NFAT1 in Adult and Cord Blood-Derived Human CD4+ T-Cells: Regulatory Implications for Graft vs. Host Disease (GVHD).

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3164-3164
Author(s):  
R.P. Weitzel ◽  
M.L. Lesnewski ◽  
Y. Huang ◽  
L.R. Fanning ◽  
Mary Laughlin

Abstract Clinical benefits of umbilical cord blood (UCB) grafts include the lower incidence of acute graft-versus-host disease (aGVHD) following allogeneic transplantation. We have previously demonstrated dramatically lower expression of the Nuclear Factor of Activated T-Cells 1 (NFAT1) protein, its associated transcripts and known binding partners, but not NFAT1 transcripts in UCB CD4+ cells following primary stimulation when compared to T-cells obtained from adult blood (AB). As NFAT1 is a key transcriptional regulator of immune responses, we hypothesize that NFAT1 and NFAT1-dependent factors play a critical role in the increased proliferation and decreased cytokine production seen in UCB, which may contribute to the lower incidence of aGVHD following UCB transplantation observed clinically. We have now demonstrated in vitro via siRNA-mediated knockdown and subsequent analysis of adult-derived primary T-cells with RT-PCR and Cytometric Bead Assay (CBA) that expression of a GVHD-promoting, Th1-type cytokine profile is dependent on expression of NFAT1, and that knockdown of NFAT1 results in a transcriptional and cytokine secretion profile similar to that seen in UCB T-cells with respect to key immunoregulatory cytokines and signal transducers following stimulation. Specifically, siRNA-mediated reduction of NFAT1 by at least 68% results in 93% and 28% reduction of IFN-γ, and TNF-αrespectively, as well as a 55% reduction in CTLA4 measured by RT-PCR. Additionally, short hairpin RNA (shRNA)-mediated stable knockdown of NFAT1 results in an enhanced and prolonged reduction of IFN-γ, IL-2, and TNF-αsecretion over transient siRNA transfection in both the Jurkat cell line and adult primary CD4+ T-cells. However, microarray and RT-PCR time point analyses indicate NFAT1 transcripts are not changed in UCB versus AB CD4+ cells. Under siRNA knockdown conditions, we observed at least a 24-hour delayed decrease in NFAT1 protein levels following mRNA reduction and an eventual rebound of both NFAT1 and its associated cytokines following transient transfection at 48 hours. This suggests a crucial regulatory role for post-translational events which may preferentially occur in UCB-derived T-cells in vivo. Indeed, we have observed ubiquitination of NFAT1 and rapid rescue of NFAT1 expression following treatment with the calpain-proteasome inhibitor Acetyl-L-Leucyl-L-Leucyl-L-Norleucinal (ALLN) in UCB-derived T-cells in vitro by co-immunoprecipitation (co-IP) in a dose-dependent manner. NFAT1 in unstimulated UCB CD4+ cells is shown by co-IP to be significantly more ubiquitinated than in AB-derived cells. Overall, our data indicates that regulation of NFAT1 through ubiquitination and proteasomal degradation, and its subsequent downstream effects may constitute a key difference between T-cells of adult and neonatal origin; specifically that this post-translational modification may serve as a method by which UCB-derived T-cells may prevent NFAT1 activity, impacting their allogeneic response.

2014 ◽  
Vol 20 (2) ◽  
pp. 192-201 ◽  
Author(s):  
Elizabeth O. Stenger ◽  
Brian R. Rosborough ◽  
Lisa R. Mathews ◽  
Huihui Ma ◽  
Markus Y. Mapara ◽  
...  

2019 ◽  
Vol 133 (17) ◽  
pp. 1901-1916 ◽  
Author(s):  
Tárcio Teodoro Braga ◽  
Wesley Nogueira Brandao ◽  
Hatylas Azevedo ◽  
Fernanda Fernandes Terra ◽  
Amanda Campelo L. Melo ◽  
...  

