The objective of this study was to assess whether peptidoglycan (

The objective of this study was to assess whether peptidoglycan (PGN) derived from Gram-positive bacteria induces trophoblast stem (TS) cell death or alters TS cell cytokine

production. Method of study  Toll-like receptor (TLR) transcript expression was assessed by RT-PCR. Protein expression was determined by confocal microscopy or flow cytometry. 7-Aminoactinomycin D (7-AAD) staining was used to assess TS cell death. Morphological features of cell death were evaluated by transmission electron microscopy. The presence of cleaved caspase-3 and high mobility group box 1 (HMGB1) protein was examined by Western blot. Cytokine levels Cisplatin mw in cell supernatants were determined using a mouse cytokine 23-plex panel. Results  Toll-like receptor 2 and TLR4 protein was expressed from the 1-cell stage through the blastocyst stage of murine embryo development. Murine TS cells expressed TLR2 and TLR6 but not TLR1 or TLR4 RNA. Only TLR2 protein was detected at the plasma membrane of TS cells.

PGN induced TS cell death by a caspase-3-independent mechanism. The cell death pathway induced by PGN was morphologically consistent with necrosis. Finally, PGN induced HMGB1 release ACP-196 chemical structure and increased MIP-1β secretion while inhibiting the constitutive release of RANTES. Conclusion  Peptidoglycan-induced TS cell necrosis and the subsequent C1GALT1 release of HMGB1 and MIP-1β may regulate an infection-induced inflammatory response at the maternal–fetal interface and thus may play a role in the pathogenesis of infection-associated pregnancy complications. “
“A good understanding of the immunological correlates of protective immunity is an important requirement for the development of effective vaccines against malaria. However,

this concern has received little attention even in the face of two decades of intensive vaccine research. Here, we review the immune response to blood-stage malaria, with a particular focus on the type of vaccine most likely to induce the kind of response required to give strong protection against infection. Malaria still causes serious illness and many deaths in some of the poorest countries in the world. Over 200–300 million new cases are reported each year with 1·2 million deaths, mainly of young children [1]. There is still no vaccine that confers strong protective immunity to infection. Gaps in our understanding both of putative vaccine antigens and of the nature of antimalarial immunity have held back the development of a protective vaccine. While some immunity is acquired to infection after several years of repeated exposure to malarial infection, it is never complete. Such partial immunity or naturally acquired immunity that does develop, in an age and exposure related manner, involves both antibody and cell-mediated immune responses.

Our data suggest that individuals with low erythrocyte CR1 are le

Our data suggest that individuals with low erythrocyte CR1 are less equipped to mop up these ICs than individuals with high erythrocyte CR1 and are more likely to develop complications as a result. This is complicated further

by the fact that individuals afflicted by some of these diseases develop low CR1 levels as a result of the infection [16,17,24]. In addition, we have Selleckchem AG 14699 reported that the level of CR1 can vary with age, and young children aged from 6 to 24 months have the lowest levels of CR1 [15,21]. This population is at greatest risk from complications due to Plasmodium falciparum infection [29]. Young children are known to produce more TNF-α during malaria infection than older children, regardless of the level of parasitaemia [30], and differential capacity to remove ICs during malaria infection may be one potential explanation. We have provided evidence for a unique role of red cells in the stimulation of TNF-α production by presenting ICs and cross-linking Fcγ receptors on macrophages. This phenomenon may be important whenever slow circulation allows close contact between erythrocytes and monocyte/macrophages, such as in the liver and the spleen, leading to local production of proinflammatory cytokines. In the setting of P. falciparum malaria,

this could also happen in capillaries of the brain and other tissues where infected erythrocytes tend to adhere to the endothelium and sequester, slowing down the circulation. This is the pathognomonic feature

of cerebral Decitabine in vitro malaria, one of the deadliest complications of this infection. In these capillaries, local production of TNF-α has been documented by immunohistochemistry [31]. We propose that presentation of ICs to monocytes/macrophages by red cells is one possible mechanism for the localized production of proinflammatory cytokines in sequestered capillaries. In addition, IC-loaded red cells in microhaemorrhages of patients with CM could stimulate microglial cells, resident macrophages that express Fcγ receptors [32]. Differential expression of CR1 on red cells is an appealing explanation for the increased susceptibility to cerebral malaria of older children compared to young children [16]. However, our this website data do not support that differences in CR1 expression level can lead to differences in the ability of red cells to stimulate macrophages. In conclusion, we have demonstrated that erythrocytes can play a dual role in immune regulation, removing ICs from circulation to prevent inflammation and at the same time being capable of stimulating an inflammatory response by presenting ICs to macrophages. Our findings justify further exploration of the role of these mechanisms in the pathology of IC-mediated diseases such as malaria. This work was supported by NIH grant HL71502 (Principle Investigator José A. Stoute). We are grateful to individuals who participated in the study.

