[89] The pathogenesis and mechanisms

[89] The pathogenesis and mechanisms mTOR inhibitor involved in vertical transmission are still not completely understood. HCMV spreads from the infected mother’s decidual cells to the fetus. Sites

of viral replication include cytotrophoblast progenitor cells in chorionic villi and differentiating/invading cytotrophoblasts.[90] Until recently, the role of dNK cells in controlling viral infection was not known. However, epidemiological studies indicate that the rate of congenital HCMV infection is often low in the first trimester of pregnancy, which coincides with high numbers of dNK cells within the decidua, which suggests that dNK cells might be involved in protection against congenital HCMV infection. Decidual NK cells express all the receptors involved MK-8669 research buy in the response to HCMV and they also contain the necessary arsenal for cell cytotoxicity (Fig. 2). In a recent work, we provided the first evidence for the involvement of dNK cells in the response against congenital HCMV infection (see Fig. 3 for visual summary).

Interestingly, dNK cells can be found in the vicinity of infected cells within floating chorionic villi, suggesting that the functional plasticity of dNK cells in response to invading pathogens is associated with modulation of their migratory phenotype.[91] Deciual NK cells respond to congenital HCMV infection by lowering the secretion of several soluble factors (CCL2, CCL4, CCL5, CXCL10, granulocyte–macrophage colony-stimulating factor and CXCL8) that are involved in trophoblast invasion. By interfering with trophoblast

invasion, dNK cells can participate actively in limiting viral spreading and congenital infection. Along the same lines, such changes within the microenvironment itself will not only limit trophoblast invasion but also induce inappropriate activation of other immune cells namely dendritic cells and T cells. The ability to cross the placental barrier is one key determinant of invasive viruses and pathogens (hepatitis viruses, HIV, Plasmodium). Montelukast Sodium Yet little is known about mechanisms underlying the fetal placenta tropism and the ability of dNK cells in the defence against these agents. Recent studies demonstrated that under certain conditions NK cells isolated from non-pregnant uterine mucosa and soluble factors secreted by decidual cells can control X4-tropic HIV-1 infection.[92, 93] Hence, it is conceivable that uterine NK and decidual NK cells act as local guardians against infection and their immune modulation might ensure efficient anti-viral protection. During the first trimester of pregnancy dNK cells display unique phenotypic and functional properties that distinguish them from other peripheral blood or tissue NK cells. They orchestrate fetal trophoblast invasion and placental vasculature remodelling, which are necessary for the maintenance of a healthy pregnancy.

Upon the autopsy, pigs were extubated and tubes were stored

Upon the autopsy, pigs were extubated and tubes were stored OTX015 at −80 °C for subsequent analysis. Then, to prevent any disruption of the biomatrix and impairment of bacterial viability, prior microscopic analyses, the ETTs were slowly unfrozen up to room temperature. The ETT exterior surface was cleaned with sterile gauzes and decontaminated through careful rinsing with 80% alcohol and saline solution. Using strict aseptic technique, two 1-cm-long sections of the distal dependent part of the ETT were excised

(Fig. 1). A 1-cm cross-section of ETT was immersed in a 1 mL phosphate buffer solution (PBS), stained with live/dead® BacLight kit™ (BacLight kit™; Invitrogen, Barcelona, Spain) for 15 min protected from the light, and then rinsed with PBS. The staining conditions were as follows: 1.5 μL of SYTO® 9 (stock 3.34 mM DMSO) and 1.5 μL propidium Apoptosis Compound Library high throughput iodide (stock 20 mM DMSO) in 1 mL PBS. During CLSM imaging, SYTO® 9 emits green fluorescence and is used to identify living microorganisms with intact membrane whereas propidium iodide (PI) emits red fluorescence and stains dead bacteria with damaged membrane. A Leica TCS SP5 laser scanning confocal system (Leica Microsystems Heidelberg GmbH, Manheim, Germany) equipped with a DMI6000 inverted microscope and a 20xPL APO numerical

