Although TLR-mediated inflammation is essential for host defence

Although TLR-mediated inflammation is essential for host defence against pathogens, TLR signalling must be tightly controlled because unrestrained TLR activation generates a chronic inflammatory milieu that can result in various chronic inflammatory disorders.9 Several TLR signalling suppressors have been described in immune cells.10 Recent studies LDK378 revealed that Tyro3, Axl and Mer (TAM) receptors play a pivotal role in negatively regulating innate immunity via the inhibition of the TLR-mediated inflammatory response and the promotion of phagocytic clearance of apoptotic cells.11–13 The TAM receptors belong to a subfamily of receptor tyrosine kinases. Of the 58

members of the receptor tyrosine kinase family,14 the TAM receptors are among the few that are specific to vertebrates. Analysis on TAM knockout mice revealed that TAM receptors play see more an essential role in the regulation of tissue homeostasis in the adult nervous, vascular and reproductive systems.15 Notably, TAM receptors have profound effects in the homeostatic regulation of innate immune responses.16,17 Two closely related proteins, the product of growth-arrest-specific gene 6 (Gas6) and Protein S (ProS), are common biological ligands of TAM receptors.18 Gas6 and ProS are two secreted soluble proteins that carry an N-terminal γ-carboxylated glutamic acid domain that confer the ability

to bind phosphatidylserine on the surface of apoptotic cells,19 and a C-terminal sex hormone-binding globulin-like module that can bind and activate TAM receptors.20 Although the Gas6/ProS-TAM Alanine-glyoxylate transaminase system has a pivotal role in regulating innate immunity, the regulation of this system remains largely unknown. In the current article, we provide evidence that TLR activation suppresses the

expression of Gas6 and ProS, which facilitates the TLR-mediated inflammatory response in macrophages. The data provide insights into the regulation of Gas6 and ProS expression and function during the inflammatory response. C57BL/6 strain mice 8–10 weeks of age were obtained from the animal facility of Peking Union Medical College (Beijing, China). The mouse mutants for TAM receptors were provided by Dr Greg Lemke (Salk Institute for Biological Studies, La Jolla, CA). These mice were housed under specific pathogen-free conditions with a 12 : 12 hr light : dark cycle and had free access to food and water. The mice were handled in compliance with the Guideline for the Care and Use of Laboratory Animals established by the Chinese Council on Animal Care. Ultra-pure S. Minnesota LPS, poly(I:C), CpG oligonucleotides, antagonists of TLR4 (tlrl-rslps) and TLR9 (tlrl-2088) were purchased from InvivoGen (San Diego, CA). Neutralizing anti-TLR3 antibody (TLR3.7) was purchased from Apotech (Geneva, Switzerland).

[25], having reviewed the early history of the parasite, strongly

[25], having reviewed the early history of the parasite, strongly recommended that N. dubius should be dropped. However, in subsequent work, it became apparent that the parasites used in laboratory studies and those parasitizing wild wood mice (Apodemus sylvaticus) in Europe were quite distinct in a number of respects. At first, it was suggested that these

were subspecies and should be referred to as H. polygyrus bakeri for the laboratory-maintained parasite and H. p. polygyrus for that in wild rodents [26], but Cable et al. [27] raised both to full species status Small molecule library supplier on the basis of molecular genetic data. This controversy about the exact taxonomic status of the parasite was reviewed again recently [28], although not everyone has accepted

www.selleckchem.com/products/Y-27632.html the change in nomenclature proposed by Cable et al. [29], and for this reason in this article, we refer to it as H. p. bakeri. In the 1960s–1970s, a key research problem with H. p. bakeri was how to induce immunity to this parasite, as primary infections appeared to be so stable for so long. Many experimenters found that removing a primary infection and then challenging the mice with a second batch of larvae just did not induce marked immunity, that is, a substantial reduction in the success of challenge infections [30-32], and it was thought at the time that oxyclozanide adult worms were not immunogenic [33]. Much effort was given therefore to devising various combinations of repeated infections, sometimes interspersed

