Increased numbers of NKG2C+, NKG2A+, and CD161+ T cells were also

Increased numbers of NKG2C+, NKG2A+, and CD161+ T cells were also associated to HCMV infection. The NKG2C deletion frequency BGJ398 mw was comparable in children with congenital HCMV infection and controls. Remarkably, the homozygous

NKG2C+/+ genotype appeared associated with increased absolute numbers of NKG2C+ NK cells. Moreover, HCMV-infected NKG2C+/+ children displayed higher absolute numbers of NKG2A+ and total NK cells than NKG2C+/− individuals. Our study provides novel insights on the impact of HCMV infection on the homeostasis of the NK-cell compartment in children, revealing a modulatory influence of NKG2C copy number. Human cytomegalovirus (HCMV) infection is highly prevalent worldwide (50–100%), and usually follows a subclinical course in healthy individuals. The virus remains in a lifelong latent state, occasionally undergoing reactivation, but may have a pathogenic GSK1120212 role in immunodeficient and immunosuppressed patients [1-3]. Moreover, HCMV has been associated

with atherosclerosis, lymphoproliferative disorders, and glioblastoma, as well as with an accelerated immunosenescence and a shorter lifespan [4-7]. Vertical transmission of HCMV during pregnancy is considered the most common cause of congenital infection worldwide, affecting ∼0.2–2% of infants and potentially causing fetal lesions [8-10]. Though most infected newborns are asymptomatic, ∼10% display a variety of clinical disorders [8, 11] potentially leading to important

sequelae such as mental retardation and deafness. The type of maternal infection (i.e., primary versus reactivation/reinfection) conditions the risk of congenital infection and the pregnancy stage at which transmission occurs is related to clinical severity [12-16]. Maternal antibodies with neutralizing activity are transferred to the fetus predominantly during the third trimester of gestation and may prevent congenital CMV disease [17]. Wilson disease protein Among other factors, fetal immune immaturity may determine the outcome of congenital infection [18, 19]. An effective defense against HCMV requires the participation of T and NK cells, and the virus has developed different immune evasion strategies [20]. Patients with congenital HCMV infection have been shown to display mature CD8+ T-cell responses [21, 22], and an expansion and differentiation of a specific TcR γδ+ cell subset has been recently reported [23]. In contrast, information on the role of NK cells in this context is rather limited [24, 25]. HCMV infection stably alters the distribution of NK-cell receptors (NKRs) in healthy adult blood donors and children.

[11] Candida hyphae have also been shown to penetrate dentinal tu

[11] Candida hyphae have also been shown to penetrate dentinal tubules along cracks of tooth surfaces, enabling the organism to invade dental hard tissues.[12] Apart from the aforementioned biological factors, the microbial cell surface hydrophobicity (CSH), which contributes to hydrophobic interactions between cells and surfaces, is thought to be an important non-biological

factor associated in the adherence of Candida to inert surfaces.[13] Studies have also shown that hydrophobic yeast are more virulent than their hydrophilic counterparts.[14, 15] Statistically significant correlations between CSH and candidal adhesion to BEC and denture acrylic surfaces have also been reported.[16, 17] Transient exposure

to antifungals may affect the aforementioned traits of Everolimus clinical trial candidal adhesion. For instances, it has been shown that foregoing attributes of Candida albicans were significantly reduced after limited exposure to subcidal concentrations of antifungal agents. The suppression of candidal growth that occurs following limited exposure to antifungal agents, as in the oral environment, GPCR Compound Library solubility dmso has been described as the postantifungal effect (PAFE). This phenomenon has been mainly studied with C. albicans isolates. It has been documented that the knowledge of PAFE, in tandem with minimum inhibitory concentration (MIC) values of a drug, would be clinically useful in evaluating new dosage regimens of a drug.[18] Furthermore, transient exposure to antifungal agents may also affect such virulence factors of Candida pertaining to their adhesion.[19, 20] Nystatin (i.e. oral suspensions, ointments, pastilles, creams) is a widely obtainable and a frequently used EGFR inhibiton antifungal agent available for topical treatment of various types of oral candidosis ranging from pseudomembranous, erythematous to denture-induced variants of oral candidosis. However, the diluents effect of saliva

and the cleansing effect of the oral musculature in the oral cavity tend to reduce the availability of nystatin below that of the effective therapeutic concentrations, thereby compromising its therapeutic efficacy. Hence, the pathogenic Candida may undergo a brief exposure to topically applied antifungal drugs, while thereafter, the drug concentration is likely to be subtherapeutic,[18] a scenario all too familiar in the niches of the oral cavity, which is similar to the phenomena as in PAFE. To our knowledge, there is no information on either the PAFE or its association with the adhesion-related attributes of oral C. dubliniensis isolates following brief exposure to subtherapeutic concentrations of nystatin. Hence, taken together the foregoing information, as well as the findings of a recent prevalence study where oral C.

