Upon aGVHD development in the group of mice receiving PBMC alone

Upon aGVHD development in the group of mice receiving PBMC alone (positive control)

(days 12–15), target organs and sera were harvested from all groups for histological analysis, serum analysis and cell characterization. All experiments were repeated two or more times with five to seven mice per group on each occasion. Target organs (lung, liver and gut) were recovered from mice (days 12 or 15) and fixed in 10% (v/v) buffered formalin, processed for histology and embedded in paraffin wax. Five-μm tissue sections were stained by haematoxylin and eosin (H&E) and coded without reference to prior treatment, blinded and then examined by two independent observers. A semi-quantitative scoring system was used to assess abnormalities in the lung, liver and gastrointestinal tract (GI) tract [30-32]. Human bone marrow mesenchymal stem cells were generated as previously described [33] in collaboration with the Regenerative HM781-36B Medicine Institute (REMEDI, NUI Galway, Ireland). Briefly, bone marrow

aspirates were taken from the iliac crest of healthy consenting adult donor patients according to an approved clinical protocol [34]. Human MSC batches used in this study conformed to the International Society for Cellular Therapy (ISCT) criteria [16] and were capable of differentiation to adipocytes, osteocytes and chondrocytes and were only used at low passage (3–8). Human MSC were cultured in complete Dulbecco’s modified Eagle’s medium (DMEM) (Invitrogen-Gibco, Dublin, Ireland) supplemented with 10 % (v/v) fetal bovine serum (FBS), 200 U/ml penicillin and 200 μg/ml streptomycin. In some instances, PF 2341066 MSC were stimulated with recombinant human IFN-γ (500 U/ml) (Peprotech, London, UK) for 48 h and washed extensively with PBS prior to their use in vitro or in vivo. For in-vitro apoptosis, PBMC (0·5 × 106/ml) were co-cultured with MSC (1·5 × 105/ml) in complete RPMI (cRPMI) in the presence or absence of 500 μg/ml cisplatin (control) (Sigma-Aldrich, Arklow, Ireland). After 24 h, PBMC were recovered by gentle aspiration

from adherent MSC and apoptosis was detected by annexin V/propidium iodide (PI) staining (BD Biosciences, Oxford, Adenosine triphosphate UK), measured by flow cytometry using a BD fluorescence activated cell sorter (FACS)Calibur cytometer with CellQuest software (BD Biosciences). For in-vivo apoptosis, in order to optimize, first, the detection of apoptosis FAM-FLIVO™ green dye (Immunochemistry Technologies, Bloomington, MN, USA) was used. As a control for the detection of FLIVO in vivo, BALB/c mice were irradiated lethally with 12 Gy gamma irradiation. After 24 h, 8 μg (100 μl) of FAM-FLIVO™ green dye was injected per mouse and left to circulate for 1 h. After 1 h (or other times, not shown), the liver was harvested and isolated cells were analysed by flow cytometry to verify detectability of apoptosis in vivo.

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 buy Ensartinib TLO development in vivo are still unclear. The majority of mechanistic data on the development of TLOs are CHIR-99021 nmr 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 Metformin cost 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.

We have reported previously the presence of

anti-M3R anti

We have reported previously the presence of

anti-M3R antibodies that recognized the second extracellular loop in SS patients but not in patients with RA or SLE, suggesting that anti-M3R antibodies could be used potentially as diagnostic markers for SS [4]. However, Kovacs et al.[14] reported the detection of anti-M3R Y-27632 cell line antibodies in 35% of their RA patients and 32% of SLE. These conflicting results emphasize the need to examine the precise prevalence of anti-M3R antibodies in other autoimmune diseases using our modified ELISA system. The correlation between anti-M3R antibodies and clinical features is still unclear. The previous study reported leukopenia was more common in anti-M3R antibody-positive than in -negative patients with primary SS [14]. Our observations in the present study showed that positivity for anti-SS-A antibody and IgG values in serum was more prevalent and higher in anti-M3R antibody-positive SS patients than -negative SS patients. The disease duration of SS was shorter among anti-M3R antibody-positive SS than -negative SS; however, there was no difference in other clinical and histological features between anti-M3R antibody-positive and -negative SS patients.