with anthelmintic treatment or just superimposed on one another. The breakthrough came when it was realized that adult worms not only failed to induce effective resistance in many mouse strains and appeared not to be susceptible to mucosal responses in some strains of immune mice [34], but actually prevented the expression of host-protective effector mechanisms operating at the mucosal level [13, 31, 35]. The larval, tissue-dwelling stages of this parasite are in fact highly immunogenic [36] and can induce immunity even in poor responder strains of mice [37], as long as the period of residence of adult worms in the gut lumen is brief, as for example after infection with irradiated infective larvae [38], following treatment with ivermectin, which kills the larvae in situ in the intestinal walls [36], or by chemotherapy immediately after their emergence from the intestinal walls 7–9 days post-infection [31, 35]. It was shown that an average of just over 3 infective larvae per mouse was sufficient to generate an 84% reduction in challenge infection worm burdens in NIH mice when the immunizing larvae were killed by ivermectin on day 6 post-infection [37].

Such tissues can rapidly form stable structures during inflammati

Such tissues can rapidly form stable structures during inflammation, and yet equally as easily regress, as seen in the dynamic development of TLOs during chronic Helicobacter pylori infection.[57] The fundamentals underpinning SLO development also lie at the heart of TLO development: inflammatory cytokine expression (LT/tumour necrosis factor-α); stromal activation and chemokine production; and high endothelial venule development.[58, 59] As seen in transplantation studies,[60,

61] activated stromal cells alone are capable of initiating TLO formation in some instances, indicating their overriding capacity to contribute to TLO development. Nevertheless, the precise signals leading to stromal activation

during selleck kinase inhibitor TLO development in vivo are still unclear. The majority of mechanistic data on the development of TLOs are selleck products derived from transgenic mice expressing molecules in ectopic sites. Although these are narrow models that lack the complexity that undoubtedly underpins in vivo TLO generation, they do offer a glimpse into TLO development that would otherwise be hard to observe. Table 2 highlights animal models of TLO development that use either LTβR signalling, homeostatic chemokine or non-homeostatic cytokine transgenic expression. If TLO and SLO development is conceptually similar, what is the source of LTα1β2 in TLO development? One possibility is that TLOs are formed by LTis in much the same way as in SLOs, but there is conflicting evidence to support this hypothesis. Interleukin-7 (a key survival factor for LTis in developing SLOs) transgenic mice develop a large number of LNs and Peyer’s patches, as well as the formation of organized TLOs after immunization with antigen, in a process that is dependent upon LTα1β2 and the LTi-associated transcription Tenofovir nmr factor retinoic

acid-related orphan receptor γt (RORγt).[62] However, a CCL21 transgenic model of TLO development lacking LTis still develops TLOs,[63] with CD3+ CD4+ T cells the first to arrive at the site of TLO development, indicating an LTi-independent mechanism that may be unique to TLOs. Formation of TLOs during inflammation of the intestine is able to occur in the absence of RORγt (and hence LTis),[64, 65] although with the recent identification of multiple innate lymphoid cell (ILC) populations, which express similar levels of LTα1β2 to their LTi cousins,[66, 67] the extent to which RORγt-independent ILCs can contribute to intestinal TLO generation requires further investigation.[68] As B and T cells both express LTα1β2,[69] are relatively much more abundant in chronically inflamed tissues than LTis (or other ILCs), and activated conventional lymphocytes are known to play a role in TLO generation in the skin,[60] it is likely that B and T cells contribute significantly to TLO development during inflammation.