LEE CHIWEI1, FUJIMURA LISA2, HIRAOKA SHUICHI3, KOSEKI HARUHIKO4,

LEE CHIWEI1, FUJIMURA LISA2, HIRAOKA SHUICHI3, KOSEKI HARUHIKO4, TOKUHISA TAKESHI5, OGAWA MAKOTO1,

YOKOSUKA OSAMU1, HATANO MASAHIKO2,6 1Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University; 2Biomedical Research Center, Chiba University, Chiba Japan; 3Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan; 4Laboratory for Developmental Genetics, Center for Integrative Medical Science, RIKEN; 5Department of Developmental Genetics, Graduate School of Medicine, Chiba University, Chiba; 6Department of Biomedical GPCR & G Protein inhibitor Science, Graduate School of Medicine, Chiba University, Chiba, Japan Introduction: Kif26a and Kif26b are unique member of kinesin superfamily proteins which belong to kinesin-11 family. Kif26b deficient (KO) mice showed impaired development of kidney while Kif26a KO mice develop a mega-colon with enteric nerve hyperplasia. Kif26a negatively Kinase Inhibitor Library in vitro regulates GDNF-Ret signaling pathways in developing enteric neurons. Since GDNF-Ret signal plays a critical role in nephrogenesis, it might be possible that Kif26a regulates kidney development. However, roles of Kif26a in kidney remain obscure. To elucidate the roles of

Kif26a in kidney, we examined the kidney of Kif26a KO and HET mice. Methods: We conducted all experiments by using BALBc mice with heterozygous(HET) and homozygous(KO) deletion of Kif26a. We investigated the histopathology of kidneys in HET and KO mice by PAS staining. We also exmamined where Kif26a expresses in kidney at developmental satge by using in situ hybridization. The number of glomeruli in each

of 4 consecutive sections adjacent to the mid-sagittal section was counted and the mean number of nephrons per section per kidney was calculated. Results: Glomerular hyperplasia and reduction of glomerulus number were observed in Kif26a KO and HET mice at 4weeks of age. Histological analysis of kidney revealed that impairment of branching and extension in collecting ducts in the KO and HET mice. Expression of Kif26a mRNA was detected in extending portion of collecting ducts in newborn mice kidney. Furthermore, secondary focal segmental glomerulosclerosis (FSGS) developed in Kif26a KO and HET mice at 25weeks of age. Conclusion: Kif26a regulates the branching and extension of collecting ducts at developmental either stage. Thus, Kif26a KO and HET mice cause oligonephronia. Kif26a KO and HET mice are useful animal model of oligonephronia and secondary FSGS. Kif26a may be one of resposible genes for familial oligonephronia. TU YUE1, SUN WEI2, WAN YI-GANG3 1Nanjing University of Chinese Medicine; 2Jiangsu Provincial Hospital of Chinese Medicine; 3Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Introduction: Dahuangfuzi decoction (DFD) is a traditionally well-prescribed formula for the treatment of renal failure (RF) in China for many years. However, little is known about its therapeutic mechanisms.

In this context, a pre-existing S  mansoni infection was shown to

In this context, a pre-existing S. mansoni infection was shown to suppress Th1 response and to impair control of L. major (28) and L. donovani (29) infection in C57BL/6 mice. Also, co-infection with tapeworm Taenia crassiceps led to increased lesions sizes upon subsequent L. major and L. mexicana infection in BALB/c mice (30). In conclusion, the helmith/Leishmania co-infection studies demonstrating impaired control of Leishmania