We could not detect any significant relationship between each B cell epitope and clinical characteristics such as saliva secretion. In conclusion, these findings support the notion of presence of several B cell epitopes on M3R in SS patients,

and that some SS patients are reactive this website to several extracellular domains of the M3R. It is possible that some anti-M3R antibodies alter salivary secretion in SS via M3R, and Acyl CoA dehydrogenase in particular antibodies against the second extracellular loop of the M3R could suppress the increase in (Ca2+)i induced by M3R agonists, resulting in reduction of salivary secretion. Therefore, anti-M3R antibodies might play pathogenic roles in salivary secretion abnormalities characteristic of patients with SS. None of the authors has any conflict of interest with the subject matter or materials discussed in the manuscript. “
“Antimicrobial resistance was studied in 100 Mycobacterium tuberculosis strains selected randomly from sputum cultures of newly diagnosed tuberculosis patients. Resistance of the isolates to rifampicin, isoniazid, and ethambutol was tested by both drug susceptibility testing (DST) and allele-specific PCR (AS-PCR). A total of 19 (19%) isolates were found resistant to at least one of the antituberculosis drugs investigated by PCR compared with 14 (14%) resistant isolates detected by DST. Eleven mutations were detected by AS-PCR in the rpoB gene (codons 516, 526, and 531), associated with rifampicin resistance, a marker of multidrug-resistant tuberculosis (MDR-TB), 14 mutations in the katG gene codon 315 that confers resistance to isoniazid, and nine mutations in the embB gene codon 306 that confers resistance to ethambutol.

The samples were then incubated with 50 µl/well of OVA-biotin (1 

The samples were then incubated with 50 µl/well of OVA-biotin (1 mg/ml; Sigma, St Louis, MO, USA) at room temperature for 1 h. Plate-bound antibody was detected by treatment

with 50 µl/well of streptavidin–horseradish peroxidase (1 : 10 000; Southern Biotechnology) for 1 h at room temperature. The colour reaction was developed by adding 100 µl/well of 200 pmol of OPD (Sigma) in pH 5·0 citrate phosphate buffer plus 0·04% H2O2 for 10 min and stopped with 50 µl of 5% sulphuric acid per well. The plates were read at 492 nm in an ELISA reader (Bio-Rad, Hercules, CA, USA). The lungs of five mice per group were removed and treated with 100 U/ml of collagenase from Clostridium histolyticum (Sigma) for 30 min at 37°C. Subsequently, the digested lung tissue was filtered through a 70 micrometre cell strainer and the red blood cells were lysed with ACK buffer (0.15 M NH4Cl, 10 mM KHCO3, 0.1 mM Na2EDTA, pH 7.2; Invitrogen, CA,

USA). The cell check details suspension was washed twice in RPMI-1640 and adjusted to 1 × 106 cells per well for surface staining and to 2·5 × 106 cells for the intracellular cytokine experiment. For CD4 and forkhead box P3 (FoxP3) staining, the cells were generally blocked with anti-mouse CD16/CD32 monoclonal antibodies (mAbs) (Fc-block) and stained for surface marker using fluorescein isothiocyanate (FITC)-labelled anti-mouse CD4 (BD Bioscience) mAb or isotype control, which were incubated for 20 min at 4°C with antibody dilution PF-01367338 manufacturer solution (PBS 0·15 M, 0·5% BSA, 2 mM NaN3). The cells were then washed with 0·15 M PBS and incubated with strepatavidin–phycoerythrin–cyanine 5 (PE-Cy5) (1 : 200) Tacrolimus (FK506) for an additional 20 min at 4°C. Surface-stained cells were washed twice with 0·15 M PBS and incubated with fixation/permeabilization buffer (eBioscience) for 30 min at