[168] Whether an initial metabolic, structural, or related defect

[168] Whether an initial metabolic, structural, or related defect leads to immune activation and a subsequent deleterious response or an initial loss of immune regulation leads directly to tissue disregulation and destruction is still a matter of debate in some circles. Selleckchem BMS-777607 Thus, the

issue of immune-mediated recurrent pregnancy loss is one that is likely amenable to iterative studies in animal models and humans. In primates, parental sharing of MHC has been correlated with decreased pregnancy success.[169] Moreover, administration of antiprogestational agents can produce early pregnancy loss, as in humans.[170] Primates have also been used to develop models of pregnancy loss related to infections.[171] A well-known mouse model of pregnancy loss involves the breeding of a CBA strain female mouse with a DBA strain male mouse. Depending on the source and housing (level of pathogens present) of the mice, pregnancies can be affected by high levels of fetal-placental degeneration (referred to as ‘resorption’)[172] and infiltration with NK and other immune cells.[173] In this model, resorption of the fetuses occurs at approximately 9–12 days of gestation.[174] AZD1208 price Contributors to increased fetal loss in this model include stress,[175] inflammation[176, 177] abnormal

cytokine milieu within the placenta/decidua,[178, 179] disrupted regulatory immune modulation,[180, 181] and abnormal placental vascular development.[182, 183] Several methods of immune modulation[184-187] have been shown to decrease fetal loss in this model, but few if any have been successfully translated to clinical care.[28] More recent models of pregnancy loss in mice involves chemically targeting[86] depletion[87] or genetic deficiency of a subpopulation[188] of regulatory T cells in normal C57Bl/6 females

mated to same strain or allogeneic males. An alternative Liothyronine Sodium immune-based models of pregnancy loss involved NK T cell activation in certain strains of mice[189] and systemic immune activation leading to ovarian insufficiency.[38] Study of the high rate of pregnancy loss in commercial pork breeds has further suggested the role of immune cells in supporting successful pregnancy.[190] Moreover, Guinea pigs (for example[191]) and Sheep[192] have been used in models of early pregnancy loss in response to infection. Finally, autoimmune-related loss, as in the antiphospholipid syndrome, has been modeled in rabbits.[193] The study of premature birth presents at least three major issues that are amenable to studies in animal models.[194] The first is the discovery of mechanisms leading to premature labor. A second pertains to delineating consequences of being born premature. Third, animal models have been employed to devise ways to better manage the premature neonate. While the factors contributing to prematurity in humans are far from understood, emerging data suggest that preterm births fall into definable categories.

Microglia contact synapses, ‘stripping’ dysfunctional ones, remov

Microglia contact synapses, ‘stripping’ dysfunctional ones, removing cell debris, and sensing and modulating neuronal activity. Hence, microglia contribute to CNS homeostasis and plasticity. Under pathological conditions, resting microglia sense activating ‘danger signals’, such as molecules expressed by infectious agents or released upon tissue damage, through diverse types of receptors, and respond rapidly towards injury displaying an alerted phenotype.

Such a shift to an activated state is accompanied by dynamic morphological, molecular and functional alterations resulting from the balance between activating inputs and calming signals. While activated microglia have been observed in many neurological diseases of diverse aetiology, ‘activation’ does not reveal the functional state of the cells, which are often engaged in highly different roles. www.selleckchem.com/products/Fludarabine(Fludara).html Indeed, microglia can play both detrimental and beneficial roles depending on inputs and feedback signals arising from the neural environment; such paradoxical

roles are associated with phenotypes that range from ‘classically activated’, with highly pro-inflammatory features, to ‘alternatively activated’ associated with a repair-oriented profile. Here, we review microglial phenotype and behaviour in health and disease and their impact on neurodegeneration; we discuss how therapeutic approaches to a neurodegenerative Selumetinib purchase disease with a predominant inflammatory component, multiple sclerosis (MS), could modulate microglia activation towards

