The reason for the efficient Cldn11 induction in BMDM is unclear,

The reason for the efficient Cldn11 induction in BMDM is unclear, although M-CSF, used to generate BMDM, and IL-4 have been shown before to co-regulate certain genes [30]. A summarized gene expression pattern of all adherence and tight junction proteins in macrophages is provided (See summary in Table 2, right columns). Although IL-4 significantly increases the mRNA levels of claudin-1, 2 and 11, this does not result in a detectable

expression of these proteins in macrophages. As a matter of fact, no reports of claudin protein expression in Selleck BAY 80-6946 macrophages exist up to now, in contrast to related cell types such as LCs and DCs. Possibly, the claudin protein expression levels in macrophages are under the detection limit of the antibodies currently used. Alternatively, we cannot exclude that post-transcriptional, such as poor

mRNA stability, and/or post-translational regulatory mechanisms preclude high claudin levels in macrophages. For example, during epithelial reorganization, claudins are ubiquitylated and undergo degradation in the lysosomes [31]. A similar mechanism might be at play in macrophages, especially if the claudins are not engaged in TJ formation. In this respect, one could imagine that claudin proteins are stabilized in vivo when intimate interactions between macrophages and epithelial cells are formed. This could help to bring macrophages in close contact with epithelial cells or with other macrophages, a phenomenon that could be relevant in several situations: (1) in tumours where BAY 73-4506 concentration fusion between macrophages and carcinoma cells might occur [32], (2) during wound healing where macrophages have to integrate in the epithelial sheet of the skin [33] and (3) during granuloma formation and the foreign body reaction where close contacts between macrophages have to be initiated to promote their fusion [29]. Interestingly, Lenzi et al. [34] reported the expression of cadherins and the tight junction–associated protein occludin during the http://www.selleck.co.jp/products/Adrucil(Fluorouracil).html process of granuloma closure. Yet, the lack of claudin proteins in our assays with IL-4-treated macrophages does not preclude their use as marker genes. Indeed, the macrophage activation status in a given pathological

condition is often evaluated by the detection of M1 versus M2 signature genes [4, 25, 26, 35]. Testing different M2 activators identified TGF-β as the most potent inducer of Cldn1 gene expression in macrophages. This finding is reminiscent of TGF-β’s central role in upregulating claudin-1 expression during IL-4-/GM-CSF-treated bone marrow cultures, ultimately giving rise to Langerhans cells [18]. The association of claudin-1 mRNA with the M2 activation status was further confirmed in vivo where high levels of Cldn1 induction were observed in TAM subsets from two mammary carcinoma models and in splenic macrophages isolated from the chronic infection stage of T. congolense infections. In both models, the implication of TGF-β seems plausible.

These conditions predominate during early childhood and do not ap

These conditions predominate during early childhood and do not appear during any other stage of life (Snyder & Merson, 1982; Hoque et al., 1994), highlighting the particular vulnerability of the intestine during early development. Infections caused Silmitasertib supplier by enteric bacterial pathogens, such as diarrheagenic enterohemorrhagic (EHEC) and enteropathogenic (EPEC) Escherichia

coli, the family of attaching and effacing (A/E) bacterial pathogens, are among the most important causative pathogens of severe infantile diarrhea (Donnenberg & Whittam, 2001; Hecht, 2001; Vallance et al.,2002). The mouse pathogen Citrobacter rodentium causes a similar A/E lesion in the murine intestine and has been used as a physiological model of human infection of EPEC and EHEC E. coli. Using the C. rodentium model, we have shown that preinoculation of murine gut with Lactobacillus acidophilus, a probiotic strain, A-769662 nmr early in life can enhance host defense against enteric bacterial infection and attenuate bacteria-mediated intestinal injury (Chen et al., 2005). We also observed that probiotic treatment stimulates regulatory cytokine expression in

the colon transforming growth factor (TGF-β) (Chen et al., 2005). In line with these observations, it has been shown that breast-fed infants have a greater resistance to enteric pathogens owing to the transfer of commensal bacteria (Fanaro et al., 2003), nondigestible oligosaccharides (Newburg et al., 2005), TGF-β in maternal milk (Saito et al., 1993), and immunoglobulins (Brandtzaeg, 2010) which enhance development of the GAI. Moreover, targeted colonization of the neonate intestine with commensal microbiota has been shown to be effective in allergy prevention in later infancy (Lodinová-Zádníková et al., 2010). More specifically,