aperture 0.7 objective were used. SYTO® 9 and PI images were acquired sequentially using 488-, 561-nm laser lines, an acousto optical beam splitter and emission detection ranges 500–550, FXR agonist 570–620 nm, respectively. The confocal pinhole set at 1 Airy units. Pixel size was 160 nm. All samples and slides were coded to ensure that the image acquisition and measurements were blinded. The first author and an experienced CLSM facility manager made all observations and pictures. We analyzed 127 CLSM images (69 for the control, 37 for the linezolid, and 21 for the vancomycin group). Biofilm

viability was computed using image j software (Wayne Rasband, NIH). Regions of interest of each image were drawn by the operator to select all bacterial aggregates and exclude areas of eukaryotic cells; selection of these regions was based on cell size, morphology, and overall consistency of these factors within the area. Then, to select and independently measure the areas of live and dead bacteria, threshold limits were set for SYTO® 9 and PI channels, respectively. Only thresholded pixels were included in area measurements. For each image, we measured total area of bacteria (comprising live and dead bacteria), area of live bacteria (green), and dead bacteria (red) to evaluate differences in bacterial presence and viability among groups of treatment. We quantified the ratio between the total area of bacteria and the area of image examined, expressed as percentage. The live/dead bacterial ratio was calculated as the ratio between the area of live bacteria and the area of dead bacteria.

Analysis of the repertoire and characteristics of Th1 enhancers i

Analysis of the repertoire and characteristics of Th1 enhancers in the absence of STAT1 or STAT4 revealed these interleukin-12 (IL-12) and interferon-γ cytokine receptor-activated ERFs to be required for almost 60% of Th1 enhancer activation. Notably, while TBET regulated the expression of a number

of Th1 genes, the levels of p300 at associated enhancers were largely independent of TBET. However, 17% of Th1 enhancer activation (p300 recruitment) was dependent on TBET. These data raise interesting questions about TBET’s mechanism of action at target this website regulatory DNA. Elegant studies from Weinmann and colleagues have demonstrated the potential for TBET to act through at least two separable mechanisms mapped to distinct protein domains – recruitment of an H3K4me2 methyltransferase and direct transactivation.[32] Therefore, it will be interesting to determine if those few Th1 enhancers that require TBET for activation rely primarily on the chromatin-modifying potential of TBET, whereas the genes whose expression is augmented by TBET, independent of extensive modification of enhancer characteristics,

rely more heavily on the transactivation domain and increased recruitment of the general transcription machinery. As in Th1 cells, it appears that Th2 cell enhancer activation is heavily reliant on ERFs, namely Inhibitor Library price STAT6 downstream of IL-4R signalling. STAT6 was required for the activation of 77% of all Th2-specific enhancers.[13] Although, like TBET, GATA3 plays a minor role in enhancer activation, when over-expressed, it is sufficient for enhancer activation at about half of STAT6-dependent enhancers. In this context, it is interesting

to consider potential GATA3 dosage effects in chromatin regulation and target gene expression, and the possibility for GATA3 to function as a ‘pioneer’-like factor in some settings. In fact, during early T-cell development, GATA3 and PU.1 binding can precede full enhancer activation and gene expression in developing Silibinin thymocytes.[33] However, during the initial events of Th cell polarization, GATA3 and TBET play a less substantial role in nucleating chromatin alterations, activating enhancers, and influencing gene expression compared with STATs. Although representing a minority, it will be interesting to better understand the enhancers and genes dependent on MRFs for activation, both in terms of their potentially distinct chromatin characteristics and functional roles. Considering the relative function of ERFs and MRFs in Th cell differentiation, a study from Littman and colleagues thoroughly explored the transcriptional programme of Th17 cells as defined by five key transcription factors: basic leucine zipper transcription factor (BATF), IRF4, STAT3, cellular musculoaponeurotic fibrosarcoma oncogene homolog (cMAF) and RORγt.