15 Administration of interleukin (IL)-10-treated DCs markedly sup

15 Administration of interleukin (IL)-10-treated DCs markedly suppressed the development of AHR, inflammation, and Th2 cytokine production.16 Similarly, activation of DCs with antibodies directed

to a member of the family of B7 costimulatory molecules PD-1 costimulatory molecule ligand ex vivo before adoptive transfer into pre-sensitized mice JNK inhibitor solubility dmso was shown to be sufficient to protect animals from inflammatory lung disease induced by subsequent repeated airway exposure to the offending antigen.17 In this study, we investigated whether transfer of histamine-treated allergen-pulsed DCs changed the course of the allergic response, in a well-defined model of OVA-induced allergic airway inflammation.18 All experiments were carried out using 2-month-old virgin female BALB/c mice raised at the National Academy of Medicine, Buenos Aires, Argentina. Mice were housed six per cage and

kept at 20 ± 2° under an automatic 12 hr light/dark schedule. Animal care was in accordance with institutional guidelines. Mice were sensitized using a standard protocol, as described previously.18 Briefly, mice were injected intraperitoneally (i.p.) with 20 μg of OVA (grade V; Sigma-Aldrich, Sigma, San Louis, MO) in 2 mg of aluminium hydroxide (alum) at days 0 and 7. Control mice received a saline injection instead of OVA/alum solution. On day 14, sensitized mice were challenged intranasally with 50 μl of phosphate-buffered MK-1775 concentration saline (PBS) containing 3% OVA for 5 days. Control mice were instillated with PBS. The procedure used to obtain DCs was as described by Inaba et al.,19 with minor modifications.20 Liothyronine Sodium Briefly, bone marrow was flushed from the long bones of the limbs using 2 ml of RPMI-1640 (Invitrogen, Carlsbad, CA) with a syringe and 25-gauge needle. Red cells were lysed with ammonium chloride. After washing, cells were suspended at a concentration of 1·5 × 106 cells/ml in 70% RPMI-1640 medium supplemented with 10% fetal calf serum (FCS), 5·5 × 10−5 mercaptoethanol (Sigma, San Louis, MO) (complete medium) and 30% J588-GM cell line supernatant. The cultures were fed every 2 days by gently swirling the plates, aspirating 50% of the medium,

and adding fresh medium with J588-GM cell line supernatant. At day 9 of the culture, > 90% of the harvested cells expressed MHC class II, CD40 and CD11c, but not Gr-1 (not shown). DCs obtained from bone marrow precursors were incubated in the absence or presence of histamine (1 μm) (DCs and DCHISs, respectively) for 30 min at 37°. Cells were then incubated for 3 hr at 37° in the presence or absence of OVA (100 μg/ml). Finally, DCs were washed and injected intratracheally (i.t.) into BALB/C mice after intranasal challenge of sensitized mice with OVA. For this purpose, mice were anaesthetized with embuthal (2% v/v in PBS), and 100 μl of PBS, DCs or DCHISs (5 × 105 cells) was injected. Lungs were cut into small pieces and treated with Type I collagenase (250 U/ml) (Roche; Bs.As.

The canonical member of the GlyAg family is polysaccharide A (PSA

The canonical member of the GlyAg family is polysaccharide A (PSA) from the capsule of B. fragilis. PSA is comprised of a tetrasaccharide repeating unit with both positively and negatively charged groups 17 that facilitate its ability to be presented by MHCII molecules 18. GlyAgs are endocytosed by professional APCs and trigger the production of NO 19, which is responsible for the oxidative cleavage of the antigen to low molecular weight fragments for MHCII-mediated presentation 20, 21. This NO-dependent oxidative X-396 processing and presentation mechanism is essential for GlyAg-specific T-cell recognition and activation. Animals lacking the iNOS

gene fail to form abscesses in response to GlyAg challenge 20. With NO-mediated oxidation at the root of GlyAg-induced abscess formation, we sought to understand the nature of the hyperresponsiveness in CGD. Using the gp91phox-deficient animal model of CGD, we discovered that the loss of a functional NADPH oxidase results in a ten-fold increase in sensitivity against GlyAg Nivolumab ic50 challenge, with