the intestinal microbial communities predominately induce the maturation of the mucosal adaptive immune system in the human neonate (Kaplan et al., 2011). Conversely, formula-fed infants lack maternal transfer of commensal bacteria, nondigestive oligosaccharides, and TGF-β which results in the modification of gut microbial communities compounding the vulnerability of the neonatal intestine to enteric pathogens (Le Huërou-Luron et al., 2010). TGF-β is a very potent negative regulator of mucosal inflammation Bupivacaine (Letterio & Roberts, 1998) inhibiting T cell activation (Letterio, 2005) vital to maintaining tolerance to innocuous antigens found within the intestine. TGF-β mediates cell signaling by ligand-dependent activation of heterodimeric transmembrane serine/threonine kinases receptors (Piek et al., 1999). Downstream, the ligand-activated receptor directly phosphorylates Smad2 and Smad3 proteins, which associate with Smad 4 and translocate to the nucleus to participate in transcriptional control of targeted genes (Heldin et al., 1997).

As shown in Fig 1A, significantly more dead

As shown in Fig. 1A, significantly more dead Acalabrutinib concentration and apoptotic cells, as judged by staining with 7-amino-actinomycin D (7-AAD) and annexin V, respectively, were presented in anti-CD3+IL-2-activated WT CD8+ T cells (54 and 72%, respectively) than in similarly activated TNFR2−/− CD8+ T cells (13 and 17%, respectively). In contrast, essentially identical 7-AAD and annexin V staining data were obtained for both WT and TNFR2−/− CD8+ T cells when monoclonal anti-CD28 antibodies were included in the AICD assays (data not shown). These results indicate that AICD in either WT or TNFR2−/− CD8+ T cells is not regulated by CD28 costimulation. We have reported previously that TNFR2−/− CD8+ T cells

undergo suboptimal proliferation relative to WT CD8+ T cells when stimulated by anti-CD3 antibodies 6. This observation is consistent with the hypothesis that TNFR2 participates in the optimal activation of anti-CD3-stimulated CD8+ T cells. Here, we found that anti-TNFR2 antibodies also inhibited the proliferation of anti-CD3 stimulated WT CD8+ T cells (Fig. 1B). The specificity of the blocking anti-TNFR2 antibody was demonstrated by its lack of effect on the proliferation of anti-CD3-activated TNFR2−/− CD8+ T cells. This result indicates that in WT CD8+ T cells, optimal proliferation after anti-CD3

stimulation is dependent on TNFR2. We next determined whether antibody-mediated blocking of TNFR2 in WT CD8+ T cells recapitulates the effect of the TNFR2−/− mutation in AICD. We found that the blocking 5-Fluoracil solubility dmso anti-TNFR2 antibody dramatically increased the resistance of activated WT CD8+ T cells to AICD (Fig. 1C). The specificity of the blocking anti-TNFR2 antibody was again demonstrated by its lack of effect on AICD of TNFR2−/− CD8+ T cells. These data indicate that TNFR2 is essential in

both the optimal proliferation of anti-CD3-activated CD8+ T cells and for the induction of AICD that terminates the proliferative response. To test the hypothesis that intracellular levels of TRAF2 regulate AICD, we determined Phenylethanolamine N-methyltransferase the expression level of TRAF2 in TNF-α-stimulated WT and TNFR2−/− CD8+ T cells. WT and TNFR2−/− CD8+ T cells were stimulated for 48 h with anti-CD3+IL-2 followed by stimulation with TNF-α for various times. Immunoblotting showed that the amount of TRAF2 protein in WT cells decreased by 6 h after adding TNF-α (Fig. 2A). In contrast, the amount of TRAF2 protein in TNFR2−/− cells remained unchanged, even after 12 h of TNF-α stimulation. Furthermore, we found that TRAF2 protein levels were lower in WT CD8+ T cells than in TNFR2−/− cells at 72 h after anti-CD3+IL-2 stimulation (Fig. 2B). These data indicate that TNFR2 signaling promotes the degradation of TRAF2 at a time when AICD occurs and suggests that intracellular levels of TRAF2 play a critical role in regulating AICD. We next determined the effect of TNFR2 blocking on intracellular TRAF2 levels.