Abstract NLRP3 inflammasome [NLR (nucleotide-binding domain, leucine-rich repeat containing protein) Pyrin-domain-containing 3 ] functions as an innate sensor of several PAMPs and DAMPs (pathogen- and damage-associated molecular patterns). It has been also reported as a transcription factor related to Th2 pattern, although its role in the adaptive immunity has been controversial, mainly because the studies were performed using gene deletion approaches. In the present study, we have investigated the NLRP3 gain-of-function in the context of encephalomyelitis autoimmune disease (EAE), considered to be a Th1- and Th17-mediated disease. We took advantage of an animal model with NLRP3 gain-of-function exclusively to T CD4+ lymphocytes (CD4CreNLRP3fl/fl). These mice presented reduced clinical score, accompanied by less infiltrating T CD4+ cells expressing both IFN-γ and IL-17 at the central nervous system (CNS) during the peak of the disease. However, besides NLRP3 gain-of-function in lymphocytes, these mice lack NLRP3 expression in non-T CD4+ cells. Therefore, in order to circumvent this deficiency, we transferred naive CD4+ T cells from WT, NLRP3−/− or CD4CreNLRP3fl/fl into Rag-1−/− mice and immunized them with MOG35–55. Likewise, the animals repopulated with CD4CreNLRP3fl/fl T CD4+ cells presented reduced clinical score and decreased IFN-γ production at the peak of the disease. Additionally, primary effector CD4+ T cells derived from these mice presented reduced glycolytic profile, a metabolic profile compatible with Th2 cells. Finally, naive CD4+ T cells from CD4CreNLRP3fl/fl mice under a Th2-related cytokine milieu cocktail exhibited in vitro an increased IL-4 and IL-13 production. Conversely, naive CD4+ T cells from CD4CreNLRP3fl/fl mice under Th1 differentiation produced less IFN-γ and T-bet. Altogether, our data evidence that the NLRP3 gain-of-function promotes a Th2-related response, a pathway that could be better explored in the treatment of multiple sclerosis.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 350-350
Author(s):  
Dongchang Zhao ◽  
Chunyan Zhang ◽  
Tangsheng Yi ◽  
Chia-Lei Lin ◽  
Ivan Todorov ◽  
...  

Abstract The αEβ7 integrin CD103 is an excellent marker for identifying in vivo activated CD4+ regulatory T (Treg) cells. CD103− naive Treg cells from donors are effective in prevention but ineffective in treatment of graft versus host disease (GVHD). It is unknown whether in vivo activated donor CD103+ Treg cells can effectively treat ongoing GVHD. We have recently reported a new chronic GVHD model, in which donor DBA/2 (H-2d) spleen cells were transferred to the irradiated BALB/c (H-2d) recipients. The recipients showed chronic GVHD-like syndrome with high-levels of serum autoantibodies, proteinuria, and hair-loss, and the disease induction required both donor CD4+ T and B cells in a cell dose dependent manner (Blood, 2006). In the current studies, we observed that the percentage of CD103+ cells among donor CD4+ T cells in the recipients without disease was up to 45% and was more than two fold higher than that of the recipients with disease. The CD103+CD4+ T cells were more than 97% FoxP3+, which was similar to natural CD25hi Treg cells, but the former expressed markedly higher levels of CCR5 as compared to the latter. The CD103+ Treg cells showed markedly stronger suppression capacity as compared to freshly isolated as well as in vitro anti-CD3-activated CD25hi natural Treg cells. When injected into ongoing chronic GVHD recipients with severe proteinuria, CD103+ Treg cells (1x106) reversed proteinuria and tissue damages in 92% (11/12) of the recipients, and the recipients survived for more than 100 days. In contrast, the in vitro activated natural Treg cells reversed disease in only 25% (2/8) of the recipients, and the freshly isolated CD25hi natural Treg cells reversed none (0/12), and the treated recipients died within 45 days, which was similar to control PBS treated recipients. Furthermore, we found that infusion of the CD103+ Treg cells significantly reduced CD138+ antibody-secreting plasma cells in spleen and reduced serum levels of autoantibodies; and that infusion of the CD103+ Treg cells also significantly reduced CD44loCD62LhiSCA-1hi post-mitotic CD4+ T memory stem cells in spleen as well as pathogenic IFN-γ+CD4+ T cells in liver and nephritogenic IFN-γ+IL-10+CD4+ T cells in kidney tissues. Our results indicate that, different from CD103− naive natural Treg cells that prevent GVHD via suppressing donor T cell activation and expansion, CD103+ Treg cells ameliorate ongoing chronic GVHD via reducing activated pathogenic T and B cells.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Qingxiao Song ◽  
Xiaoning Wang ◽  
Xiwei Wu ◽  
Tae Hyuk Kang ◽  
Hanjun Qin ◽  
...  