CGD abscesses being consistently larger compared with WT C57BL/6 (WT) controls. Ex vivo experiments further reveal an earlier and more robust T-cell activation response against GlyAg that correlated with increased NO and iNOS protein production in CGD animals and increased GlyAg processing in CGD APCs. Remarkably, CGD hyperresponsiveness was transferrable to WT animals through adoptive transfer of neutrophil-depleted CGD APCs, demonstrating that increased abscess formation was a result of aberrant APC function and the resulting downstream T-cell activation, rather than changes in neutrophil or T-cell activity resulting from Cediranib (AZD2171) changes in ROS production. Perhaps most significantly, we discovered that attenuation of iNOS activity with 1400W (N-(3-(aminomethyl)benzyl)acetamidine, 2HCl) effectively and safely reduced the incidence and severity of abscesses in CGD. These findings reveal that the abscess hyperresponsiveness in CGD is mediated at least in part through greater sensitivity to GlyAg

via an increase in NO-dependent T-cell activation and that treatment with 1400W could represent a novel approach to improving infection outcomes for CGD patients. GlyAg-mediated abscess formation in rodent models of sepsis is dependent upon MHCII presentation 20, 22, 23 and CD4+ T-cell activation 16, 23–26, while being exquisitely sensitive to NO production in responding APCs 19–21, 23. Given the dependence upon oxidation, we measured the impact of the CGD mutation on GlyAg-specific responses. CGD and WT mice were challenged i.p. with either 200 μg GlyAg containing undiluted sterile cecal contents (SCC) (dilution=1), SCC alone, or dilutions of each inoculum. On day 7, the number of mice with at least one abscess was scored (Fig. 1A). CGD animals were ten-fold more sensitive to GlyAg challenge compared with WT control animals (C1/2=four-fold dilution for WT; 40 for CGD).

Here, we more closely evaluate, in an in vivo setting in immunoco

Here, we more closely evaluate, in an in vivo setting in immunocompetent mice, the checkpoints at which polyclonal Treg cells exert their inhibitory function. We evaluated the role of Treg cells in the well-characterized model of myelin oligodendrocyte glycoprotein (MOG)-induced EAE. As previous studies 9 have shown that administration of polyclonal Treg cell to normal mice can partially inhibit the development of EAE, we transferred into recipient mice either Treg cells that had been purified from normal mice and expanded in vitro by stimulation with

anti-CD3 and IL-2 or Treg cells that had been generated from Foxp3− T cells by stimulation in vitro with TGF-β. One day following transfer, the mice were immunized for the induction of EAE. Both groups of Treg cell-treated mice displayed significantly reduced clinical

severity mTOR inhibitor as compared with the control group (Fig. 1A, right panel). Endogenous Treg cells also control the development of EAE as mice treated with a partially depleting or inactivating anti-CD25 antibody 10 3 days prior to immunization consistently exhibited an exacerbated disease course (Fig. 1A, left panel). Overall, these studies demonstrate that merely altering the number of Treg cells see more in vivo can dramatically alter the course of an autoimmune disease. To more thoroughly understand the mechanism(s) for the reduction of disease severity by enhancement of Treg cell numbers, we evaluated the phenotype of the Teff cells that had trafficked into the brain. We isolated the cellular infiltrate from the spinal cords of mice with EAE that had either received or had not received Treg cells, re-stimulated them in vitro with PMA/ionomycin, and evaluated cytokine production

by intracellular Elongation factor 2 kinase staining. Mice that had received Treg cells had a two-fold reduction in the percentage of central nervous system infiltrating CD4+ Teff cells (Fig. 1B, top), but on a per cell basis, the cytokine profile of these cells was almost identical between the two groups (Fig. 1B, bottom; the two-fold difference in IFN-γ+IL-17+ cells was not a consistently reproducible result). No differences were observed in the production of IL-2, IL-4, or TNF-α, or in the expression of memory/activation markers such as CD44, CD25, or CD69 (data not shown). Thus, the reduced clinical disease most strongly correlates with the reduced percentage of Teff cells that invade the CNS rather than Treg cell-mediated inhibition of Th1/Th17 differentiation or induction of immune deviation leading to the development of a less pathogenic Th2 phenotype.