NK cells after HSCT express high levels of CD56 27–30, 32, 33 Th

NK cells after HSCT express high levels of CD56 27–30, 32, 33. This has often been used as an argument that ptCD56bright are immature 29, 31, 32, 34. Here, we report that ptCD56bright have only few characteristics

of immature NK cells and are indistinguishable from cytokine-activated CD56bright. We show that ptCD56bright are CD11b+CD27−, a phenotype characteristic of mature NK cells and that CD11b+CD27+CD56bright become CD11b+CD27− after stimulation with IL-15. Both PXD101 datasheet ptCD56bright and NKIL-15 were CCR7−, HLA-DR and perforin-positive and readily produced IFN-γ after stimulation with IL-12. We also found that after culture in the absence of cytokines, ptCD56bright and NKIL-15 upregulated c-kit, CD127 but not CCR7. Hence, stimulation with IL-15 induces many of the features characteristic of ptCD56bright on CD56bright and because both cell types also regulated the expression of c-kit, CD127 and CCR7 in a similar manner, we believe that ptCD56bright are mature CD56bright that have expanded after being stimulated by the elevated cytokine levels that have been observed

in the serum of transplanted patients 27–29. The finding that the number of ptCD56bright was not correlated with the level of hematopoiesis supported this hypothesis further. We found that the number of ptCD56bright was highest in patients with low numbers of Talazoparib order T cells. During the first month after transplantation, T cells are generated by peripheral expansion rather than through the hematopoiesis-dependent thymic pathway 44–46. This expansion is driven by IL-7 and IL-15 of which the latter also regulates the homeostasis of NK cells 47, 48. CD8+ memory effector T cells are known to restrict IL-15-dependent homeostasis

of γδ-T cells 49, 50. Furthermore, NK cells and CD8+ T cells compete for IL-15 in lymphopenic mice 51. Therefore, it is conceivable that CD8+ T cells that represent the major T-cell Selleckchem Neratinib population after transplantation also compete with NK cells for the elevated levels of IL-15 present in transplanted patients 27–29. Because IL-15 also induces the ptCD56bright phenotype in CD56bright, we think that IL-15 is most likely to be the cytokine with the most impact on the post-transplant NK-cell compartment. The correlations between the number of NK cells and the plasma levels of IL-15 after HSCT have been reported as absent 29, weak 27 or strong 28. We have not measured IL-15 serum levels in our cohort because we believed that there would be too many reasons why the relationship between IL-15 levels and NK-cell expansion may remain hidden. First, most IL-15 is presented in trans in tissues 52 and could effectively stimulate NK cells also when serum levels are low.

Oral administration of azithromycin to recipient mice for 5 days

Oral administration of azithromycin to recipient mice for 5 days during major-histoincompatible BMT suppressed lethal GVHD selleckchem significantly, whereas ex-vivo lymphocyte function was not affected by the drug. These data suggest that azithromycin has potential as a novel prophylactic drug for lethal GVHD. Haematopoietic stem cell transplantation from an allogeneic donor provides curative therapy

for patients with malignant and non-malignant haematological diseases. However, acute graft-versus-host disease (GVHD) is still a major cause of morbidity and mortality after allogeneic bone marrow transplantation (BMT). GVHD is initiated by donor T lymphocytes that recognize host histocompatibility antigens that distinguish host from this website donor. To date, most therapeutic approaches designed to attenuate GVHD have focused on suppressing donor T lymphocytes

[1-5]. These approaches, however, often result in incomplete GVHD attenuation, especially in histoincompatible transplants. Recent murine studies have shown that interactions between donor T lymphocytes and host antigen-presenting cells (APCs) are essential for triggering GVHD [6-11]. Dendritic cells (DCs) derived from haematopoietic stem cells are distributed ubiquitously in blood, lymphoid and peripheral tissues and play important roles in the immune system by stimulating naive T lymphocytes and secreting cytokines that initiate the immune response [12]. The state of DC maturation influences their functions. Various factors, including bacteria-derived antigens such as Metformin price lipopolysaccharide (LPS), viral products, inflammatory cytokines and conditioning regimens such as total body irradiation (TBI) can induce maturation of DCs, which is characterized by up-regulation of major histocompatibility complex (MHC) class II, co-stimulatory molecules and essential chemokine receptors.

