341
0.92
0.67 - 1.27
0.620
1.10
0.80 - 1.51
0.571
1.50
0.67 - 3.40
0.396
0.98
0.50 - 1.94
0.329
0.98
0.78 - 1.24
0.859
0.93
0.76 - 1.13
0.430
 
IL-8
0.68
0.27 - 1.71
0.646
0.77
0.34 - 1.73
0.809
0.95
0.79 - 1.14
0.580
0.98
0.82 - 1.16
0.781
0.72
0.32 - 1.62
0.402
0.61
0.30 - 1.22
0.100
0.95
0.80 - 1.12
0.543
0.97
0.84 - 1.11
0.633
For each time interval (5 to 12 months and 12 to 24 months) P values correspond to each one of the analyses methods used.
* Incidence rate ratio in high tertile relative to low tertile
Incidence rate ratio in two-fold increment
Adjusted by treatment group, gender, previous episodes of clinical malaria, current infection, neighborhood
§ Negative binomial regression model using likelihood ratio test. In bold, P values that remained statistically significant (P < 0.05) or border line significant (P < 0.095) after Bonferroni correction for multiple comparisons. Corrected P values (in order from top to bottom, left to right): intracellular IFN-γ, 0.083, 0.052; intracellular IL-10, 0.076; plasma IFN-γ, 0.022, 0.008; plasma IL4, 0.077; IL-5, 0.027, 0.031; IL-13, 0.093; plasma IL-10, 0.008; G-CSF, 0.018; MCP-1, 0.019, 0.002, 0.053.
Associations between individual plasma cytokines and incidence of malaria were observed throughout the two years, but mostly in the second year. High concentrations of IL-10 and MCP-1 in plasma were associated with higher malaria incidence in both time intervals and by all analysis methods (Table 4). High IL-13 was also positively associated with incidence of malaria as analysed by all methods but only during the second year. A few disperse significant associations for plasma IFN-γ, IL-1β, IL-4, IL-5, IL-7, G-CSF and GM-CSF were also observed during the second year, when adjusted the analyses by the variables included in the multivariate model; all were in the same direction of higher concentrations correlating with increased malaria risk (Table 4). After correction for multiple comparisons, some statistically significant associations remained for MCP-1, IFN-γ, IL-10 and IL-5 (Table 4).

Multiple cytokines and incidence of malaria

We did a multiple regression analysis in which we used a stepwise procedure to determine the variables that were independently associated with incidence of malaria. All cytokines were included in the list of variables taken into account for the stepwise procedure. Significant associations were only observed between higher levels of intracellular IFN-γ at 5 months and higher malaria incidence in the follow up interval 5-12 months (IRR 1.28, 95% CI 1.03-1.58, P = 0.0234). Similarly, higher concentrations of plasma IFN-γ (IRR 1.15, 95% CI 1.05-1.27, P = 0.0040) and MCP-1 (IRR 1.60, 95% CI 1.22-2.10, P = 0.0007) at 12 months were associated with higher malaria risk during the 12-24 month period. In contrast, in this analysis higher plasma concentrations of IL-17 at 12 months were associated with lower malaria incidence during the interval 12-24 months (IRR 0.80, 95% CI 0.69-0.93, P = 0.0035). Other co-variates that were significantly associated with malaria incidence, independently of the cytokine concentrations, were having had a previous episode in the second year of life (IRR 4.63, 95% CI 2.59-8.26, P < 0.0001), and the child's neighborhood of residence in both time intervals (P < 0.0001).
When antibody variables from prior studies were also included in the stepwise model together with all cytokines and co-variates, significant associations were only observed for higher intracellular IFN-γ at 5 months and higher malaria incidence during the follow up interval 5-12 months (IRR 1.26, 95% CI 1.01-1.57, P = 0.0381). At 12 months, higher plasma IFN-γ (IRR 1.26, 95% CI 1.11-1.44, P = 0.0004) and IL-5 (IRR 1.27, 95% CI 1.04-1.55, P = 0.0203) correlated with higher incidence of malaria up to 24 months, while higher IL-17 plasma concentrations were associated with lower malaria risk during the 12-24 month period (IRR 0.84, 95% CI 0.71-0.98, P = 0.0300). Likewise, the additional variables that independently showed significant associations with malaria risk were having had a previous episode in the second year of life (IRR 3.16, 95% CI 1.64-6.09, P < 0.0006), and the neighborhood in both time intervals (P < 0.0001).
Finally, no significant differences in cytokine or chemokine levels were found in relation to parasite density (data not shown). Levels of cytokines or chemokines could not be analysed in relation to incidence of severe malaria due to its low incidence (Additional file 2, Table S2).

Discussion

In the context of a clinical trial evaluating the safety and efficacy of IPTi-SP as a malaria control tool during infancy, we studied prospectively the cytokine responses to P. falciparum antigens and the concentrations of plasma cytokines and chemokines at multiple time points during a 2 year follow up period. The longitudinal design and the established demographic and morbidity surveillance systems allowed for a rigorous and complete documentation of clinical malaria cases that could provide insights into the acquisition of cytokine/chemokine responses in children living in a malaria endemic area, how they are influenced by age and P. falciparum exposure, and the correlation between these responses and incidence of symptomatic malaria.
One of the main findings of this study was that overall there were no differences in the magnitude of intracellular or plasma cytokines/chemokines between IPTi treatment groups, except for some plasma cytokines and chemokines in which children receiving placebo had higher concentrations than those receiving SP (IL-12 at 9 months, IL-7 at 12 months and G-CSF at 24 months). In these cases, we hypotesize that placebo recipients, who suffered more P. falciparum infections than SP recipients owing to the efficacy of IPTi [57] had more systemic immune activation and higher levels of some plasma cytokines. However, of these, only IL-12 was directly associated to previous episodes of malaria in the multivariate analysis; to our knowledge, there is no data in the literature on the influence of P. falciparum infection or malaria interventions in the concentrations of IL-7 or G-CSF in peripheral blood. The levels of these cytokines/chemokines at those time points were not clearly associated with subsequent incidence of malaria.
Among the factors that significantly affected the levels of cytokines (both intracellular and plasma), age was the most prominent. Without exception, all cytokines varied significantly in magnitude from the first visit to at least one of the following visits. In all cases, there was a decrease of cytokine values from 5 to 9 months, and some had the highest levels at 24 months. This fluctuation is not surprising as cross-sectional samplings were not scheduled to pick up bursts of production upon infection and most plasma cytokines' half-lives are short [34], thus may not represent a long lasting response. In addition, cytokine production is stimulated and regulated by many different factors within the cellular responses network, and while some are specific to infection, other are innate or are produced in response to acute or systemic inflammatory conditions.
In contrast to antibody responses that are strongly affected by past or present malaria infection [59], parasite exposure appeared to have a minor effect on the concentrations of cytokines. Plasma IL-10 increased significantly in children with previous episodes or current infection, and higher levels of IL-12 and IL-13 were also associated with previous episodes. Each of these cytokines belongs to a different family (immuno-regulatory, Th1 and Th2 respectively) therefore it seems that the limited effect of exposure to P. falciparum infection was generalized and not biased towards a specific type. Similar observations have been reported [9] where malaria patients responded to P. falciparum infected erythrocytes with significant increases in the percentage of IL-2, IFN-γ, and TNF, but also IL-10, positive cells.
We analyzed the correlations and ratios among some representative cytokine families, as they have synergistic and antagonistic effects on each other to regulate the immune system. First, we observed no correlation between intracellular and plasma IL-4, IL-10 or IFN-γ, which is not uncommon as they represent two different measurements. Intracellular cytokines were produced by CD3+ cells after antigen stimulation in vitro (recall response) whereas plasma cytokines reflect what is present ex-vivo in peripheral blood (non malaria specific), and may be produced by many types of cells. In another study [66], the strength of associations between serum and cellular cytokines varied greatly, suggesting that serum cytokines at best only weakly reflect peripheral blood cell cytokine production and balances. We observed a high ratio between prototype Th1 (IFN-γ) and Th2 (IL-4) cytokines at all ages, and this proportion increased when looking only at the placebo group possibly indicating more parasite exposure. We also observed the relative transition from Th2 to Th1 responses with age [69].
We also assessed whether cytokines and chemokines were associated with subsequent incidence of malaria, and what factors affected these associations. Overall, high cytokine responses were not associated with a reduced incidence of clinical malaria when analyzed individually. Only when all cytokines where analyzed together in relation to risk of malaria, a significant association towards a decreased risk was found for IL-17 in the second year of life. IL-17 is a Th17 cytokine and its relation to various infectious agents has been described [70]. However to our knowledge, there has not been any study reporting a role for IL- 17 in protection against human malaria.
In fact, the most common finding was that there were no associations, or that higher levels of some cytokines correlated with increased malaria incidence. In particular, IL-10 (intracellular and plasma) and MCP-1 were more consistently associated with incidence of malaria, and this association was not explained by age or previous episodes. IL-10, an immunoregulatory cytokine, is extensively reported in relation to malaria immunopathogenesis [9, 12, 20, 26, 27] and not so much associated with immunity [38]. Higher IL-10 has been associated with less effective parasite clearance [71]. To our knowledge nothing has been reported for chemokine MCP-1 in children in relation to immunity, and it does not correlate with any of the other cytokines associated with increased incidence of malaria. In pregnant women, MCP-1 concentrations were higher in the placentas of primiparous women (more susceptible to malaria) than in those of multiparous women [46].
In addition, IFN-γ (intracellular and plasma) and IL-13 were also associated with malaria incidence, but these were dependent on age and/or previous episodes. A few other disperse associations were observed during the second year for plasma IL-1β, IL-4, IL-5, IL-7, G-CSF, GM-CSF, when adjusted by previous episodes. These results suggest that the responses measured were not necessarily part of an acquired protective response, but rather could be interpreted as biomarkers of physiopathological processes. In other studies, IL-13, IL-4 and IL-5, Th2-type cytokines, have been associated with reduced parasite killing [24]. Production of IL- 4 but not IFN-γ by activated human T cells has been associated with elevated antibodies to malaria antigens [72], and consistent with this, our previous studies also found correlations between higher antibody levels and increased malaria risk (Dobaño et al. in press).
Responses associated with reduced malaria incidence would more likely be expected in the antigen-specific cytokines, as measured by ICS. Blood cells from donors in malaria endemic areas stimulated with Plasmodium antigens are known to activate many types of cells, with production of both IFN-γ (Th1 type) and IL-4 (Th2 type) [72]. IFN-γ is thought to be a central mediator of protective immune responses against blood stages of malaria [73]. Surprisingly, in our study, increased frequency of IFN-γ positive CD3+ cells at 5 months was associated with higher incidence of malaria up to 1 year. This is in contrast with other studies since IFN-γ is more often reported to confer protection [30, 3236] than pathology [9, 12, 74]. In most of these studies IFN-γ was measured in older children or adults and it is likely that the immature immune system of the infant responds differently. In the second year, however, there were no significant associations between intracellular cytokines and incidence of malaria. At a population level, memory-like IFN-γ responses have been measured following malaria infection [75] and it has been reported that IFN-γ responses are both more prevalent and of greater magnitude at the end of the rainy malaria transmission season [45], thus showing that IFN-γ might also be a marker of exposure.
The apparent predisposition of children with higher cytokine responses to increased malaria risk might also be influenced by genetic factors. Many studies have shown associations between genetic polymorphisms, immunoregulation, phenotypes and disease risk [7691], but unfortunately this was not assessed in this study. Other factors not evaluated that could help explain these associations would be malnutrition [92, 93], co-infection with HIV or other pathogens [94], or prenatal exposure [95]. The duration and/or the nature of antigen exposure in utero appears to govern the outcome with respect to neonatal immune responses, such that placental malaria induce antigen-specific IL-10-producing regulatory T cells that can inhibit Th1-type responses, while antigen-specific IFN-γ production predominate in babies born to mothers successfully treated for malaria during gestation [95]. Prenatal infection could thus contribute to the P. falciparum-specific IFN-γ and IL-10 response pattern in 5-month old children seen in this study.