The authors would like to thank Ane M Rulykke for excellent techn

The authors would like to thank Ane M Rulykke for excellent technical assistance. We would like to thank Jesper Jurlander for sharing reagents and ideas. Anti-CD20 antibodies were a kind gift from Mark S. Cragg and Claude H.T. Chan, whom we would also like to thank for scientific discussions. We would like to thank Esben G. Schmidt for technical support and Morten Rasch for advice on protease inhibition. This work was made possible by the University of Copenhagen, Faculty of Health Sciences and The Neye Foundation. The authors declare to have no financial conflicts or interest. “
“Formation EPZ-6438 price of immune synapses (IS) between T cells and

APC requires multiple rearrangements in the actin cytoskeleton and selective receptor accumulation in supramolecular activation

clusters (SMAC). The inner cluster (central SMAC) contains the TCR/CD3 complex. The outer cluster (peripheral SMAC) contains the integrin LFA-1 and Talin. Molecular mechanisms selectively stabilizing receptors in the IS remained largely unknown. Here, we demonstrate that sustained LFA-1 clustering in the IS is a consequence of the combined activities of the actin-bundling protein L-plastin (LPL) and calmodulin. Thus, upon antigen-recognition of T cells, LPL accumulated predominantly in the peripheral SMAC. siRNA-mediated knock-down of LPL led to a failure of LFA-1 and Talin redistribution – however, not TCR/CD3 relocalization – into the IS. As a result of this LPL knock-down, the T-cell/APC interface became smaller over time and T-cell proliferation was inhibited. Importantly, this website binding of calmodulin to LPL was required

for the maintenance of LPL in the IS and consequently inhibition of calmodulin also prevented stable accumulation of LFA-1 and Talin, but not CD3, in the IS. During the activation of T cells Protein kinase N1 the immune synapse (IS) is formed at the area of interaction between T cells and APC 1, 2. The IS is involved in enhancing, directing and terminating the T-cell immune response (for review, see 3–7). Within the IS, surface receptors as well as intracellular signaling and scaffolding proteins are organized in distinct structures, which are called supramolecular activation clusters (SMAC). The inner cluster (central SMAC or cSMAC) contains PKCΘ and the TCR/CD3 complex. The outer cluster (peripheral SMAC or pSMAC) is composed of the integrin LFA-1 (CD11a/CD18) and Talin 8. It is clear that for the development of an IS the actin cytoskeleton is of special importance 2, 9–11. For construction of an actin meshwork, as it is found in the IS, crosslinking and bundling of F-actin is indispensable to support F-actin rigidity. Here, we demonstrate that the actin-bundling protein L-plastin (LPL) is an important component to orchestrate the ordered formation of a mature IS. LPL is a leukocyte-specific protein.

We observed chitin-mediated inhibition of T-cell proliferation in

We observed chitin-mediated inhibition of T-cell proliferation in cultures from WT mice, whereas only weak inhibition was observed in cultures from B7-H1-deficient mice (Fig. 5A and B). Indeed, chitin-induced inhibition of T-cell proliferation was four times less efficient in cultures with cells from B7-H1-deficient

mice as compared with Sorafenib cultures with cells from WT mice (Fig. 5C). Therefore, we conclude that chitin-induced inhibition of T-cell proliferation was largely mediated by B7-H1. We found that chitin does neither induce nor inhibit Th2-cell polarization but rather reduces the proliferation of T cells mainly via upregulation of B7-H1 on macrophages. Based on our previous

observation that chitin induced recruitment of innate IL-4-producing effector cells 9, we would have expected to find more Th2 cells in LN and lung of OVA/chitin-challenged PLX-4720 mouse mice compared with controls which received OVA alone. However, the recruitment of eosinophils and basophils is a transient and rather late process that follows an earlier recruitment of neutrophils and macrophages which may in fact counteract the potential Th2-polarizing activity of eosinophils and basophils 9, 18. Although we have not addressed whether the transferred T cells acquire a Th1, Th17 or Treg phenotype, we clearly observed a reduced frequency of these cells in OVA/chitin-treated mice compared with controls. This finding is consistent with the in vitro experiments which demonstrated that chitin blocks T-cell proliferation indirectly by conditioning accessory cells for contact-dependent RVX-208 inhibition. These accessory cells can be macrophages, as we demonstrated by direct coculture of macrophages and sorted T cells, although other cell types may also contribute