Mature DCs (mDCs) promote antigen-specific T cell activation and proliferation. Moreover, following CD40 ligation or Toll-like receptor ligation, mDCs secrete interleukin (IL)-12 p70, which induces interferon (IFN)-γ-producing T helper type 1 (Th1) cells that are considered a pivotal pathogenic factor in acute GVHD [12-15]. Nuclear factor (NF)-κB is a rapid response transcription factor in various cells involved in immune and inflammatory reactions and exerts its effect by inducing expression of cytokines, chemokines, cell adhesion molecules and growth factors [16, 17]. NF-κB is sequestered normally in the cytoplasm of non-stimulated cells and is translocated into the nucleus in response to a variety of stimuli, such as bacterial lipopolysaccharide (LPS) and tumour necrosis factor (TNF)-α. Because it also plays a crucial role in DC maturation [18, 19], NF-κB in DCs might be a rational target for preventing GVHD.

In a CD4-dependent

model of GVHD, Warren and Mark Shlomch

In a CD4-dependent

model of GVHD, Warren and Mark Shlomchik and colleagues from Yale 8 were the first to show that irradiated allogeneic recipients of either total CD4+ T cells or CD44− CD62L+ TN cells developed severe GVHD, whereas AUY-922 datasheet recipients of CD44+CD62L− TMP cells remained entirely well and free of GVHD. Furthermore, rapid reconstitution of the peripheral T-cell compartment in the BMT recipients by TMP cells permitted robust recall immunity to third party antigens, indicative that responses to persistent and acquired infections might be preserved. Importantly, protection from GVHD did not rely upon the presence of regulatory T cells within the TMP-cell fraction. Several other groups have since confirmed and extended these findings in experimental BMT by selecting for TMP cells in the bulk T-cell population or from individual CD4+ or CD8+ T-cell subsets of unprimed mice, and by using BMT models that involve MHC mismatches or that are MHC-matched but mismatched for multiple minor histocompatibility (H) antigens 9–13. In general, the results have been broadly similar, although whether CD44+ CD62L+ TMP cells are as disabled as CD44+CD62L− TMP cells in inducing GVHD is less clear 11, 14. Caution is required, however, before assuming

that transfer of human memory T cells can successfully be applied to the clinic because the TMP-cell populations in mice housed under specific pathogen-free conditions are likely to be distinct in several respects from the memory T-cell populations found in humans. In such mice, TMP Midostaurin solubility dmso cells arise many as a result of lymphopenia-induced proliferation as new thymic emigrants enter the periphery of neonatal mice 15

or represent the proliferation of cells in response to environmental antigens or allergens. In humans, T cells expressing memory markers will include a greater proportion of cells that have been primed previously following exposure to pathogens. A very high proportion of human memory T-cell lines or clones specific for viruses such as EBV or CMV demonstrate cross-reactivity with allogeneic peptide:HLA, a consequence of the degenerate TCR recognition of peptide:HLA ligands 16. Although alloreactivity is also demonstrable in the human TN-cell pool 17, memory populations, which contain unprimed TMP cells as well as primed effector memory T (TEM) cells, could be potentially more harmful than TN cells since they are less stringent in their requirements for TCR stimulation or costimulation than their TN-cell counterparts 1. Reduced susceptibility to apoptosis or to peripheral tolerance mechanisms in the host might also make such human memory T-cell populations more dangerous than TN cells 1. This increased alloreactivity could also be relevant in cases where the donors and recipients are HLA-matched, but the donors are female and have previously been primed to male antigens as a result of pregnancy 18.

These proteases may cleave extracellular matrix proteins and inju

These proteases may cleave extracellular matrix proteins and injure the endothelium. Lu et al. demonstrated that

ANCA-activated neutrophils released serine proteases, but not superoxide when co-cultured with EC, and that serine proteases mediated EC damage resulting in von Willebrand factor (vWF) release [78]. Serine proteases that are packaged in ANCA-induced neutrophil microparticles or in neutrophil extracellular check details traps (NETs) possibly also participate in endothelial damage [79,80]. Together, ANCA induce a variety of neutrophil responses in vitro. Some of these were shown to be significant in vivo, such as p38 MAPK, PI3Kγ, C5a and serine proteases. Others that are thought to be important await further in-vivo proof, including the role of ANCA-induced reactive oxygen generation. The neutrophil is both the cell that expresses target

ANCA antigens and a major effector cell in ANCA-induced small vessel vasculitis. The ANCA antigens PR3 and MPO differ substantially in their expression pattern on the neutrophil plasma membrane. ANCA bind to membrane expressed target antigens and initiate intracellular signalling events. The PR3–NB1–Mac-1 membrane complex is one example showing that larger signalling complexes with transmembrane molecules exist. Distinct signalling pathways triggered by ANCA F(ab)2 and the intact ANCA IgG molecule were identified and co-operate in neutrophil activation. Detailed characterization LY2606368 supplier of the activation process will identify novel treatment targets that need to be tested in animal models and subsequently in patients. Ralph Kettritz was supported by grants from the Deutsche Forschungsgemeinschaft and the Experimental and Clinical Research Center, a joint co-operation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Elongation factor 2 kinase Medicine Berlin-Buch. Nothing to declare. “
“Cytomegalovirus (CMV) -specific immunity is often estimated by the number of in vitro CMV antigen-inducible interferon-γ-positive