4°C. Anti-FoxP3-PE-labelled antibodies in permeabilization buffer (eBioscience) were added to cells and then incubated for 30 min at 4°C. Cells were washed twice with 150 µl of permeabilization buffer (eBioscience) and fixed with 2% paraformaldehyde. For IL-10 and FoxP3 intracellular staining, cells were cultured for 14 h in medium or OVA (25 µg/ml). After this stimulation period, 1 mg/ml of brefeldin A was added to the cell culture, which was incubated for an additional 4 h in a CO2 incubator at 37°C. Before CD4 staining, the cells were treated with anti-CD16/CD32 (Fc-block). Cell surface and intracellular staining were performed as described above for surface experiments; however, the cells were stained for CD4, IL-10 and FoxP3 using anti-CD4 FITC-labelled, anti-IL-10 PE-labelled, and anti-FoxP3 biotin-labelled plus streptavidin–PE-Cy5 antibodies. Data acquisition was performed using fluorescence activated cell sorter (FACScan) (Becton Dickinson, San Jose, CA, USA). Data analysis was performed using a FlowJO interface (Becton Dickinson). Statistical analysis was performed using the software GraphPad Prism (GraphPad Software, San Diego, CA, USA). The mean ± standard deviation (s.d.

We co-cultured the human gastric cancer cell line AGS with H pyl

We co-cultured the human gastric cancer cell line AGS with H. pylori exposed to IFN-γ; both phosphorylated CagA and nonphosphorylated CagA in AGS cells were downregulated by IFN-γ, and the proportion of cells with the ‘hummingbird’ phenotype was also decreased. Thus, IFN-γ can help control H. pylori infection indirectly through the virulence factor CagA. Helicobacter pylori is one of the most frequently seen pathogens in gastric mucosa and colonizes the stomachs of more than half of the world’s population Alpelisib datasheet today (Suerbaum & Josenhans, 2007). The main consequences include chronic gastritis, stomach and duodenal ulcers, gastric carcinoma and mucosa-associated lymphoid

tissue lymphoma. Gastric carcinoma is the fourth most common of all cancers. Helicobacter

find more pylori was classified as a class I carcinogenic factor by the World Health Organization in 1994. Helicobacter pylori has a cytotoxin-associated gene (Cag) pathogenicity island, a 40-kb DNA that encodes a type IV secretion system (T4SS). This T4SS can inject a virulence factor such as CagA protein into the host cells (Covacci & Rappuoli, 2000) and augment the gastric carcinoma risk (Franco et al., 2008). CagA protein is one of the most important virulent factors in H. pylori, and its expression is regulated by many environmental factors, including iron (Ernst et al., 2005), acid (Karita et al., 1996; Merrell et al., 2003; Shao et al., 2008b), sodium chloride (Loh et al., 2007; Gancz

et al., 2008), bile (Shao et al., 2008a) and nitric oxide (Qu et al., 2009). Interleukin-1b (IL-1b) (Porat et al., 1991), tumor necrosis factor-α (TNF-α; Luo et al., 1993), IL-2 and granulocyte-macrophage colony-stimulating factor (Denis et al., 1991) can affect the growth and virulence properties of a Protirelin virulent strain of Escherichia coli, and interferon-γ (IFN-γ) can upregulate the main virulence of Pseudomonas aeruginosa (Wu et al., 2005). However, no study has investigated IFN-γ altering the properties of H. pylori, or more particularly, the effect on the virulence protein CagA. IFN-γ is a proinflammatory cytokine secreted predominantly by CD4+CD25− effector T-helper cells in response to many stimuli, including endotoxin and Gram-negative bacteria. Clinical samples show a significantly higher level of IFN-γ in H. pylori-infected human gastric mucosa than in uninfected mucosa (Shimizu et al., 2004; Pellicanòet al., 2007), as do animal models (Cinque et al., 2006; Sayi et al., 2009). In addition, peripheral blood mononuclear cells produced IFN-γ when exposed to an H. pylori component (Meyer et al., 2000). IFN-γ was produced by natural killer cells in response to an H. pylori component (Yun et al., 2005). Although Shimizu et al. (2004) found no significant correlation between IFN-γ levels and inflammatory cell infiltrations in children with H.