to inhibition. The in vitro-cultured chitin-induced macrophages do not acquire an alternatively activated phenotype as they do not express Fizz1, a highly specific marker for AAM in mice 27, although they express low levels of Arg1, a gene that is generally associated with alternative activation but can also be induced by Stat6-independent signals 25. Chitin-exposed macrophages appeared to express higher levels of the inhibitory ligand B7-H1 as compared with glass- or PBS-treated macrophages. B7-H1 is expressed on many cell types, whereas expression of the closely related ligand B7-DC (PD-L2) is restricted to macrophages and DC 28. LPS, IFN, GM-CSF or IL-4 can upregulate B7-H1 on macrophages 29, 30. The potent inhibitory activity of B7-H1 against T cells has been demonstrated in autoimmune, infection and tumor models 31–33. B7-H1-deficient mice show spontaneous accumulation of activated CD8 T cells in the liver, suggesting a role for maintenance of immune tolerance under steady-state conditions 34.

Here, the ChAdV68 Gag alone and in combination

with other

Here, the ChAdV68.Gag alone and in combination

with other vectors elicited T cells capable of producing multiple intercellular signaling molecules and degranulation. While it is difficult to discern among the individual regimens in terms of the overall quality, responses after challenge appeared proportionally more polyfunctional relative to prechallenge. While inability of a vaccine to elicit polyfunctional T cells would likely result in “no-go” decision for further development and impaired T cells are not likely to control HIV-1 infection, T-cell polyfunctionality www.selleckchem.com/products/AZD2281(Olaparib).html during acute HIV-1 infection was not associated with selection of escape mutants Torin 1 molecular weight [49, 50]. Thus, in the absence of clear functional T-cell correlates of protection in humans, we showed that ChAdV68.GagB alone and in heterologous

combinations with plasmid DNA and recombinant MVA vaccines induced potent T-cell responses capable of decreasing virus loads of a surrogate EcoHIV/NDK challenge. These responses did so at their peak frequencies and 4 months later indicating development of effector memory T cells. Conferred immunity through development of protective T-cell memory together with the proven mucosal homing to the important makes ChAdVs highly attractive vectors for anti-HIV-1 vaccine development. Finally, the work presented here parallels similar vaccine studies in rhesus macaques [11, 19, 21] and a site of

HIV-1 replication phase I/IIa clinical trial in human volunteers (EUdraCT 2010–018439-16). Both in mouse here and rhesus macaque, the DNA-ChAdV-MVA regimen induced robust Tg-specific responses. In future when the human data are complete, this will allow to compare immunogenicity of similar vaccine regimens between mice, non-human primates, and humans, the three important species most commonly used in HIV-1 vaccine development for iterative, stepwise improvements 6-phosphogluconolactonase of vaccine designs. The WT isolate SAdV-25 was obtained from ATCC, propagated in HEK293 cells and purified by double CsCl gradient ultracentrifugation according to standard practice. Viral genomic DNA was isolated by phenol extraction. Based on the GenBank RefSeq for SAdV-25, PCR primers were designed for amplification of flanking regions for recombination-based cloning of the viral genome into a BAC vector, pBACe3.6, a method we have also applied to another chimpanzee [40]. Two full-genome clones were transferred into the SW102 strain for precise deletion of E1 and E3 by GalK recombineering [42] and a single nonfermenting colony from each original clone was amplified for verification by restriction mapping and the whole genome of one clone of E1- and E3-deleted ChAdV68-BAC was shotgun sequenced (Eurofins MWG Operon). ChAdV68.

Many TAA-specific T and B lymphocytes have been identified in can

Many TAA-specific T and B lymphocytes have been identified in cancer patients 4, 9, but these TAA-specific cells are often found in an unresponsive or anergised state. Moreover, tumours may also evade the immune system by interacting actively with host immune cells to block their functions 1, 8, 10. It has become a central question in tumour immunology as to how these TAA-specific clones are tolerated or suppressed, and whether they can

be re-activated to induce effective anti-tumour immunity 11, Vemurafenib clinical trial 12. The initial idea of DC-based tumour immunotherapy was prompted by the understanding that DC could be a potent antigen presenting cell (APC) for T-cell activation 11. Owing to their unique immunobiological properties, DC serve as a buy Palbociclib crucial link between the innate and adaptive immune systems. DC are widely distributed in various tissues and