(IFN-γ+) T cells. However, recent work indicates that simultaneous production of IFN-γ, tumour necrosis factor-α (TNF-α) and interleukin-2 (IL-2) (referred to as ‘polyfunctionality’) is more relevant for anti-viral protection. Here, we compared polyfunctionality of CMV-specific T cells (pp65 and IE-1 proteins) in 23 solid-organ transplant patients and seven healthy controls by flow cytometry. The proportions of TNF-α+/IFN-γ+/IL-2 cells among the activated cells were significantly reduced in transplant patients but not the frequencies of IFN-γ+ CD8+ T cells. Immunosuppression reduces polyfunctionality, which reflects the increased infection risk in this patient group. In healthy individuals, CD4+ and CD8+ T cells restrain many infectious pathogens but in transplant patients these mechanisms are weakened by the immunosuppressive medication required to prevent graft rejection.

In this respect, the ASC-probe technique offers two main advantag

In this respect, the ASC-probe technique offers two main advantages to the use of

serum antibodies. The first is its ability to recognize antigens that have low immunogenicity or are only transiently exposed to the immune system, which is likely to be the case for surface or secreted antigens from migrating schistosomula. By analysing the local antibody response, it may be possible to identify antigens that are not seen when using serum as a probe, as in the previously referred studies with H. contortus, A. suum and F. hepatica. The second main advantage of lymph node-derived ASC probes is the capacity to focus on particular tissue compartments in isolation from more immunologically dominant infection https://www.selleckchem.com/products/Gefitinib.html sites. Eberl et al. (78) showed that YAP-TEAD Inhibitor 1 concentration even a fairly significant number (2000) of schistosome cercariae used to infect chimpanzees provides a low antigenic stimulus in serum and that the major cause of antibody production in schistosomiasis was egg deposition in the liver and intestine. Therefore, to be able to focus on the antibody response caused by schistosomula alone, in

isolation from that caused by the egg deposition (and even the potentially irrelevant adult response), would be a significant advantage. Despite the value of this technique, it has not been applied hitherto to any of the important human helminthiases, including schistosomiasis; however, preliminary studies we have undertaken suggest that it can be used to great effect for novel, stage-specific antigen discovery and for studying the natural or protective immune response (79; McWilliam H.E.G.,

Piedrafita D., Driguez P., McManus D.P. and Meeusen E.N.T., unpublished data). Once the desired next tissue-specific ASC probes are obtained, there are various techniques available to identify their target molecules, including one- and two-dimensional Western blotting and screening of recombinant expression libraries. A promising new approach, however, is to make use of both protein or carbohydrate arrays that are becoming increasingly available and provide promising new tools to study the immunome. These applications are further elaborated in the following sections. The publication of the schistosome genomes (57,63), along with the wealth of new proteomic and transcriptomic data available (58,59,61), has opened the door to novel protein discovery. These information-rich biological datasets, when combined with high-throughput experimental methods, can revolutionize vaccine and diagnostics research. For example, we have developed an immunomics protein microarray for vaccine antigen discovery for S. japonicum and S. mansoni (80).

24,25 An FcR-mediated activity of a broadly reactive HIV neutrali

24,25 An FcR-mediated activity of a broadly reactive HIV neutralizing monoclonal antibody (mAb) has also been shown to contribute to protective efficacy in a macaque challenge model,26 further invoking a role of NK cells. Moreover, the recent modest success of

the RV144 HIV clinical vaccine trial in Thailand27 has been suggested to be partly the result of ADCC activity elicited by the vaccine regimen.28 Hence, there is heightened interest in the HIV vaccine field in NK-cell-mediated effector functions. Despite the potential role played by NK cells during innate and adaptive immune responses against HIV/SIV, and the utility of rhesus macaque models, the variety and function of roles Y-27632 order of different macaque NK cell subpopulations have not been exhaustively explored. Previous reports have described macaque circulatory NK cells as CD3− CD8α+ CD20−/dim NKG2A+ cells that can be further divided into four subpopulations based on their CD56 and CD16 expression patterns.29–31 However, CD8α expression on different human NK cell subsets is variable,32,33 and therefore CD8α expression selleck kinase inhibitor is not necessarily a requisite marker for NK cell phenotyping. In this regard, a minor subset of CD8α− NK cells has been recently identified in healthy and HIV-infected chimpanzees.34 Furthermore, it has been shown that peripheral