For example, T-bet, the transcription factor that controls IFN-γ

For example, T-bet, the transcription factor that controls IFN-γ production,[42] is expressed by the majority of iNKT cells. Most of the liver and spleen iNKT cells that are Th1-like express T-bet, are NK1.1+ and produce IFN-γ. The iNKT cells can also express Gata3, which is a major transcription factor involved in inducing Th2 cytokines, especially IL-4, and in suppressing Th1 responses.[43] T helper type 2-like iNKT cells express IL-17RB, CD4 and Gata3, and mainly produce IL-13 and Th2 cytokines after stimulation with IL-25.[44] However, iNKT cells can simultaneously produce both IFN-γ and IL-4, and can express both T-bet and Gata3. Therefore the ‘master-regulator’ concept

in which cells express particular transcription factors mTOR inhibitor that control their Th1 or Th2 polarization is more complicated with iNKT cells, which can be both Th1 and Th2 producers simultaneously. There is also a population of IL-17RB+ iNKT cells that do not express CD4 and primarily produce

IL-17 due to their expression of the transcription factor RORγT. These Th17 iNKT cells respond to IL-23 and represent a distinct population in the thymus, and are enriched in lung and skin.[41] Other functional differences have been described for iNKT cells based on location. Adoptive transfer of hepatic iNKT cells mediates BYL719 tumour rejection, whereas thymus-derived iNKT cells do not. Furthermore, IMP dehydrogenase this anti-tumour function is unique to hepatic CD4− iNKT cells.[45] These studies emphasize the importance of considering the iNKT cell source and phenotype when studying iNKT cells. Invariant NKT cells resident in adipose tissue have a unique phenotype in terms of surface marker expression and function. While the majority of iNKT cells in the periphery are CD4 and have up-regulated NK1.1, adipose iNKT cells are mainly CD4− and a large proportion of adipose iNKT do not express NK1.1.[3,

7] This could imply that adipose iNKT cells are more immature than iNKT cells in liver and spleen and have yet to up-regulate NK1.1. It could also suggest that adipose iNKT cells are constitutively activated, as NK1.1 is transiently down-regulated following activation.[46] The lack of NK1.1 on many adipose iNKT cells also highlights the need to use CD1d-αGalCer tetramers to identify and study adipose iNKT cells, rather than the earlier and less specific method using CD3+ NK1.1+ markers. Adipose iNKT cells have a different cytokine profile compared with iNKT elsewhere. Although adipose iNKT cells express T-bet (L. Lynch & M. Brenner, unpublished data) and are capable of producing IFN-γ when stimulated with potent activators like PMA and Ionomycin they produce significantly less IFN-γ than iNKT cells elsewhere when activated with lipid antigens.[3] They also produce more IL-4 and IL-13 than splenic iNKT cells when stimulated with αGalCer.

This post-hoc analysis supports the hypothesis that failure to ac

This post-hoc analysis supports the hypothesis that failure to achieve target haemoglobin or hypo-responsiveness to ESA contributes to

poor outcomes. The Correction of Haemoglobin and Outcomes in Renal Insufficiency Trial compared the effect of two haemoglobin target groups (135 g/L vs 113 g/L) on the composite end-point of death, congestive heart failure, stroke and myocardial infarction in 1432 pre-dialysis CKD patients.12 The trial was terminated on the second interim learn more analysis, even though neither the efficacy nor the futility boundaries had been crossed. The composite event rates at median follow up of 16 months were higher in the high haemoglobin group (HR 1.34, 95% CI 1.03–1.74). Because the conditional power for demonstrating a benefit for the high haemoglobin group by the scheduled end of the study was less than 5% for all plausible values of the true effect for the remaining data, the trial was stopped early. This excess of primary end-point was predominantly due to death (total 88 events (6%) HR 1.48, 95% CI 0.97–2.27, P = 0.07) and heart failure (total 111 events (8%), 3-deazaneplanocin A HR 1.41, 95% CI 0.97–2.05, P = 0.07). Only 12 patients in each group (1.7%) developed stroke and the risk of stroke was comparable between the two groups (HR 1.01, 95% CI 0.45–2.25, P = 0.98). Two post-hoc analyses were performed at 4 and 9 months after randomization comparing high versus low haemoglobin (135 g/L vs 113 g/L)