Conclusions

Our work has further confirmed that IPTi-SP is a safe intervention that does not compromise the development of a wide range of cytokines and chemokines thought to be major contributors to anti-malarial immunity in infancy. This study has also given some insights as to how these responses are acquired upon age and exposure to the P. falciparum parasite. Due to the multiple comparisons performed, isolated findings are interpreted with caution, and only those associations that showed consistency and biological plausibility are considered here as potential true findings. Despite the low and heterogeneous antigen-specific cytokine responses observed, infant field studies such as the current one help to advance the understanding of the relationship between innate and adaptive cellular immune responses. Eventually, an effective orchestration of both types of immune responses is necessary for the generation of an efficient and non-pathogenic resolution of the malarial disease.

Acknowledgements

We thank the volunteer children and their mothers and guardians for their participation in the study. We are very grateful to the following investigators from the Naval Medical Research Center, USA, for invaluable assistance in the cellular assays: G.T. Brice and M. Stefaniak for the development of the ICS protocols and flow cytometry data analysis, and C. Moch, D. Haynes and N. Martinez-Alier, for the preparation and shipment of the P. falciparum schizont lysate. We thank D. Naniche and C. Romagosa for assistance in flow cytometry setup in Manhiça, L. Quimice, N. José and A.R. Manhiça for technical support in sample processing, J. Campo for analytical assistance, and G. Moncunill for critically reviewing the manuscript.
This work was funded by the Bill and Melinda Gates Foundation (grant IPTi03-0), and the Banco de Bilbao-Vizcaya-Argentaria Foundation (grant BBVA 02-0). DQ was supported by WHO/TDR (grant OD/TS-07-00017), CD is supported by the Ministerio de Ciencia e Innovación (RYC-2008-02631), and DLD is supported by a Pfizer Australia Senior Research Fellowship. The Centro de Investigaçao em Saúde da Manhiça receives core support from the Spanish Agency for International Cooperation and Development (AECID).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Authors' contributions