blood mononuclear cells (PBMCs) from HIV-infected mothers and their infants that strongly respond to HIV-1 peptide stimulation [by up-regulating interferon-γ (IFN-γ) and interleukin-2 (IL-2) production in both CD3− CD8− and CD3− CD8+ cells] are less likely to transmit and acquire infection, respectively.35 For the reasons mentioned above, in the present study we evaluated the presence of NK cell lineage markers on macaque CD3− CD14− CD20−/dim CD8α− PBMCs, and the potential of these cells to mediate functional responses. Using multi-parametric flow cytometry, we identified a subpopulation of

circulatory CD8α− NK cells in naive and SIV-infected macaques that expressed the CD56 and/or CD16 NK cell lineage markers. A subset of these CD3− CD14− CD20−/dim CD8α− cells (from now on referred to as CD8α− NK cells) also co-expressed granzyme B, perforin, NKG2D and KIR2D. Upon cytokine Aldol condensation stimulation, CD8α− NK cells up-regulated CD69 expression and IFN-γ mRNA transcription and produced low levels of tumour necrosis factor-α (TNF-α). Importantly, enriched CD8α− NK cells were capable of mediating direct cell lysis as well as antibody-dependent killing, suggesting a potential for contributing to both innate and adaptive immune responses. Rhesus macaques (n = 30, 17 naive and 13 chronically infected with SIV) used in this study were housed at the National Institutes of Health (NIH) Division of Veterinary Resources (Bethesda, MD), at Bioqual, Inc.

The resultant final diagnosis was enteric hyperoxaluria complicat

The resultant final diagnosis was enteric hyperoxaluria complicated by an acute irreversible oxalate nephropathy. Management

consisted of a low-oxalate diet and intensification Rapamycin manufacturer of pancreatic enzyme supplements to limit malabsorption. In addition, calcium carbonate and subsequently Sevelamer were added in order to reduce the enteric absorption of oxalate. Reduction in systemic oxalate load was attempted by the use of daily haemodiafiltration via a tunnelled internal jugular catheter and it is notable that he did not suffer any systemic manifestation of oxalate deposition such as heart block, arthropathy or neuropathy. The patient was managed as an outpatient and received tacrolimus, mycophenolate and steroids

and remained free of pulmonary rejection with Forced expiratory volume (FEV1) maintained above 3.0 L. The patient was distressed and angry at the need for regular haemodialysis and the impact it made on his life despite the renewed benefit of his lung transplantation. Options for renal transplantation were considered and his mother was assessed as a potential kidney donor. Ten months post lung transplant he underwent a renal transplant with Basiliximab and methylprednisolone induction with maintenance of standard tacrolimus this website and mycophenolate dosing. There was immediate graft function and no complication. Calcium and Sevelamer supplementation were initially ceased, but were recommenced because of early hyperoxaluria with restoration of adequate glomerular

filtration and tubular flow. The patient was advised to maintain a urine output of 3 L a day (see Fig. 3). Elongation factor 2 kinase Urinary oxalate excretion was monitored regularly in order to adjust pancreatic supplementation and oral oxalate binders. Initially very high levels may have reflected an elevated systemic burden and it is notable the urinary oxalate declined to the normal range after 3 months. A 2-week post-transplant renal biopsy showed no evidence of recurrent oxalate deposition. In the months following his renal transplant, intermittent episodes of diarrhoea related to antibiotics or mycophenolate use precipitated episodes of acute renal failure. However, these diarrhoeal episodes improved on switching mycophenolate to azathioprine. At 8 months post renal transplant he has a creatinine of 122 µmol/L with an eGFR of 60 mL/min per 1.73 m2. His lung function remains stable and he is gainfully employed as an electrician. Oxalate is a ubiquitous molecule found in the soil and taken up by plants and vegetables such as spinach, rhubarb and nuts. Concentrations in foods vary widely depending on the soil and water conditions they were grown in, making quantification in feeds difficult. Approximately2 20–40% of oxalate is obtained from the diet where it is absorbed in the colon.