ELISA antibody pairs were used to detect IL-12p70 levels (R&D Bio

ELISA antibody pairs were used to detect IL-12p70 levels (R&D Biosystems) and IL-23 levels (R&D Biosystems) in supernatants. PGE2 quantitative ELISA was performed in DC culture supernatants according to the manufacturer’s protocol (R&D Biosystems) and serum PGE2 levels determined using DetectX PGE2 kit validated

for mouse serum (Arbor Assays). RNA was extracted as described previously, reverse transcribed, and amplified using FAM-labeled LY2157299 chemical structure probe and primers on the ABI Prism 7900 detection system 23. Fold increase in signal over that derived from control samples was determined using the ΔΔct calculation. In some cases, the levels of mRNA relative

to housekeeping gene (GAPDH) were calculated. The primer and probes sequences have been published previously 23 or were commercially purchased (ABI Biosystems). Single-cell suspensions were stained with fluorochrome-labeled antibodies specific for CD3 (17A2), CD4 (RM4-5), and Trametinib CD8 (53–6.7). Intracellular staining was performed by using anti-IFN-γ (XMG1.2) on cells stimulated with PMA and ionomycin as per the method described 12. To sort for a purified DLN cell population, stained cells were sorted on BD FACS Aria flow cytometer as CD3+ CD4+ (purity, >94%). For analysis, FlowJo (Tree Star, CA) was used. Differences between the means of groups were analyzed using the two-tailed Tacrolimus (FK506) Student’s t-test in GraphPad Prism 5 (La Jolla, CA). Inherently, logarithmic data from bacterial growth and RT-PCR were transformed for statistical analyses. This work was supported by funds from Children’s Hospital of Pittsburgh, to S. A. K., Research Advisory Committee Grant from Children’s Hospital of Pittsburgh of the University

of Pittsburgh Medical Center Health System to Y. L. and S. S., a UICC American Cancer Society Beginning Investigators Fellowship funded by the American Cancer Society to NO. Grants from National Institute of Heath, USA – A1083541, HL105427-01 to S. A. K. and CA132714 to P. K. The authors thank Dr. A. Cooper, Trudeau Institute for providing M. bovis BCG stocks, Dr. N. Ghilardi, Genentech Inc, for providing il23p19−/− mice. il17ra−/− mice were a kind gift from Amgen Inc. The authors thank Dr. J. Kolls, Dr. T. Darville, Dr. S. Gaffen, and Dr. J. Alcorn for critical reading of the manuscript. Conflict of nterest: The authors declare no financial or commercial conflict of interest. Detailed facts of importance to specialist readers are published as ”Supporting Information”. Such documents are peer-reviewed, but not copy-edited or typeset. They are made available as submitted by the authors.

The ability of the DNA vaccine constructs to elicit cellular immu

The ability of the DNA vaccine constructs to elicit cellular immune responses makes them an attractive weapon as a safer vaccine candidate for preventive and therapeutic applications against tuberculosis. Tuberculosis (TB) is a major local, regional and global infectious disease problem with about 9 million new cases and

2 million deaths every year [1]. Mycobacterium tuberculosis kills more adults each year than any other single pathogen. The vaccination with Mycobacterium bovis bacille Calmette Guerin (BCG) is considered to be the most important tool to protect against TB [2]. In spite of its widespread use and many advantages like being inexpensive, safe at birth, given as a single shot and provision of some protection against leprosy, BCG vaccination remains controversial [2–4]. EPZ-6438 solubility dmso The protection afforded by BCG vaccination has shown wide variations in different parts of the world, and its impact on the global problem of TB remains unclear [5]. Estimates of protection given by BCG against pulmonary TB vary greatly [4]. For example, a trial in British school children, in 1952, showed about 80% efficacy, whereas the Chingleput trial in India showed zero efficacy

of protection against adult pulmonary BMN 673 supplier TB, after BCG vaccination [4, 6]. This variability has been attributed to various factors including strain variation in BCG preparations, environmental influences such as sunlight exposure, poor cold-chain maintenance, genetic or nutritional differences between populations and exposure Protein kinase N1 to environmental mycobacterial infections etc. [5]. In addition, because of sharing most of the antigens, BCG vaccination induces a delayed-type hypersensitivity skin response to the purified protein derivative of M. tuberculosis (the stimulus used to test the individuals for tuberculous infection), which cannot be distinguished from exposure to M. tuberculosis [7]. This makes the use