and high- versus low-dose erythropoietin (≥20 000 U/week vs <20 000 U/week).13 In the 4 months analysis, more patients in the high haemoglobin group failed to achieve target haemoglobin than the low haemoglobin group (37.5% vs 4.7%).

Also, more patients in the high haemoglobin group required high-dose erythropoietin than the low haemoglobin group (35.1% vs 9.6%). Requirement of high-dose erythropoietin among non-achievers was greater in the high haemoglobin group than in the low haemoglobin group (64.2% vs 11.2%). The 9 months analysis showed a similar finding. The initial Cox proportional hazard model demonstrated more harm in the high haemoglobin arm (4 months analysis HR 1.44, 95% CI 1.05–1.97 and 9 months analysis HR 1.62, 95% CI 1.09–2.40). In the subsequent models, composite event rates among the high haemoglobin arm were no longer statistically significant when the additional variables of not Avelestat (AZD9668) achieving haemoglobin target and requirement of high-dose ESA were added either alone or together (4 months analysis HR 1.21, 95% CI 0.85–1.71 and 9 months analysis HR 1.28, 95% CI 0.82–2.00). These results indicate that the poor outcomes observed could have been due to either toxicities related to high-dose ESA or patient-level factors underpinning ESA hypo-responsiveness or a combination of both. In the CREATE trial, 603 pre-dialysis CKD patients were randomly assigned to target haemoglobin value in the normal range (130–150 g/L) or the subnormal range (105–115 g/L).

The skilful technical assistance of Virpi Fisk and Merja Esselstr

The skilful technical assistance of Virpi Fisk and Merja Esselström is gratefully acknowledged. This study was financially

supported by Kuopio University Hospital (project no. 5021605) and the Väinö and Laina Kivi foundation. AK performed the research and analysed the results. AK, TK and TV wrote the manuscript. TK and TV designed the research https://www.selleckchem.com/products/Roscovitine.html study. WWK, JR and MRN provided essential reagents or resources for the research and critically reviewed the manuscript. The authors declare that they have no competing interests. “
“Autoantibodies to double-stranded (ds) DNA represent a serological hallmark of systemic lupus erythematosus (SLE) and may critically contribute to the pathogenesis of lupus nephritis. Self-reactive antibodies might be partially produced by long-lived plasma cells (PCs), which mainly reside within the bone marrow and spleen. In contrast to short-lived PCs, long-lived selleck chemicals PCs are extremely resistant to therapy and may sustain refractory disease courses. Recently, antibody-secreting cells were found within the inflamed kidneys of New Zealand black/white (NZB/W) F1 lupus mice as well as of patients with SLE. To analyze the longevity of the IgG-producing cells

present in nephritic kidneys of NZB/W F1 mice we performed in vivo BrdU-labeling. We identified a higher frequency of long-lived than short-lived renal PCs, indicating that survival niches for long-lived PCs also exist within inflamed kidneys. Using ELISPOT assays, we found that on average 31% of renal IgG-producing cells reacted with dsDNA and 24% with nucleolin. Moreover, the frequencies of IgG-secreting cells specific for the autoantigens dsDNA and nucleolin were higher in the kidneys compared with those in the spleen and bone marrow. Autoantibodies critically contribute to the pathogenesis of various diseases including immune thrombocytopenia, autoimmune hemolytic anemia, myasthenia gravis and systemic lupus erythematosus (SLE). The latter