an alternative phenotype favouring Sodium butyrate neuroprotection, with the potential to modify the outcome of neurological diseases. Monitoring of microglial morphology in the intact brain by two-photon microscopy has shown that ‘resting’ microglia are highly active, extending and retracting motile processes through which they survey their microenvironment and interact dynamically with surrounding cells.[1, 2] Through this dynamic sensing of their environment, microglia perceive ‘danger signals’ upon changes of the CNS microenvironment or upon injury and become activated, undergoing morphological changes through an intermediate amoeboid form with several short, thickened processes to a round ‘over-activated’ profile. The functional role of the immediate microglial response upon injury has not been fully elucidated, but might be related to a shielding of the injured area, with the number of responding microglia apparently dependent upon the severity of the injury, to preserve a stable environment in the vicinity of nearby neurons and thereby minimize ensuing damage.

7B) TdTom-transduced cells expressed red tdTom protein spread th

7B). TdTom-transduced cells expressed red tdTom protein spread throughout the cytoplasm (Fig. 1B-iv) and similarly to untransduced CTLs (Supporting Information Fig. 7A) relocalized GZMB-containing granules expressing Lamp-1 to the CTL/target contact zone (Fig. 1B-iv). Mathematical analyses showed that GZMB-tdTom colocalized with Lamp-1 and GZMB (Pearson’s Rr coefficient around 0.55) whereas tdTom did not show any colocalization (Rr 0.1) (Supporting Information Fig. 7C). Following TCR/antigen

engagement, calcium flux and PKC activation are important signals for gene activation and granule migration to the CTL/target contact zone preceding degranulation 4, 8. CTLs preloaded with Fluo-4 were used to monitor by video microscopy the Ca++ fluxes and the redistribution of GZMB-tdTom-containing granules. When GZMB-tdTom-transduced selleck products P14-TCR CTLs faced a specific target, an attachment signal preceded a rapid Ca++ flux (10–20 s) and granule translocation to the contact zone occurring at various times (20–480 s) (Fig. 1C-i and ii, Supporting Information Fig. 7D, Video 1). No significant signal was observed when the CTLs were facing control targets (Fig. 1C-iii and iv, Video 2). These kinetics are in agreement with published studies using CTL clones 6, 9. We used the Lamp-1 exposure method to assess CTL degranulation in response to antigenic stimulation and

to observe the fate of GZMB-tdTom during that process. GZMB-tdTom-transduced P14-TCR CTLs exposed Lamp-1

in response to gp33-loaded RMA-S, the extent of Selleckchem GSI-IX degranulation being dependent on peptide concentration (Fig. 2A). The percent of GZMB-tdTom fluorescent Mannose-binding protein-associated serine protease CTLs markedly decreased (from 20% for non-stimulated or control-peptide stimulated CTLs to 13% for CTLs activated with 10−6 M gp33-loaded RMA-S), with a level of GZMB-tdTom fluorescence much lower in Lamp-1–positive (MRFI 422 (MRFI, mean relative fluorescence intensity)) as compared to Lamp-1–negative (607) CTLs. GZMB expression as measured on fixed and permeabilized cells were also reduced (about 50%) in the antigen-activated CTLs (data not shown). These results suggest that the whole GZMB-tdTom fusion protein was released during degranulation. Similarly, analysis of GZMB-tdTom-transduced OT1-TCR-Gzmb-KO (Gzmb, GZMB-encoding gene) CTLs, in which the only source of GZMB is GZMB-tdTom, showed that expression of GZMB-tdTom as well as GZMB was markedly decreased upon CTL activation with OVA-expressing cells (Supporting Information Fig. 8). We also found that the capacity of GZMB-tdTom-transducted P14-TCR CTLs to kill specific targets was not affected as compared to that of untransduced CTLs (Fig. 2B). To our knowledge, two attempts at expressing fluorescent GZMB fusion proteins have been reported, but they were not expressed in CTLs 10, 11.