(28–30) used helminths that induced long-lasting or even chronic infections while the ones including our own, reporting still intact host defence upon co-infection used either transient or semi-permissive helminth infection models (22,23,31). One study describing efficient control of L. major in BALB/c and C57BL/6 mice carrying previous S. mansoni infections used Gefitinib in vivo an extremely high L. major infection dose (4 × 107 promastigotes) that might have functioned as a very potent Th1 inducer, even in the presence of chronic helminth infection (31). The diversity of these results highlights the importance of all protagonists involved for the final outcome of co-infections that is, as pointed out above, helminth species, Leishmania infection doses and genetic background of the host mice (32). As this reflects the diversity of human population and their parasites, we argue that important knowledge YAP-TEAD Inhibitor 1 research buy is extracted from all these

different co-infection models, despite heterogeneous results. Regarding the reciprocal impact of L. major infection on the nematode infection, we did observe a suppression of the local S. ratti-specific Th2 response. To our surprise, this suppression was detectable in the mesLN after 2 days of subsequent co-infection with L. major but not if L. major infection preceded S. ratti infection by 14 days. This clearly shows that the establishment of a protective local S. ratti-specific Th2 response was not impaired if an S. ratti co-infection took place in

mice with a fully established L. major-specific Th1 response. The S. ratti-specific Th2 response in the mesLN, however, is transient and starts to decline by day 8 p.i. (10). From next this data we conclude that an L. major co-infection that was established at day 6 post-S. ratti infection accelerated the decline of the S. ratti-specific response, thus resulting in the observed reduction in Th2 cytokines in co-infected mice. Here, it is of special interest that a local infection such as L. major is usually restricted to the draining, i.e. the popLN displayed a systemic effect by changing cytokine responses in the mesLN. Interestingly, the reduced S. ratti-specific Th2 response observed upon L. major co-infection was still sufficient to allow efficient nematode expulsion, as we showed by unchanged worm burden. The artificial interference with Strongyloides-induced Th2 polarization, in contrast, has been shown to interfere with host defence.

Furthermore, FISH is not a stand-alone technique in the diagnosti

Furthermore, FISH is not a stand-alone technique in the diagnostic setting, as culture is still used for antibiotic susceptibility testing. While traditionally the probes for FISH were based on single MLN0128 ic50 stranded DNA, another set of probes increasingly used in diagnostics are based on a polyamide ‘peptide’ backbone (Egholm et al., 1993; Bjarnsholt et al., 2008). PNA FISH probes abide by Watson/Crick

pairing but possess unique hybridization characteristics because of their uncharged chemical backbone, including rapid and stronger binding to complementary targets compared with traditional DNA probes. PNA probes can also be used with unfixed biological samples; however, only a limited number of probes are currently available, restricting the use of PNA FISH for the present. CLSM and FISH emphasize that demonstrating biofilm spatial organization is extremely important to: (1) identify whether the bacteria present are aggregated, (2) indicate a polymicrobial nature of a biofilm, (3) indicate the extent of biofilm on a surface that CFU may vastly underestimate, and (4) to show biofilm EPS that may comprise a greater

part of the biofilm than cells alone. On nonbiological, flat surfaces, biofilm spatial organization can best be measured by various parameters using image analysis software. The most common program is comstat that yields a number of spatial parameters including thickness, biovolume, selleck and roughness (Heydorn et al., 2000). Quantification of biofilm spatial organization is harder Lepirudin however in clinical specimens that usually have a complicated and convoluted surface geometry, and currently is largely descriptive

or qualitative in these samples – that is, data showing cells or clusters per unit area without a good method to quantify spatial dimensions. As comstat thresholding does not work well on tissue backgrounds, quantifying the biofilm involves a manual rendering of biofilm images in other software to resolve bacteria and laborious cell counting, particularly if NA probes are used because they stain host cell nuclei as well as bacterial DNA (Nistico et al., 2011). Resolving biofilm spatial organization is also made more difficult because of the spatial scales involved. For example to be able to resolve individual bacteria in an image, the field of view needs to be on the order of 100 μm2, while the specimen might be on the order of cm2 (1 million fields) for tissue or even 100s of cm2 (over 100 million fields) for large orthopedic implants making microscopic data from a small proportion of the sample often the only practical method to demonstrate biofilm in situ. Finally, because biofilms may also be extremely localized, it is difficult to quantify by averaging several images on the surface, because heterogeneity leads to extensive sample variability.