AbstractEfforts to improve the prognosis of steroid-resistant gut acute graft-versus-host-disease (SR-Gut-aGVHD) have suffered from poor understanding of its pathogenesis. Here we show that the pathogenesis of SR-Gut-aGVHD is associated with reduction of IFN-γ+ Th/Tc1 cells and preferential expansion of IL-17−IL-22+ Th/Tc22 cells. The IL-22 from Th/Tc22 cells causes dysbiosis in a Reg3γ-dependent manner. Transplantation of IFN-γ-deficient donor CD8+ T cells in the absence of CD4+ T cells produces a phenocopy of SR-Gut-aGVHD. IFN-γ deficiency in donor CD8+ T cells also leads to a PD-1-dependent depletion of intestinal protective CX3CR1hi mononuclear phagocytes (MNP), which also augments expansion of Tc22 cells. Supporting the dual regulation, simultaneous dysbiosis induction and depletion of CX3CR1hi MNP results in full-blown Gut-aGVHD. Our results thus provide insights into SR-Gut-aGVHD pathogenesis and suggest the potential efficacy of IL-22 antagonists and IFN-γ agonists in SR-Gut-aGVHD therapy.


1992 ◽  
Vol 176 (5) ◽  
pp. 1431-1437 ◽  
Author(s):  
M Croft ◽  
D D Duncan ◽  
S L Swain

Because of the low frequency of T cells for any particular soluble protein antigen in unprimed animals, the requirements for naive T cell responses in specific antigens have not been clearly delineated and they have been difficult to study in vitro. We have taken advantage of mice transgenic for the V beta 3/V alpha 11 T cell receptor (TCR), which can recognize a peptide of cytochrome c presented by IEk. 85-90% of CD4+ T cells in these mice express the transgenic TCR, and we show that almost all such V beta 3/V alpha 11 receptor-positive cells have a phenotype characteristic of naive T cells, including expression of high levels of CD45RB, high levels of L-selectin (Mel-14), low levels of CD44 (Pgp-1), and secretion of interleukin 2 (IL-2) as the major cytokine. Naive T cells, separated on the basis of CD45RB high expression, gave vigorous responses (proliferation and IL-2 secretion) to peptide antigen presented in vitro by a mixed antigen-presenting cell population. At least 50% of the T cell population appeared to respond, as assessed by blast transformation, entry into G1, and expression of increased levels of CD44 by 24 h. Significant contributions to the response by contaminating memory CD4+ cells were ruled out by demonstrating that the majority of the CD45RB low, L-selectin low, CD44 high cells did not express the V beta 3/V alpha 11 TCR and responded poorly to antigen. We find that proliferation and IL-2 secretion of the naive CD4 cells is minimal when resting B cells present peptide antigen, and that both splenic and bone marrow-derived macrophages are weak stimulators. Naive T cells did respond well to high numbers of activated B cells. However, dendritic cells were the most potent stimulators of proliferation and IL-2 secretion at low cell numbers, and were far superior inducers of IL-2 at higher numbers. These studies establish that naive CD4 T cells can respond vigorously to soluble antigen and indicate that maximal stimulation can be achieved by presentation of antigen on dendritic cells. This model should prove very useful in further investigations of activation requirements and functional characteristics of naive helper T cells.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5011-5011
Author(s):  
Haiping He ◽  
Atsuko Takahashi ◽  
Yuki Yamamoto ◽  
Akiko Hori ◽  
Yuta Miharu ◽  
...  