The CBMCs were obtained by Ficoll–Hypaque density gradient centri

The CBMCs were obtained by Ficoll–Hypaque density gradient centrifugation. We separated the mononuclear cells from peripheral blood of adults and then isolated

CD8+ CD45RA+ T cells as naive CD8+ T cells and CD8+ CD45RO+ T cells DAPT concentration as memory CD8+ T cells. Peripheral blood mononuclear cells (PBMCs) were isolated from blood using Ficoll–Hypaque density gradient centrifugation. Cells were resuspended at a concentration of 2 × 106/ml in complete RPMI-1640 medium (Gibco, Grand Island, NY) supplemented with 10% fetal calf serum (Sijiqing, China), 100 U/ml penicillin, 100 μg/ml streptomycin, 50 μm 2-mercaptoethanol and 2 mm l-glutamine (all from Gibco). Naive CD8+ T cells were isolated from CBMCs by positive selection with anti-CD8 microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany). To purify naive and memory CD8+ T cells from PBMCs, CD8+ T cells were negatively isolated from

PBMCs p38 MAPK inhibitor using a biotin–antibody cocktail (Miltenyi Biotec). Subsequently, purified CD8+ T cells were incubated with anti-CD45RA and anti-CD45RO microbeads (Miltenyi Biotec) respectively. CD8+ CD45RA+ and CD8+ CD45RO+ cells were obtained by positively selecting from the column. The purity of cells, assessed by flow cytometry (FACSCalibur; Becton Dickinson, San Jose, CA) exceeded 97% for each T subset. Cells were resuspended at a concentration of 0·5 × 106/ml in complete RPMI-1640 medium. The CBMCs were stimulated with soluble anti-CD3 (0·2 μg/ml) plus anti-CD28 (1 μg/ml) in the presence of various doses of IL-21 (Peprotech, Rocky Hill, NJ, USA) for 4 days. CD8+ CD45RA+ or CD8+ CD45RO+ T cells were stimulated with plate-bound anti-CD3 (1 μg/ml) plus anti-CD28 (1 μg/ml) in the presence or absence of IL-21 (50 ng/ml) or IL-15 (20 U/ml) for 4 days. Naive CD8+ T cells from CBMCs were stimulated with anti-CD3 plus anti-CD28 in the presence or absence of IL-21 (50 ng/ml), IL-15 (20 U/ml; Peprotech), IL-2 (50 U/ml; Peprotech)

or IL-21 plus transforming growth factor-β (TGF-β; 1 ng/ml; Peprotech) for 4 days. Culture supernatants were collected for the assay of cytokines by ELISA. The cells were harvested and rested in the presence of IL-2 (10 U/ml) for 3 days and restimulated with PMA (20 ng/ml; Flavopiridol (Alvocidib) Sigma-Aldrich, Saint Louis, MO, USA) + ionomycin (1 μg/ml; Sigma-Aldrich) and used for flow cytometry analysis or RNA extraction. Culture supernatants for 72 hr were used for cytokine measurement by ELISA. Purified CD8+ T cells from CBMCs or CD8+ CD45RA+ T cells from PBMCs were resuspended in complete RPMI-1640 medium at 107 cells/ml. Carboxyfluorescein diacetate succinimidyl ester (CFSE; Invitrogen, Carlsbad, CA) was added at a final concentration of 5 μm, and the cells were incubated for 10 min at 37° in 5% CO2.

001) In contrast, scores for both cored and diffuse SP for each

001). In contrast, scores for both cored and diffuse SP for each region (except for diffuse SP in occipital cortex: X2 = 11.7, P = 0.008) did not significantly differ across the four pathological phenotypes (cored-frontal: X2 = 1.8, P = 0.609; temporal: X2 = 3.5, P = 0.318; occipital: X2 = 7.1, P = 0.07) (diffuse-frontal: X2 = 2.4, P = 0.495; temporal: X2 = 2.2, P = 0.534). Post-hoc analysis for diffuse SP in occipital cortex revealed a significant difference between group 1 and group

2 (P < 0.001). There were no significant differences between the four groups with regard to the proportion selleck products of patients with ‘typical’ vs. ‘focal’ variants of AD. A statistically significant (X2 = 4.1, P = 0.042) difference in gender proportions was observed between group 1 and group 2 (Figure 3) such that women (64.7%) made up a greater proportion of group 1 than men, but a lesser proportion of group 2 (43.4%). There were no statistically significant differences in the distribution of cases with a positive family history PD-L1 inhibitor of AD across the four pathological phenotypes.