is a prototypic G protein-coupled receptor kinase autoimmune disease, which can affect virtually all organs. Lupus nephritis is a frequent and serious complication. Anti-dsDNA antibody titers correlate with the clinical activity of the disease and there is accumulating evidence that anti-dsDNA antibodies are crucially involved in the pathogenesis of lupus nephritis 1, 2. Anti-dsDNA and anti-nucleosome autoantibodies co-localize within the glomerular deposits in nephritic kidneys 2. These immune complexes cause complement activation with the release of chemotactic factors, which is linked to recruitment of leukocytes 3. Infiltrating inflammatory cells get further activated by FcγR-mediated mechanisms and essentially contribute to inflammatory organ destruction. These mechanisms lead to extensive inflammation and eventually renal lesions.

[118, 119] Similar to

[118, 119] Similar to see more some of the EAE models, stimulation of type I NKT cells with αGalCer results in disease exacerbation associated with a Th1 cytokine release profile.[118-121] In the latter cases, type I NKT cell activation by αGalCer or its analogues may lead to the tolerization of APC populations. In turn, this outcome may inhibit the activity of most Th1/Th17/Th2 secreting effector cells and thereby lead to protection from autoimmune disease. Generally, activation of type II NKT cells with self-glycolipid

sulphatide may control both antigen-induced and spontaneously arising autoimmune disease. During EAE, sulphatide-reactive type II NKT cells, but not type I NKT cells, are increased several fold in the CNS. This greater abundance of type II NKT cells in the CNS inverts the usual ratio of type II : type I NKT cells (type II NKT cells, 3–4%; and type I NKT cells, 0·6–0·9%) and affords BIBW2992 molecular weight protection from EAE.[27, 61] Furthermore, administration of sulphatide to activate type II NKT cells decreases the number of IFN-γ- and IL-17-secreting myelin basic protein and proteolipid protein-reactive encephalitogenic CD4+

T cells. The net outcome is protection from EAE via a CD1d-dependent regulatory pathway (Maricic et al., submitted). This type II NKT-mediated immunoregulatory pathway results in (i) inactivation of type I NKT cells that now function as regulatory T cells, (ii) tolerization

of conventional DCs, (iii) tolerization of microglia in the CNS and (iv) inhibition of the effector Benzatropine functions of pathogenic MHC-restricted CD4+ T cells. As APCs that activate pathogenic Th1 and Th17 cells in lymphoid organs and the CNS are tolerized following sulphatide administration, activation of type II NKT cells induced by sulphatide is much more potent in the regulation of autoimmune demyelination than only the inactivation of type I NKT cells by αGalCer (Maricic et al., submitted). Activation of type II NKT cells by sulphatide was recently reported to protect NOD mice from type 1 diabetes.[28, 89] Pre-treatment of NOD mice with the C24:0 but not C18:0 sulphatide analogue was found to protect against the transfer of type 1 diabetes.[89] These data suggest that the longer C24:0 sulphatide analogue should be examined for its therapeutic value in clinical trials in human subjects at risk for or newly diagnosed with type 1 diabetes. Our preliminary studies suggest that activation of type II NKT cells following administration of sulphatide significantly prevents lupus nephritis in (NZB × NZW) F1 mice, indicating that the protective capacity of sulphatide activated type II NKT cells can counteract potentially pathogenic type I NKT cells.