Background: Mesenchymal stromal cells (MSC) are known to have the immunosuppressive ability and have been applied in clinic to treat acute graft-versus-host disease (GVHD), as one of severe complications after hematopoietic stem cells transplantation (HSCT) in Japan. However, MSC are activated to suppress the immune system only upon the stimulation of inflammatory cytokines and the clinical results of MSC therapies for acute GVHD are varied. It is ideal that MSC are primed to be activated and ready to suppress the immunity (=priming) before administration in vivo. Triptolide (TPL) is a diterpene triepoxide purified from a Chinese herb - Tripterygium Wilfordii Hook F (TWHF). It has been shown to possess anti-inflammatory and immunosuppressive properties in vitro. In this study, we aim to use TPL as the activator for umbilical cord-derived MSC (UC-MSC) to entry stronger immunosuppressive status. Methods: The proliferation of UC-MSC with TPL at the indicated concentrations for different time of 24, 48, 72, and 96 hours. Cell counting kit-8(CCK-8) was added in the culture medium to detect cell toxicity and the absorbance was measured using microplate reader. Flow cytometry was used to identify the MSC surface markers expression. TPL-primed UC-MSC were once replaced with fresh medium and co-culture with mixed lymphocyte reaction (MLR) consisted with mononuclear cells (MNCs) stained with CFSE and irradiated allogenic dendritic cell line (PMDC05) in RPMI 1640 medium supplemented with 10 % FBS (complete medium). IDO-1, SOD1, and TGF-β gene expression in TPL-primed UC-MSC and UC-MSC induced by 10 ng/ml IFN-γ and/or 15 ng/ml TNF-α were evaluated by RT-PCR. PDL1 and PDL2 expression in TPL-primed UC-MSC and UC-MSC in response to IFN-γ and/or TNF-α were checked by Flowjo. Results: Exposure of TPL for UC-MSC for 72hour at the concentration above 0.1 μM resulted in the cell damage significantly. Therefore, we added TPL in UC-MSC at 0.01μM of TPL for up to 48 hours, then washed thourouphly for the following culture for experiments. To evaluate the influence of TPL on the surface markers of UC-MSC, we cultured UC-MSC for 4 hours in complete medium following culture with 0.01μM of TPL for 20 hours (TPL-primed UC-MSC). TPL-primed UC-MSC revealed positive for CD105, CD73, and CD90, negative for CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR surface molecules as same as the non-primed UC-MSC. In MLR suppression by UC-MSC, the TPL-primed UC-MSC activity revealed stronger anti-proliferative effect on the CD4+ and CD8+ T cells activated by allogeneic DC than those of non-primed UC-MSC in MLR. Furthermore, the TPL-primed UC-MSC promoted the expression of IDO-1, SOD1 and TGF-β in response to IFN-γ+/-TNF-α by RT-PCR and enhanced the expression of PD-L1 by FACS analysis. Discussion:In this study, we found the TPL-primed UC-MSC showed stronger antiproliferative potency on CD4+ and CD8+ T cells compared with non-primed UC-MSC. TPL-primed UC-MSC promoted the expression of IDO-1, SOD1 and TGF-β stimulated by IFN-γ+/-TNF-α, although TPL alone did not induce these factors. Furthermore, we found that the PD1 ligand (PD-L1) was induced in TPL-primed UC-MSC, likely IFN-γ enhanced the PD-L1 expression, evaluated by flowcytometry. These results suggested that TPL-primed UC-MSC seemed more sensitive to be activated as the immunosuppressant. Here, we firstly report the new function of TPL to induce the upregulation of immunosuppressive effect, although the mechanisms of TPL inhibition to MSC need to be explore. Conclusively, TPL-primed UC-MSC might be applied for the immunosuppressive inducer of MSC. Figure Disclosures He: SASAGAWA Medical Scholarship: Research Funding; IMSUT Joint Research Project: Research Funding. Nagamura:AMED: Research Funding. Tojo:AMED: Research Funding; Torii Pharmaceutical: Research Funding. Nagamura-Inoue:AMED: Research Funding.