PLA, CM, EM and PA carried out the IPTi clinical trial. PLA, CM and CD conceived, designed and coordinated the study. DQ, TN, ES and CD participated in the sample collection and processing. CD and DLD designed and supervised the immunoassays. DQ and LP carried out the immunoassays. LQ, SS and JJA performed the statistical analyses. DQ and CD drafted the first version of the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
4.
Zurück zum Zitat Cohen S, McGregor IA, Carrington S: Gamma-globulin and acquired immunity to human malaria. Nature. 1961, 192: 733-737. 10.1038/192733a0.PubMedCrossRef Cohen S, McGregor IA, Carrington S: Gamma-globulin and acquired immunity to human malaria. Nature. 1961, 192: 733-737. 10.1038/192733a0.PubMedCrossRef
5.
Zurück zum Zitat Pombo DJ, Lawrence G, Hirunpetcharat C, Rzepczyk C, Bryden M, Cloonan N, Anderson K, Mahakunkijcharoen Y, Martin LB, Wilson D, Elliott S, Eisen DP, Weinberg JB, Saul A, Good MF: Immunity to malaria after administration of ultra-low doses of red cells infected with Plasmodium falciparum. Lancet. 2002, 360: 610-617. 10.1016/S0140-6736(02)09784-2.PubMedCrossRef Pombo DJ, Lawrence G, Hirunpetcharat C, Rzepczyk C, Bryden M, Cloonan N, Anderson K, Mahakunkijcharoen Y, Martin LB, Wilson D, Elliott S, Eisen DP, Weinberg JB, Saul A, Good MF: Immunity to malaria after administration of ultra-low doses of red cells infected with Plasmodium falciparum. Lancet. 2002, 360: 610-617. 10.1016/S0140-6736(02)09784-2.PubMedCrossRef
6.
Zurück zum Zitat Good MF, Xu H, Wykes M, Engwerda CR: Development and regulation of cell-mediated immune responses to the blood stages of malaria: implications for vaccine research. Annual Review of Immunology. 2005, 23: 69-99. 10.1146/annurev.immunol.23.021704.115638.PubMedCrossRef Good MF, Xu H, Wykes M, Engwerda CR: Development and regulation of cell-mediated immune responses to the blood stages of malaria: implications for vaccine research. Annual Review of Immunology. 2005, 23: 69-99. 10.1146/annurev.immunol.23.021704.115638.PubMedCrossRef
7.
Zurück zum Zitat Fritsche G, Larcher C, Schennach H, Weiss G: Regulatory interactions between iron and nitric oxide metabolism for immune defense against Plasmodium falciparum infection. The Journal of Infectious Diseases. 2001, 183: 1388-94. 10.1086/319860.PubMedCrossRef Fritsche G, Larcher C, Schennach H, Weiss G: Regulatory interactions between iron and nitric oxide metabolism for immune defense against Plasmodium falciparum infection. The Journal of Infectious Diseases. 2001, 183: 1388-94. 10.1086/319860.PubMedCrossRef
8.
Zurück zum Zitat Tsuji M, Rodrigues EG, Nussenzweig S: Progress toward a malaria vaccine: efficient induction of protective anti-malaria immunity. Biological Chemistry. 2001, 382: 553-70. 10.1515/BC.2001.069.PubMedCrossRef Tsuji M, Rodrigues EG, Nussenzweig S: Progress toward a malaria vaccine: efficient induction of protective anti-malaria immunity. Biological Chemistry. 2001, 382: 553-70. 10.1515/BC.2001.069.PubMedCrossRef
9.
Zurück zum Zitat Ramharter M, Willheim M, Winkler H, Wahl K, Lagler H, Graninger W, Winkler S: Cytokine profile of Plasmodium falciparum-specific T cells in non-immune malaria patients. Parasite Immunology. 2003, 25: 211-219. 10.1046/j.1365-3024.2003.00628.x.PubMedCrossRef Ramharter M, Willheim M, Winkler H, Wahl K, Lagler H, Graninger W, Winkler S: Cytokine profile of Plasmodium falciparum-specific T cells in non-immune malaria patients. Parasite Immunology. 2003, 25: 211-219. 10.1046/j.1365-3024.2003.00628.x.PubMedCrossRef
10.
Zurück zum Zitat Luty AJ, Perkins DJ, Lell B, Schmidt-Ott R, Lehman LG, Luckner D, Greve B, Matousek P, Herbich K, Schmid D, Weinberg JB, Kremsner PG: Low interleukin-12 activity in severe Plasmodium falciparum malaria. Infection and Immunity. 2000, 68: 3909-15. 10.1128/IAI.68.7.3909-3915.2000.PubMedPubMedCentralCrossRef Luty AJ, Perkins DJ, Lell B, Schmidt-Ott R, Lehman LG, Luckner D, Greve B, Matousek P, Herbich K, Schmid D, Weinberg JB, Kremsner PG: Low interleukin-12 activity in severe Plasmodium falciparum malaria. Infection and Immunity. 2000, 68: 3909-15. 10.1128/IAI.68.7.3909-3915.2000.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Perkins DJ, Weinberg JB, Kremsner PG: Reduced interleukin-12 and transforming growth factor-beta1 in severe childhood malaria: relationship of cytokine balance with disease severity. The Journal of Infectious Diseases. 2000, 182: 988-992. 10.1086/315762.PubMedCrossRef Perkins DJ, Weinberg JB, Kremsner PG: Reduced interleukin-12 and transforming growth factor-beta1 in severe childhood malaria: relationship of cytokine balance with disease severity. The Journal of Infectious Diseases. 2000, 182: 988-992. 10.1086/315762.PubMedCrossRef
12.
Zurück zum Zitat Day NP, Hien TT, Schollaardt T, Loc PP, Chuong LV, Chau TT, Mai NT, Phu NH, Sinh DX, White NJ, Ho M: The prognostic and pathophysiologic role of pro- and antiinflammatory cytokines in severe malaria. The Journal of Infectious Diseases. 1999, 180: 1288-1297. 10.1086/315016.PubMedCrossRef Day NP, Hien TT, Schollaardt T, Loc PP, Chuong LV, Chau TT, Mai NT, Phu NH, Sinh DX, White NJ, Ho M: The prognostic and pathophysiologic role of pro- and antiinflammatory cytokines in severe malaria. The Journal of Infectious Diseases. 1999, 180: 1288-1297. 10.1086/315016.PubMedCrossRef
13.
Zurück zum Zitat Brown H, Turner G, Rogerson S, Tembo M, Mwenechanya J, Molyneux M, Taylor T: Cytokine expression in the brain in human cerebral malaria. The Journal of Infectious Diseases. 1999, 180: 1742-6. 10.1086/315078.PubMedCrossRef Brown H, Turner G, Rogerson S, Tembo M, Mwenechanya J, Molyneux M, Taylor T: Cytokine expression in the brain in human cerebral malaria. The Journal of Infectious Diseases. 1999, 180: 1742-6. 10.1086/315078.PubMedCrossRef
14.
Zurück zum Zitat Kern P, Hemmer CJ, Van Damme J, Gruss HJ, Dietrich M: Elevated tumor necrosis factor alpha and interleukin-6 serum levels as markers for complicated Plasmodium falciparum malaria. The American Journal of Medicine. 1989, 87: 139-43. 10.1016/S0002-9343(89)80688-6.PubMedCrossRef Kern P, Hemmer CJ, Van Damme J, Gruss HJ, Dietrich M: Elevated tumor necrosis factor alpha and interleukin-6 serum levels as markers for complicated Plasmodium falciparum malaria. The American Journal of Medicine. 1989, 87: 139-43. 10.1016/S0002-9343(89)80688-6.PubMedCrossRef
15.
Zurück zum Zitat Molyneux ME, Taylor TE, Wirima JJ, Grau GE: Tumour necrosis factor, interleukin-6, and malaria. Lancet. 1991, 337: 1098-10.1016/0140-6736(91)91745-G.PubMedCrossRef Molyneux ME, Taylor TE, Wirima JJ, Grau GE: Tumour necrosis factor, interleukin-6, and malaria. Lancet. 1991, 337: 1098-10.1016/0140-6736(91)91745-G.PubMedCrossRef
16.
Zurück zum Zitat Grau GE, Piguet PF, Vassalli P, Lambert PH: Tumor-necrosis factor and other cytokines in cerebral malaria: experimental and clinical data. Immunological Reviews. 1989, 112: 49-70. 10.1111/j.1600-065X.1989.tb00552.x.PubMedCrossRef Grau GE, Piguet PF, Vassalli P, Lambert PH: Tumor-necrosis factor and other cytokines in cerebral malaria: experimental and clinical data. Immunological Reviews. 1989, 112: 49-70. 10.1111/j.1600-065X.1989.tb00552.x.PubMedCrossRef
17.
Zurück zum Zitat Kwiatkowski D, Cannon JG, Manogue KR, Cerami A, Dinarello CA, Greenwood BM: Tumour necrosis factor production in Falciparum malaria and its association with schizont rupture. Clinical and Experimental Immunology. 1989, 77: 361-6.PubMedPubMedCentral Kwiatkowski D, Cannon JG, Manogue KR, Cerami A, Dinarello CA, Greenwood BM: Tumour necrosis factor production in Falciparum malaria and its association with schizont rupture. Clinical and Experimental Immunology. 1989, 77: 361-6.PubMedPubMedCentral
18.
Zurück zum Zitat Kwiatkowski D, Hill AV, Sambou I, Twumasi P, Castracane J, Manogue KR, Cerami A, Brewster DR, Greenwood BM: TNF concentration in fatal cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria. Lancet. 1990, 336: 1201-4. 10.1016/0140-6736(90)92827-5.PubMedCrossRef Kwiatkowski D, Hill AV, Sambou I, Twumasi P, Castracane J, Manogue KR, Cerami A, Brewster DR, Greenwood BM: TNF concentration in fatal cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria. Lancet. 1990, 336: 1201-4. 10.1016/0140-6736(90)92827-5.PubMedCrossRef
19.
Zurück zum Zitat Körner H, McMorran B, Schlüter D, Fromm P: The role of TNF in parasitic diseases: Still more questions than answers. International Journal for Parasitology. 2010, 40: 879-88. 10.1016/j.ijpara.2010.03.011.PubMedCrossRef Körner H, McMorran B, Schlüter D, Fromm P: The role of TNF in parasitic diseases: Still more questions than answers. International Journal for Parasitology. 2010, 40: 879-88. 10.1016/j.ijpara.2010.03.011.PubMedCrossRef
20.
Zurück zum Zitat Othoro C, Lal AA, Nahlen B, Koech D, Orago AS, Udhayakumar V: A low interleukin-10 tumor necrosis factor-alpha ratio is associated with malaria anemia in children residing in a holoendemic malaria region in western Kenya. The Journal of Infectious Diseases. 1999, 179: 279-282. 10.1086/314548.PubMedCrossRef Othoro C, Lal AA, Nahlen B, Koech D, Orago AS, Udhayakumar V: A low interleukin-10 tumor necrosis factor-alpha ratio is associated with malaria anemia in children residing in a holoendemic malaria region in western Kenya. The Journal of Infectious Diseases. 1999, 179: 279-282. 10.1086/314548.PubMedCrossRef