[14] As a result of the decrease in recent thymic emigrants, it h

[14] As a result of the decrease in recent thymic emigrants, it has been suggested that peripheral T cells in individuals with DS undergo increased homeostatic proliferation in comparison to the general population.[14]

Because of mixed genetic background in Ts65Dn mice, differences in recent thymic emigrants cannot be reliably measured. Nevertheless, the selleck chemicals llc data are consistent with a loss of thymic precursors in the Ts65Dn mice leading to altered peripheral T-cell populations. Defects in Ts65Dn peripheral T-cell function are most evident in the decreased proliferation in response to polyclonal stimulation. This loss of function may be consistent with immune dysfunction in DS, as lymphocytes from individuals with DS have also been shown to exhibit a decreased proliferative response to polyclonal stimuli such as phytohaemagglutinin,[47, 48] in addition to the documented decrease in responses in some individuals with DS to vaccinations.[49, 50] Vaccine studies have shown that IL-7 and TCR signalling can synergize to promote antigen-specific effector cell generation, especially when using subdominant antigens.[51] Therefore decreased IL-7Rα expression as well as the deficient Trichostatin A mw proliferation in response to TCR stimulation may

contribute to the T-cell dysfunction observed in DS. It is tempting to speculate that the impaired proliferation in the immature thymocyte subsets as a consequence of decreased IL-7Rα expression 4��8C may be one of the causes of accelerated thymic involution as well as decreased thymic output in DS. In turn, the increased, possibly excessive, homeostatic cycling of peripheral T

cells in individuals with DS may result in premature senescence and impaired function. The changes in lymphocyte responses were not limited to T cells as B-cell proliferation was also diminished in response to antigen receptor stimulation, but not lipopolysaccharide. This is consistent with an anergic/senescent phenotype in the peripheral lymphocyte pools. However, in contrast to thymic development, B-cell progenitors in the bone marrow and IL-7Rα expression on those cells were not altered in the Ts65Dn mice, suggesting a selective effect on T-lymphocyte precursors. It is interesting, but unclear, why the previously reported decrease in CLP in Ts65Dn bone marrow[6] only results in diminished T-cell progenitors. One postulate is that decreases in Notch signalling, due to BACH1-mediated inhibition of Nrf2 or increased DYRK1a[52] in DS, leads to impaired T-cell specification, but not B-cell development. The resultant changes in mature B-cell function and spleen subsets may be, as has been proposed previously,53 due to altered T-cell help.

This review will focus on biophysical properties and biogenesis o

This review will focus on biophysical properties and biogenesis of exosomes, their pathophysiological roles and their potential

as biomarkers and therapeutics in kidney diseases. Intercellular communication is vital for the regulation and coordination of many different processes within multicellular organisms. Extracellular membrane-bound vesicles are emerging as a novel and significant mechanism of cell signalling and communication. Exosomes are a specific subset of membrane-bound vesicles of endosomal origin, which are released into the extracellular environment by many cells from different tissues and organs. Exosomes exist in Silmitasertib cost a wide range of biological fluids, including blood and urine. The ubiquitous nature of exosomes has highlighted them as significant vehicles of cellular communication, with many important biological and pathophysiological implications. Exosomes are defined as small vesicles between 30 and 100 nm in diameter, consisting of a limiting lipid bilayer, transmembrane proteins and a hydrophilic core containing proteins, mRNAs and microRNAs (miRNA). They are distinguished from other microparticles by

their size and the fact that they are formed intracellularly within multivesicular endosomes (multivesicular bodies; MVB), while microvesicles (100 to 1000 nm in diameter) MLN8237 supplier are shed from the plasma membrane surface[1] (see Table 1). Cellular breakdown Release from cellular blebs during apoptosis Exosomes contain a defined set of proteins, which varies according to the cell of origin.[6] Common components of exosomes are proteins involved with endosomal trafficking, membrane trafficking and fusion proteins, tetraspanins (CD63, CD81, CD9, CD82), heat shock proteins (HSP70, HSP90), metabolic enzymes, adhesion molecules, signal transduction proteins, lipid rafts and cytoskeletal proteins, in addition to cell type-specific

proteins, such as major histocompatibility complex (MHC) class I and II, α-synuclein, and the A33 antigen.[6] Exosomes have a specific lipid composition distinct from their Calpain parental MVB, although they do reflect their cell of origin, and can also contain bioactive lipids such as prostaglandins, which may contribute to their function.[7] Exosomes contain mRNAs and miRNAs, and RNA profiling of exosomal fractions has identified significant differences to parental cellular RNA.[8, 9] Both mRNAs and miRNAs present in the exosomal fraction maintain their function when transferred to other cells,[8, 10] demonstrating that exosomal RNA transfer may be an important route for epigenetic signalling between cells. However, recent studies suggested that many extracellular miRNAs may not be contained within exosomes, but can be complexed with circulating Argonaute-2 or other ribonucleoprotein complexes.[11-13] Exosomes are formed by the intraluminal budding of late endosomal compartments to create MVB, containing intraluminal vesicles.