phagocytophilum

phagocytophilum selleck inhibitor surface protein (Rikihisa, 2010), and examined by confocal microscopy. Comparable to observations of infected HL-60 cells, 61.0% ± 6.2% AVMs in RF/6A

cells were FK2-positive (Fig. 2a–c and g). Notably, fewer AVMs exhibited detectable ubiquitination in ISE6 cells, as only 13.8% ± 0.4% were FK2-positive (Fig. 2d–g). After binding to the HL-60 cell surface, the majority of A. phagocytophilum organisms enter to reside in ApVs within 4 h (Carlyon et al., 2004; IJdo & Mueller, 2004; Borjesson et al., 2005), after which they replicate by binary fission for approximately 24 h and subsequently exit the host cell to initiate a second round of infection (Troese & Carlyon, 2009). Reinfection occurs between 24 and 36 h following a synchronous infection (Troese & Carlyon, 2009). To assess the temporal association of ubiquitinated conjugates with the AVM over the course of a synchronous infection, A. phagocytophilum organisms were added to HL-60 cells and allowed to bind for 40 min followed find more by the removal of unbound bacteria. At various postinfection time points over a 48-h period, aliquots were screened with FK2 and anti-Msp2 (P44) and examined by confocal microscopy (Fig. 3). At 4 and 6 h, a time period during which nascent ApVs

form, ubiquitin association with 22.1% ± 0.8% and 27.1% ± 0.4% AVMs was detected as aggregative and/or punctate staining patterns surrounding intravacuolar A. phagocytophilum organisms (Figs 3a–f and 4). AVM ubiquitination consistently increased over the next 12 h, as 35.2% ± 6.7%, 41.3% ± 5.7%, and 52.6% ± 4.2% exhibited FK2 staining at 8, 12, and 18 h (Fig. 4). The aggregative FK2 staining pattern on most of the AVMs continually increased over the duration of infection (Fig. 3). By and after 12 h, these many AVMs were completely decorated such that a ring-like staining pattern surrounding

the bacteria resulted (Fig. 3j–D). By 24 h, AVM ubiquitination began to decline, as 46.2% ± 5.0% and 38.9% ± 10.1% of AVMs were FK2 positive at 24 and 30 h (Fig. 4). Beginning at 36 h, the percentages of ubiquitinated AVMs began to increase once again. At 30 and 36 h, in addition to large ApVs full of A. phagocytophilum bacteria, many HL-60 cells also harbored small ApVs that contained one to a few organisms (Fig. 3s–x). The small ApVs exhibited punctate FK2 staining reminiscent of the staining patterns observed at 4 and 6 h (Fig. 3a–f), thereby indicating that reinfection had occurred between 24 and 36 h and that the infection had become asynchronous. Because mono- and polyubiquitination differentially dictate the subcellular trafficking of downstream processes in which protein substrates participate (Raasi et al., 2005; Chen & Sun, 2009; Dikic & Dotsch, 2009), we next determined whether mono- or polyubiquitinated proteins accumulate on the AVM. Accordingly, we stained A.

Because we found no significant change in phosphorylation at Tyr-

Because we found no significant change in phosphorylation at Tyr-505 of Lck under the ephrin-Bs costimulation (data not shown), the association between Eph and CD45 may not be involved. Wu and colleagues [[18-20]] have previously reported that EphB receptors and TCR were located closely in aggregated rafts and ephrin-B ligand simply enhanced TCR signaling, in which p38 and p44/42 MAPK activations were essential parts of ephrin-B1/B2/B3 costimulation. However, in our study, the suppressive phase in

primary T-cell proliferation induced by solid-phase ephrin-B ligands with CD3 stimulation PKC412 purchase has been newly revealed. Cytokine assay also showed the different costimulation effects from Wu and colleagues’ previous data. In their studies, the lymphokinetic pattern induced by ephrin-B1, B2, and B3 ligand costimulation was different from that of CD28 in T-cell proliferation; Lapatinib datasheet wherein, it remarkably stimulated production

of IFN-γ but not IL-2 possibly due to the absence of Akt activation. In our assay, IL-2 production, as well as IFN-γ and TNF-α, is regulated biphasically by costimulation with ephrin-B1/B2, and was simply promoted by ephin-B3. This implies that IL-2 secretion is evident, as well as IFN-γ and TNF-α, in ephrin-B costimulation. In the promotion phase, EphB receptor functions as one of the costimulatory molecules like CD28. We speculate that the discrepancy between the results may be due to the differences in the origin and concentration Docetaxel of ephrin-B ligands (Wu and colleagues utilized their own ephrin-B-Fc chimeric proteins, while we purchased from