2021 ◽  
Author(s):  
N Bhaskaran ◽  
E Schneider ◽  
F Faddoul ◽  
A Paes da Silva ◽  
R Asaad ◽  
...  

AbstractResidual systemic inflammation and mucosal immune dysfunction persist in people living with HIV (PLWH) despite treatment with combined anti-retroviral therapy (cART), but the underlying immune mechanisms are poorly understood. Here we report an altered immune landscape involving upregulation of TLR- and inflammasome signaling, localized CD4+ T cell hyperactivation, and counterintuitively, an enrichment of CD4+CD25+FOXP3+ regulatory T cells (Tregs) in the oral mucosa of HIV+ patients on therapy. Using human oral tonsil cultures, we found that HIV infection causes an increase in a unique population of FOXP3+ cells expressing PD-1, IFN-γ, Amphiregulin (AREG), and IL-10. These cells persisted even in the presence of the anti-retroviral drug and underwent further expansion driven by TLR-2 ligands and IL-1β. IL-1β also promoted PD-1 upregulation in AKT1 dependent manner. PD-1 stabilized FOXP3 and AREG expression in these cells through a mechanism requiring the activation of Asparaginyl Endopeptidase (AEP). Importantly, these FOXP3+ cells were incapable of suppressing CD4+ T cells in vitro. Concurrently, HIV+ patients harbored higher levels of PD-1, IFN-γ, Amphiregulin (AREG), and IL-10 expressing FOXP3+ cells, which strongly correlated with CD4+ T cell hyperactivation, suggesting an absence of CD4+ T cell regulation in the oral mucosa. Taken together, this study provides insights into a novel mechanism of FOXP3+ cell dysregulation and reveals a critical link in the positive feedback loop of oral mucosal immune activation events in HIV+ patients on therapy.One Sentence SummaryHIV-induced immune dysfunction in lymphoid and mucosal tissues


2017 ◽  
Vol 3 (2) ◽  
pp. 28
Author(s):  
Desie Dwi Wisudanti

Kefir is a functional foodstuff of probiotics, made from fermented milk with kefir grains containing various types of beneficial bacteria and yeast. There have been many studies on the effects of oral kefir on the immune system, but few studies have shown the effect of bioactive components from kefir (peptides and exopolysaccharides/ kefiran), on immune responses. The purpose of this study was to prove the effect of kefir supernatant from milk goat on healthy immune volunteer response in vitro. The study was conducted on 15 healthy volunteers, then isolated PBMC from whole blood, then divided into 5 groups (K-, P1, P2, P3 and P4) before culture was done for 4 days. The harvested cells from culture were examined for the percentage of CD4+ T cells, CD8+ T cells, IFN-γ, IL-4 using flowsitometry and IL-2 levels, IL-10 using the ELISA method. The results obtained that kefir do not affect the percentage of CD4+ T cells and CD8+ T cells. The higher the concentration of kefir given, the higher levels of secreted IFN- γ and IL-4, but a decrease in IL-2 levels. Significant enhancement occurred at levels of IL-10 culture PBMC given kefir with various concentrations (p <0.01), especially at concentrations of 1%. These results also show the important effects of kefir bioactive components on immune responses. The conclusion of this study is that kefir can improve the immune response, through stimulation of IL-10 secretion in vitro.


Sign in / Sign up

Export Citation Format

Share Document