, 2009), it is not limited to the hospital environment; community

, 2009), it is not limited to the hospital environment; community-acquired CDI and asymptomatic carriage are also prevalent (Limbago et al., 2009; Freeman et al., 2010). Production of toxins and spores is the most important virulence determinant of C. difficile. The toxins are highly immunogenic. They have been shown to induce the production of pro-inflammatory cytokines such as IL-1α, IL-1β, IL-6, IL-8 and TNF-α (Flegel et al., 1991; Linevsky et al., 1997; Melo Filho et al., 1997; Johal et al., 2004; Canny et al., 2006) and are responsible for the acute inflammatory response in C. difficile infection (Savidge et al., 2003), which is characterized by pseudomembrane click here formation (Knoop et al., 1993; Castagliuolo &

LaMont, 1999). However, a large number of proteins are released along with the toxins during growth of C. difficile (Mukherjee et al., 2002). These include several

surface-associated proteins such as surface-layer proteins (SLPs), flagella, cell wall proteins like Cwp66 and Cwp84, GroEL and fibronectin-binding protein 68, which are involved in adhesion (Hennequin et al., Obeticholic Acid datasheet 2001b; Tasteyre et al., 2001; Waligora et al., 2001; Calabi et al., 2002) and have also been found to elicit immune responses within the host (Péchiné et al., 2005a, b; Wright et al., 2008). Serum IgG to such surface proteins have been detected in patients and healthy adults in several studies (Pantosti et al., 1989; Mulligan et al., 1993; Sánchez-Hurtado et al., 2008), and in many, a correlation between lower levels of antibodies to somatic antigens and the occurrence or recurrences of disease was identified (Mulligan et al., 1993; Kyne et al., 2000; Péchiné et al., 2005a). Interestingly, in one study, the toxins appeared to be less immunogenic than the somatic antigens, suggesting that surface

adhesins were able to induce Digestive enzyme a host immune response during the course of infection and the intensity of this response affected the outcome of infection (Péchiné et al., 2005a). It has thus been suggested that antibodies against toxin as well as nontoxin antigens may determine the outcome of infection with C. difficile (Kelly & Kyne, 2011). These observations have led to the investigation of different surface-associated proteins such as FliD and FliC, SLPs, Cwp84 and Cwp66 as vaccine components (O’Brien et al., 2005; Péchiné et al., 2007, 2011). GroEL and Cwp66 are heat-shock proteins (HSPs) of C. difficile that are strongly induced and expressed on the cell surface following heat shock at 42 and 60 °C, respectively (Hennequin et al., 2001a; Waligora et al., 2001). The primary aim of this study was to assess the production of immunomodulatory cytokines by a macrophage cell line when challenged with different C. difficile proteins. These included SLPs, flagella, HSPs induced at 42 and 60 °C, and cell-free culture supernatants collected at different stages during the growth of C. difficile.

In our previous studies, the use of cationic solid–lipid nanopart

In our previous studies, the use of cationic solid–lipid nanoparticle (cSLN) formulation as a delivery system has revealed comparable efficiency: cytotoxicity ratio with linear PEI-25 kDa–pDNA polyplexes, protected CPA, CPB and CPB−CTE genes from extracellular enzymatic degradation and also exhibited considerable low cytotoxicity [22]. Hence, cSLNs can be considered as suitable adjuvant and/or delivery system for designing third-generation cocktail vaccines. Also, these characterized formulations of cocktail vaccine candidates could immunize BALB/c

mice against cutaneous leishmaniasis [23]. In this study, we evaluated the potency of the BI 2536 order A2–CPA–CPB−CTE trifusion gene delivered using either a physical method (electroporation) or a chemical delivery system (cSLN) as a candidate vaccine against L. infantum infection and assessed its immune induction potential in BALB/c mice. The A2 gene (with Kozak sequence) was subcloned from pUC57 vector (synthesized by ShineGene Molecular Biotech, Inc., Shanghai, China‎‏) into pGEM7zf(+)vector (Promega, Madison, WI, USA). Both pGEM-cpa and pGEM-cpb were available from our previous studies [11], and CPA and CPB−CTE fragments were subcloned