of tuberculin skin test difficult for diagnostic or epidemiological purposes. Furthermore, BCG vaccination cannot be used in all groups of people, e.g. WHO has recommended that children with symptoms of HIV or AIDS should receive all the vaccines except BCG. This is because BCG is a live attenuated vaccine that might cause disease in immuno-compromised people rather than giving immunity [8]. Thus, there is an urgent need to develop M. tuberculosis-specific and safer vaccines against TB [6, 9]. The development of a better BCG vaccine or alternative vaccines needs the identification and evaluation of antigens recognized by protective immune responses [9]. In previous studies, we have identified RD1 PE35 (Rv3872), PPE68 (Rv3873), EsxA (Rv3874), EsxB (Rv3875) and RD9 EsxV (Rv3619c) as M. tuberculosis-specific antigens [10–13]. Furthermore, in vitro studies in patients with TB and healthy subjects infected with M. tuberculosis have shown that these antigens induced cellular immune responses that correlate with protection [9].

We report here that B lymphocytes from SLE-afflicted mice express

We report here that B lymphocytes from SLE-afflicted mice express relatively elevated levels of CD74, compared with B cells

from healthy mice. CD74 is a receptor found in complex with CD44, and it binds the pro-inflammatory cytokine MIF. The latter components were also up-regulated in B cells from the diseased mice, and treatment with hCDR1 resulted in their down-regulation and in reduced B-cell survival. Furthermore, up-regulation of CD74 and Venetoclax concentration CD44 expression was detected in brain hippocampi and kidneys, two target organs in SLE. Treatment with hCDR1 diminished the expression of those molecules to the levels determined for young healthy mice. These results suggest that the CD74/MIF pathway plays an important role in lupus pathology. Systemic lupus

erythematosus (SLE) is an autoimmune disease characterized by impaired B-cell and T-cell functions and it is associated with serological and clinical manifestations that involve multiple organ systems.1 Because B and T cells play a pivotal role in SLE pathogenesis, successful treatment strategies for the disease should optimally target both cell types. For a specific treatment of SLE, a peptide designated hCDR1,2 which is based on the sequence of the complementarity-determining region (CDR) -1 of an autoantibody,3 was designed and shown to ameliorate lupus manifestations in both spontaneous and induced models of SLE.4,5 The mechanisms underlying the beneficial effects of hCDR1 are manifested through the induction of CD4+ CD25+6 MK-1775 chemical structure and CD8+ CD28−7 regulatory T cells, immunomodulation of cytokines,4 Ribose-5-phosphate isomerase apoptosis8 and induction of regulatory molecules.9–11 Serologically, SLE is characterized by the presence of high titres of autoantibodies and abnormal B-cell activation and differentiation.12 The regulation of mature B-cell survival involves multiple mechanisms. The B-cell receptor provides survival

signals essential for maintaining the mature B-cell pool. In addition, the B-cell activating factor (BAFF) is required for successful survival and maturation of splenic B cells.13 We demonstrated that BAFF, which was found to be elevated in sera from patients with SLE and lupus-prone mice,14,15 was down-regulated following treatment with hCDR1 in SLE-afflicted mice.16 Recently, we described an additional mechanism that regulates B-cell survival, which depends on CD74 (the cell surface form of invariant chain, li).17–19 CD74 is a type II integral membrane protein containing a transmembrane region and a luminal domain that functions as a MHC class II chaperone.20 Part of the CD74 molecule, modified by the addition of chondroitin sulphate, is expressed on antigen-presenting cells, monocytes and B cells, and interacts with CD44.21,22 Macrophage migration inhibitory factor (MIF) binds to the CD74 extracellular domain on macrophages, consequently initiating a signalling pathway.