21.
Zurück zum Zitat Scuderi P, Sterling KE, Lam KS, Finley PR, Ryan KJ, Ray CG, Petersen E, Slymen DJ, Salmon SE: Raised serum levels of tumour necrosis factor in parasitic infections. Lancet. 1986, 2: 1364-5.PubMedCrossRef Scuderi P, Sterling KE, Lam KS, Finley PR, Ryan KJ, Ray CG, Petersen E, Slymen DJ, Salmon SE: Raised serum levels of tumour necrosis factor in parasitic infections. Lancet. 1986, 2: 1364-5.PubMedCrossRef
22.
Zurück zum Zitat Shaffer N, Grau GE, Hedberg K, Davachi F, Lyamba B, Hightower AW, Breman JG, Phuc ND: Tumor necrosis factor and severe malaria. The Journal of Infectious Diseases. 1991, 163: 96-101. 10.1093/infdis/163.1.96.PubMedCrossRef Shaffer N, Grau GE, Hedberg K, Davachi F, Lyamba B, Hightower AW, Breman JG, Phuc ND: Tumor necrosis factor and severe malaria. The Journal of Infectious Diseases. 1991, 163: 96-101. 10.1093/infdis/163.1.96.PubMedCrossRef
23.
Zurück zum Zitat Eisenhut M: The role of interleukin-4 in the immune response to Plasmodium falciparum. Parasite Immunology. 2010, 32: 470-471. 10.1111/j.1365-3024.2010.01214.x.PubMedCrossRef Eisenhut M: The role of interleukin-4 in the immune response to Plasmodium falciparum. Parasite Immunology. 2010, 32: 470-471. 10.1111/j.1365-3024.2010.01214.x.PubMedCrossRef
24.
Zurück zum Zitat Kumaratilake LM, Ferrante A: IL-4 inhibits macrophage-mediated killing of Plasmodium falciparum in vitro. A possible parasite-immune evasion mechanism. Journal of Immunology. 1992, 149: 194-199. Kumaratilake LM, Ferrante A: IL-4 inhibits macrophage-mediated killing of Plasmodium falciparum in vitro. A possible parasite-immune evasion mechanism. Journal of Immunology. 1992, 149: 194-199.
25.
Zurück zum Zitat Troye-Blomberg M: Human T-cell responses to blood stage antigens in Plasmodium falciparum malaria. Immunology Letters. 1994, 41: 103-107. 10.1016/0165-2478(94)90116-3.PubMedCrossRef Troye-Blomberg M: Human T-cell responses to blood stage antigens in Plasmodium falciparum malaria. Immunology Letters. 1994, 41: 103-107. 10.1016/0165-2478(94)90116-3.PubMedCrossRef
26.
Zurück zum Zitat Kurtzhals JA, Adabayeri V, Goka BQ, Akanmori BD, Oliver-Commey JO, Nkrumah FK, Behr C, Hviid L: Low plasma concentrations of interleukin 10 in severe malarial anaemia compared with cerebral and uncomplicated malaria. Lancet. 1998, 351: 1768-1772. 10.1016/S0140-6736(97)09439-7.PubMedCrossRef Kurtzhals JA, Adabayeri V, Goka BQ, Akanmori BD, Oliver-Commey JO, Nkrumah FK, Behr C, Hviid L: Low plasma concentrations of interleukin 10 in severe malarial anaemia compared with cerebral and uncomplicated malaria. Lancet. 1998, 351: 1768-1772. 10.1016/S0140-6736(97)09439-7.PubMedCrossRef
27.
Zurück zum Zitat Winkler S, Willheim M, Baier K, Schmid D, Aichelburg A, Graninger W, Kremsner PG: Reciprocal regulation of Th1- and Th2-cytokine-producing T cells during clearance of parasitemia in Plasmodium falciparum malaria. Infection and Immunity. 1998, 66: 6040-4.PubMedPubMedCentral Winkler S, Willheim M, Baier K, Schmid D, Aichelburg A, Graninger W, Kremsner PG: Reciprocal regulation of Th1- and Th2-cytokine-producing T cells during clearance of parasitemia in Plasmodium falciparum malaria. Infection and Immunity. 1998, 66: 6040-4.PubMedPubMedCentral
28.
Zurück zum Zitat Ochiel DO, Awandare GA, Keller CC, Hittner JB, Kremsner PG, Weinberg JB, Perkins DJ: Differential regulation of beta-chemokines in children with Plasmodium falciparum malaria. Infection and Immunity. 2005, 73: 4190-7. 10.1128/IAI.73.7.4190-4197.2005.PubMedPubMedCentralCrossRef Ochiel DO, Awandare GA, Keller CC, Hittner JB, Kremsner PG, Weinberg JB, Perkins DJ: Differential regulation of beta-chemokines in children with Plasmodium falciparum malaria. Infection and Immunity. 2005, 73: 4190-7. 10.1128/IAI.73.7.4190-4197.2005.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Perkins DJ, Kremsner PG, Weinberg JB: Inverse relationship of plasma prostaglandin E2 and blood mononuclear cell cyclooxygenase-2 with disease severity in children with Plasmodium falciparum malaria. The Journal of Infectious Diseases. 2001, 183: 113-118. 10.1086/317660.PubMedCrossRef Perkins DJ, Kremsner PG, Weinberg JB: Inverse relationship of plasma prostaglandin E2 and blood mononuclear cell cyclooxygenase-2 with disease severity in children with Plasmodium falciparum malaria. The Journal of Infectious Diseases. 2001, 183: 113-118. 10.1086/317660.PubMedCrossRef
30.
Zurück zum Zitat Dodoo D, Omer FM, Todd J, Akanmori BD, Koram KA, Riley EM: Absolute levels and ratios of proinflammatory and anti-inflammatory cytokine production in vitro predict clinical immunity to Plasmodium falciparum malaria. The Journal of Infectious Diseases. 2002, 185: 971-979. 10.1086/339408.PubMedCrossRef Dodoo D, Omer FM, Todd J, Akanmori BD, Koram KA, Riley EM: Absolute levels and ratios of proinflammatory and anti-inflammatory cytokine production in vitro predict clinical immunity to Plasmodium falciparum malaria. The Journal of Infectious Diseases. 2002, 185: 971-979. 10.1086/339408.PubMedCrossRef
31.
Zurück zum Zitat Sedegah M, Finkelman F, Hoffman SL: Interleukin 12 induction of interferon gamma-dependent protection against malaria. Proceedings of the National Academy of Sciences of the United States of America. 1994, 91: 10700-2. 10.1073/pnas.91.22.10700.PubMedPubMedCentralCrossRef Sedegah M, Finkelman F, Hoffman SL: Interleukin 12 induction of interferon gamma-dependent protection against malaria. Proceedings of the National Academy of Sciences of the United States of America. 1994, 91: 10700-2. 10.1073/pnas.91.22.10700.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Torre D, Speranza F, Giola M, Matteelli A, Tambini R, Biondi G: Role of Th1 and Th2 cytokines in immune response to uncomplicated Plasmodium falciparum malaria. Clinical and Diagnostic Laboratory Immunology. 2002, 9: 348-51.PubMedPubMedCentral Torre D, Speranza F, Giola M, Matteelli A, Tambini R, Biondi G: Role of Th1 and Th2 cytokines in immune response to uncomplicated Plasmodium falciparum malaria. Clinical and Diagnostic Laboratory Immunology. 2002, 9: 348-51.PubMedPubMedCentral
33.
Zurück zum Zitat Chizzolini C, Grau GE, Geinoz A, Schrijvers D: T lymphocyte interferon-gamma production induced by Plasmodium falciparum antigen is high in recently infected non-immune and low in immune subjects. Clinical and Experimental Immunology. 1990, 79: 95-9.PubMedPubMedCentralCrossRef Chizzolini C, Grau GE, Geinoz A, Schrijvers D: T lymphocyte interferon-gamma production induced by Plasmodium falciparum antigen is high in recently infected non-immune and low in immune subjects. Clinical and Experimental Immunology. 1990, 79: 95-9.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Harpaz R, Edelman R, Wasserman SS, Levine MM, Davis JR, Sztein MB: Serum cytokine profiles in experimental human malaria. Relationship to protection and disease course after challenge. The Journal of Clinical Investigation. 1992, 90: 515-23. 10.1172/JCI115889.PubMedPubMedCentralCrossRef Harpaz R, Edelman R, Wasserman SS, Levine MM, Davis JR, Sztein MB: Serum cytokine profiles in experimental human malaria. Relationship to protection and disease course after challenge. The Journal of Clinical Investigation. 1992, 90: 515-23. 10.1172/JCI115889.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Luty AJ, Lell B, Schmidt-Ott R, Lehman LG, Luckner D, Greve B, Matousek P, Herbich K, Schmid D, Migot-Nabias F, Deloron P, Nussenzweig RS, Kremsner PG: Interferon-gamma responses are associated with resistance to reinfection with Plasmodium falciparum in young African children. The Journal of Infectious Diseases. 1999, 179: 980-8. 10.1086/314689.PubMedCrossRef Luty AJ, Lell B, Schmidt-Ott R, Lehman LG, Luckner D, Greve B, Matousek P, Herbich K, Schmid D, Migot-Nabias F, Deloron P, Nussenzweig RS, Kremsner PG: Interferon-gamma responses are associated with resistance to reinfection with Plasmodium falciparum in young African children. The Journal of Infectious Diseases. 1999, 179: 980-8. 10.1086/314689.PubMedCrossRef
36.
Zurück zum Zitat Riley EM, Allen SJ, Wheeler JG, Blackman MJ, Bennett S, Takacs B, Schönfeld HJ, Holder AA, Greenwood BM: Naturally acquired cellular and humoral immune responses to the major merozoite surface antigen (PfMSP1) of Plasmodium falciparum are associated with reduced malaria morbidity. Parasite Immunology. 1992, 14: 321-37. 10.1111/j.1365-3024.1992.tb00471.x.PubMedCrossRef Riley EM, Allen SJ, Wheeler JG, Blackman MJ, Bennett S, Takacs B, Schönfeld HJ, Holder AA, Greenwood BM: Naturally acquired cellular and humoral immune responses to the major merozoite surface antigen (PfMSP1) of Plasmodium falciparum are associated with reduced malaria morbidity. Parasite Immunology. 1992, 14: 321-37. 10.1111/j.1365-3024.1992.tb00471.x.PubMedCrossRef
37.
Zurück zum Zitat Bouharoun-Tayoun H, Oeuvray C, Lunel F, Druilhe P: Mechanisms underlying the monocyte-mediated antibody-dependent killing of Plasmodium falciparum asexual blood stages. The Journal of Experimental Medicine. 1995, 182: 409-18. 10.1084/jem.182.2.409.PubMedCrossRef Bouharoun-Tayoun H, Oeuvray C, Lunel F, Druilhe P: Mechanisms underlying the monocyte-mediated antibody-dependent killing of Plasmodium falciparum asexual blood stages. The Journal of Experimental Medicine. 1995, 182: 409-18. 10.1084/jem.182.2.409.PubMedCrossRef