There were no significant differences between the four pathological phenotypes for either the mean age of onset (F3,96 = 1.248, P = 0.297), mean age at death (F3,117 = 1.364, P = 0.257), mean disease duration (F3,97 = 11.786, P = 0.277) or mean brain weight (F3,111 = 0.370, P = 0.775) (Table 1). The frequency of APOE alleles and genotype within each pathological phenotypic group are shown in Table 2. There was a statistically significant difference between the genotype groups with the ε4/ε4 genotype frequency being significantly higher in group 3 compared with group

1 (χ2 = 9.6, P = 0.002) and the ε3/ε3 genotype frequency consequently being significantly lower in group 3 compared with group 1 (χ2 = 4.5, P = 0.033). The APOE ε4 allele frequency was significantly higher in group 3 than group 1 χ2 = 9.7, P = 0.002), but only tended to be higher in group 2 compared with group 3 χ2 = 3.6, P = 0.057). No significant differences in ε2 allele frequency were found between any of the four pathological groups. Seven cases were identified where the pathological phenotype was not clearly assignable, although these most closely resembled the type 2 phenotype (Table 3). All had Aβ deposition in the form of numerous Unoprostone SP and CAA in leptomeningeal and cortical vessels which, while present in the frontal and/or temporal lobe, and in contrast to ‘typical’ type 2 cases, was NOT present within the occipital lobe. No significant differences were seen in either the age of onset (P = 0.716), age at death (P = 0.930), disease duration (P = 0.630) or brain weight (P = 0.952) were found between these and the typical group 2 cases. There was no significant difference in the proportion of APOE ε4 allele bearers between the typical group 2 cases and the group 2 ‘outliers’.

Flow fluorescence in-situ hybridization was performed to determin

Flow fluorescence in-situ hybridization was performed to determine the telomere length of CD4+ and CD8+ T cells. The isolated PBMCs were stained with either CD4-biotin (Beckman-Coulter, BV, Woerden, the Netherlands) or CD8-biotin (Biolegend, Europe BV, Uithoorn, the Netherlands) followed by staining with streptavidin-cyanin 5 (Cy5) (Biolegend). The

PBMCs were fixed and permeabilized (Invitrogen Life Technologies, Bleiswijk, the Netherlands) and then, using the telomere PNA-kit/fluorescein isothiocyanate (FITC) (Zebra Bioscience BV, Enschede, BAY 80-6946 research buy the Netherlands), we determined the relative telomere length. The subcell-line 1301 of CCRF-CEM, which is known to have long telomeres, was used to calculate the relative telomere length (RTL) BAY 73-4506 in vivo of the CD4+ and

CD8+ T cells using the following formula [18]: In addition, PBMCs of five elderly CMV-seropositive ESRD patients were sorted into a purified CD28+ or CD28null CD4+ or CD8+ T cell fraction to examine whether or not the relative telomere length differed in these sorted T cell fractions. For this purpose, PBMCs (20 × 106) were stained with AmCyan-labelled anti-CD3 (BD Biosciences, Erembodegem, Belgium), Pacific Blue-labelled anti-CD4 (BD Biosciences), allophycocyanin (APC)-labelled anti-CD8 (BD Biosciences), phycoerythrin (PE)-labelled anti-CD28 (BD Biosciences) and with 7-aminoactinomycin D (7AAD) (BD Biosciences). Sorting was performed on a FACSAria II SORP (BD Biosciences). All fractions had a purity of more than 95%. The activity of the telomerase enzyme was measured in five CMV-seropositive and five age-matched CMV-seronegative ESRD patients using the TRAPeze®