These three emm genotypes are frequently isolated in clinical pra

These three emm genotypes are frequently isolated in clinical practice. Sequencing of the csrRS gene, one of the two-component signal transduction systems implicated in virulence, was performed on 25 strains bearing different amounts of M protein. CsrS mutations, in contrast to CsrR protein, were detected in 11 strains. The M protein-high producer strain of emm1 type carried two amino acid substitutions, whereas the other

three emm1 strains carried only one substitution each. The M protein-high producer expressed its emm gene more strongly than the corresponding M protein-low producer did according to TaqMan RT-PCR. These observations suggest that the accumulation of amino acid substitutions in CsrS protein may contribute, at least in part, to the large amount of M protein production seen in selleck chemicals several emm genotypes. Streptococcus pyogenes is an important human pathogen with several different clinical Selleck JNK inhibitor manifestations. Pharyngitis among school-age children is one of the most common conditions caused by S. pyogenes. In addition, S. pyogenes has been responsible for severe invasive diseases such as sepsis and STSS throughout the world, particularly during the last 20 years (1). Streptococcus

pyogenes produces a virulence determinant, known as M protein, which occurs on the cell surface and has a dimeric alpha-helical coiled-coil structure. Since identification of the species by Rebecca Lancefield, this protein has possibly been one of the best-studied molecules among the known streptococcal virulence determinants. Over the last few decades, possible roles suggested for M protein in streptococcal infection have included: (i) effecting an antiphagocytic function against human neutrophils (2, 3); (ii) promoting of adhesion to, and invasion into, epithelial cells (4);

(iii) enabling size variation of the N-terminal region for the purpose of escaping recognition by human antibodies (5, 6); and (iv) forming, through biological reactions, a complex with fibrinogen which triggers vascular leakage (7). Though the role of the M protein as an important virulence factor in S. pyogenes has already been thoroughly characterized, no quantitative assay of clinical isolates has been performed to date. Yet the information that could be provided by this kind of analysis is critical: recent reports have demonstrated that S. pyogenes strains express a number of virulence-related determinants more abundantly after in-vivo passage (8–10), suggesting that quantitative measurement of M protein is essential to our understanding of the mechanisms underlying severe streptococcal infection. Here, we performed a quantitative assay of M protein in 141 field isolates with various emm genotypes and assessed the relationship between the amount of M protein and CsrRS proteins, which have been reported to be involved in the expression of many virulence factors of S.

Failure to mount this protective Th2 response exacerbates infecti

Failure to mount this protective Th2 response exacerbates infection (11,12). Leishmania spp. are obligate intracellular parasites that cause a wide range of diseases such as cutaneous, mucocutaneous

and visceral leishmaniasis and worldwide an estimated 12 million people are infected (13). The murine model of cutaneous L. major infection has been well characterized and results in a localized cutaneous lesion whose resolution depends on the development of IL-12-induced Th1 response and production of IFN-γ. Initiation of a Th2-type response, characterized by the production of IL-4 and IL-10 as found Ridaforolimus cost in susceptible BALB/c mice, in contrast, is associated with the development of large non-healing lesions after L. major infection (14–17).

As Th1 and Th2 responses are counterregulatory, we investigated the interaction of these two parasites in vivo by co-infecting C57BL/6 mice with S. ratti and L. major and comparing disease progression, parasite-specific humoral as well as cellular immune response in the lymph nodes (LN) draining the sites of infection. We show that concurrent S. ratti infection did not interfere with the efficient control of L. major infection in C57BL/6 mice. Also, the Th2 response induced by S. ratti infection did not alter the Th1 biased responses to L. major. In contrast, the Th1 response induced click here by L. major resulted in partial suppression of S. ratti-induced Th2 response in the mesenteric LN draining the gut. Control Sulfite dehydrogenase of S. ratti infection, however, was not significantly impaired. Taken together, co-existence of the two parasites within the same host modulated the immune response to each species to a certain degree without affecting parasite clearance. All in vivo experiments were carried out at the animal facility of the Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, with permission of the Federal Health Authorities of the state of Hamburg, Germany. Female C57BL/6 mice were obtained from the University Hospital Eppendorf, and wistar rats were purchased from

Charles River (Sulzfeld, Germany). Animals were kept in individually ventilated cages and used at the age of 8–12 weeks (mice) or 4–8 weeks (rats). The S. ratti life cycle was kindly provided by Dr. Utzinger (Swiss Tropical Institute) and maintained by serial passage of S. ratti through wistar rats. iL3 of S. ratti were purified from charcoal faeces cultures as described before (5). Prior to infection, iL3 were stored overnight in PBS supplemented with penicillin (100 U/mL) and streptomycin (100 μg/mL). Strongyloides antigen lysate was prepared as described (10). The cloned virulent L. major isolate (MHOM/IL/81/FE/BNI) was propagated in vitro in blood agar cultures as described previously (18). To prepare L. major parasites for infection experiments, stationary phase promastigotes from the third to seventh in vitro passage were harvested, washed four times and resuspended in sterile PBS.