38.
Zurück zum Zitat John CC, Sumba PO, Ouma JH, Nahlen BL, King CL, Kazura JW: Cytokine responses to Plasmodium falciparum liver-stage antigen 1 vary in rainy and dry seasons in highland Kenya. Infection and Immunity. 2000, 68: 5198-204. 10.1128/IAI.68.9.5198-5204.2000.PubMedPubMedCentralCrossRef John CC, Sumba PO, Ouma JH, Nahlen BL, King CL, Kazura JW: Cytokine responses to Plasmodium falciparum liver-stage antigen 1 vary in rainy and dry seasons in highland Kenya. Infection and Immunity. 2000, 68: 5198-204. 10.1128/IAI.68.9.5198-5204.2000.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Prakash D, Fesel C, Jain R, Cazenave PA, Mishra GC, Pied S: Clusters of cytokines determine malaria severity in Plasmodium falciparum-infected patients from endemic areas of Central India. The Journal of Infectious Diseases. 2006, 194: 198-207. 10.1086/504720.PubMedCrossRef Prakash D, Fesel C, Jain R, Cazenave PA, Mishra GC, Pied S: Clusters of cytokines determine malaria severity in Plasmodium falciparum-infected patients from endemic areas of Central India. The Journal of Infectious Diseases. 2006, 194: 198-207. 10.1086/504720.PubMedCrossRef
40.
Zurück zum Zitat Deloron P, Lepers JP, Coulanges P: Evolution of the levels of soluble interleukin-2 receptors during Plasmodium falciparum and P. vivax malaria. Journal of Clinical Microbiology. 1989, 27: 1887-9.PubMedPubMedCentral Deloron P, Lepers JP, Coulanges P: Evolution of the levels of soluble interleukin-2 receptors during Plasmodium falciparum and P. vivax malaria. Journal of Clinical Microbiology. 1989, 27: 1887-9.PubMedPubMedCentral
41.
Zurück zum Zitat Kremsner PG, Zotter GM, Feldmeier H, Graninger W, Rocha RM, Jansen-Rosseck R, Bienzle U: Immune response in patients during and after Plasmodium falciparum infection. The Journal of Infectious Diseases. 1990, 161: 1025-8. 10.1093/infdis/161.5.1025.PubMedCrossRef Kremsner PG, Zotter GM, Feldmeier H, Graninger W, Rocha RM, Jansen-Rosseck R, Bienzle U: Immune response in patients during and after Plasmodium falciparum infection. The Journal of Infectious Diseases. 1990, 161: 1025-8. 10.1093/infdis/161.5.1025.PubMedCrossRef
42.
Zurück zum Zitat Nguyen-Dinh P, Greenberg AE: Increased levels of released interleukin-2 receptors in Plasmodium falciparum malaria. The Journal of Infectious Diseases. 1988, 158: 1403-4. 10.1093/infdis/158.6.1403.PubMedCrossRef Nguyen-Dinh P, Greenberg AE: Increased levels of released interleukin-2 receptors in Plasmodium falciparum malaria. The Journal of Infectious Diseases. 1988, 158: 1403-4. 10.1093/infdis/158.6.1403.PubMedCrossRef
43.
Zurück zum Zitat Rhodes-Feuillette A, Bellosguardo M, Druilhe P, Ballet JJ, Chousterman S, Canivet M, Périès J: The interferon compartment of the immune response in human malaria: II. Presence of serum-interferon gamma following the acute attack. Journal of Interferon Research. 1985, 5: 169-78. 10.1089/jir.1985.5.169.PubMedCrossRef Rhodes-Feuillette A, Bellosguardo M, Druilhe P, Ballet JJ, Chousterman S, Canivet M, Périès J: The interferon compartment of the immune response in human malaria: II. Presence of serum-interferon gamma following the acute attack. Journal of Interferon Research. 1985, 5: 169-78. 10.1089/jir.1985.5.169.PubMedCrossRef
44.
Zurück zum Zitat Peyron F, Vuillez JP, Barbe G, Boudin C, Picot S, Ambroise-Thomas P: Plasma levels of tumor necrosis factor during a longitudinal survey in an endemic area of malaria. Acta Tropica. 1990, 47: 47-51. 10.1016/0001-706X(90)90006-L.PubMedCrossRef Peyron F, Vuillez JP, Barbe G, Boudin C, Picot S, Ambroise-Thomas P: Plasma levels of tumor necrosis factor during a longitudinal survey in an endemic area of malaria. Acta Tropica. 1990, 47: 47-51. 10.1016/0001-706X(90)90006-L.PubMedCrossRef
45.
Zurück zum Zitat Riley EM, Morris-Jones S, Blackman MJ, Greenwood BM, Holder AA: A longitudinal study of naturally acquired cellular and humoral immune responses to a merozoite surface protein (MSP1) of Plasmodium falciparum in an area of seasonal malaria transmission. Parasite Immunology. 1993, 15: 513-524. 10.1111/j.1365-3024.1993.tb00639.x.PubMedCrossRef Riley EM, Morris-Jones S, Blackman MJ, Greenwood BM, Holder AA: A longitudinal study of naturally acquired cellular and humoral immune responses to a merozoite surface protein (MSP1) of Plasmodium falciparum in an area of seasonal malaria transmission. Parasite Immunology. 1993, 15: 513-524. 10.1111/j.1365-3024.1993.tb00639.x.PubMedCrossRef
46.
Zurück zum Zitat Bouyou-Akotet MK, Kombila M, Kremsner PG, Mavoungou E: Cytokine profiles in peripheral, placental and cord blood in pregnant women from an area endemic for Plasmodium falciparum. European Cytokine Network. 2004, 15: 120-125.PubMed Bouyou-Akotet MK, Kombila M, Kremsner PG, Mavoungou E: Cytokine profiles in peripheral, placental and cord blood in pregnant women from an area endemic for Plasmodium falciparum. European Cytokine Network. 2004, 15: 120-125.PubMed
47.
Zurück zum Zitat Breitling LP, Fendel R, Mordmueller B, Adegnika AA, Kremsner PG, Luty AJF: Cord blood dendritic cell subsets in African newborns exposed to Plasmodium falciparum in utero. Infection and Immunity. 2006, 74: 5725-9. 10.1128/IAI.00682-06.PubMedPubMedCentralCrossRef Breitling LP, Fendel R, Mordmueller B, Adegnika AA, Kremsner PG, Luty AJF: Cord blood dendritic cell subsets in African newborns exposed to Plasmodium falciparum in utero. Infection and Immunity. 2006, 74: 5725-9. 10.1128/IAI.00682-06.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Brustoski K, Kramer M, Möller U, Kremsner PG, Luty AJF: Neonatal and maternal immunological responses to conserved epitopes within the DBL-gamma3 chondroitin sulfate A-binding domain of Plasmodium falciparum erythrocyte membrane protein 1. Infection and Immunity. 2005, 73: 7988-95. 10.1128/IAI.73.12.7988-7995.2005.PubMedPubMedCentralCrossRef Brustoski K, Kramer M, Möller U, Kremsner PG, Luty AJF: Neonatal and maternal immunological responses to conserved epitopes within the DBL-gamma3 chondroitin sulfate A-binding domain of Plasmodium falciparum erythrocyte membrane protein 1. Infection and Immunity. 2005, 73: 7988-95. 10.1128/IAI.73.12.7988-7995.2005.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Brustoski K, Moller U, Kramer M, Hartgers FC, Kremsner PG, Krzych U, Luty AJF: Reduced cord blood immune effector-cell responsiveness mediated by CD4+ cells induced in utero as a consequence of placental Plasmodium falciparum infection. The Journal of Infectious Diseases. 2006, 193: 146-54. 10.1086/498578.PubMedCrossRef Brustoski K, Moller U, Kramer M, Hartgers FC, Kremsner PG, Krzych U, Luty AJF: Reduced cord blood immune effector-cell responsiveness mediated by CD4+ cells induced in utero as a consequence of placental Plasmodium falciparum infection. The Journal of Infectious Diseases. 2006, 193: 146-54. 10.1086/498578.PubMedCrossRef
50.
Zurück zum Zitat Brustoski K, Moller U, Kramer M, Petelski A, Brenner S, Palmer DR, Bongartz M, Kremsner PG, Luty AJ, Krzych U: IFN-gamma and IL-10 mediate parasite-specific immune responses of cord blood cells induced by pregnancy-associated Plasmodium falciparum malaria. Journal of Immunology. 2005, 174: 1738-1745.CrossRef Brustoski K, Moller U, Kramer M, Petelski A, Brenner S, Palmer DR, Bongartz M, Kremsner PG, Luty AJ, Krzych U: IFN-gamma and IL-10 mediate parasite-specific immune responses of cord blood cells induced by pregnancy-associated Plasmodium falciparum malaria. Journal of Immunology. 2005, 174: 1738-1745.CrossRef
51.
Zurück zum Zitat Fievet N, Ringwald P, Bickii J, Dubois B, Maubert B, Le Hesran JY, Cot M, Deloron P: Malaria cellular immune responses in neonates from Cameroon. Parasite Immunology. 1996, 18: 483-90. 10.1046/j.1365-3024.1996.d01-19.x.PubMedCrossRef Fievet N, Ringwald P, Bickii J, Dubois B, Maubert B, Le Hesran JY, Cot M, Deloron P: Malaria cellular immune responses in neonates from Cameroon. Parasite Immunology. 1996, 18: 483-90. 10.1046/j.1365-3024.1996.d01-19.x.PubMedCrossRef
52.
Zurück zum Zitat King CL, Malhotra I, Wamachi A, Kioko J, Mungai P, Wahab SA, Koech D, Zimmerman P, Ouma J, Kazura JW: Acquired immune responses to Plasmodium falciparum merozoite surface protein-1 in the human fetus. Journal of Immunology. 2002, 168: 356-64.CrossRef King CL, Malhotra I, Wamachi A, Kioko J, Mungai P, Wahab SA, Koech D, Zimmerman P, Ouma J, Kazura JW: Acquired immune responses to Plasmodium falciparum merozoite surface protein-1 in the human fetus. Journal of Immunology. 2002, 168: 356-64.CrossRef
53.
Zurück zum Zitat Malhotra I, Mungai P, Muchiri E, Ouma J, Sharma S, Kazura JW, King CL: Distinct Th1- and Th2-Type prenatal cytokine responses to Plasmodium falciparum erythrocyte invasion ligands. Infection and Immunity. 2005, 73: 3462-3470. 10.1128/IAI.73.6.3462-3470.2005.PubMedPubMedCentralCrossRef Malhotra I, Mungai P, Muchiri E, Ouma J, Sharma S, Kazura JW, King CL: Distinct Th1- and Th2-Type prenatal cytokine responses to Plasmodium falciparum erythrocyte invasion ligands. Infection and Immunity. 2005, 73: 3462-3470. 10.1128/IAI.73.6.3462-3470.2005.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Metenou S, Long C, Leke RG, Taylor DW: Fetal Immune Responses to Plasmodium falciparum Antigens in a Malaria-Endemic Region of Cameroon. Journal of Immunology. 2007, 178: 2770-2777.CrossRef Metenou S, Long C, Leke RG, Taylor DW: Fetal Immune Responses to Plasmodium falciparum Antigens in a Malaria-Endemic Region of Cameroon. Journal of Immunology. 2007, 178: 2770-2777.CrossRef