These data indicate the critical role of B cells not only for aut

These data indicate the critical role of B cells not only for autoantibody production, but also for CD4+ T cell priming as professional antigen-presenting cells. B cells are therefore an ideal therapeutic target in terms

of not only lowering activities of pathogenic antibodies, but also dampening pathogenic autoimmune responses per se in autoimmune diseases. However, B cell KO mice have a serious problem, in that these mice have major qualitative and quantitative abnormalities in the immune system [7,8]. By contrast, B cell depletion may be a feasible approach to study the function of B cells in autoimmune diseases. Indeed monoclonal antibodies to B cell-specific cell surface molecules such as CD19, CD20, CD79 and to a B cell-surviving factor (B cell lymphocyte stimulator, BLyS) have been used successfully Ixazomib chemical structure to deplete B cells in vivo and to treat numerous autoimmune and malignant haematopoietic diseases in humans and mice [2,9,10]. Transient depletion of B cells by these means can distinguish between the role of B cells during immune development and during immune responses. CD20 is a B cell-specific

molecule that is expressed on the cell surface during the transition of pre-B to immature B cells but is lost upon plasma cell differentiation [11]. In human autoimmune diseases, rituximab, a chimeric anti-human Obeticholic Acid manufacturer CD20 monoclonal antibody, has proved to be effective for treatment of autoimmune diseases, including rheumatoid arthritis, SLE, idiopathic thrombocytopenic purpura, haemolytic anaemia and pemphigus vulgaris [12]. In addition, preliminary clinical studies have shown the therapeutic efficacy of rituximab in a small fraction of Graves’ patients with mild hyperthyroidism [13–16]. In mice, anti-mouse CD20 monoclonal antibodies (anti-mCD20 mAbs) which efficiently eliminate mouse B cells in vivo have been isolated recently

[11,17], and used to treat mouse models of autoimmune thyroiditis, systemic sclerosis, collagen- or proteoglycan-induced Lepirudin arthritis, Sjögren’s syndrome, SLE and type 1 diabetes [17–22]. Moreover, the soluble decoy receptor-Fc fusion proteins to block B cell surviving factors [BLyS and a proliferation-inducing ligand (APRIL)] reduced TSAb activities and thyroxine (T4) levels in a mouse model of Graves’ disease [23]. In the present study, we evaluated the efficacy of anti-mCD20 mAb in a mouse model of Graves’ disease we have established previously [23]. We found that this approach depleted B cells efficiently and that B cell depletion by this agent was effective for preventing Graves’ hyperthyroidism. Our results indicate the requirement of antibody production and T cell activation by B cells in the early phase of disease initiation for the disease pathogenesis. Female BALB/c mice (6 weeks old) were purchased from Charles River Japan Laboratory Inc. (Tokyo, Japan) and were kept in a specific pathogen-free facility.

The clinical and immunological patterns of this unique chronic in

The clinical and immunological patterns of this unique chronic infectious disease clearly demonstrate a continuous scale of changes in histological lesions. Disease classification is defined within two poles (tuberculoid to lepromatous) with transitions between these clinical forms. While typical epithelioid

macrophages predominate at the paucibacillary tuberculoid pole of the disease, inactivated foamy macrophages predominate at the lepromatous end [1]. In lepromatous leprosy (LL), the lack of systemic inflammatory signals and corresponding local ones strongly indicates that a complex anti-inflammatory network is at work. In this regard, neuroendocrine system involvement, in conjunction with the existence of multiple suppressive pathways under the control of the innate and adaptive immune Selleck Osimertinib response, has been reported [2-7]. We have suggested that IDO may play a role in a hitherto unknown suppressive mechanism in leprosy [6]. It has also been reported that accumulated oxidized host phospholipids in lepromatous macrophages downregulate the innate immune response [8]. Foamy macrophages seem to sustain intracellular mycobacteria in a physiological state similar to a nonreplicating

vegetative one [9]. In this context, Montoya et al. [10] demonstrated that lepromatous macrophages Midostaurin purchase exhibit a high expression of the cysteine-rich superfamily scavenger receptor (SRCR), which increases the phagocytic capacity of macrophages and leads to a reduction in bactericidal activity. CD163, a receptor only expressed in monocytes and macrophages, is a member of the class B SRCR superfamily with immunomodulatory Resveratrol properties. Likewise, CD163 is a receptor of hemoglobin (Hb) and hemoglobin–haptoglobin (Hp, Hb–Hp) complexes. The metabolites resulting from intracellular Hb degradation exhibit potent antioxidative