1 This encapsulated yeast is also able to persist in healthy host

1 This encapsulated yeast is also able to persist in healthy hosts, thus causing dormant infections that may later be reactivated under an immunosuppressive disease.2 Cryptococcal infections in rats have been shown to have similarities with human cryptococcosis,

revealing a strong granulomatous response and a low susceptibility to disseminated infections.3 T-cell-mediated immunity is a critical component of protective immunity against infection with C. neoformans. Both CD4+ and CD8+ T cells are required for effective immune pulmonary clearance and prevention of extrapulmonary dissemination.4 The cells recruited during the inflammatory response include neutrophils, eosinophils, Selleck FK228 monocyte/macrophages (Mφ), dendritic cells and lymphocytes [CD4+ T cells, CD8+ T cells, B cells

and natural killer (NK) cells]. Of these cells, activated Mφ, neutrophils and lymphocytes are all capable of in vitro killing or growth inhibition of C. neoformans.5 Related to this, previous studies in our laboratory have shown that Mφ from infected rats appear to be able to kill C. neoformans, principally by generating nitric oxide (NO).6 Moreover, the Proteasome activity NADPH oxidase system was also found to be very important in the mechanism of C. neoformans killing by rat peritoneal cells, with the superoxide anion, hydrogen peroxide (H2O2) Amylase and the hydroxyl radical being involved in this process.7 Eosinophils,

in contrast, are implicated as effector cells in helminthic infections, releasing their many cytoplasmic granules, containing toxic molecules, in response to antigenic stimuli.8 Moreover, they notably contribute to allergic inflammation at airway mucosal sites.9 Recent studies have also demonstrated that eosinophils are able to function as antigen-presenting cells (APCs). The eosinophils express major histocompatibility complex (MHC) class I and class II, and the costimulatory molecules CD28, CD40, CD80 and CD86, suggesting that these cells can directly communicate with T cells to regulate immune responses. In addition, eosinophils also secrete a range of cytokines that are not only proinflammatory, but also function as growth factors, stimulants and chemoattractants [e.g. interleukin (IL)-2, IL-4, IL-5, IL-10, IL-12, IL-16, interferon-γ (IFN-γ) and regulated on activation, normal, T-cell expressed, and secreted (RANTES)] for T cells.10 In this sense, eosinophils were demonstrated to present antigens to primed T cells, thus increasing T-helper 2 (Th2) cytokine production.10–14 Furthermore, antigen-loaded eosinophils migrate into local lymph nodes and localize in the T-cell-rich paracortical zones, where they stimulate the expansion of CD4+ T cells.

CRP is a specific but not sensitive marker in the early stages of

CRP is a specific but not sensitive marker in the early stages of neonatal sepsis, while the WBC count appears to be unreliable [4, 5]. The neonatal immune response, however, includes increased production of other inflammatory mediators, the assessment of which may improve diagnostic accuracy in suspected sepsis [2, 6]. Cytokines are endogenous chemical mediators that play an important role in the

inflammatory cascade. They participate in the development of both innate (natural) and adaptive immunity. Interleukin-1 (IL-1), IL-6 and TNF-α are interleukins that have been tested in neonatal sepsis as indices that could increase the accuracy of its diagnosis [7–10]. The mortality and morbidity of patients Roxadustat with sepsis is influenced by a dysregulation of the immune response to the infection, and for this reason, research

efforts into sepsis have been learn more focussed on immune mechanisms. Studies in adults with sepsis have shown considerable changes in the subsets of lymphocytes, and especially in the T-helper cells, B cells and natural killer (NK) cells [11–13]. There are indications of a special role of NK cells as a component of the innate immune system [11]. It is known that the defence of neonates is initially dependent on innate immunity, as antigen-specific immunity develops later in life. Little data are available on these factors in infected neonates, while reference values for healthy neonates at various Immune system chronological ages have not been fully established. This study was designed to investigate certain factors of the immune system in full-term neonates with

sepsis and in healthy control subjects, to evaluate possible changes in levels of these factors during the course of neonatal sepsis. The study included 95 full-term neonates born in the regional hospital during the same period, classified into three groups, matched for chronological age and sex. Neonates were included in the sepsis group (n = 25) when sepsis was confirmed by a positive blood culture accompanied by compatible signs and symptoms. Neonates with signs and symptoms of infection, but whose blood cultures were negative, comprised the group with suspected infection (n = 20). For matching purposes, for each neonate with sepsis, the next neonate admitted with suspected infection and of the same chronological age and sex was recruited. The control group comprised 50 healthy neonates without clinical findings or maternal risk factors for infection admitted to the neonatal intensive care unit (NICU) for minor problems or nursed in the neonatal ward.