55.
Zurück zum Zitat Hesran JYL, Fievet N, Thioulouse J, Personne P, Maubert B, M'bidias S, Etye'ale D, Cot M, Deloron P: Development of cellular immune responses to Plasmodium falciparum blood stage antigens from birth to 36 months of age in Cameroon. Acta Tropica. 2006, 98: 261-269. 10.1016/j.actatropica.2006.05.010.PubMedCrossRef Hesran JYL, Fievet N, Thioulouse J, Personne P, Maubert B, M'bidias S, Etye'ale D, Cot M, Deloron P: Development of cellular immune responses to Plasmodium falciparum blood stage antigens from birth to 36 months of age in Cameroon. Acta Tropica. 2006, 98: 261-269. 10.1016/j.actatropica.2006.05.010.PubMedCrossRef
56.
Zurück zum Zitat Mshana RN, Boulandi J, Mshana NM, Mayombo J, Mendome G: Cytokines in the pathogenesis of malaria: levels of IL-I beta, IL-4, IL-6, TNF-alpha and IFN-gamma in plasma of healthy individuals and malaria patients in a holoendemic area. Journal of Clinical & Laboratory Immunology. 1991, 34: 131-9. Mshana RN, Boulandi J, Mshana NM, Mayombo J, Mendome G: Cytokines in the pathogenesis of malaria: levels of IL-I beta, IL-4, IL-6, TNF-alpha and IFN-gamma in plasma of healthy individuals and malaria patients in a holoendemic area. Journal of Clinical & Laboratory Immunology. 1991, 34: 131-9.
57.
Zurück zum Zitat Macete E, Aide P, Aponte JJ, Sanz S, Mandomando I, Espasa M, Sigauque B, Dobaño C, Mabunda S, Dgedge M, Alonso P, Menendez C: Intermittent preventive treatment for malaria control administered at the time of routine vaccinations in mozambican infants: a randomized, placebo-controlled trial. The Journal of Infectious Diseases. 2006, 194: 276-285. 10.1086/505431.PubMedCrossRef Macete E, Aide P, Aponte JJ, Sanz S, Mandomando I, Espasa M, Sigauque B, Dobaño C, Mabunda S, Dgedge M, Alonso P, Menendez C: Intermittent preventive treatment for malaria control administered at the time of routine vaccinations in mozambican infants: a randomized, placebo-controlled trial. The Journal of Infectious Diseases. 2006, 194: 276-285. 10.1086/505431.PubMedCrossRef
58.
Zurück zum Zitat Quelhas D, Jiménez A, Quintó L, Serra-Casas E, Mayor A, Cisteró P, Puyol L, Wilson D, Richards J, Nhampossa T, Macete E, Aide P, Mandomando I, Sanz S, Aponte J, Alonso P, Beeson J, Menéndez C, Dobaño C: IgG against Plasmodium falciparum variant surface antigens and growth inhibitory antibodies in Mozambican children receiving intermittent preventive treatment with Sulfadoxine-Pyrimethamine. Immunobiology. 2011, 216: 793-802. 10.1016/j.imbio.2010.12.010.PubMedCrossRef Quelhas D, Jiménez A, Quintó L, Serra-Casas E, Mayor A, Cisteró P, Puyol L, Wilson D, Richards J, Nhampossa T, Macete E, Aide P, Mandomando I, Sanz S, Aponte J, Alonso P, Beeson J, Menéndez C, Dobaño C: IgG against Plasmodium falciparum variant surface antigens and growth inhibitory antibodies in Mozambican children receiving intermittent preventive treatment with Sulfadoxine-Pyrimethamine. Immunobiology. 2011, 216: 793-802. 10.1016/j.imbio.2010.12.010.PubMedCrossRef
59.
Zurück zum Zitat Quelhas D, Puyol L, Quinto L, Serra-Casas E, Nhampossa T, Macete E, Aide P, Mayor A, Mandomando I, Sanz S, Aponte JJ, Chauhan VS, Chitnis CE, Alonso PL, Menendez C, Dobaño C: Impact of intermittent preventive treatment with sulfadoxine-pyrimethamine on antibody responses to erythrocytic-stage Plasmodium falciparum antigens in infants in Mozambique. Clinical and Vaccine Immunology. 2008, 15: 1282-1291. 10.1128/CVI.00044-08.PubMedPubMedCentralCrossRef Quelhas D, Puyol L, Quinto L, Serra-Casas E, Nhampossa T, Macete E, Aide P, Mayor A, Mandomando I, Sanz S, Aponte JJ, Chauhan VS, Chitnis CE, Alonso PL, Menendez C, Dobaño C: Impact of intermittent preventive treatment with sulfadoxine-pyrimethamine on antibody responses to erythrocytic-stage Plasmodium falciparum antigens in infants in Mozambique. Clinical and Vaccine Immunology. 2008, 15: 1282-1291. 10.1128/CVI.00044-08.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Otoo LN, Riley EM, Menon A, Byass P, Greenwood BM: Cellular immune responses to Plasmodium falciparum antigens in children receiving long term anti-malarial chemoprophylaxis. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1989, 83: 778-782. 10.1016/0035-9203(89)90324-6.PubMedCrossRef Otoo LN, Riley EM, Menon A, Byass P, Greenwood BM: Cellular immune responses to Plasmodium falciparum antigens in children receiving long term anti-malarial chemoprophylaxis. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1989, 83: 778-782. 10.1016/0035-9203(89)90324-6.PubMedCrossRef
61.
Zurück zum Zitat Friedman JF, Phillips-Howard PA, Hawley WA, Terlouw DJ, Kolczak MS, Barber M, Okello N, Vulule JM, Duggan C, Nahlen BL, ter Kuile FO: Impact of permethrin-treated bed nets on growth, nutritional status, and body composition of primary school children in western Kenya. The American Journal of Tropical Medicine and Hygiene. 2003, 68: 78-85.PubMed Friedman JF, Phillips-Howard PA, Hawley WA, Terlouw DJ, Kolczak MS, Barber M, Okello N, Vulule JM, Duggan C, Nahlen BL, ter Kuile FO: Impact of permethrin-treated bed nets on growth, nutritional status, and body composition of primary school children in western Kenya. The American Journal of Tropical Medicine and Hygiene. 2003, 68: 78-85.PubMed
62.
Zurück zum Zitat Aponte JJ, Schellenberg D, Egan A, Breckenridge A, Carneiro I, Critchley J, Danquah I, Dodoo A, Kobbe R, Lell B: Efficacy and safety of intermittent preventive treatment with sulfadoxine-pyrimethamine for malaria in African infants: a pooled analysis of six randomised, placebo-controlled trials. The Lancet. 2009, 374: 1533-1542. 10.1016/S0140-6736(09)61258-7.CrossRef Aponte JJ, Schellenberg D, Egan A, Breckenridge A, Carneiro I, Critchley J, Danquah I, Dodoo A, Kobbe R, Lell B: Efficacy and safety of intermittent preventive treatment with sulfadoxine-pyrimethamine for malaria in African infants: a pooled analysis of six randomised, placebo-controlled trials. The Lancet. 2009, 374: 1533-1542. 10.1016/S0140-6736(09)61258-7.CrossRef
63.
Zurück zum Zitat Barbosa A, Naniche D, Aponte JJ, Manaca MN, Mandomando I, Aide P, Sacarlal J, Renom M, Lafuente S, Ballou WR, Alonso PL: Plasmodium falciparum-specific cellular immune responses after immunization with the RTS,S/AS02D candidate malaria vaccine in infants living in an area of high endemicity in Mozambique. Infection and Immunity. 2009, 77: 4502-4509. 10.1128/IAI.00442-09.PubMedPubMedCentralCrossRef Barbosa A, Naniche D, Aponte JJ, Manaca MN, Mandomando I, Aide P, Sacarlal J, Renom M, Lafuente S, Ballou WR, Alonso PL: Plasmodium falciparum-specific cellular immune responses after immunization with the RTS,S/AS02D candidate malaria vaccine in infants living in an area of high endemicity in Mozambique. Infection and Immunity. 2009, 77: 4502-4509. 10.1128/IAI.00442-09.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Struik SS, Omer FM, Artavanis-Tsakonas K, Riley EM: Uninfected erythrocytes inhibit Plasmodium falciparum-induced cellular immune responses in whole-blood assays. Blood. 2004, 103: 3084-92. 10.1182/blood-2003-08-2867.PubMedCrossRef Struik SS, Omer FM, Artavanis-Tsakonas K, Riley EM: Uninfected erythrocytes inhibit Plasmodium falciparum-induced cellular immune responses in whole-blood assays. Blood. 2004, 103: 3084-92. 10.1182/blood-2003-08-2867.PubMedCrossRef
65.
Zurück zum Zitat Walther M, Woodruff J, Edele F, Jeffries D, Tongren JE, King E, Andrews L, Bejon P, Gilbert SC, Souza JBD, Sinden R, Hill AV, Riley EM: Innate immune responses to human malaria: heterogeneous cytokine responses to blood-stage Plasmodium falciparum correlate with parasitological and clinical outcomes. Journal of Immunology. 2006, 177: 5736-5745.CrossRef Walther M, Woodruff J, Edele F, Jeffries D, Tongren JE, King E, Andrews L, Bejon P, Gilbert SC, Souza JBD, Sinden R, Hill AV, Riley EM: Innate immune responses to human malaria: heterogeneous cytokine responses to blood-stage Plasmodium falciparum correlate with parasitological and clinical outcomes. Journal of Immunology. 2006, 177: 5736-5745.CrossRef
66.
Zurück zum Zitat Jason J, Archibald LK, Nwanyanwu OC, Byrd MG, Kazembe PN, Dobbie H, Jarvis WR: Comparison of serum and cell-specific cytokines in humans. Clinical and Diagnostic Laboratory Immunology. 2001, 8: 1097-1103.PubMedPubMedCentral Jason J, Archibald LK, Nwanyanwu OC, Byrd MG, Kazembe PN, Dobbie H, Jarvis WR: Comparison of serum and cell-specific cytokines in humans. Clinical and Diagnostic Laboratory Immunology. 2001, 8: 1097-1103.PubMedPubMedCentral
67.
Zurück zum Zitat Abacassamo F, Enosse S, Aponte JJ, Gomez-Olive FX, Quinto L, Mabunda S, Barreto A, Magnussen P, Ronn AM, Thompson R, Alonso PL: Efficacy of chloroquine, amodiaquine, sulphadoxine-pyrimethamine and combination therapy with artesunate in Mozambican children with non-complicated malaria. Tropical Medicine & International Health. 2004, 9: 200-208. 10.1046/j.1365-3156.2003.01182.x.CrossRef Abacassamo F, Enosse S, Aponte JJ, Gomez-Olive FX, Quinto L, Mabunda S, Barreto A, Magnussen P, Ronn AM, Thompson R, Alonso PL: Efficacy of chloroquine, amodiaquine, sulphadoxine-pyrimethamine and combination therapy with artesunate in Mozambican children with non-complicated malaria. Tropical Medicine & International Health. 2004, 9: 200-208. 10.1046/j.1365-3156.2003.01182.x.CrossRef