organs throughout the body, and are very efficient in antigen uptake, processing and presentation 13. DC also constitutively express MHC class I and class II molecules, which can be highly up-regulated on mature or activated DC, and are able to present antigens effectively to both CD4+ (helper) and CD8+ (cytotoxic) T cells. Importantly, unlike tissue macrophages, DC naturally exhibit migratory properties. Upon uptake of antigens in the peripheral non-lymphoid tissues, DC migrate to the T-cell areas of secondary

lymphoid organs, where naïve T cells preferentially home to. In other words, DC are in PAK5 the position, and in theory the only cell type, capable of activating naïve T cells in vivo, and are thus crucial in the initiation of adaptive immune responses 14. These, together with the fact that DC or DC-like cells could be generated in vitro in large numbers 15–17, and readily loaded with either defined or even un-defined tumour antigens 18, led to the attractive concept of using DC therapeutically as an immunogenic cell vector for vaccine delivery 11, 19–23. Over the past two decades, the DC therapy has attracted intense interest in cancer research. Despite some favourable findings from studies in various experimental models, clinical application has thus far been limited by a lack of achievable general efficacy and consistency, and the outcomes from many clinical trials had not been met with initial expectations 24, 25. Indeed, since the early proof-of-concept studies in animal models reported nearly two decades ago 11, 19, 20, the promise remains to be delivered clinically. In a recent update by Gerold Schuler, current progress and several important issues regarding clinical applications of DC in cancer therapy have been discussed 26.

Indeed, their immunologically “innate” status needs to be questio

Indeed, their immunologically “innate” status needs to be questioned if NK cells are incapable of independently responding to PfRBC in the absence of “adaptive” T cells. However, the insights presented do provide encouraging implications for malaria vaccine development, since they suggest that by inducing classical memory Roscovitine in vitro T-cell responses vaccination will simultaneously achieve enhanced NK responses “into the bargain”. Protocols for and clinical course of stringently controlled experimental human malaria infections at our centre have been described in detail earlier 12, 13. Briefly, after providing written informed consent, five healthy malaria-naïve Dutch volunteers were infected with malaria by exposure

to the bites of five P. falciparum-infected mosquitoes and followed-up closely for symptoms and signs of malaria. As soon as a standard microscopic thick smear of peripheral blood became positive for malaria parasites, volunteers were treated with a standard curative regimen of the anti-malarial drug artemether–lumefantrine. The study

was approved by the Institutional Review Board of the Radboud University Nijmegen Medical Centre (CMO 2006/207). Preparations of mature parasitized RBC (PfRBC) and mock-cultured uninfected erythrocytes (uRBC) were obtained by routine methods as described previously 12 and cryopreserved at 150×106/mL in 15% glycerol/PBS in aliquots for use in stimulation assays. Cryopreserved PfRBC form almost as strong a stimulus as fresh PfRBC and have identical stimulatory characteristics drug discovery (Supporting Information Fig. 2). Their use in large experiments has logistical advantages, in addition to reducing confounding due

to inter-batch variation. One single large batch of cryopreserved PfRBC was used for the entire follow-up study described above. Venous whole blood was collected into citrated CPT vacutainers (Becton and Dickinson, Basel, Switzerland) prior to challenge (day C−1), during blood-stage malaria infection (day C+9), 3 wk after treatment (day C+35) and again 20 wk after challenge (day C+140). PBMC were obtained by density gradient centrifugation, washed 3× in cold PBS, enumerated, frozen down in 10% DMSO/FBS and stored in liquid nitrogen. Immediately prior to use, cells were thawed, washed twice in RPMI and resuspended in complete culture medium (RPMI 1640 containing click here 2 mM glutamine, 1 mM pyruvate, 50 μg/mL gentamycine and 10% v/v human A+ serum, Sanquin, Nijmegen) for a final concentration of 2.5×106/mL. PBMC were transferred into 96-well round-bottom plates and were stimulated in duplo wells with either 5×106/mL cryopreserved PfRBC or uRBC. PBMC were stimulated for 24 h at 37°C/5%CO2; 4 h prior to cell harvest, 100 μL/well supernatant was collected and stored at −80°C for subsequent cytokine measurement and replaced with 100 μL/well fresh culture medium containing brefeldin A (Sigma) for a final concentration of 10 μg/mL.