into pAT153 vector (Boca Scientific, Boca Raton, FL, USA), respectively. Then CPA–CPB−CTE was cloned downstream of the A2 gene in pGEM7zf(+) vector. The A2–CPA–CPB−CTE fragment was subcloned into pcDNA3·1(−) (Invitrogen, Grand Island, NY, USA) vector to generate pcDNA–A2–CPA–CPB−CTE as a DNA vaccine. pcDNA–A2–CPA–CPB−CTE plasmids were purified by ion exchange chromatography with QIAGEN (Hilden, Germany) Endofree Mega C646 molecular weight kit and then confirmed by PCR and digestion (data not shown). The cSLN suspension was manufactured by a validated technique previously Suplatast tosilate described by Doroud et al. [22]. cSLN–pDNA complexes were prepared by adding volumes corresponding to 1200 μg of purified pDNA (pcDNA–A2–CPA–CPB−CTE) to cSLN suspension at DOTAP: pDNA ratio of 6 : 1 (w/w) and at 60 min incubation at room temperature separately [22, 24]. Complete condensation and complexation

of pDNAs with cSLN were analysed by agarose gel electrophoresis, as previously demonstrated [22, 24]. Size and zeta potential measurements, gel retardation analysis and DNase I protection study were all performed according to the conditions demonstrated in our previous study [22, 24]. The physicochemical stability of the formulations was assessed during 1 month and reported previously [22]. In this study, the characteristics of the formulations, that is, the mean diameter, polydispersity index, zeta potential and retardation ability, were assessed according the ICH guidelines. For this purpose, nanoparticles containing pDNAs were stored at high temperatures and relative humidity (25 ± 2°C/60% RH) in a qualified stability analysis chamber (accuracy: ±0·2°C; humidity uniformity: ±3% RH) over a period of 12 months, at dark and regular time intervals.

Concomitant with the upregulation of IL-10 production, recently a

Concomitant with the upregulation of IL-10 production, recently activated Th17 clones switched off IL-17 production that was regained only at later time points. Mechanistically,

the loss of IL-17 production was explained by the downregulation of RORγt in recently activated Th17 cells and by the induction, in response to autocrine IL-2, of phosphorylated STAT5, which competes with STAT3 for binding to the IL-17 promoter [49]. These studies reveal a novel aspect of Th17 biology, namely that IL-17 production is strongly elicited in effector and memory Th17 cells within a few hours after antigenic stimulation, while it is actively suppressed at later time points when anti-inflammatory mechanisms, such as the production of IL-10, are induced to prevent excessive immunopathology. Time- and activation-dependent regulation of cytokine gene Cabozantinib nmr expression selleck chemicals llc has been described in other cell types such as dendritic cells where different genes are activated with different kinetics over several hours after the initial stimulus [50].

In this context, human Th17 cells may provide an attractive model system to study the contribution of reversible and dynamic chromatic changes in T-cell activation [51]. In this review, we have discussed how the study of cytokine production, homing capacity, antigenic specificity, and activation state can be a useful approach to understand the complex physiology of effector and memory human T cells. We are starting to understand mechanistically some of this complexity, for instance in the Th17 field we are now appreciating the role of IL-1β and IL-12 in the induction of IL-17/IFN-γ double-producing T cells, a phenotype

that is frequently observed in pathological conditions. Furthermore, we are beginning to appreciate the role of the Th17-cell activation state and cytokine milieu in modulating inflammatory from and anti-inflammatory cytokine production. These findings thus reveal new targets and rationale for therapeutic intervention of inflammatory diseases. Several years ago, studies performed in the human system demonstrated that the vast majority of memory Th cells maintain both memory and flexibility of cytokine gene expression. For instance, Th1 and Th2 cells could be induced to simultaneously produce IFN-γ and IL-4 when stimulated in opposite polarizing conditions, that is, in the presence of IL-4 or IL-12, respectively [52]. At the time, the general consensus from mouse studies was that Th cells were undergoing a rapid and irreversible commitment to their lineage and that the silenced cytokine genes were repositioned to heterochromatin in order to maintain cell identity.