68.
Zurück zum Zitat Saúte F, Aponte J, Almeda J, Ascaso C, Abellana R, Vaz N, Dgedge M, Alonso P: Malaria in southern Mozambique: malariometric indicators and malaria case definition in Manhiça district. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2003, 97: 661-6. 10.1016/S0035-9203(03)80098-6.PubMedCrossRef Saúte F, Aponte J, Almeda J, Ascaso C, Abellana R, Vaz N, Dgedge M, Alonso P: Malaria in southern Mozambique: malariometric indicators and malaria case definition in Manhiça district. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2003, 97: 661-6. 10.1016/S0035-9203(03)80098-6.PubMedCrossRef
69.
Zurück zum Zitat PrabhuDas M, Adkins B, Gans H, King C, Levy O, Ramilo O, Siegrist CA: Challenges in infant immunity: implications for responses to infection and vaccines. Nature Immunology. 2011, 12: 189-194.PubMedCrossRef PrabhuDas M, Adkins B, Gans H, King C, Levy O, Ramilo O, Siegrist CA: Challenges in infant immunity: implications for responses to infection and vaccines. Nature Immunology. 2011, 12: 189-194.PubMedCrossRef
70.
Zurück zum Zitat van de Veerdonk FL, Gresnigt MS, Kullberg BJ, van der Meer JWM, Joosten LAB, Netea MG: Th17 responses and host defense against microorganisms: an overview. BMB Reports. 2009, 42: 776-87. 10.5483/BMBRep.2009.42.12.776.PubMedCrossRef van de Veerdonk FL, Gresnigt MS, Kullberg BJ, van der Meer JWM, Joosten LAB, Netea MG: Th17 responses and host defense against microorganisms: an overview. BMB Reports. 2009, 42: 776-87. 10.5483/BMBRep.2009.42.12.776.PubMedCrossRef
71.
Zurück zum Zitat Hugosson E, Montgomery SM, Premji Z, Troye-Blomberg M, Bjorkman A: Higher IL-10 levels are associated with less effective clearance of Plasmodium falciparum parasites. Parasite Immunology. 2004, 26: 111-117. 10.1111/j.0141-9838.2004.00678.x.PubMedCrossRef Hugosson E, Montgomery SM, Premji Z, Troye-Blomberg M, Bjorkman A: Higher IL-10 levels are associated with less effective clearance of Plasmodium falciparum parasites. Parasite Immunology. 2004, 26: 111-117. 10.1111/j.0141-9838.2004.00678.x.PubMedCrossRef
72.
Zurück zum Zitat Troye-Blomberg M, Riley EM, Kabilan L, Holmberg M, Perlmann H, Andersson U, Heusser CH, Perlmann P: Production by activated human T cells of interleukin 4 but not interferon-gamma is associated with elevated levels of serum antibodies to activating malaria antigens. Proceedings of the National Academy of Sciences of the United States of America. 1990, 87: 5484-5488. 10.1073/pnas.87.14.5484.PubMedPubMedCentralCrossRef Troye-Blomberg M, Riley EM, Kabilan L, Holmberg M, Perlmann H, Andersson U, Heusser CH, Perlmann P: Production by activated human T cells of interleukin 4 but not interferon-gamma is associated with elevated levels of serum antibodies to activating malaria antigens. Proceedings of the National Academy of Sciences of the United States of America. 1990, 87: 5484-5488. 10.1073/pnas.87.14.5484.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat McCall MBB, Sauerwein RW: Interferon-{gamma}--central mediator of protective immune responses against the pre-erythrocytic and blood stage of malaria. Journal of Leukocyte Biology. 2010, 88: 1131-43. 10.1189/jlb.0310137.PubMedCrossRef McCall MBB, Sauerwein RW: Interferon-{gamma}--central mediator of protective immune responses against the pre-erythrocytic and blood stage of malaria. Journal of Leukocyte Biology. 2010, 88: 1131-43. 10.1189/jlb.0310137.PubMedCrossRef
74.
Zurück zum Zitat Miller LH, Baruch DI, Marsh K, Doumbo OK: The pathogenic basis of malaria. Nature. 2002, 415: 673-679. 10.1038/415673a.PubMedCrossRef Miller LH, Baruch DI, Marsh K, Doumbo OK: The pathogenic basis of malaria. Nature. 2002, 415: 673-679. 10.1038/415673a.PubMedCrossRef
75.
Zurück zum Zitat McCall MBB, Roestenberg M, Ploemen I, Teirlinck A, Hopman J, de Mast Q, Dolo A, Doumbo OK, Luty A, van der Ven AJAM, Hermsen CC, Sauerwein RW: Memory-like IFN-γ response by NK cells following malaria infection reveals the crucial role of T cells in NK cell activation by P. falciparum. European Journal of Immunology. 2010, 40: 3472-7. 10.1002/eji.201040587.PubMedCrossRef McCall MBB, Roestenberg M, Ploemen I, Teirlinck A, Hopman J, de Mast Q, Dolo A, Doumbo OK, Luty A, van der Ven AJAM, Hermsen CC, Sauerwein RW: Memory-like IFN-γ response by NK cells following malaria infection reveals the crucial role of T cells in NK cell activation by P. falciparum. European Journal of Immunology. 2010, 40: 3472-7. 10.1002/eji.201040587.PubMedCrossRef
76.
Zurück zum Zitat Allison AC: Genetic control of resistance to human malaria. Current opinion in immunology. 2009, 21: 499-505. 10.1016/j.coi.2009.04.001.PubMedCrossRef Allison AC: Genetic control of resistance to human malaria. Current opinion in immunology. 2009, 21: 499-505. 10.1016/j.coi.2009.04.001.PubMedCrossRef
77.
Zurück zum Zitat Allison AC: Protection afforded by sickle-cell trait against subtertian malareal infection. British Medical Journal. 1954, 1: 290-4. 10.1136/bmj.1.4857.290.PubMedPubMedCentralCrossRef Allison AC: Protection afforded by sickle-cell trait against subtertian malareal infection. British Medical Journal. 1954, 1: 290-4. 10.1136/bmj.1.4857.290.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Aubouy A, Migot-Nabias F, Deloron P: Correlations between treatment outcome and both anti-MSP119 antibody response and erythrocyte-related genetic factors in Plasmodium falciparum malaria. Journal of molecular epidemiology and evolutionary genetics in infectious diseases. 2007, 7: 147-54.PubMedCrossRef Aubouy A, Migot-Nabias F, Deloron P: Correlations between treatment outcome and both anti-MSP119 antibody response and erythrocyte-related genetic factors in Plasmodium falciparum malaria. Journal of molecular epidemiology and evolutionary genetics in infectious diseases. 2007, 7: 147-54.PubMedCrossRef
79.
Zurück zum Zitat Awandare GA, Ouma C, Keller CC, Were T, Otieno R, Ouma Y, Davenport GC, Hittner JB, Ong'echa JM, Ferrell R, Perkins DJ: A macrophage migration inhibitory factor promoter polymorphism is associated with high-density parasitemia in children with malaria. Genes and Immunity. 2006, 7: 568-75. 10.1038/sj.gene.6364332.PubMedCrossRef Awandare GA, Ouma C, Keller CC, Were T, Otieno R, Ouma Y, Davenport GC, Hittner JB, Ong'echa JM, Ferrell R, Perkins DJ: A macrophage migration inhibitory factor promoter polymorphism is associated with high-density parasitemia in children with malaria. Genes and Immunity. 2006, 7: 568-75. 10.1038/sj.gene.6364332.PubMedCrossRef
80.
Zurück zum Zitat Basu M, Maji AK, Chakraborty A, Banerjee R, Mullick S, Saha P, Das S, Kanjilal SD, Sengupta S: Genetic association of Toll-like-receptor 4 and tumor necrosis factor-alpha polymorphisms with Plasmodium falciparum blood infection levels. Journal of molecular epidemiology and evolutionary genetics in infectious diseases. 2010, 10: 686-96.PubMedCrossRef Basu M, Maji AK, Chakraborty A, Banerjee R, Mullick S, Saha P, Das S, Kanjilal SD, Sengupta S: Genetic association of Toll-like-receptor 4 and tumor necrosis factor-alpha polymorphisms with Plasmodium falciparum blood infection levels. Journal of molecular epidemiology and evolutionary genetics in infectious diseases. 2010, 10: 686-96.PubMedCrossRef
81.
Zurück zum Zitat Duah NO, Weiss HA, Jepson A, Tetteh KKA, Whittle HC, Conway DJ: Heritability of antibody isotype and subclass responses to Plasmodium falciparum antigens. PloS One. 2009, 4: e7381-10.1371/journal.pone.0007381.PubMedPubMedCentralCrossRef Duah NO, Weiss HA, Jepson A, Tetteh KKA, Whittle HC, Conway DJ: Heritability of antibody isotype and subclass responses to Plasmodium falciparum antigens. PloS One. 2009, 4: e7381-10.1371/journal.pone.0007381.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Hill AV: The immunogenetics of human infectious diseases. Annual Review of Immunology. 1998, 16: 593-617. 10.1146/annurev.immunol.16.1.593.PubMedCrossRef Hill AV: The immunogenetics of human infectious diseases. Annual Review of Immunology. 1998, 16: 593-617. 10.1146/annurev.immunol.16.1.593.PubMedCrossRef
83.
Zurück zum Zitat McNicholl JM, Downer MV, Udhayakumar V, Alper CA, Swerdlow DL: Host-pathogen interactions in emerging and re-emerging infectious diseases: a genomic perspective of tuberculosis, malaria, human immunodeficiency virus infection, hepatitis B, and cholera. Annual Review of Public Health. 2000, 21: 15-46. 10.1146/annurev.publhealth.21.1.15.PubMedCrossRef McNicholl JM, Downer MV, Udhayakumar V, Alper CA, Swerdlow DL: Host-pathogen interactions in emerging and re-emerging infectious diseases: a genomic perspective of tuberculosis, malaria, human immunodeficiency virus infection, hepatitis B, and cholera. Annual Review of Public Health. 2000, 21: 15-46. 10.1146/annurev.publhealth.21.1.15.PubMedCrossRef
84.