R&D systems) and the genetic background of the mouse. One could argue that the unique modification patterns that we observed might be due to the replacement of anti-CD3 antibody by high-dose ephrin-Bs during the coating procedure. But it is very unlikely because of following three reasons, (i) each concentration of normal human IgG instead of ephrin-Bs leads to no inhibition of the anti-CD3 induced T-cell proliferation (Fig. 1A), (ii) high dose of ephrin-B3 did not inhibit (rather promoted) the proliferation in the same culture system, (iii) SHP1 recruitment by EphB4 (Fig. 6A), but not by EphA4 (Fig. 6B) or EphB6 (Supporting Information Fig. 7), reasonably explains the functional inhibition of TCR signaling. We also conducted the culture with wells coated with ephrin-Bs in the presence of soluble anti-CD3 antibody. In this assay, however, the modification patterns by ephrin-Bs were not observed (Supporting Information Fig. 8).

However, the design of the present study with a restricted number

However, the design of the present study with a restricted number of available very old 3xTg mice does not allow more detailed biochemical and immunohistochemical analyses as well as additional behavioural testing. Such studies might include links between abnormal phosphorylation and conformational changes of tau, possibly also affecting GSK 3β known as an important enzyme for the generation of phospho-tau epitopes [65] and shown here in cells co-labelled with AT8. These

desirable efforts are mainly forced by the widely accepted crucial role of abnormal tau and tangle formation in AD and other tauopathies such as corticobasal degeneration, argyrophilic grain disease, progressive supranuclear palsy, Pick’s disease and fronto-temporal dementia (for reviews on

tauopathies see [6, 66, 67]). In AD patients, the number of Fostamatinib in vivo counted tangles post mortem correlated with the severity of dementia in lifetime [56]. Transgenic models already contributed to novel concepts for a better understanding of AD and other neurodegenerative disorders as summarized by several reviews [12, 14, 68, 69]. However, the incomplete nature of these models allows recapitulation of only a few aspects of the complexity in AD [1, 15, 70, 71]. Furthermore, models with less limitations might also settle controversies of whether deleterious effects of tau pathologies result from toxic gain-of-function by pathological tau or from critically affected tau function in the Talazoparib disease state [72]. Improved models might verify the proposed, partly neuroprotective role of tau phosphorylation [73]. Notably, Western blotting has revealed considerably enhanced Rebamipide hippocampal GFAP levels in 7-month-old immunolesioned 3xTg versus naive mice. This might partially model the general and dramatic glial reaction as already described by Delacourte

in 1990 [74]. While fluorescence labelling also suggested enhanced gliosis associated with strong Aβ-immunoreactivity, it only allows for qualitative estimation. The observed activated glia surrounding plaques resembles the relationships of microglia and astrocytes to Aβ deposits in AD and in mouse models with age-dependent β-amyloidosis such as Tg2576 [75]. Microglia as active sensors and versatile effector cells in pathologically altered brain [76] have been reported as a driving force in plaque formation, whereas astrocytes were found as a leading factor in their degradation [77]. On the other hand, the proposed role for microglia in plaque maintenance [78] could not be confirmed by Grathwohl et al. [79]. However, microglia were found to be increasingly dysfunctional during ageing, possibly contributing to age-dependent neurodegeneration [80], which remains a promising target for therapeutic intervention.

Solt et al demonstrated very similar effects with the synthetic

Solt et al. demonstrated very similar effects with the synthetic RORγt ligand SR1001, which prevented Th17-cell differentiation and ameliorated EAE [[68]]. In a model for inflammatory bowel disease, RORγt-dependent ILCs can mediate pathology [[41]]. Together these

results suggest that the RORγt antagonist SR1001 may be utilised therapeutically to target pathogenic ILCs. Interestingly, in addition to RORγt, SR1001 also inhibits the activity of the type 2 ILC-related transcription factor RORα [[68]] This opens up the possibility of using ROR antagonists such as SR1001 in the treatment of type 2 ILC-related immune pathologies, including airway hyperreactivity in allergic asthma, Selleckchem Smoothened Agonist as well as those mediated by RORγt-dependent ILCs. However, the application of ROR agonists and antagonists needs to be carefully assessed in view of the known beneficial roles of ILCs. Future work needs to reveal how RORα/γt antagonism affects ILC functions, and how this can be applied in the clinical settings. In addition to RORγt and RORα, AhR plays a prominent role in the survival and function of the ILC22 population. The AhR agonist FICZ increases the number of intestinal IL-22-producing ILCs, cells that are crucial for clearing C. rodentium infection [[54]]. This role in the gut makes AhR an interesting target for the treatment of inflammatory bowel disease, a disease in which ILC-derived IL-22 plays a protective