XL telomerase detection kit (Millipore, Temecula, CA, USA), according to the manufacturer’s instructions. Briefly, PBMCs (20 × 106) were sorted into purified and viable CD4+ and CD8+ T cell fractions (according to the sort protocol described briefly under Telomere length assay). The sorted T cell fractions (all with a purity of more than 95%) were stimulated with anti-CD3/CD28 beads (25 μl/1 ml; Invitrogen Selleck Fluorouracil Life Technologies) for 3 days at 37°C. Next, cells were resuspended in CHAPS lysis buffer (provided in the kit) and cell extractions were made (10–750 μg). Protein levels were determined by using the Bio-Rad protein assay (Bio-Rad, München, Germany). This assay is based on the capacity of a test sample to amplify a telomere template. The activity is expressed in total product-generated (TPG) units, which is calculated using the TSR8 standard curve (provided in the kit). A whole blood staining was performed to determine the T cell differentiation status [10, 11, 14]. Briefly, whole blood was stained with AmCyan-labelled anti-CD3 (BD Biosciences) in combination with Pacific Blue-labelled anti-CD4 (BD Biosciences) and APC-Cy70-labelled anti-CD8 (BD Biosciences).

4 vs 15 5 days) at dialysis initiation were higher in the usual c

4 vs 15.5 days) at dialysis initiation were higher in the usual care group. Estimated medical costs during 3 months before dialysis till dialysis initiation, the MDC group yielded a reduction of NT$ 59 251 for each patient (P < 0.001). Patient mortality was not significantly different. Multidisciplinary care intervention for pre-ESRD patients could not only significantly improve the quality of disease care and clinical outcome, but also reduce medical costs. "
“Date written: December 2008 Final submission: March 2009 No recommendations possible ABC294640 purchase based on Level I or II evidence (Suggestions

are based on Level III and IV evidence) Registry data and data from observational cohort studies suggest that coexisting vascular disease, whether it be coronary artery disease (CAD), peripheral vascular disease (PVD) or cerebrovascular disease is associated with Decitabine in vivo increased mortality risk for patients on dialysis. Limited studies have addressed the effect of different levels of disease severity. Dialysis itself is associated with a significantly increased risk of worsening vascular disease and nephrologists should consider these factors when a decision is being made to commence dialysis and the patient

should be adequately informed regarding the outcomes in people with these comorbidities. No recommendation. Databases searched: MeSH terms and text words for cardiovascular disease, coronary disease and myocardial ischaemia were combined with MeSH

terms and text words for renal replacement therapy and dialysis. The search was carried out in Medline (1950–March, Week 3, 2008). The Cochrane Renal Group Trials Register was also searched for trials not indexed in Medline. Date of search/es: 2 April 2008. Patients with end-stage kidney disease (ESKD) are at high risk of developing cardiovascular disease (CVD), which is considered the leading cause of mortality and morbidity in dialysis patients, accounting for 40–50% of deaths.1 Although there have only been a few studies of CVD in a population with mild renal insufficiency, several authors have reported ADAMTS5 an elevated prevalence of CVD in patients starting dialysis compared with the general population.2–5 On admission to dialysis, patients have a high prevalence of cardiovascular risk factors. According to the Lombardy registry,6 it was estimated that 17.4% of the incident patients admitted to dialysis have CAD (9.8%) or myocardial infarction (7.6%). Congestive cardiac failure (CCF) was reported in the same study to be 8.3%. In the United States Renal Data System Registry (USRDS),7 the prevalence of CVD in incident ESKD patients should be proportionally higher, as there is a higher proportion of diabetics, however, the proportion of patients affected by ischaemic heart disease is 3 times higher (40.0%) and the proportion of patients affected by CCF is 5 times higher at 36.0%.