and anti-inflammatory effects. It has been described that the binding of Hb to CD163 induces the release of IL-10 and other anti-inflammatory mediators from macrophages in vivo [11]. It has also been demonstrated that IL-10 enhances CD163 expression by creating a feedback arm of regulation [12, 13] and that the CD163 levels in plasma inversely correlate with the expression of CD163 in blood monocytes [14]. In addition, increased CD163 shedding seems to be associated with the immunosuppressive control of inflammation [15]. The role of CD163 as a bacterial sensor has also been proposed, raising the possibility that a different extracellular domain in this receptor is responsible for triggering proinflammatory cytokines, in contrast to what has been considered its traditional endocytic role [16]. Recent reports have demonstrated ongoing interaction between CD163 and IDO in bone marrow-derived dendritic cells (BMDCs), perhaps indicating that different CD163 signals lead to IDO expression [17].

By excluding the results of the filariasis samples, the

s

By excluding the results of the filariasis samples, the

specificities of the IgG4- ELISA and both of the IgG-ELISAs increased to 100% and Raf inhibitor 98%, respectively. Thus, although the IgG4-ELISA is less sensitive than the IgG-ELISAs, the former is more specific. To determine whether the cross-reactivity with filariasis patient sera was influenced by the abundance of antifilarial antibodies, titrations of IgG4 were performed on the filariasis patient serum samples, followed by an analysis of the correlation with the results of the Strongyloides IgG4-ELISA (Figure 3). The two parameters were found to be weakly correlated (Spearman rho = 0·4544; P = 0·0294). Although previous investigators had reported cross-reactivity between strongyloidiasis and filariasis [4, 13, 27], this selleck chemicals llc study demonstrated that the binding of the Strongyloides antigen to the antifilarial antibodies was not much influenced by the titre of the latter. It is thus highly recommended that, in filariasis endemic area, positive serological cases of strongyloidiasis should also be tested for filariasis before confirming the serodiagnosis. For brugian filariasis, a commercially

available test called Brugia Rapid (Reszon Diagnostics International Sdn. Bhd., Selangor, Malaysia) can be used to assist with this differential diagnosis because the test has been shown to be highly specific (>95%) when tested with serum samples from patients with strongyloidiasis [28, 29]. In this regard, a 31-kDa Strongyloides recombinant antigen (NIE) has been reported to be specific against antibodies to nonlymphatic and lymphatic filariasis [27, 30, 31] and thus is potentially useful as a diagnostic reagent. In conclusion, because the detection of parasite-specific IgG4 antibodies is more specific but less sensitive than the detection of parasite-specific IgG antibodies, the combined use of IgG and IgG4 assays would be helpful in improving the serodiagnosis of strongyloidiasis.

Efforts to develop field-applicable rapid tests using recombinant antigen(s) that do not cross-react with antibodies to lymphatic and nonlymphatic filaria should be encouraged. This study was funded by Universiti Sains Malaysia Research University grant, No: 1001/CIPPM/812078 Microbiology inhibitor and USM short-term grant No. 304/PPSP/61312089. We gratefully acknowledge the contributions of Madihah Basuni and Dr Khoo Boon Yin in this study. “
“This study aimed to examine the frequency of different subsets of circulating B and T follicular helper (Tfh) cells in patients with new-onset rheumatoid arthritis (RA) and following standard therapies. Twenty-five RA patients and 15 healthy controls (HC) were recruited for characterizing the frequency of CD27+, immunoglobulin (Ig)D+, CD86+, CD95+, Toll-like receptor (TLR)-9+ B cells and inducible T cell co-stimulator (ICOS) and programmed death 1 (PD-1)-positive Tfh cells and the level of serum interleukin (IL)-21.