selleck role [[28, 30]]. In summary, as discussed in this review, the transcriptional programs that govern the development of the various branches of the ILC family, including RORγt and RORα dependent ILCs, are

beginning to be unraveled. Future studies should aim to address the precise requirements of specific transcription factors at different stages of ILC development and to unravel how these transcription factors are regulated, what the effects of antagonism are, and how the potential interactions between PI-1840 the various transcription factors affect ILC development and function. With such knowledge, attention can be turned to specific therapeutics based on regulating these family members. “
“The function of IL-10 producing regulatory B cells (Breg) during gestation is unknown. Here, we aimed to understand their participation in early pregnancy. CD19+CD24hiCD27+B cell frequency, measured by flow cytometry, increased with pregnancy onset but not in the case of spontaneous abortions. B cells from non-pregnant women cultured with serum from normal pregnant women produced higher IL-10 levels than those cultured with serum from spontaneous abortion patients or autologous serum. CD19+-activated B cells from pregnant women strongly suppressed TNF-a production by CD4+T cells when cocultured. We identified hCG as an important factor regulating the number and function of Breg during pregnancy. Breg emerge as important players in pregnancy; they suppress undesired immune responses from maternal T cells and are therefore important for tolerance acquisition.

[74] Intravenous administration of miR-124 at the effector phase

[74] Intravenous administration of miR-124 at the effector phase of disease ameliorated EAE and reduced neuroinflammation probably through its effect on macrophages, whereby miR-124 is able to promote a phenotypic switch from classically to alternatively activated macrophage, through indirect down-regulation of transcription factor PU.1, and thereby decreased expression of activation markers CD45, MHC class II and CD86, via inhibition of C/EBP-α.[74] Such a function is probably also Midostaurin cost at play in the maintenance of a quiescent microglial phenotype in the normal CNS. Alternatively activated microglia can secrete a wide range of molecules that can have a neuroprotective effect

in MS/EAE, either directly, such as insulin-like growth factor 1, which promotes proliferation and differentiation of neural progenitor cells,[75, 76] or indirectly through their anti-inflammatory effect, such as the anti-inflammatory cytokines

IL-4, IL-10 and TGF-β. In vitro studies have shown that IL-4-stimulated microglia are able to instruct neural progenitor cells to differentiate into oligodendrocytes, at least in part through release of insulin-like growth factor 1.[75] A number of disease-modifying drugs that have been, or are in the process of being, approved for MS, can potentially affect microglial phenotype directly or indirectly. We shall address this issue for the two most used first-line treatments for relapsing–remitting MS, IFN-β and glatiramer acetate (GA), and for the recently approved fingolimod and dimethyl fumarate (DMF). The precise mechanisms 3-MA solubility dmso through which IFN-β exerts its immunomodulatory effect in

MS are still uncertain, but generally include inhibition and apoptosis of autoreactive T cells, induction of regulatory T cells, inhibition of leucocyte extravasation through the BBB, and modulation of cytokine expression.[77] Its effect on microglia has, as yet, been poorly investigated, with only scant in vitro studies reported. Kim et al.[78] showed that IFN-β induced the expression of chemokines such as RANTES and MIP-1b in primary human microglia, through activation of at least three different partially interconnected signalling cascades Tolmetin including nuclear factor-κB, activator protein-1 and Janus kinase/signal transducer and activator of transcription. Kawanokuchi et al.[79] addressed the effect of IFN-β on murine microglial functions such as antigen presentation and secretion of inflammatory mediators; they showed that IFN-β inhibits the antigen-presenting function of microglia through suppression of IFN-γ-induced MHC class II expression and down-regulation of the co-stimulatory molecule B7-1, and suppresses differentiation of pathogenic autoreactive T helper type 1 T cells through down-regulation of microglial IL-12 production. Surprisingly, and in accordance with the study of Dasgupta et al.