Zurück zum Zitat Mockenhaupt FP, Cramer JP, Hamann L, Stegemann MS, Eckert J, Oh NR, Otchwemah RN, Dietz E, Ehrhardt S, Schröder NWJ, Bienzle U, Schumann RR: Toll-like receptor (TLR) polymorphisms in African children: Common TLR-4 variants predispose to severe malaria. Proceedings of the National Academy of Sciences of the United States of America. 2006, 103: 177-82. 10.1073/pnas.0506803102.PubMedPubMedCentralCrossRef Mockenhaupt FP, Cramer JP, Hamann L, Stegemann MS, Eckert J, Oh NR, Otchwemah RN, Dietz E, Ehrhardt S, Schröder NWJ, Bienzle U, Schumann RR: Toll-like receptor (TLR) polymorphisms in African children: Common TLR-4 variants predispose to severe malaria. Proceedings of the National Academy of Sciences of the United States of America. 2006, 103: 177-82. 10.1073/pnas.0506803102.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Modiano D, Luoni G, Sirima BS, Lanfrancotti A, Petrarca V, Cruciani F, Simporé J, Ciminelli BM, Foglietta E, Grisanti P, Bianco I, Modiano G, Coluzzi M: The lower susceptibility to Plasmodium falciparum malaria of Fulani of Burkina Faso (west Africa) is associated with low frequencies of classic malaria-resistance genes. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2001, 95: 149-52. 10.1016/S0035-9203(01)90141-5.PubMedCrossRef Modiano D, Luoni G, Sirima BS, Lanfrancotti A, Petrarca V, Cruciani F, Simporé J, Ciminelli BM, Foglietta E, Grisanti P, Bianco I, Modiano G, Coluzzi M: The lower susceptibility to Plasmodium falciparum malaria of Fulani of Burkina Faso (west Africa) is associated with low frequencies of classic malaria-resistance genes. Transactions of the Royal Society of Tropical Medicine and Hygiene. 2001, 95: 149-52. 10.1016/S0035-9203(01)90141-5.PubMedCrossRef
86.
Zurück zum Zitat Modiano D, Petrarca V, Sirima BS, Nebié I, Diallo D, Esposito F, Coluzzi M: Different response to Plasmodium falciparum malaria in west African sympatric ethnic groups. Proceedings of the National Academy of Sciences of the United States of America. 1996, 93: 13206-11. 10.1073/pnas.93.23.13206.PubMedPubMedCentralCrossRef Modiano D, Petrarca V, Sirima BS, Nebié I, Diallo D, Esposito F, Coluzzi M: Different response to Plasmodium falciparum malaria in west African sympatric ethnic groups. Proceedings of the National Academy of Sciences of the United States of America. 1996, 93: 13206-11. 10.1073/pnas.93.23.13206.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Ockenhouse CF, chung Hu W, Kester KE, Cummings JF, Stewart A, Heppner DG, Jedlicka AE, Scott AL, Wolfe ND, Vahey M, Burke DS: Common and divergent immune response signaling pathways discovered in peripheral blood mononuclear cell gene expression patterns in presymptomatic and clinically apparent malaria. Infection and Immunity. 2006, 74: 5561-73. 10.1128/IAI.00408-06.PubMedPubMedCentralCrossRef Ockenhouse CF, chung Hu W, Kester KE, Cummings JF, Stewart A, Heppner DG, Jedlicka AE, Scott AL, Wolfe ND, Vahey M, Burke DS: Common and divergent immune response signaling pathways discovered in peripheral blood mononuclear cell gene expression patterns in presymptomatic and clinically apparent malaria. Infection and Immunity. 2006, 74: 5561-73. 10.1128/IAI.00408-06.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Paganotti GM, Palladino C, Modiano D, Sirima BS, Råberg L, Diarra A, Konaté A, Coluzzi M, Walliker D, Babiker HA: Genetic complexity and gametocyte production of Plasmodium falciparum in Fulani and Mossi communities in Burkina Faso. Parasitology. 2006, 132: 607-14.PubMedCrossRef Paganotti GM, Palladino C, Modiano D, Sirima BS, Råberg L, Diarra A, Konaté A, Coluzzi M, Walliker D, Babiker HA: Genetic complexity and gametocyte production of Plasmodium falciparum in Fulani and Mossi communities in Burkina Faso. Parasitology. 2006, 132: 607-14.PubMedCrossRef
89.
Zurück zum Zitat Riley EM: The role of MHC- and non-MHC-associated genes in determining the human immune response to malaria antigens. Parasitology. 1996, 112 (Suppl): S39-51.PubMed Riley EM: The role of MHC- and non-MHC-associated genes in determining the human immune response to malaria antigens. Parasitology. 1996, 112 (Suppl): S39-51.PubMed
90.
Zurück zum Zitat Stirnadel HA, Al-Yaman F, Genton B, Alpers MP, Smith TA: Assessment of different sources of variation in the antibody responses to specific malaria antigens in children in Papua New Guinea. International Journal of Epidemiology. 2000, 29: 579-86. 10.1093/ije/29.3.579.PubMedCrossRef Stirnadel HA, Al-Yaman F, Genton B, Alpers MP, Smith TA: Assessment of different sources of variation in the antibody responses to specific malaria antigens in children in Papua New Guinea. International Journal of Epidemiology. 2000, 29: 579-86. 10.1093/ije/29.3.579.PubMedCrossRef
91.
Zurück zum Zitat Traoré Y, Rihet P, Traoré-Leroux T, Aucan C, Gazin P, Coosemans M, Smith A, Abel L, Tall F, Nacro B, Traoré A, Fumoux F: Analysis of the genetic factors controlling malarial infection in man. Santé. 1999, 9: 53-9.PubMed Traoré Y, Rihet P, Traoré-Leroux T, Aucan C, Gazin P, Coosemans M, Smith A, Abel L, Tall F, Nacro B, Traoré A, Fumoux F: Analysis of the genetic factors controlling malarial infection in man. Santé. 1999, 9: 53-9.PubMed
92.
Zurück zum Zitat Mbugi EV, Meijerink M, Veenemans J, Jeurink PV, McCall M, Olomi RM, Shao JF, Chilongola JO, Verhoef H, Savelkoul HFJ: Effect of nutrient deficiencies on in vitro Th1 and Th2 cytokine response of peripheral blood mononuclear cells to Plasmodium falciparum infection. Malaria Journal. 2010, 9: 162-10.1186/1475-2875-9-162.PubMedPubMedCentralCrossRef Mbugi EV, Meijerink M, Veenemans J, Jeurink PV, McCall M, Olomi RM, Shao JF, Chilongola JO, Verhoef H, Savelkoul HFJ: Effect of nutrient deficiencies on in vitro Th1 and Th2 cytokine response of peripheral blood mononuclear cells to Plasmodium falciparum infection. Malaria Journal. 2010, 9: 162-10.1186/1475-2875-9-162.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Mbugi EV, Meijerink M, Veenemans J, Jeurink PV, McCall M, Olomi RM, Shao JF, Verhoef H, Savelkoul HF: Alterations in early cytokine-mediated immune responses to Plasmodium falciparum infection in Tanzanian children with mineral element deficiencies: a cross-sectional survey. Malaria Journal. 2010, 9: 130-10.1186/1475-2875-9-130.PubMedPubMedCentralCrossRef Mbugi EV, Meijerink M, Veenemans J, Jeurink PV, McCall M, Olomi RM, Shao JF, Verhoef H, Savelkoul HF: Alterations in early cytokine-mediated immune responses to Plasmodium falciparum infection in Tanzanian children with mineral element deficiencies: a cross-sectional survey. Malaria Journal. 2010, 9: 130-10.1186/1475-2875-9-130.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Chaisavaneeyakorn S, Moore JM, Otieno J, Chaiyaroj SC, Perkins DJ, Shi YP, Nahlen BL, Lal AA, Udhayakumar V: Immunity to placental malaria. III. Impairment of interleukin(IL)-12, not IL-18, and interferon-inducible protein-10 responses in the placental intervillous blood of human immunodeficiency virus/malaria-coinfected women. The Journal of Infectious Diseases. 2002, 185: 127-131. 10.1086/338013.PubMedCrossRef Chaisavaneeyakorn S, Moore JM, Otieno J, Chaiyaroj SC, Perkins DJ, Shi YP, Nahlen BL, Lal AA, Udhayakumar V: Immunity to placental malaria. III. Impairment of interleukin(IL)-12, not IL-18, and interferon-inducible protein-10 responses in the placental intervillous blood of human immunodeficiency virus/malaria-coinfected women. The Journal of Infectious Diseases. 2002, 185: 127-131. 10.1086/338013.PubMedCrossRef
95.
Zurück zum Zitat Broen K, Brustoski K, Engelmann I, Luty AJF: Placental Plasmodium falciparum infection: causes and consequences of in utero sensitization to parasite antigens. Molecular and Biochemical Parasitology. 2007, 151: 1-8. 10.1016/j.molbiopara.2006.10.001.PubMedCrossRef Broen K, Brustoski K, Engelmann I, Luty AJF: Placental Plasmodium falciparum infection: causes and consequences of in utero sensitization to parasite antigens. Molecular and Biochemical Parasitology. 2007, 151: 1-8. 10.1016/j.molbiopara.2006.10.001.PubMedCrossRef
Metadaten
Titel
Intermittent preventive treatment with sulfadoxine-pyrimethamine does not modify plasma cytokines and chemokines or intracellular cytokine responses to Plasmodium falciparum in Mozambican Children
verfasst von
Diana Quelhas
Laura Puyol
Llorenç Quintó
Tacilta Nhampossa
Elisa Serra-Casas
Eusébio Macete
Pedro Aide
Sergi Sanz
John J Aponte
Denise L Doolan
Pedro L Alonso
Clara Menéndez
Carlota Dobaño
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
BMC Immunology / Ausgabe 1/2012
Elektronische ISSN: 1471-2172
DOI
https://doi.org/10.1186/1471-2172-13-5

Weitere Artikel der Ausgabe 1/2012

BMC Immunology 1/2012 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

RAS-Blocker bei Hyperkaliämie möglichst nicht sofort absetzen

14.05.2024 Hyperkaliämie Nachrichten

Bei ausgeprägter Nierenfunktionsstörung steigen unter der Einnahme von Renin-Angiotensin-System(RAS)-Hemmstoffen nicht selten die Serumkaliumspiegel. Was in diesem Fall zu tun ist, erklärte Prof. Jürgen Floege beim diesjährigen Allgemeinmedizin-Update-Seminar.

Gestationsdiabetes: In der zweiten Schwangerschaft folgenreicher als in der ersten

13.05.2024 Gestationsdiabetes Nachrichten

Das Risiko, nach einem Gestationsdiabetes einen Typ-2-Diabetes zu entwickeln, hängt nicht nur von der Zahl, sondern auch von der Reihenfolge der betroffenen Schwangerschaften ab.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.