This achieved a Teff- to Treg-cell ratio of 5:1, a dose that we h

This achieved a Teff- to Treg-cell ratio of 5:1, a dose that we have previously used to detect functional differences between allospecific Treg-cell lines [28, 30]. Animals were monitored for cGVHD symptoms for 7 weeks as described earlier. At the experimental endpoint, find more all Treg cells were found to have significantly inhibited cGVHD-induced splenomegaly (Fig. 3A). This was associated

with a significant reduction in both the donor cell compartment (Fig. 3B and C) (cGVHD 8.6 ± 4.1% H-2Kd− cells of total splenocytes versus auto-Treg cells 2.8 ± 2.3%, indirect Treg cells 3.8 ± 1.8% and direct Treg cells 1.8 ± 1.2%) and recipient T-cell composition of the spleen (Fig. 3C and D) (cGVHD 29.9 ± 14.6% CD4+H-2Kd+ cells of total splenocytes versus auto-Treg cells 20.3 ± 0.5%, indirect Treg cells 20.2 ± 2%, direct Treg cells 17.6 ± 1.6% and PBS control animals 20.2 ± 0.9%). This suggests that recipient lymphocyte hyperactivity induced by alloreactive Kinase Inhibitor Library cell line donor cells had also been efficiently controlled by Treg-cell transfer. Of importance, auto-Treg cells completely prevented donor cell engraftment, with no residual donor T cells being

detectable within the splenic tissue of any treated animals, however indirect- and direct allospecific Treg cells were able to permit donor-derived T-cell engraftment (cGVHD 3.8 ± 1.6% CD4+H-2Kd− cells of total splenocytes versus indirect Treg cells 0.5 ± 0.4% and direct Treg cells 0.2 ± 0.2%), demonstrating that cGVHD disease could be fully prevented by allospecific Treg cells despite the sustained engraftment of donor cells. This suggests that while auto-Treg cells may have suppressed initial donor

T-cell proliferation and engraftment, allospecific Treg cells may have specifically regulated donor alloreactive T-cell proliferation. As serum autoantibodies are indicative of SLE-type immunopatholgy, sera from cGVHD and Treg-treated animals were screened for both Th1 (IgG2a) and Th2 (IgE, IgG1) associated immunoglobulins. Elevated levels of both IgG autoantibodies and IgE induced by cGVHD after 7 weeks were significantly and equally inhibited by all Treg-cell Sodium butyrate lines (Fig. 4A–C), resulting in autoantibody levels similar to PBS controls (PBS versus Treg-treated groups: ns). Co-transfer of Treg cells further abrogated IgG immune complex deposition within kidney glomeruli (Fig. 4D and E). Our finding that Treg cells with autoreactivity, indirect or direct allospecificity were all able to prevent cGVHD immune pathology, suggests that although a combination of dysregulated autoimmune reactivity and alloimmunity plays a critical role in the progression of cGVHD, inhibition of either can control disease progression.

[27] consistent with a role for phagocytosis in the disappearance

[27] consistent with a role for phagocytosis in the disappearance of virion–IgG complexes in Fiebig Stage IV.[27] This hypothesis is supported by the finding that phagocytosis by both monocytes and dendritic cells is increased in acute

infection and impaired in chronic infection.[27] The impairment in chronic infection was tightly associated with down-regulation of FcγR2a and FcγR3a on monocytes and dendritic cells.[27] The expansion of circulating natural killer cells expressing FcγR3 in Fiebig Stages II and III,[56] immediately before selleck or at the beginning of seroconversion, suggests that ADCC responses might occur concomitant with emergence of free IgG antibodies to gp41 and gp120. The involvement of Fc-mediated effector function before Fiebig Stage V where ADCC responses are first detectable[24, 26] is hypothetical and based on indirect indications. This hypothesis can be tested readily with infection R428 mw models in NHPs where effector cells and antibodies can

be quantified at defined times post-infection. Despite the uncertainty about the role of Fc-mediated effector function in acute infection, a large body of data has accumulated over the years demonstrating correlations between clinical outcome and ADCC titres in HIV-infected individuals. These studies are summarized in Table 1. The earliest report of a correlation between ADCC titres and clinical stage appeared in 1987[57] and studies with similar conclusions continue to appear PI3K inhibitor up to the time of writing.[58] Of the 19 studies listed in Table 1, three failed to detect correlations between ADCC and clinical outcomes whereas the other 16 reported correlations between ADCC and positive clinical outcomes. Further, the negative studies were in the early years of the epidemic when methodology

was more challenging. The 15 positive studies, spanning 26 years and involving different cohorts and methods, provide compelling support for the involvement of Fc-mediated effector function, particularly ADCC, and post-infection control of HIV. This conclusion is supported also by similar studies in NHPs, although they are fewer in number. The first NHP study, which appeared in 2002, reported an inverse correlation between ADCC titres and progression to simian AIDS in the simian immunodeficiency virus model of infection.[59] A second study appeared in 2011 and reported similar conclusions in the same model.[60] A third study reported an inverse correlation between another Fc-mediated effector function, antibody-dependent cellular viral inhibition (ADCVI),[24, 61] which has elements similar to ADCC, and viral control.[62] Collectively, studies in both HIV-infected individuals and simian immunodeficiency virus-infected rhesus macaques strongly support a role for Fc-mediated effector function, and ADCC in particular, in post-infection control of viraemia.

The authors declare no conflict of interest “
“Bile acids (

The authors declare no conflict of interest. “
“Bile acids (BAs) play important roles not only in lipid metabolism, but also in signal transduction. TGR5, a transmembrane receptor of BAs, is an immunomodulative factor, but its detailed mechanism remains unclear. Here, we aimed to delineate how BAs operate in immunological responses via the TGR5 pathway in human mononuclear cell lineages. We examined TGR5 expression in human peripheral blood monocytes, several types of in vitro differentiated macrophages (Mϕs) and dendritic cells. Mϕs differentiated with macrophage colony-stimulating factor and interferon-γ (Mγ-Mϕs), which are similar to the human intestinal lamina propria CD14+ Mϕs that contribute

to Crohn’s disease (CD) pathogenesis by production of pro-inflammatory cytokines, highly expressed TGR5 compared with any other type of differentiated Mϕ and dendritic cells. We also showed that a TGR5 agonist and

two types of BAs, Lorlatinib deoxycholic acid and lithocholic acid, could inhibit tumour necrosis factor-α production in Mγ-Mϕs stimulated by commensal bacterial antigen or lipopolysaccharide. This inhibitory effect was mediated by the TGR5–cAMP pathway to induce phosphorylation of c-Fos that regulated nuclear factor-κB p65 activation. Next, we analysed TGR5 levels in lamina propria mononuclear cells (LPMCs) obtained from the intestinal mucosa of patients with CD. Compared with non-inflammatory bowel disease, inflamed CD LPMCs contained more TGR5 transcripts. Among LPMCs, FK228 solubility dmso isolated CD14+

intestinal Mϕs from patients with CD expressed TGR5. Anacetrapib In isolated intestinal CD14+ Mϕs, a TGR5 agonist could inhibit tumour necrosis factor-α production. These results indicate that TGR5 signalling may have the potential to modulate immune responses in inflammatory bowel disease. “
“Both iron-deficient anemia (IDA) and malaria remain a threat to children in developing countries. Children with IDA are resistant to malaria, but the reasons for this are unknown. In this study, we addressed the mechanisms underlying the protection against malaria observed in IDA individuals using a rodent malaria parasite, Plasmodium yoelii (Py). We showed that the intra-erythrocytic proliferation and amplification of Py parasites were not suppressed in IDA erythrocytes and immune responses specific for Py parasites were not enhanced in IDA mice. We also found that parasitized IDA cells were more susceptible to engulfment by phagocytes in vitro than control cells, resulting in rapid clearance of parasitized cells and that protection of IDA mice from malaria was abrogated by inhibiting phagocytosis. One possible reason for this rapid clearance might be increased exposure of phosphatidylserine at the outer leaflet of parasitized IDA erythrocytes. The results of this study suggest that parasitized IDA erythrocytes are eliminated by phagocytic cells, which sense alterations in the membrane structure of parasitized IDA erythrocytes.